1
|
Cheng G, Zhang M, Zhang J, Teng S, Wang Z, Cui T, Xiao S. E se tea aqueous-ethanol extract ameliorates D-galactose induced oxidative stress and inflammation via the Nrf2 signal pathway. Food Res Int 2025; 209:116323. [PMID: 40253213 DOI: 10.1016/j.foodres.2025.116323] [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: 10/15/2024] [Revised: 02/01/2025] [Accepted: 03/17/2025] [Indexed: 04/21/2025]
Abstract
E Se tea is a traditional herbal tea produced by traditional green tea processing technique from the tender leaves of Malus toringoides (Rehd.) Hughes with anti-inflammatory and antioxidant activities. This study investigated the inhibitory effect of the aqueous-ethanol extract of E Se tea against oxidative stress induced damage on D-galactose (D-gal) induced mice. UPLC-ESI-HRMS/MS analysis resulted in the identification of eleven compounds inclusive of 1 isoflavone (9), 1 phenolic acid (2), 2 flavanols (1 and 10), 3 dihydrochalcones (5, 8, and 11), and 4 flavones (3, 4, 6, and 7). The quantitative analysis demonstrated that phlorizin (8) had the highest content, followed by phloretin (11) and kaempferol-3-O-glucoside (7). The aqueous-ethanol extract of E Se tea significantly increased the total antioxidant capacity (T-AOC) in serum, reduced MDA level, and enhanced SOD activity and GSH level in brain and liver tissues. In addition, this extract also remarkably decreased the levels of inflammatory cytokines (IL-6 and IL-1β) in serum, and inhibited the AchE activity in brain. The possible mechanism might be related to the upregulation of Nrf2, HO-1, and NQO1 the expressions by using western blotting experiment. The pearson correlation analysis revealed that phloretin was the possible antioxidant and anti-inflammatory compound, and coumaroyl quinic acid was the active compound on AChE enzyme. These findings indicated that E Se tea extract had the protective effect on D-gal induced oxidative stress damage in mice via activating the Nrf2 signal pathway.
Collapse
Affiliation(s)
- Guiguang Cheng
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650550, China; Yunnan International Joint Laboratory of Green Food Processing, Kunming 650500, China
| | - Meng Zhang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650550, China; Yunnan International Joint Laboratory of Green Food Processing, Kunming 650500, China
| | - Jinke Zhang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650550, China; Yunnan International Joint Laboratory of Green Food Processing, Kunming 650500, China
| | - Sifan Teng
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650550, China; Yunnan International Joint Laboratory of Green Food Processing, Kunming 650500, China
| | - Zhengxuan Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650550, China; Yunnan International Joint Laboratory of Green Food Processing, Kunming 650500, China
| | - Tianqi Cui
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650550, China; Yunnan International Joint Laboratory of Green Food Processing, Kunming 650500, China.
| | - Shanshan Xiao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650550, China; Yunnan International Joint Laboratory of Green Food Processing, Kunming 650500, China.
| |
Collapse
|
2
|
Jin M, Lou Y, Wang X, Lv J, Wu Y, Huang G. Hyperoside suppresses NSCLC progression by inducing ATG13-mediated autophagy and apoptosis. Cell Immunol 2025; 411-412:104947. [PMID: 40250077 DOI: 10.1016/j.cellimm.2025.104947] [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: 02/22/2025] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Lung cancer is a leading cause for cancer-related mortality across the globe. In the last decade, significant advancements have been made in the research of non-small cell lung cancer (NSCLC). However, new biotherapeutic drugs urgently need to be developed. This study investigated the regulating effect of hyperoside on NSCLC progression. METHODS The colony formation assay and Cell Counting Kit-8 were used to detect cell proliferation. The Transwell assay was used to monitor cell migration. NSCLC growth in vivo was examined using a subcutaneous xenograft model. Proteomics, immunohistochemistry, and immunofluorescence analyses were used to detect anticancer regulatory mechanisms. RESULTS The results showed that hyperoside treatment inhibited cell migration, proliferation, and tumor growth in NSCLC in vivo and in vitro. Also, hyperoside treatment promoted apoptosis and cell cycle S-phase arrest. Proteomics, immunohistochemistry, and immunofluorescence detection also showed that hyperoside treatment promoted autophagy-related protein 13 (ATG13)-mediated autophagy, which further increased NSCLC apoptosis. CONCLUSION In summary, the findings illustrated that hyperoside treatment suppressed NSCLC progression by promotingATG13 expression and enhancing autophagy activation, finally promoting autophagy and apoptosis.
Collapse
Affiliation(s)
- Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yuqing Lou
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Xiaoshuo Wang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jia Lv
- Department of Obstetrics and Gynecology, Shanghai Fourth People's hospital, School of Medicine, Tongji University, Shanghai 200434, China.
| | - Yue Wu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China..
| | - Gang Huang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201318, China..
| |
Collapse
|
3
|
Karakas H, Cagman Z, Kizilarslan-Hancer C, Erol E. Isolation and Characterization of Secondary Metabolites from Endemic and Edible Polygonum sivasicum with In Vitro Antioxidant and Cytotoxic Activities. ACS OMEGA 2025; 10:9756-9767. [PMID: 40092762 PMCID: PMC11904701 DOI: 10.1021/acsomega.5c00438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
Polygonum sivasicum Kit Tan and Yildiz, one of the eight endemic Polygonum species in Türkiye, belongs to the Polygonaceae family. Preliminary phytochemical investigation of methanol and hexane extracts of P. sivasicum resulted in four compounds, namely, annphenone (1), hyperoside (2), daucosterol (3), and β-sitosterol (4). Their structures were elucidated by 1D-, 2D-NMR, and HRESIMS analyses. This study signifies the first isolation of annphenone from the Polygonum genus. Antioxidant capabilities of different extracts of P. sivasicum were carried out using DPPH·, ABTS·+, CUPRAC, metal chelating, and β-carotene linoleic acid bleaching assays, and their effectiveness was quantified through IC50 values. Furthermore, 27 phenolic compounds were identified using LC-HRESIMS from methanol extract, which has the highest antioxidant activity among the P. sivasicum extracts. The major phenolic constituents identified were hyperoside (4535.0 μg/g extract), rutin (4387.4 μg/g extract), and chlorogenic acid (3306.6 μg/g extract). GC-MS analysis determined palmitic acid, α-linolenic acid, and 8,11-octadecadieonic acid as major fatty acids in the hexane extract. The cell viability profile of P. sivasicum methanol extract and its isolates hyperoside, annphenone, and daucosterol was evaluated on fibroblast (CCD-1079Sk), breast carcinoma (MCF-7) and lung carcinoma (A549) cell lines. Annphenone exhibited IC50 values of 0.25 ± 0.01 mg/mL against the A549 cell line and 0.36 ± 0.02 mg/mL against the MCF-7 cell line. The selective cytotoxicity observed for daucosterol against the A549 cell line, with a high selectivity index of 1.44, underscores its potential as a promising candidate for drug development. The study establishes a framework integrating phytochemical profiling with biological assays to identify therapeutic agents from endemic plants.
Collapse
Affiliation(s)
- Humeyra Karakas
- Department
of Pharmacognosy and Natural Products Chemistry, Health Sciences Institute, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Zeynep Cagman
- Department
of Biochemistry, Faculty of Pharmacy, Bezmialem
Vakif University, 34093 Istanbul, Turkey
| | - Cagla Kizilarslan-Hancer
- Department
of Pharmaceutical Botany, Faculty of Pharmacy, Bezmialem Vakif University, 34093 Istanbul, Turkey
| | - Ebru Erol
- Department
of Analytical Chemistry, Faculty of Pharmacy, Bezmialem Vakif University, 34093 Istanbul, Turkey
| |
Collapse
|
4
|
Zhang W, Wang R, Guo R, Yi Z, Wang Y, Wang H, Li Y, Li X, Song J. The multiple biological activities of hyperoside: from molecular mechanisms to therapeutic perspectives in neoplastic and non-neoplastic diseases. Front Pharmacol 2025; 16:1538601. [PMID: 40098612 PMCID: PMC11911483 DOI: 10.3389/fphar.2025.1538601] [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: 12/03/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
In recent years, hyperoside (quercetin 3-O-β-D-galactopyranoside) has garnered significant attention due to its diverse biological effects, which include vasoprotective, antioxidant, anti-inflammatory, and anti-tumor properties. Notably, hyperoside has shown remarkable potential in cancer therapy by targeting multiple mechanisms; it induces apoptosis, inhibits proliferation, blocks angiogenesis, and reduces the metastatic potential of cancer cells. Furthermore, hyperoside enhances the sensitivity of cancer cells to chemotherapy by modulating key signaling pathways. Beyond neoplastic diseases, hyperoside also presents promising therapeutic applications in managing non-cancerous conditions such as diabetes, Alzheimer's disease, and pulmonary fibrosis. This review comprehensively examines the molecular mechanisms underlying hyperoside's anti-cancer effects and highlights its role in the treatment of cancers, including lung and colorectal cancers. Additionally, it explores the latest research on hyperoside's potential in addressing non-neoplastic conditions, such as pulmonary fibrosis, diabetes, and Parkinson's disease. By summarizing current findings, this review underscores the unique therapeutic value of hyperoside and its potential as a multifunctional treatment in both neoplastic and non-neoplastic contexts.
Collapse
Affiliation(s)
- Weisong Zhang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Rui Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Rongqi Guo
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Zhongquan Yi
- Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Yihao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Hao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Yangyang Li
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Xia Li
- Department of General Medicine, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Jianxiang Song
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| |
Collapse
|
5
|
Chen J, Zhao Y, Wang X, Zang L, Yin D, Tan S. Hyperoside Inhibits RNF8-mediated Nuclear Translocation of β-catenin to Repress PD-L1 Expression and Prostate Cancer. Anticancer Agents Med Chem 2024; 24:464-476. [PMID: 38305391 DOI: 10.2174/0118715206289246240110044931] [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: 10/26/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Hyperoside is a flavonol glycoside isolated from Hypericum perforatum L. that has inhibitory effects on cancer cells; however, its effects on prostate cancer (PCa) remain unclear. Therefore, we studied the anti-PCa effects of hyperoside and its underlying mechanisms in vitro and in vivo. AIM This study aimed to explore the mechanism of hyperoside in anti-PCa. METHODS 3-(4,5-Dimethyl-2-Thiazolyl)-2,5-Diphenyl Tetrazolium Bromide (MTT), transwell, and flow cytometry assays were used to detect PCa cell growth, invasion, and cell apoptosis. Immunoblot analysis, immunofluorescence, immunoprecipitation, and quantitative real-time PCR (qRT-PCR) were used to analyze the antitumor mechanism of hyperoside. RESULTS Hyperoside inhibited PCa cell growth, invasion, and cell cycle and induced cell apoptosis. Furthermore, RING finger protein 8 (RNF8), an E3 ligase that assembles K63 polyubiquitination chains, was predicted to be a direct target of hyperoside and was downregulated by hyperoside. Downregulation of RNF8 by hyperoside impeded the nuclear translocation of β-catenin and disrupted the Wnt/β-catenin pathway, which reduced the expression of the target genes c-myc, cyclin D1, and programmed death ligand 1 (PD-L1). Decreased PD-L1 levels contributed to induced immunity in Jurkat cells in vitro. Finally, in vivo studies demonstrated that hyperoside significantly reduced tumor size, inhibited PD-L1 and RNF8 expression, and induced apoptosis in tumor tissues of a subcutaneous mouse model. CONCLUSION Hyperoside exerts its anti-PCa effect by reducing RNF8 protein, inhibiting nuclear translocation of β-catenin, and disrupting the Wnt/β-catenin pathway, in turn reducing the expression of PD-L1 and improving Jurkat cell immunity.
Collapse
Affiliation(s)
- Jie Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Yi Zhao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Xiaoli Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Long Zang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Song Tan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| |
Collapse
|
6
|
Wang K, Zhang H, Yuan L, Li X, Cai Y. Potential Implications of Hyperoside on Oxidative Stress-Induced Human Diseases: A Comprehensive Review. J Inflamm Res 2023; 16:4503-4526. [PMID: 37854313 PMCID: PMC10581022 DOI: 10.2147/jir.s418222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/27/2023] [Indexed: 10/20/2023] Open
Abstract
Hyperoside is a flavonol glycoside mainly found in plants of the genera Hypericum and Crataegus, and also detected in many plant species such as Abelmoschus manihot, Ribes nigrum, Rosa rugosa, Agrostis stolonifera, Apocynum venetum and Nelumbo nucifera. This compound exhibits a multitude of biological functions including anti-inflammatory, antidepressant, antioxidative, vascular protective effects and neuroprotective effects, etc. This review summarizes the quantification, original plant, chemical structure and property, structure-activity relationship, pharmacologic effect, pharmacokinetics, toxicity and clinical application of hyperoside, which will be significant for the exploitation for new drug and full utilization of this compound.
Collapse
Affiliation(s)
- Kaiyang Wang
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Huhai Zhang
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Lie Yuan
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Research Laboratory for Drug Metabolism, College of Pharmacy, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yongqing Cai
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
7
|
Kalaycı B, Şimşek Özek N, Aysin F, Özbek H, Kazaz C, Önal M, Güvenalp Z. Evaluation of cytotoxic and apoptotic effects of the extracts and phenolic compounds of Astragalus globosus Vahl and Astragalus breviflorus DC. Saudi Pharm J 2023; 31:101682. [PMID: 37448848 PMCID: PMC10336577 DOI: 10.1016/j.jsps.2023.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
Astragalus L. is a genus member of the Fabaceae family, representing about 3,000 species all over the world and 380 species in Turkey. Astragalus species have been used in traditional medicine for many years. Astragalus globosus Vahl, known as "top geven", is a dwarf, scapose, perennial herb, Astragalus breviflorus DC., known as "yünlü geven", is an extremely spiny dwarf shrub. These endemic species grow in the Turkish cities of Erzurum, Kars, and Van. This is the first phytochemical and cytotoxic investigation of Astragalus globosus Vahl and Astragalus breviflorus DC. The main extracts and sub-fractions from the plants were evaluated for in vitro cytotoxic and apoptotic activities. The IC50 values of dichloromethane, n-butanol, and water extracts of the aerial parts of A. globosus against the MCF-7 cell line were determined as 28.39, 868.60, and 1753.00 µg/mL. The values for the MDA-MB-231 cell line were 264.00, 620.30, and 1300.50 µg/mL, respectively. From A. globosus, the following were isolated: a flavone glycoside, diosmetin-7-O-rutinoside (1); and two flavonol glycosides, isorhamnetin-3-O-rutinoside (2) and quercetin-3-O-galactoside (3). From A. breviflorus, two phenolic acids, caffeic acid (4) and chlorogenic acid (5), and a flavan-3-ol, catechin (6), were isolated. Diosmetin-7-O-rutinoside was isolated from Astragalus species for the first time and showed the highest cytotoxic activities on the MCF-7 and MDA-MB-231 breast cancer cell lines with IC50 values of 13.65 and 12.89 μg/mL, respectively. Moreover, we observed that diosmin exerts cytotoxic effects by causing cell necrosis.
Collapse
Affiliation(s)
- Benan Kalaycı
- Department of Pharmacognosy, Faculty of Pharmacy, Ağrı İbrahim Çeçen University, Ağrı, Turkey
| | - Nihal Şimşek Özek
- Department of Biology, Faculty of Science, Atatürk University, Erzurum, Turkey
| | - Ferhunde Aysin
- Department of Biology, Faculty of Science, Atatürk University, Erzurum, Turkey
| | - Hilal Özbek
- Department of Pharmacognosy, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
- Medicinal Aromatic Plant and Drug Research Center, Atatürk University, Erzurum 25240, Turkey
| | - Cavit Kazaz
- Department of Chemistry, Faculty of Science, Atatürk University, Erzurum, Turkey
| | - Mehmet Önal
- Non-wood Products Chief Engineering, Regional Directorate of Forestry, Erzurum, Turkey
| | - Zühal Güvenalp
- Department of Pharmacognosy, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
- Medicinal Aromatic Plant and Drug Research Center, Atatürk University, Erzurum 25240, Turkey
| |
Collapse
|
8
|
Crosstalk between xanthine oxidase (XO) inhibiting and cancer chemotherapeutic properties of comestible flavonoids- a comprehensive update. J Nutr Biochem 2022; 110:109147. [PMID: 36049673 DOI: 10.1016/j.jnutbio.2022.109147] [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: 06/03/2021] [Revised: 12/17/2021] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Gout is an inflammatory disease caused by metabolic disorder or genetic inheritance. People throughout the world are strongly dependent on ethnomedicine for the treatment of gout and some receive satisfactory curative treatment. The natural remedies as well as established drugs derived from natural sources or synthetically made exert their action by mechanisms that are closely associated with anticancer treatment mechanisms regarding inhibition of xanthine oxidase, feedback inhibition of de novo purine synthesis, depolymerization and disappearance of microtubule, inhibition of NF-ĸB activation, induction of TRAIL, promotion of apoptosis, and caspase activation and proteasome inhibition. Some anti-gout and anticancer novel compounds interact with same receptors for their action, e.g., colchicine and colchicine analogues. Dietary flavonoids, i.e., chrysin, kaempferol, quercetin, fisetin, pelargonidin, apigenin, luteolin, myricetin, isorhamnetin, phloretinetc etc. have comparable IC50 values with established anti-gout drug and effective against both cancer and gout. Moreover, a noticeable number of newer anticancer compounds have already been isolated from plants that have been using by local traditional healers and herbal practitioners to treat gout. Therefore, the anti-gout plants might have greater potentiality to become selective candidates for screening of newer anticancer leads.
Collapse
|
9
|
Xia J, Wan Y, Wu JJ, Yang Y, Xu JF, Zhang L, Liu D, Chen L, Tang F, Ao H, Peng C. Therapeutic potential of dietary flavonoid hyperoside against non-communicable diseases: targeting underlying properties of diseases. Crit Rev Food Sci Nutr 2022; 64:1340-1370. [PMID: 36073729 DOI: 10.1080/10408398.2022.2115457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Non-communicable diseases (NCDs) are a global epidemic with diverse pathogenesis. Among them, oxidative stress and inflammation are the most fundamental co-morbid features. Therefore, multi-targets and multi-pathways therapies with significant anti-oxidant and anti-inflammatory activities are potential effective measures for preventing and treating NCDs. The flavonol glycoside compound hyperoside (Hyp) is widely found in a variety of fruits, vegetables, beverages, and medicinal plants and has various health benefits, especially excellent anti-oxidant and anti-inflammatory properties targeting nuclear factor erythroid 2-related factor 2 (Nrf2) and nuclear factor-κB (NF-κB) signaling pathways. In this review, we summarize the pathogenesis associated with oxidative stress and inflammation in NCDs and the biological activity and therapeutic potential of Hyp. Our findings reveal that the anti-oxidant and anti-inflammatory activities regulated by Hyp are associated with numerous biological mechanisms, including positive regulation of mitochondrial function, apoptosis, autophagy, and higher-level biological damage activities. Hyp is thought to be beneficial against organ injuries, cancer, depression, diabetes, and osteoporosis, and is a potent anti-NCDs agent. Additionally, the sources, bioavailability, pharmacy, and safety of Hyp have been established, highlighting the potential to develop Hyp into dietary supplements and nutraceuticals.
Collapse
Affiliation(s)
- Jia Xia
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiao-Jiao Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jin-Feng Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Wang Q, Wei HC, Zhou SJ, Li Y, Zheng TT, Zhou CZ, Wan XH. Hyperoside: A review on its sources, biological activities, and molecular mechanisms. Phytother Res 2022; 36:2779-2802. [PMID: 35561084 DOI: 10.1002/ptr.7478] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 12/22/2022]
Abstract
Hyperoside is a natural flavonol glycoside in various plants, such as Crataegus pinnatifida Bge, Forsythia suspensa, and Cuscuta chinensis Lam. Medical research has found that hyperoside possesses a broad spectrum of biological activities, including anticancer, anti-inflammatory, antibacterial, antiviral, antidepressant, and organ protective effects. These pharmacological properties lay the foundation for its use in treating multiple diseases, such as sepsis, arthritis, colitis, diabetic nephropathy, myocardial ischemia-reperfusion, pulmonary fibrosis, and cancers. Hyperoside is obtained from the plants and chemical synthesis. This study aims to provide a comprehensive overview of hyperoside on its sources and biological activities to provide insights into its therapeutic potential, and to provide a basis for high-quality studies to determine the clinical efficacy of this compound.
Collapse
Affiliation(s)
- Qi Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Hao-Cheng Wei
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Sheng-Jun Zhou
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Ying Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Ting-Ting Zheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Chang-Zheng Zhou
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Xin-Huan Wan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, China
| |
Collapse
|
11
|
Lam BQ, Srivastava R, Morvant J, Shankar S, Srivastava RK. Association of Diabetes Mellitus and Alcohol Abuse with Cancer: Molecular Mechanisms and Clinical Significance. Cells 2021; 10:cells10113077. [PMID: 34831299 PMCID: PMC8620339 DOI: 10.3390/cells10113077] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/28/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus (DM), one of the metabolic diseases which is characterized by sustained hyperglycemia, is a life-threatening disease. The global prevalence of DM is on the rise, mainly in low- and middle-income countries. Diabetes is a major cause of blindness, heart attacks, kidney failure, stroke, and lower limb amputation. Type 2 diabetes mellitus (T2DM) is a form of diabetes that is characterized by high blood sugar and insulin resistance. T2DM can be prevented or delayed by a healthy diet, regular physical activity, maintaining normal body weight, and avoiding alcohol and tobacco use. Ethanol and its metabolites can cause differentiation defects in stem cells and promote inflammatory injury and carcinogenesis in several tissues. Recent studies have suggested that diabetes can be treated, and its consequences can be avoided or delayed with proper management. DM has a greater risk for several cancers, such as breast, colorectal, endometrial, pancreatic, gallbladder, renal, and liver cancer. The incidence of cancer is significantly higher in patients with DM than in those without DM. In addition to DM, alcohol abuse is also a risk factor for many cancers. We present a review of the recent studies investigating the association of both DM and alcohol abuse with cancer incidence.
Collapse
Affiliation(s)
- Bao Q. Lam
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (B.Q.L.); (S.S.)
| | - Rashmi Srivastava
- Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
| | - Jason Morvant
- Department of Surgery, Ochsner Health System, 120 Ochsner Boulevard, Gretna, LA 70056, USA;
- A.B. Freeman School of Business, Tulane University, New Orleans, LA 70118, USA
| | - Sharmila Shankar
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (B.Q.L.); (S.S.)
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- John W. Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA
| | - Rakesh K. Srivastava
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (B.Q.L.); (S.S.)
- A.B. Freeman School of Business, Tulane University, New Orleans, LA 70118, USA
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Correspondence:
| |
Collapse
|
12
|
Kim A, Ha J, Kim J, Cho Y, Ahn J, Cheon C, Kim SH, Ko SG, Kim B. Natural Products for Pancreatic Cancer Treatment: From Traditional Medicine to Modern Drug Discovery. Nutrients 2021; 13:nu13113801. [PMID: 34836055 PMCID: PMC8625071 DOI: 10.3390/nu13113801] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer, the seventh most lethal cancer around the world, is considered complicated cancer due to poor prognosis and difficulty in treatment. Despite all the conventional treatments, including surgical therapy and chemotherapy, the mortality rate is still high. Therefore, the possibility of using natural products for pancreatic cancer is increasing. In this study, 68 natural products that have anti-pancreatic cancer effects reported within five years were reviewed. The mechanisms of anti-cancer effects were divided into four types: apoptosis, anti-metastasis, anti-angiogenesis, and anti-resistance. Most of the studies were conducted for natural products that induce apoptosis in pancreatic cancer. Among them, plant extracts such as Eucalyptus microcorys account for the major portion. Some natural products, including Moringa, Coix seed, etc., showed multi-functional properties. Natural products could be beneficial candidates for treating pancreatic cancer.
Collapse
Affiliation(s)
- Ahyeon Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (A.K.); (J.H.); (J.K.)
| | - Jiwon Ha
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (A.K.); (J.H.); (J.K.)
| | - Jeongeun Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (A.K.); (J.H.); (J.K.)
| | - Yongmin Cho
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (Y.C.); (J.A.); (S.-H.K.)
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (C.C.); (S.-G.K.)
| | - Jimyung Ahn
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (Y.C.); (J.A.); (S.-H.K.)
| | - Chunhoo Cheon
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (C.C.); (S.-G.K.)
| | - Sung-Hoon Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (Y.C.); (J.A.); (S.-H.K.)
| | - Seong-Gyu Ko
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (C.C.); (S.-G.K.)
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (A.K.); (J.H.); (J.K.)
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (Y.C.); (J.A.); (S.-H.K.)
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea; (C.C.); (S.-G.K.)
- Correspondence: ; Tel.: +82-2-961-9217
| |
Collapse
|
13
|
Zhang Y, Zhang Y, Gao W, Zhou R, Liu F, Ng TB. A novel antitumor protein from the mushroom Pholiota nameko induces apoptosis of human breast adenocarcinoma MCF-7 cells in vivo and modulates cytokine secretion in mice bearing MCF-7 xenografts. Int J Biol Macromol 2020; 164:3171-3178. [DOI: 10.1016/j.ijbiomac.2020.08.187] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/22/2020] [Accepted: 08/23/2020] [Indexed: 11/15/2022]
|
14
|
Benvenuto M, Albonici L, Focaccetti C, Ciuffa S, Fazi S, Cifaldi L, Miele MT, De Maio F, Tresoldi I, Manzari V, Modesti A, Masuelli L, Bei R. Polyphenol-Mediated Autophagy in Cancer: Evidence of In Vitro and In Vivo Studies. Int J Mol Sci 2020; 21:E6635. [PMID: 32927836 PMCID: PMC7555128 DOI: 10.3390/ijms21186635] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
One of the hallmarks of cellular transformation is the altered mechanism of cell death. There are three main types of cell death, characterized by different morphological and biochemical features, namely apoptosis (type I), autophagic cell death (type II) and necrosis (type III). Autophagy, or self-eating, is a tightly regulated process involved in stress responses, and it is a lysosomal degradation process. The role of autophagy in cancer is controversial and has been associated with both the induction and the inhibition of tumor growth. Autophagy can exert tumor suppression through the degradation of oncogenic proteins, suppression of inflammation, chronic tissue damage and ultimately by preventing mutations and genetic instability. On the other hand, tumor cells activate autophagy for survival in cellular stress conditions. Thus, autophagy modulation could represent a promising therapeutic strategy for cancer. Several studies have shown that polyphenols, natural compounds found in foods and beverages of plant origin, can efficiently modulate autophagy in several types of cancer. In this review, we summarize the current knowledge on the effects of polyphenols on autophagy, highlighting the conceptual benefits or drawbacks and subtle cell-specific effects of polyphenols for envisioning future therapies employing polyphenols as chemoadjuvants.
Collapse
Affiliation(s)
- Monica Benvenuto
- Saint Camillus International University of Health and Medical Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy;
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (L.A.); (C.F.); (S.C.); (L.C.); (F.D.M.); (I.T.); (V.M.); (A.M.)
| | - Loredana Albonici
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (L.A.); (C.F.); (S.C.); (L.C.); (F.D.M.); (I.T.); (V.M.); (A.M.)
| | - Chiara Focaccetti
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (L.A.); (C.F.); (S.C.); (L.C.); (F.D.M.); (I.T.); (V.M.); (A.M.)
- Department of Human Science and Promotion of the Quality of Life, San Raffaele University Rome, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Sara Ciuffa
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (L.A.); (C.F.); (S.C.); (L.C.); (F.D.M.); (I.T.); (V.M.); (A.M.)
| | - Sara Fazi
- Department of Experimental Medicine, University of Rome “Sapienza”, Viale Regina Elena 324, 00161 Rome, Italy; (S.F.); (L.M.)
| | - Loredana Cifaldi
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (L.A.); (C.F.); (S.C.); (L.C.); (F.D.M.); (I.T.); (V.M.); (A.M.)
- Academic Department of Pediatrics (DPUO), Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy
| | - Martino Tony Miele
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
| | - Fernando De Maio
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (L.A.); (C.F.); (S.C.); (L.C.); (F.D.M.); (I.T.); (V.M.); (A.M.)
| | - Ilaria Tresoldi
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (L.A.); (C.F.); (S.C.); (L.C.); (F.D.M.); (I.T.); (V.M.); (A.M.)
| | - Vittorio Manzari
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (L.A.); (C.F.); (S.C.); (L.C.); (F.D.M.); (I.T.); (V.M.); (A.M.)
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (L.A.); (C.F.); (S.C.); (L.C.); (F.D.M.); (I.T.); (V.M.); (A.M.)
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome “Sapienza”, Viale Regina Elena 324, 00161 Rome, Italy; (S.F.); (L.M.)
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (L.A.); (C.F.); (S.C.); (L.C.); (F.D.M.); (I.T.); (V.M.); (A.M.)
| |
Collapse
|
15
|
Qiu J, Zhang T, Zhu X, Yang C, Wang Y, Zhou N, Ju B, Zhou T, Deng G, Qiu C. Hyperoside Induces Breast Cancer Cells Apoptosis via ROS-Mediated NF-κB Signaling Pathway. Int J Mol Sci 2019; 21:ijms21010131. [PMID: 31878204 PMCID: PMC6981893 DOI: 10.3390/ijms21010131] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/17/2019] [Accepted: 12/21/2019] [Indexed: 01/10/2023] Open
Abstract
Hyperoside (quercetin 3-o-β-d-galactopyranoside) is one of the flavonoid glycosides with anti-inflammatory, antidepressant, and anti-cancer effects. But it remains unknown whether it had effects on breast cancer. Here, different concentrations of hyperoside were used to explore its therapeutic potential in both breast cancer cells and subcutaneous homotransplant mouse model. CCK-8 and wound healing assays showed that the viability and migration capability of Michigan Cancer Foundation-7 (MCF-7) and 4T1 cells were inhibited by hyperoside, while the apoptosis of cells were increased. Real-time quantitative PCR (qRT-PCR) and western blot analysis were used to detect mRNA and the protein level, respectively, which showed decreased levels of B cell lymphoma-2 (Bcl-2) and X-linked inhibitor of apoptosis (XIAP), and increased levels of Bax and cleaved caspase-3. After exploration of the potential mechanism, we found that reactive oxygen species (ROS) production was reduced by the administration of hyperoside, which subsequently inhibited the activation of NF-κB signaling pathway. Tumor volume was significantly decreased in subcutaneous homotransplant mouse model in hyperoside-treated group, which was consistent with our study in vitro. These results indicated that hyperoside acted as an anticancer drug through ROS-related apoptosis and its mechanism included activation of the Bax-caspase-3 axis and the inhibition of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jinxia Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.Q.); (T.Z.); (X.Z.); (C.Y.); (N.Z.); (B.J.); (T.Z.); (G.D.)
| | - Tao Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.Q.); (T.Z.); (X.Z.); (C.Y.); (N.Z.); (B.J.); (T.Z.); (G.D.)
| | - Xinying Zhu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.Q.); (T.Z.); (X.Z.); (C.Y.); (N.Z.); (B.J.); (T.Z.); (G.D.)
| | - Chao Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.Q.); (T.Z.); (X.Z.); (C.Y.); (N.Z.); (B.J.); (T.Z.); (G.D.)
| | - Yaxing Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Ning Zhou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.Q.); (T.Z.); (X.Z.); (C.Y.); (N.Z.); (B.J.); (T.Z.); (G.D.)
| | - Bingxin Ju
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.Q.); (T.Z.); (X.Z.); (C.Y.); (N.Z.); (B.J.); (T.Z.); (G.D.)
| | - Tianhong Zhou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.Q.); (T.Z.); (X.Z.); (C.Y.); (N.Z.); (B.J.); (T.Z.); (G.D.)
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.Q.); (T.Z.); (X.Z.); (C.Y.); (N.Z.); (B.J.); (T.Z.); (G.D.)
| | - Changwei Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (J.Q.); (T.Z.); (X.Z.); (C.Y.); (N.Z.); (B.J.); (T.Z.); (G.D.)
- Correspondence: ; Tel.: +86-180-8661-7267
| |
Collapse
|
16
|
Guo W, Yu H, Zhang L, Chen X, Liu Y, Wang Y, Zhang Y. Effect of hyperoside on cervical cancer cells and transcriptome analysis of differentially expressed genes. Cancer Cell Int 2019; 19:235. [PMID: 31516392 PMCID: PMC6734331 DOI: 10.1186/s12935-019-0953-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background Hyperoside (Hy) is a plant-derived quercetin 3-d-galactoside that exhibits inhibitory activities on various tumor types. The objective of the current study was to explore Hy effects on cervical cancer cell proliferation, and to perform a transcriptome analysis of differentially expressed genes. Methods Cervical cancer HeLa and C-33A cells were cultured and the effect of Hy treatment was determined using the Cell Counting Kit-8 (CCK-8) assay. After calculating the IC50 of Hy in HeLa and C-33A cells, the more sensitive to Hy treatment cell type was selected for RNA-Seq. Differentially expressed genes (DEGs) were identified by comparing gene expression between the Hy and control groups. Candidate genes were determined through DEG analysis, protein interaction network (PPI) construction, PPI module analysis, transcription factor (TF) prediction, TF-target network construction, and survival analysis. Finally, the key candidate genes were verified by RT-qPCR and western blot. Results Hy inhibited HeLa and C33A cell proliferation in a dose- and time-dependent manner, as determined by the CCK-8 assay. Treatment of C-33A cells with 2 mM Hy was selected for the subsequent experiments. Compared with the control group, 754 upregulated and 509 downregulated genes were identified after RNA-Seq. After functional enrichment, 74 gene ontology biological processes and 43 Kyoto Encyclopedia of Genes and Genomes pathways were obtained. According to the protein interaction network (PPI), PPI module analysis, TF-target network construction, and survival analysis, the key genes MYC, CNKN1A, PAX2, TFRC, ACOX2, UNC5B, APBA1, PRKACA, PEAR1, COL12A1, CACNA1G, PEAR1, and CCNA2 were detected. RT-qPCR was performed on the key genes, and Western blot was used to verify C-MYC and TFRC. C-MYC and TFRC expressions were lower and higher than the corresponding values in the control group, respectively, in accordance with the results from the RNA-Seq analysis. Conclusion Hy inhibited HeLa and C-33A cell proliferation through C-MYC gene expression reduction in C-33A cells and TFRC regulation. The results of the current study provide a theoretical basis for Hy treatment of cervical cancer.
Collapse
Affiliation(s)
- Weikang Guo
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Hui Yu
- 2Department of Cardiopulmonary Function, Harbin Medical University Cancer Hospital, Harbin, 150081 Heilongjiang Province China
| | - Lu Zhang
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Xiuwei Chen
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Yunduo Liu
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Yaoxian Wang
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Yunyan Zhang
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| |
Collapse
|
17
|
Swanepoel B, Venables L, Olaru OT, Nitulescu GM, van de Venter M. In Vitro Anti-proliferative Activity and Mechanism of Action of Anemone nemorosa. Int J Mol Sci 2019; 20:ijms20051217. [PMID: 30862032 PMCID: PMC6429291 DOI: 10.3390/ijms20051217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 01/17/2023] Open
Abstract
Anemone nemorosa is part of the Ranunculaceae genus Anemone (order Ranunculales) which comprises more than 150 species. Various parts of the plant have been used for the treatment of numerous medical conditions such as headaches, tertian agues, rheumatic gout, leprosy, lethargy, eye inflammation as well as malignant and corroding ulcers. The Anemone plants have been found to contain various medicinal compounds with anti-cancer, immunomodulatory, anti-inflammatory, anti-oxidant and anti-microbial activities. To date there has been no reported evidence of its use in the treatment of cancer. However, due to the reported abundance of saponins which usually exert anti-cancer activity via cell cycle arrest and the induction of apoptosis, we investigated the mode of cell death induced by an aqueous A. nemorosa extract by using HeLa cervical cancer cells. Cisplatin was used as a positive control. With a 50% inhibitory concentration (IC50) of 20.33 ± 2.480 µg/mL, treatment with A. nemorosa yielded a delay in the early mitosis phase of the cell cycle. Apoptosis was confirmed through fluorescent staining with annexin V-FITC. Apoptosis was more evident with A. nemorosa treatment compared to the positive control after 24 and 48 h. Tetramethylrhodamine ethyl ester staining showed a decrease in mitochondrial membrane potential at 24 and 48 h. The results obtained imply that A. nemorosa may have potential anti-proliferative properties.
Collapse
Affiliation(s)
- Bresler Swanepoel
- Department of Biochemistry and Microbiology, PO Box 77000, Nelson Mandela University, Port Elizabeth 6031, South Africa.
| | - Luanne Venables
- Department of Biochemistry and Microbiology, PO Box 77000, Nelson Mandela University, Port Elizabeth 6031, South Africa.
| | - Octavian Tudorel Olaru
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, Traian Vuia 6, Bucharest 020956, Romania.
| | - George Mihai Nitulescu
- Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, Traian Vuia 6, Bucharest 020956, Romania.
| | - Maryna van de Venter
- Department of Biochemistry and Microbiology, PO Box 77000, Nelson Mandela University, Port Elizabeth 6031, South Africa.
| |
Collapse
|
18
|
Ma C, Wen B, Zhang Q, Shao P, Gu W, Qu K, Shi Y, Wang B. Polydatin Regulates the Apoptosis and Autophagy of Fibroblasts Obtained from Patients with Ankylosing Spondylitis. Biol Pharm Bull 2019; 42:50-56. [PMID: 30333377 DOI: 10.1248/bpb.b18-00522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The development of ankylosing spondylitis (AS) occurs due to excessive proliferation of fibroblasts. Polydatin, a monomeric compound isolated from a traditional Chinese medicine Polygonum cuspidatum, exhibits anti-inflammatory and anti-arthritic effects. However, the mechanisms underlying the regulatory effects of polydatin on the proliferation, apoptosis and autophagy of fibroblasts obtained from patients with AS remain unclear. The aim of this study was to investigate the therapeutic effects of polydatin on symptoms associated with AS. Multiple cellular and molecular biology experiments were performed in the present study, such as cell viability assay, Western blotting, flow cytometry, monodansylcadaverine (MDC) staining and immunofluorescence assays. In the present study, the results revealed that polydatin induced the apoptosis of fibroblasts isolated from patients with AS by upregulating the expression of active caspase-3 and Bax, and downregulating the expression of Bcl-2. Meanwhile, polydatin was revealed to enhance the autophagy of fibroblasts by increasing the expression levels of LC3II, Beclin 1 and Atg5. The results of MDC and immunofluorescence assays further demonstrated that polydatin significantly induced the formation of autophagosomes in fibroblasts. Furthermore, polydatin-induced apoptosis and autophagy were markedly inhibited following treatment with the autophagy inhibitor, 3-methyladenine (3-MA). In conclusion, the results of the present study indicated that polydatin induces the apoptosis and autophagy of fibroblasts obtained from patients suffering from AS, and that polydatin may represent a therapeutic agent for the future treatment of patients with AS.
Collapse
Affiliation(s)
- Cong Ma
- Department of Rheumatology and Immunology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University
| | - Bo Wen
- Department of Rheumatology and Immunology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University
| | - Qin Zhang
- Department of Rheumatology and Immunology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University
| | - Peipei Shao
- Department of Rheumatology and Immunology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University
| | - Wen Gu
- Department of Rheumatology and Immunology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University
| | - Kun Qu
- Department of Rheumatology and Immunology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University
| | - Yang Shi
- Department of Rheumatology and Immunology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University
| | - Bei Wang
- Department of Rheumatology and Immunology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University
| |
Collapse
|
19
|
Bonucci M, Pastore C, Ferrera V, Fiorentini C, Fabbri A. Integrated Cancer Treatment in the Course of Metastatic Pancreatic Cancer: Complete Resolution in 2 Cases. Integr Cancer Ther 2018; 17:994-999. [PMID: 29478350 PMCID: PMC6142071 DOI: 10.1177/1534735418755479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pancreatic cancer (PC) has a very low average survival, but its prognosis is
further reduced in the case of metastatic spread. Medical therapy in these cases
is the only applicable methodology in the international guidelines. During
anticancer treatments, common side effects are nausea, vomiting, arthralgia,
neuropathy, and alopecia as well as a myelosuppressive effect. The toxicity of
various drugs not only affects the quality of life of the patient, but often its
severity requires a reduction in if not the termination of drug administration.
Scientific studies have shown that a combined use of chemotherapy and certain
natural substances, in the form of standardized extracts, can lead to an
enhancement of the action of the chemotherapy. Here, we describe 2 cases of
metastatic PC. The first case concerns the integrated treatment of a patient
with cancer of the pancreas tail with metastatic involvement ab initio of
peripancreatic lymph nodes and liver parenchyma, with numerous secondary lesions
greater than 9.5 cm. The second case concerns the integrated treatment of a
patient with cancer of the pancreatic body with metastatic involvement of the
liver parenchyma, with a small secondary lesion. In both cases, an integrated
cancer treatment approach, combining chemotherapy with natural remedies,
extracts, and hyperthermia, induced a notable remission of primary and
metastatic lesions.
Collapse
|
20
|
Hwang HV, Tran DT, Rebuffatti MN, Li CS, Knowlton AA. Investigation of quercetin and hyperoside as senolytics in adult human endothelial cells. PLoS One 2018; 13:e0190374. [PMID: 29315311 PMCID: PMC5760026 DOI: 10.1371/journal.pone.0190374] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/13/2017] [Indexed: 01/09/2023] Open
Abstract
Quercetin has been reported to act as a senolytic by selectively removing senescent endothelial cells, and thus it would seem quercetin could revolutionize the field of gerontology. However, given quercetin's narrow therapeutic index reported in work done with human umbilical vein endothelial cells (HUVECs), we hypothesized that quercetin is not innocuous for non-senescent adult human vascular endothelial cells at concentrations that have been reported to be safe for proliferating HUVECs. Furthermore, we investigated quercetin 3-D-galactoside (Q3G; hyperoside), an inactive quercetin derivative that needs to be cleaved by beta-galactosidase overexpressed in senescent cells to release quercetin, as a potential safer senolytic. We compared the effectiveness of quercetin and Q3G in primary human coronary artery endothelial cells (HCAEC), which are adult microvascular cells. We found that quercetin caused cell death in non-senescent endothelial cells at a concentration that has been reported to selectively remove senescent cells, and that Q3G was not cytotoxic to either young or senescent cells. Thus, in primary adult human endothelial cells, quercetin and Q3G are not senolytics. Earlier work reporting positive results was done with HUVECs, and given their origin and the disparate findings from the current study, these may not be the best cells for evaluating potential senolytics in clinically relevant endothelial cells.
Collapse
Affiliation(s)
- HyunTae V. Hwang
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
| | - Darlene Thuy Tran
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
| | - Michelle Nicole Rebuffatti
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
| | - Chin-Shang Li
- Division of Biostatistics, Department of Public Health Sciences, University of California-Davis, Davis, CA, United States of America
| | - Anne A. Knowlton
- Molecular & Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California-Davis, Davis, CA, United States of America
- VA Medical Center, Sacramento, CA, United States of America
- Pharmacology Department, University of California-Davis, Davis, CA, United States of America
- * E-mail:
| |
Collapse
|
21
|
Niu C, Ma M, Han X, Wang Z, Li H. Hyperin protects against cisplatin-induced liver injury in mice. Acta Cir Bras 2017; 32:633-640. [PMID: 28902939 DOI: 10.1590/s0102-865020170080000005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/18/2017] [Indexed: 12/30/2022] Open
Affiliation(s)
- Chengwei Niu
- Jiangsu Food and Pharmaceutical Science College, China
| | - Man Ma
- Jiangsu Food and Pharmaceutical Science College, China
| | - Xiao Han
- Huai an Hospital of Huai’an, China
| | - Zimin Wang
- Jiangsu Food and Pharmaceutical Science College, China
| | - Hangyan Li
- Jiangsu Food and Pharmaceutical Science College, China
| |
Collapse
|
22
|
Zhu X, Ji M, Han Y, Guo Y, Zhu W, Gao F, Yang X, Zhang C. PGRMC1-dependent autophagy by hyperoside induces apoptosis and sensitizes ovarian cancer cells to cisplatin treatment. Int J Oncol 2017; 50:835-846. [PMID: 28197632 DOI: 10.3892/ijo.2017.3873] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/12/2016] [Indexed: 11/06/2022] Open
Abstract
Cisplatin treatment some times leads to chemoresistance, which is now acknowledged partially due to the inductive expression of progesterone receptor membrane component (PGRMC)1 in ovarian cancer cells. PGRMC1 enhances autophagy, activates cytochrome p450, and inveigles signaling pathways to promote cell survival and reduce the effect of drug treatments. In this study, we give first line evidence that hyperoside inhibits cell viability, triggers autophagy and apoptosis in ovarian cancer cell lines. Mechanistically, PGRMC1-dependent autophagy was utilized by hyperoside to induce apoptotic cell death. Hyperoside induced the conversion of LC3B-I to LC3B-II and the formation of autophagosomes in ovarian cancer cells. Notably, PGRMC1 colocolized with LC3B‑II, and PGRMC1 overexpression enhanced hyperoside-induced autophagy and apoptosis, while PGRMC1 knockdown abrogated the action. Additionally, AKT signaling and Bcl-2 family were also involved in the hyperoside-induced autophagy and apoptosis. Importantly, in cisplatin-resistant ovarian cancer cells where PGRMC1 was overexpressed, hyperoside sensitized the cells to cisplatin treatment. Together these findings indicate hyperoside functions as a complementary therapy for ovarian cancer patients receiving platinum-based therapy.
Collapse
Affiliation(s)
- Xiaofei Zhu
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Mingde Ji
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Yue Han
- Department of Gynecology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Yuanyuan Guo
- Department of Biochemistry, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Wenqiang Zhu
- Department of Surgical Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Feng Gao
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Xuewen Yang
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Chunbing Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|