1
|
Tian F, Sun S, Ge Z, Ge Y, Ge X, Shi Z, Qian X. Understanding the Anticancer Effects of Phytochemicals: From Molecular Docking to Anticarcinogenic Signaling. J Nutr 2025; 155:431-444. [PMID: 39581266 DOI: 10.1016/j.tjnut.2024.11.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024] Open
Abstract
As nontraditional nutrients, the biological activity of phytochemicals have been extensively studied for their antioxidant, anti-inflammatory, and apoptosis-promoting effects in various diseases. The general anticancer benefits of phytochemicals have been demonstrated in both basic researches and clinical trials. However, researchers understanding of how phytochemicals target cancer-related signaling pathways is still in its infancy. Molecular docking simulation analyses have yielded a large amount of cellular target molecules of phytochemicals. Herein, we review the potential signaling pathways that may be involved in the phytochemical-driven cancer benefits. We expect these findings to help in the design of potential cancer treatments designed by manipulating the binding modes and sites of these plant chemicals.
Collapse
Affiliation(s)
- Fuwei Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuhong Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zehe Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuqian Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Ge
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhumei Shi
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurosurgery of the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xu Qian
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Neurosurgery of the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
2
|
El Mahi Y, Nizami ZN, Wali AF, Al Neyadi A, Magramane M, Al Azzani M, Arafat K, Attoub S, Eid AH, Iratni R. Rhus coriaria induces autophagic and apoptotic cell death in pancreatic cancer cells. Front Pharmacol 2024; 15:1412565. [PMID: 39139643 PMCID: PMC11319293 DOI: 10.3389/fphar.2024.1412565] [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: 04/05/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Background:Pancreatic cancer is a leading cause of cancer-related mortality worldwide with increasing global incidence. We previously reported the anticancer effect of Rhus coriaria ethanolic extract (RCE) in triple negative breast and colon cancer cells. Herein, we investigated the anticancer effect of RCE on human pancreatic cancer cells. Methods: Cell viability was measured using Cell Titer-Glo and staining of viable and dead cells based on differential permeability to two DNA binding dyes. Cell cycle distribution and annexin V staining was carried out in Muse cell analyzer. Protein level was determined by Western blot. Tumor growth was assessed by in ovo chick embryo chorioallantoic membrane assay. Results: We found that RCE significantly inhibited the viability and colony growth of pancreatic cancer cells (Panc-1, Mia-PaCa-2, S2-013, AsPC-1). The antiproliferative effects of RCE in pancreatic cancer cells (Panc-1 and Mia-PaCa-2) were mediated through induction of G1 cell cycle arrest, Beclin-1-independent autophagy, and apoptosis. RCE activated both the extrinsic and intrinsic pathways of apoptosis and regulated the Bax/Bcl-2 apoptotic switch. Mechanistically, we found that RCE inhibited the AKT/mTOR pathway, downstream of which, inactivation of the cell cycle regulator p70S6K and downregulation of the antiapoptotic protein survivin was observed. Additionally, we found that RCE-induced autophagy preceded apoptosis. Further, we confirmed the anticancer effect of RCE in a chick embryo xenograft model and found that RCE inhibited the growth of pancreatic cancer xenografts without affecting embryo survival. Conclusion: Collectively, our findings demonstrate that Rhus coriaria exerts potent anti-pancreatic cancer activity though cell cycle impairment, autophagy, and apoptosis, and is hence a promising source of anticancer phytochemicals.
Collapse
Affiliation(s)
- Yassine El Mahi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Adil Farooq Wali
- Department of Pharmaceutical Chemistry, RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Aysha Al Neyadi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohamed Magramane
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mazoun Al Azzani
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Kholoud Arafat
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Samir Attoub
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
3
|
Gu M, Liu Y, Xin P, Guo W, Zhao Z, Yang X, Ma R, Jiao T, Zheng W. Fundamental insights and molecular interactions in pancreatic cancer: Pathways to therapeutic approaches. Cancer Lett 2024; 588:216738. [PMID: 38401887 DOI: 10.1016/j.canlet.2024.216738] [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: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/18/2024] [Indexed: 02/26/2024]
Abstract
The gastrointestinal tract can be affected by a number of diseases that pancreatic cancer (PC) is a malignant manifestation of them. The prognosis of PC patients is unfavorable and because of their diagnosis at advanced stage, the treatment of this tumor is problematic. Owing to low survival rate, there is much interest towards understanding the molecular profile of PC in an attempt in developing more effective therapeutics. The conventional therapeutics for PC include surgery, chemotherapy and radiotherapy as well as emerging immunotherapy. However, PC is still incurable and more effort should be performed. The molecular landscape of PC is an underlying factor involved in increase in progression of tumor cells. In the presence review, the newest advances in understanding the molecular and biological events in PC are discussed. The dysregulation of molecular pathways including AMPK, MAPK, STAT3, Wnt/β-catenin and non-coding RNA transcripts has been suggested as a factor in development of tumorigenesis in PC. Moreover, cell death mechanisms such as apoptosis, autophagy, ferroptosis and necroptosis demonstrate abnormal levels. The EMT and glycolysis in PC cells enhance to ensure their metastasis and proliferation. Furthermore, such abnormal changes have been used to develop corresponding pharmacological and nanotechnological therapeutics for PC.
Collapse
Affiliation(s)
- Ming Gu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Yang Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Peng Xin
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Zimo Zhao
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Xu Yang
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Ruiyang Ma
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Taiwei Jiao
- Department of Gastroenterology and Endoscopy, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Wenhui Zheng
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
4
|
Bojňanská T, Kolesárová A, Čech M, Tančinová D, Urminská D. Extracts with Nutritional Potential and Their Influence on the Rheological Properties of Dough and Quality Parameters of Bread. Foods 2024; 13:382. [PMID: 38338518 PMCID: PMC10855696 DOI: 10.3390/foods13030382] [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: 12/19/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Formulating basic food to improve its nutritional profile is one potential method for food innovation. One option in formulating basic food such as bread is to supplement flours with specified amounts of non-bakery raw materials with high nutritional benefits. In the research presented here, we studied the influence of the addition of curcumin and quercetin extracts in amounts of 2.5% and 5% to wheat flour (2.5:97.5; 5:95). The analysis of the rheological properties of dough was carried out using a Mixolab 2. A Rheofermentometer F4 was used to assess the dough's fermentation, and a Volscan was used to evaluate the baking trials. The effect of the extracts on the rheological properties of dough was measured and found to be statistically significant, with curcumin shortening both dough development time and dough stability. Doughs made with greater quantities of extract had a greater tendency to early starch retrogradation, which negatively affects the shelf life of the end products. The addition of extracts did not significantly affect either the ability to form gas during fermentation or its retention, which is important because this gas is prerequisite to forming a final product with the required volume and porosity of crumb. Less favourable results were found on sensory evaluation, wherein the trial bread was significantly worse than the control wheat bread. The panel's decision-making might have been influenced by the atypical colour of the bread made with additives, and in case of a trial bread made with quercetin, by a bitter taste. From the technological point of view, the results confirmed that the composite flours prepared with the addition of extracts of curcumin and quercetin in amounts of 2.5% and 5% can be processed according to standard procedures. The final product will be bread with improved nutritional profile and specific sensory properties, specifically an unconventional and attractive colour.
Collapse
Affiliation(s)
- Tatiana Bojňanská
- Institute of Food Sciences, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda A. Hlinku 2, 949 76 Nitra, Slovakia; (A.K.); (M.Č.)
| | - Anna Kolesárová
- Institute of Food Sciences, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda A. Hlinku 2, 949 76 Nitra, Slovakia; (A.K.); (M.Č.)
| | - Matej Čech
- Institute of Food Sciences, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda A. Hlinku 2, 949 76 Nitra, Slovakia; (A.K.); (M.Č.)
| | - Dana Tančinová
- Institute of Biotechnology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda A. Hlinku 2, 949 76 Nitra, Slovakia; (D.T.); (D.U.)
| | - Dana Urminská
- Institute of Biotechnology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Trieda A. Hlinku 2, 949 76 Nitra, Slovakia; (D.T.); (D.U.)
| |
Collapse
|
5
|
Ashrafizadeh M, Luo K, Zhang W, Reza Aref A, Zhang X. Acquired and intrinsic gemcitabine resistance in pancreatic cancer therapy: Environmental factors, molecular profile and drug/nanotherapeutic approaches. ENVIRONMENTAL RESEARCH 2024; 240:117443. [PMID: 37863168 DOI: 10.1016/j.envres.2023.117443] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/17/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023]
Abstract
A high number of cancer patients around the world rely on gemcitabine (GEM) for chemotherapy. During local metastasis of cancers, surgery is beneficial for therapy, but dissemination in distant organs leads to using chemotherapy alone or in combination with surgery to prevent cancer recurrence. Therapy failure can be observed as a result of GEM resistance, threatening life of pancreatic cancer (PC) patients. The mortality and morbidity of PC in contrast to other tumors are increasing. GEM chemotherapy is widely utilized for PC suppression, but resistance has encountered its therapeutic impacts. The purpose of current review is to bring a broad concept about role of biological mechanisms and pathways in the development of GEM resistance in PC and then, therapeutic strategies based on using drugs or nanostructures for overcoming chemoresistance. Dysregulation of the epigenetic factors especially non-coding RNA transcripts can cause development of GEM resistance in PC and miRNA transfection or using genetic tools such as siRNA for modulating expression level of these factors for changing GEM resistance are suggested. The overexpression of anti-apoptotic proteins and survival genes can contribute to GEM resistance in PC. Moreover, supportive autophagy inhibits apoptosis and stimulates GEM resistance in PC cells. Increase in metabolism, glycolysis induction and epithelial-mesenchymal transition (EMT) stimulation are considered as other factors participating in GEM resistance in PC. Drugs can suppress tumorigenesis in PC and inhibit survival factors and pathways in increasing GEM sensitivity in PC. More importantly, nanoparticles can increase pharmacokinetic profile of GEM and promote its blood circulation and accumulation in cancer site. Nanoparticles mediate delivery of GEM with genes and drugs to suppress tumorigenesis in PC and increase drug sensitivity. The basic research displays significant connection among dysregulated pathways and GEM resistance, but the lack of clinical application is a drawback that can be responded in future.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong, 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Kuo Luo
- Department of Oncology, Chongqing Hyheia Hospital, Chongqing, 4001331, China
| | - Wei Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Xianbin Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
6
|
Luiz-Ferreira A, Pacifico T, Cruz ÁC, Laudisi F, Monteleone G, Stolfi C. TRAIL-Sensitizing Effects of Flavonoids in Cancer. Int J Mol Sci 2023; 24:16596. [PMID: 38068921 PMCID: PMC10706592 DOI: 10.3390/ijms242316596] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) represents a promising anticancer agent, as it selectively induces apoptosis in transformed cells without altering the cellular machinery of healthy cells. Unfortunately, the presence of TRAIL resistance mechanisms in a variety of cancer types represents a major hurdle, thus limiting the use of TRAIL as a single agent. Accumulating studies have shown that TRAIL-mediated apoptosis can be facilitated in resistant tumors by combined treatment with antitumor agents, ranging from synthetic molecules to natural products. Among the latter, flavonoids, the most prevalent polyphenols in plants, have shown remarkable competence in improving TRAIL-driven apoptosis in resistant cell lines as well as tumor-bearing mice with minimal side effects. Here, we summarize the molecular mechanisms, such as the upregulation of death receptor (DR)4 and DR5 and downregulation of key anti-apoptotic proteins [e.g., cellular FLICE-inhibitory protein (c-FLIP), X-linked inhibitor of apoptosis protein (XIAP), survivin], underlying the TRAIL-sensitizing properties of different classes of flavonoids (e.g., flavones, flavonols, isoflavones, chalcones, prenylflavonoids). Finally, we discuss limitations, mainly related to bioavailability issues, and future perspectives regarding the clinical use of flavonoids as adjuvant agents in TRAIL-based therapies.
Collapse
Affiliation(s)
- Anderson Luiz-Ferreira
- Inflammatory Bowel Disease Research Laboratory, Department of Biological Sciences, Institute of Biotechnology, Federal University of Catalão (UFCAT), Catalão 75704020, GO, Brazil;
| | - Teresa Pacifico
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Álefe Cardoso Cruz
- Inflammatory Bowel Disease Research Laboratory, Department of Biological Sciences, Institute of Biotechnology, Federal University of Catalão (UFCAT), Catalão 75704020, GO, Brazil;
| | - Federica Laudisi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| |
Collapse
|
7
|
Aghababaei F, Hadidi M. Recent Advances in Potential Health Benefits of Quercetin. Pharmaceuticals (Basel) 2023; 16:1020. [PMID: 37513932 PMCID: PMC10384403 DOI: 10.3390/ph16071020] [Citation(s) in RCA: 157] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/08/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Quercetin, a flavonoid found in fruits and vegetables, has been a part of human diets for centuries. Its numerous health benefits, including antioxidant, antimicrobial, anti-inflammatory, antiviral, and anticancer properties, have been extensively studied. Its strong antioxidant properties enable it to scavenge free radicals, reduce oxidative stress, and protect against cellular damage. Quercetin's anti-inflammatory properties involve inhibiting the production of inflammatory cytokines and enzymes, making it a potential therapeutic agent for various inflammatory conditions. It also exhibits anticancer effects by inhibiting cancer cell proliferation and inducing apoptosis. Finally, quercetin has cardiovascular benefits such as lowering blood pressure, reducing cholesterol levels, and improving endothelial function, making it a promising candidate for preventing and treating cardiovascular diseases. This review provides an overview of the chemical structure, biological activities, and bioavailability of quercetin, as well as the different delivery systems available for quercetin. Incorporating quercetin-rich foods into the diet or taking quercetin supplements may be beneficial for maintaining good health and preventing chronic diseases. As research progresses, the future perspectives of quercetin appear promising, with potential applications in nutraceuticals, pharmaceuticals, and functional foods to promote overall well-being and disease prevention. However, further studies are needed to elucidate its mechanisms of action, optimize its bioavailability, and assess its long-term safety for widespread utilization.
Collapse
Affiliation(s)
- Fatemeh Aghababaei
- Centre d'Innovació, Recerca i Transferència en Tecnologia dels Aliments (CIRTTA), TECNIO-UAB, XIA, Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, UAB-Campus, 08193 Bellaterra, Spain
| | - Milad Hadidi
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| |
Collapse
|
8
|
Homayoonfal M, Gilasi H, Asemi Z, Mahabady MK, Asemi R, Yousefi B. Quercetin modulates signal transductions and targets non-coding RNAs against cancer development. Cell Signal 2023; 107:110667. [PMID: 37023996 DOI: 10.1016/j.cellsig.2023.110667] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/22/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
In recent decades, various investigations have indicated that natural compounds have great potential in the prevention and treatment of different chronic disorders including different types of cancer. As a bioactive flavonoid, Quercetin (Qu) is a dietary ingredient enjoying high pharmacological values and health-promoting effects due to its antioxidant and anti-inflammatory characterization. Conclusive in vitro and in vivo evidence has revealed that Qu has great potential in cancer prevention and development. Qu exerts its anticancer influences by altering various cellular processes such as apoptosis, autophagy, angiogenesis, metastasis, cell cycle, and proliferation. In this way, Qu by targeting numerous signaling pathways as well as non-coding RNAs regulates several cellular mechanisms to suppress cancer occurrence and promotion. This review aimed to summarize the impact of Qu on the molecular pathways and non-coding RNAs in modulating various cancer-associated cellular mechanisms.
Collapse
Affiliation(s)
- Mina Homayoonfal
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamidreza Gilasi
- Department of Biostatistics and Epidemiology, Kashan University of Medical Sciences, Kashan, Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Reza Asemi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Hu Y, Li R, Jin J, Wang Y, Ma R. Quercetin improves pancreatic cancer chemo-sensitivity by regulating oxidative-inflammatory networks. J Food Biochem 2022; 46:e14453. [PMID: 36181395 DOI: 10.1111/jfbc.14453] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 01/14/2023]
Abstract
Chemotherapy is the main method for controlling pancreatic cancer metastasis but the prevalent chemotherapy resistance limits its utilization. The response of oxidation and inflammation often promotes pancreatic cancer progression and chemo-resistance. It is critical to explore the potential natural products with few side effects to control inflammatory responses and understand the related mechanisms. Quercetin is a flavonoid widely found in numerous vegetables, fruits, and foods and is thought to have antioxidant and anti-inflammatory properties, which may be associated with improvement of chemotherapy sensitivity during pancreatic cancer treatment. Quercetin may sensitize pancreatic cancer cells to the chemotherapeutic agents, including bromodomain and extraterminal domain inhibitors (BETI), daunorubicin, gemcitabine, sulforaphane, doxorubicin, and tumor necrosis factor-related signaling apoptosis-inducing ligand (TRAIL). Meanwhile, during the chemo-resistance therapy, many signaling molecules are involved with toll-like receptor 4 (TLR4)-mediated oxidative and inflammatory pathway. The effects of quercetin on other oxidative and inflammatory pathways were also explored. Quercetin may exert antitumor activity during the prevention of pancreatic cancer progression by regulating oxidative and inflammatory networks, which can promote immune escape of cancer cells by inducing immunosuppressive cytokines. Studying these patterns will help us to better understand the functional role of quercetin in the improvement of pancreatic cancer chemo-sensitivity. PRACTICAL APPLICATIONS: Chemotherapy is the major way for treating pancreatic cancer metastasis but the prevalent chemotherapy resistance caused by oxidative and inflammatory responses limits its utilization. It is necessary to explore the potential natural products with few side effects to prevent the oxidative and inflammatory responses. Quercetin is a flavonoid widely found in numerous vegetables, fruits, and foods and is thought to have antioxidant and anti-inflammatory properties, which may be associated with improvement of chemotherapy sensitivity of pancreatic cancer treatment by sensitizing pancreatic cancer cells to various chemotherapeutic agents via the regulation of oxidative and inflammatory networks. Studying these patterns will help us to better understand the functional role of quercetin in the improvement of pancreatic cancer chemo-sensitivity.
Collapse
Affiliation(s)
- Yaoyuan Hu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Junyi Jin
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yihui Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Rui Ma
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Michala AS, Pritsa A. Quercetin: A Molecule of Great Biochemical and Clinical Value and Its Beneficial Effect on Diabetes and Cancer. Diseases 2022; 10:37. [PMID: 35892731 PMCID: PMC9326669 DOI: 10.3390/diseases10030037] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/01/2023] Open
Abstract
Quercetin belongs to the broader category of polyphenols. It is found, in particular, among the flavonols, and along with kaempferol, myricetin and isorhamnetin, it is recognized as a foreign substance after ingestion in contrast to vitamins. Quercetin occurs mainly linked to sugars with the most common compounds being quercetin-3-O-glucoside or as an aglycone, especially in the plant population. The aim of this review is to present a recent bibliography on the mechanisms of quercetin absorption and metabolism, bioavailability, and antioxidant and the clinical effects in diabetes and cancer. The literature reports a positive effect of quercetin on oxidative stress, cancer, and the regulation of blood sugar levels. Moreover, research-administered drug dosages of up to 2000 mg per day showed mild to no symptoms of overdose. It should be noted that quercetin is no longer considered a carcinogenic substance. The daily intake of quercetin in the diet ranges 10 mg-500 mg, depending on the type of products consumed. This review highlights that quercetin is a valuable dietary antioxidant, although a specific daily recommended intake for this substance has not yet been determined and further studies are required to decide a beneficial concentration threshold.
Collapse
Affiliation(s)
| | - Agathi Pritsa
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University (IHU), P.O. 141 Sindos, 57400 Thessaloniki, Greece;
| |
Collapse
|
11
|
Mirazimi SMA, Dashti F, Tobeiha M, Shahini A, Jafari R, Khoddami M, Sheida AH, EsnaAshari P, Aflatoonian AH, Elikaii F, Zakeri MS, Hamblin MR, Aghajani M, Bavarsadkarimi M, Mirzaei H. Application of Quercetin in the Treatment of Gastrointestinal Cancers. Front Pharmacol 2022; 13:860209. [PMID: 35462903 PMCID: PMC9019477 DOI: 10.3389/fphar.2022.860209] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/02/2022] [Indexed: 02/06/2023] Open
Abstract
Many cellular signaling pathways contribute to the regulation of cell proliferation, division, motility, and apoptosis. Deregulation of these pathways contributes to tumor cell initiation and tumor progression. Lately, significant attention has been focused on the use of natural products as a promising strategy in cancer treatment. Quercetin is a natural flavonol compound widely present in commonly consumed foods. Quercetin has shown significant inhibitory effects on tumor progression via various mechanisms of action. These include stimulating cell cycle arrest or/and apoptosis as well as its antioxidant properties. Herein, we summarize the therapeutic effects of quercetin in gastrointestinal cancers (pancreatic, gastric, colorectal, esophageal, hepatocellular, and oral).
Collapse
Affiliation(s)
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Tobeiha
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Shahini
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Raha Jafari
- Department of Medicine, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran
| | - Mehrad Khoddami
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Hossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Parastoo EsnaAshari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Hossein Aflatoonian
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fateme Elikaii
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Melika Sadat Zakeri
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Mohammad Aghajani
- Infectious Disease Research Center, School of Nursing and Midwifery, Kashan University of Medical Sciences, Kashan, Iran
| | - Minoodokht Bavarsadkarimi
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
12
|
An P, Zhang LJ, Peng W, Chen YY, Liu QP, Luan X, Zhang H. Natural products are an important source for proteasome regulating agents. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 93:153799. [PMID: 34715511 DOI: 10.1016/j.phymed.2021.153799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/14/2021] [Accepted: 10/07/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Natural medicines have a long history in the prevention and treatment of various diseases in East Asian region, especially in China. Modern research has proved that the pharmacological effects of numerous natural medicines involve the participation of ubiquitin proteasome system (UPS). UPS can degrade the unwanted and damaged proteins widely distributed in the nucleus and cytoplasm of various eukaryotes. PURPOSE The objective of the present study was to review and discuss the regulatory effects of natural products and extracts on proteasome components, which may help to find new proteasome regulators for drug development and clinical applications. METHODS The related information was compiled using the major scientific databases, such as CNKI, Elsevier, ScienceDirect, PubMed, SpringerLink, Wiley Online, and GeenMedical. The keywords "natural product" and "proteasome" were applied to extract the literature. Nature derived extracts, compounds and their derivatives involved in proteasome regulation were included, and the publications related to synthetic proteasome agents were excluded. RESULTS The pharmacological effects of more than 80 natural products and extracts derived from phytomedicines related to the proteasome regulation were reviewed. These natural products were classified according to their chemical properties. We also summarized some laws of action of natural products as proteasome regulators in the treatment of diseases, and listed the action characteristics of the typical natural products. CONCLUSION Natural products derived from nature can induce the degradation of damaged proteins through UPS or act as regulators to directly regulate the activity of proteasome. But few proteasome modulators are applied clinically. Summary of known rules for proteasome modulators will contribute to discover, modify and synthesize more proteasome modulators for clinical applications.
Collapse
Affiliation(s)
- Pei An
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China
| | - Li-Jun Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China
| | - Wei Peng
- School of pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu-Ying Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China
| | - Qiu-Ping Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China.
| | - Hong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China.
| |
Collapse
|
13
|
Asgharian P, Tazehkand AP, Soofiyani SR, Hosseini K, Martorell M, Tarhriz V, Ahangari H, Cruz-Martins N, Sharifi-Rad J, Almarhoon ZM, Ydyrys A, Nurzhanyat A, Yessenbekova A, Cho WC. Quercetin Impact in Pancreatic Cancer: An Overview on Its Therapeutic Effects. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4393266. [PMID: 34777687 PMCID: PMC8580629 DOI: 10.1155/2021/4393266] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/09/2021] [Accepted: 10/16/2021] [Indexed: 12/11/2022]
Abstract
Pancreatic cancer (PC) is a lethal malignancy cancer, and its mortality rates have been increasing worldwide. Diagnosis of this cancer is complicated, as it does not often present symptoms, and most patients present an irremediable tumor having a 5-year survival rate after diagnosis. Regarding treatment, many concerns have also been raised, as most tumors are found at advanced stages. At present, anticancer compounds-rich foods have been utilized to control PC. Among such bioactive molecules, flavonoid compounds have shown excellent anticancer abilities, such as quercetin, which has been used as an adjunctive or alternative drug to PC treatment by inhibitory or stimulatory biological mechanisms including autophagy, apoptosis, cell growth reduction or inhibition, EMT, oxidative stress, and enhancing sensitivity to chemotherapy agents. The recognition that this natural product has beneficial effects on cancer treatment has boosted the researchers' interest towards more extensive studies to use herbal medicine for anticancer purposes. In addition, due to the expensive cost and high rate of side effects of anticancer drugs, attempts have been made to use quercetin but also other flavonoids for preventing and treating PC. Based on related studies, it has been found that the quercetin compound has significant effect on cancerous cell lines as well as animal models. Therefore, it can be used as a supplementary drug to treat a variety of cancers, particularly pancreatic cancer. This review is aimed at discussing the therapeutic effects of quercetin by targeting the molecular signaling pathway and identifying antigrowth, cell proliferation, antioxidative stress, EMT, induction of apoptotic, and autophagic features.
Collapse
Affiliation(s)
- Parina Asgharian
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Pirpour Tazehkand
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saiedeh Razi Soofiyani
- Clinical Research Development Unit of Sina Educational, Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kamran Hosseini
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy and Centre for Healthy Living, University of Concepción, 4070386 Concepción, Chile
| | - Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Ahangari
- Department of Food Science and Technology, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Natália Cruz-Martins
- Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
- Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal
| | | | - Zainab M. Almarhoon
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Alibek Ydyrys
- Biomedical Research Centre, Al-Farabi Kazakh National University, Al-Farabi Av. 71, 050040 Almaty, Kazakhstan
| | - Ablaikhanova Nurzhanyat
- Department of Biophysics, Biomedicine and Neuroscience, Al-Farabi Kazakh National University, Al-Farabi Av. 71, 050040 Almaty, Kazakhstan
| | - Arailym Yessenbekova
- Department of Biophysics, Biomedicine and Neuroscience, Al-Farabi Kazakh National University, Al-Farabi Av. 71, 050040 Almaty, Kazakhstan
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China
| |
Collapse
|
14
|
Razeghian E, Suksatan W, Sulaiman Rahman H, Bokov DO, Abdelbasset WK, Hassanzadeh A, Marofi F, Yazdanifar M, Jarahian M. Harnessing TRAIL-Induced Apoptosis Pathway for Cancer Immunotherapy and Associated Challenges. Front Immunol 2021; 12:699746. [PMID: 34489946 PMCID: PMC8417882 DOI: 10.3389/fimmu.2021.699746] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/05/2021] [Indexed: 01/04/2023] Open
Abstract
The immune cytokine tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted rapidly evolving attention as a cancer treatment modality because of its competence to selectively eliminate tumor cells without instigating toxicity in vivo. TRAIL has revealed encouraging promise in preclinical reports in animal models as a cancer treatment option; however, the foremost constraint of the TRAIL therapy is the advancement of TRAIL resistance through a myriad of mechanisms in tumor cells. Investigations have documented that improvement of the expression of anti-apoptotic proteins and survival or proliferation involved signaling pathways concurrently suppressing the expression of pro-apoptotic proteins along with down-regulation of expression of TRAILR1 and TRAILR2, also known as death receptor 4 and 5 (DR4/5) are reliable for tumor cells resistance to TRAIL. Therefore, it seems that the development of a therapeutic approach for overcoming TRAIL resistance is of paramount importance. Studies currently have shown that combined treatment with anti-tumor agents, ranging from synthetic agents to natural products, and TRAIL could result in induction of apoptosis in TRAIL-resistant cells. Also, human mesenchymal stem/stromal cells (MSCs) engineered to generate and deliver TRAIL can provide both targeted and continued delivery of this apoptosis-inducing cytokine. Similarly, nanoparticle (NPs)-based TRAIL delivery offers novel platforms to defeat barricades to TRAIL therapeutic delivery. In the current review, we will focus on underlying mechanisms contributed to inducing resistance to TRAIL in tumor cells, and also discuss recent findings concerning the therapeutic efficacy of combined treatment of TRAIL with other antitumor compounds, and also TRAIL-delivery using human MSCs and NPs to overcome tumor cells resistance to TRAIL.
Collapse
Affiliation(s)
- Ehsan Razeghian
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Suleimanyah, Suleimanyah, Iraq
- Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaimaniyah, Iraq
| | - Dmitry O. Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russia
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahboubeh Yazdanifar
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
15
|
Hosseinzadeh E, Hassanzadeh A, Marofi F, Alivand MR, Solali S. Flavonoid-Based Cancer Therapy: An Updated Review. Anticancer Agents Med Chem 2021; 20:1398-1414. [PMID: 32324520 DOI: 10.2174/1871520620666200423071759] [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: 04/09/2019] [Revised: 10/27/2019] [Accepted: 11/06/2019] [Indexed: 12/24/2022]
Abstract
As cancers are one of the most important causes of human morbidity and mortality worldwide, researchers try to discover novel compounds and therapeutic approaches to decrease survival of cancer cells, angiogenesis, proliferation and metastasis. In the last decade, use of special phytochemical compounds and flavonoids was reported to be an interesting and hopeful tactic in the field of cancer therapy. Flavonoids are natural polyphenols found in plant, fruits, vegetables, teas and medicinal herbs. Based on reports, over 10,000 flavonoids have been detected and categorized into several subclasses, including flavonols, anthocyanins, flavanones, flavones, isoflavones and chalcones. It seems that the anticancer effect of flavonoids is mainly due to their antioxidant and anti inflammatory activities and their potential to modulate molecular targets and signaling pathways involved in cell survival, proliferation, differentiation, migration, angiogenesis and hormone activities. The main aim of this review is to evaluate the relationship between flavonoids consumption and cancer risk, and discuss the anti-cancer effects of these natural compounds in human cancer cells. Hence, we tried to collect and revise important recent in vivo and in vitro researches about the most effective flavonoids and their main mechanisms of action in various types of cancer cells.
Collapse
Affiliation(s)
- Elham Hosseinzadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Hassanzadeh
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Novel therapeutic strategies and perspectives for pancreatic cancer: Autophagy and apoptosis are key mechanisms to fight pancreatic cancer. Med Oncol 2021; 38:74. [PMID: 34019188 DOI: 10.1007/s12032-021-01522-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/10/2021] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer (PC) is the most lethal malignancy of the gastrointestinal tract. The poor prognosis of patients with PC is primarily due to lack of effective treatments against its progressive and metastatic behavior. Hence, figuring out the mechanisms underlying PC development and putting up with effective targeted therapies are of great significance to improve the prognosis of patients with PC. Apoptosis and autophagy serve to maintain tissue homoeostasis. Escaping from apoptosis or autophagy is one of the features of malignancy. PC is seriously resistant to autophagy and apoptosis, which explains its invasiveness and resistance to conventional treatment. Recently, several biological activities and pharmacological functions found in natural product extracts have been reported to inhibit PC progression. The current review focuses on understanding natural product extracts and their derivatives as one kind of novel treatments through affecting the apoptosis or autophagy in PC.
Collapse
|
17
|
Guo Y, Tong Y, Zhu H, Xiao Y, Guo H, Shang L, Zheng W, Ma S, Liu X, Bai Y. Quercetin suppresses pancreatic ductal adenocarcinoma progression via inhibition of SHH and TGF-β/Smad signaling pathways. Cell Biol Toxicol 2020; 37:479-496. [PMID: 33070227 DOI: 10.1007/s10565-020-09562-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is an aggressive type of malignant tumor with a poor prognosis and high mortality. Aberrant activation of hedgehog signaling plays a crucial role in the maintenance and progression of PDA. Here, we report that the dietary bioflavonoid quercetin has therapeutic potential for PDA by targeting sonic hedgehog (SHH) signaling. The effects of quercetin on the proliferation, apoptosis, migration, and invasion of pancreatic cancer cells (PCCs) and tumor growth and metastasis in PDA xenograft mouse models were evaluated. Additionally, SHH signaling activity was determined. Quercetin significantly inhibited PCC proliferation by downregulating c-Myc expression. In addition, quercetin suppressed epithelial-mesenchymal transition (EMT) by reducing TGF-β1 level, which resulted in inhibition of PCC migration and invasion. Moreover, quercetin induced PCC apoptosis through mitochondrial and death receptor pathways. In nude mouse models, PDA growth and metastasis were reduced by quercetin treatment. Mechanically, quercetin exerts its therapeutic effects on PDA by decreasing SHH activity. Interestingly, quercetin-induced SHH inactivation is mainly dependent on Gli2, but not Gli1. Enhance SHH activity by recombinant Shh protein abolished the quercetin-mediated inhibition of PCC proliferation, migration, and invasion. Furthermore, Shh activated TGF-β1/Smad2/3 signaling and promoted EMT by inducing the expression of Zeb2 and Snail1 that eventually resulted in a partial reversal of quercetin-mediated inhibition of PCC migration and invasion. We conclude that quercetin inhibited the growth, migration, and invasion and induced apoptosis of PCCs by antagonizing SHH and TGF-β/Smad signaling pathways. Thus, quercetin may be a potential candidate for PDA treatment.
Collapse
Affiliation(s)
- Yangyang Guo
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yu Tong
- Department of Laboratory Medicine, People's Hospital of Wenzhou City, Wenzhou, 325000, China
| | - Hengyue Zhu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.,Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yanyi Xiao
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Hangcheng Guo
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lumeng Shang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.,Department of Laboratory Medicine, People's Hospital of Wenzhou City, Wenzhou, 325000, China
| | - Wenjing Zheng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.,Department of Laboratory Medicine, People's Hospital of Wenzhou City, Wenzhou, 325000, China
| | - Shumei Ma
- Platform for Radiation Protection and Emergency Preparedness, School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325000, China.,Center for Health Assessment, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaodong Liu
- Platform for Radiation Protection and Emergency Preparedness, School of Public Health and Management, Wenzhou Medical University, Wenzhou, 325000, China. .,Center for Health Assessment, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China. .,Center for Health Assessment, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
18
|
Lai M, Ge Y, Chen M, Sun S, Chen J, Cheng R. Saikosaponin D Inhibits Proliferation and Promotes Apoptosis Through Activation of MKK4-JNK Signaling Pathway in Pancreatic Cancer Cells. Onco Targets Ther 2020; 13:9465-9479. [PMID: 33061432 PMCID: PMC7522527 DOI: 10.2147/ott.s263322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction Pancreatic cancer remains one of the most lethal malignancies and has few treatment options. Saikosaponin D (SSD), a major bioactive triterpene saponin isolated from Bupleurum chinense, has been reported to exert cytotoxicity properties toward many cancer cells. However, the effects of SSD on pancreatic cancer have been little scrutinized. Methods Here, we investigated the effect of SSD on the proliferation and apoptosis of human pancreatic cancer BxPC3 and PANC1 cells and the mouse pancreatic cancer cell line Pan02. Cell viability was determined by MTT assays and cell apoptosis analyzed by DAPI staining and flow cytometry. Expression levels of apoptosis-regulating markers and activity of the MKK4–JNK signaling pathway were determined by Western blotting. The inhibitor SP600125 was applied to confirm the role of the JNK pathway in SSD efficiency. Results SSD significantly inhibited the proliferation of BxPC3, PANC1, and Pan02 cells in a concentration- and time-dependent manner. Flow-cytometry analysis indicated obvious apoptosis induction after SSD exposure. Furthermore, SSD significantly triggered cleavage of caspase 3 and caspase 9 proteins and increased the expression of FoxO3a. In addition, activity of the MKK4–JNK pathway was dramatically increased after treatment with SSD in BxPC3 cells. SSD obviously stimulated phosphorylation of JNK, cJun, and SEK1/MKK4 proteins within 30 minutes. The addition of SP600125 blocked the activation of SSD on the MKK4–JNK regulatory pathway and reversed the effects of SSD on proliferation inhibition and apoptosis induction in BxPC3 cells. Conclusion These results revealed that SSD was capable of suppressing tumor growth and promoting apoptosis of pancreatic cancer cells via targeting the MKK4–JNK signaling pathway, indicating the possibility of further developing SSD as a potential therapeutic candidate for pancreatic cancer.
Collapse
Affiliation(s)
- Mengru Lai
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Yuqing Ge
- First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310006, People's Republic of China
| | - Meng Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Siya Sun
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Jianzhen Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Rubin Cheng
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| |
Collapse
|
19
|
Gao Y, Chen S, Sun J, Su S, Yang D, Xiang L, Meng X. Traditional Chinese medicine may be further explored as candidate drugs for pancreatic cancer: A review. Phytother Res 2020; 35:603-628. [PMID: 32965773 DOI: 10.1002/ptr.6847] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer is a disease with a high mortality rate. Although survival rates for different types of cancers have improved in recent years, the five-year survival rate of pancreatic cancer stands at 8%. Moreover, the current first-line therapy, gemcitabine, results in low remission rates and is associated with drug resistance problems. Alternative treatments for pancreatic cancer such as surgery, chemotherapy and radiation therapy provide marginal remission and survival rates. This calls for the search of more effective drugs or treatments. Traditional Chinese medicine contains numerous bioactive ingredients some of which show activity against pancreatic cancer. In this review, we summarize the mechanisms of five types of traditional Chinese medicine monomers. In so-doing, we provide new potential drug candidates for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yue Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyu Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayi Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyu Su
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Xiang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
20
|
Thyagarajan A, Forino AS, Konger RL, Sahu RP. Dietary Polyphenols in Cancer Chemoprevention: Implications in Pancreatic Cancer. Antioxidants (Basel) 2020; 9:651. [PMID: 32717779 PMCID: PMC7464582 DOI: 10.3390/antiox9080651] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/26/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023] Open
Abstract
Naturally occurring dietary agents present in a wide variety of plant products, are rich sources of phytochemicals possessing medicinal properties, and thus, have been used in folk medicine for ages to treat various ailments. The beneficial effects of such dietary components are frequently attributed to their anti-inflammatory and antioxidant properties, particularly in regards to their antineoplastic activities. As many tumor types exhibit greater oxidative stress levels that are implicated in favoring autonomous cell growth activation, most chemotherapeutic agents can also enhance tumoral oxidative stress levels in part via generating reactive oxygen species (ROS). While ROS-mediated imbalance of the cellular redox potential can provide novel drug targets, as a consequence, this ROS-mediated excessive damage to cellular functions, including oncogenic mutagenesis, has also been implicated in inducing chemoresistance. This remains one of the major challenges in the treatment and management of human malignancies. Antioxidant-enriched natural compounds offer one of the promising approaches in mitigating some of the underlying mechanisms involved in tumorigenesis and metastasis, and therefore, have been extensively explored in cancer chemoprevention. Among various groups of dietary phytochemicals, polyphenols have been extensively explored for their underlying chemopreventive mechanisms in other cancer models. Thus, the current review highlights the significance and mechanisms of some of the highly studied polyphenolic compounds, with greater emphasis on pancreatic cancer chemoprevention.
Collapse
Affiliation(s)
- Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of medicine Wright State University, Dayton, OH 45435, USA
| | - Andrew S. Forino
- Department of Anatomy and Physiology, Boonshoft School of medicine Wright State University, Dayton, OH 45435, USA;
| | - Raymond L. Konger
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Ravi P. Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of medicine Wright State University, Dayton, OH 45435, USA
| |
Collapse
|
21
|
Vafadar A, Shabaninejad Z, Movahedpour A, Fallahi F, Taghavipour M, Ghasemi Y, Akbari M, Shafiee A, Hajighadimi S, Moradizarmehri S, Razi E, Savardashtaki A, Mirzaei H. Quercetin and cancer: new insights into its therapeutic effects on ovarian cancer cells. Cell Biosci 2020; 10:32. [PMID: 32175075 PMCID: PMC7063794 DOI: 10.1186/s13578-020-00397-0] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 02/29/2020] [Indexed: 12/25/2022] Open
Abstract
Ovarian cancer is known as a serious malignancy that affects women's reproductive tract and can considerably threat their health. A wide range of molecular mechanisms and genetic modifications have been involved in ovarian cancer pathogenesis making it difficult to develop effective therapeutic platforms. Hence, discovery and developing new therapeutic approaches are required. Medicinal plants, as a new source of drugs, could potentially be used alone or in combination with other medicines in the treatment of various cancers such as ovarian cancer. Among various natural compounds, quercetin has shown great anti-cancer and anti-inflammatory properties. In vitro and in vivo experiments have revealed that quercetin possesses a cytotoxic impact on ovarian cancer cells. Despite obtaining good results both in vitro and in vivo, few clinical studies have assessed the anti-cancer effects of quercetin particularly in the ovarian cancer. Therefore, it seems that further clinical studies may introduce quercetin as therapeutic agent alone or in combination with other chemotherapy drugs to the clinical setting. Here, we not only summarize the anti-cancer effects of quercetin but also highlight the therapeutic effects of quercetin in the ovarian cancer.
Collapse
Affiliation(s)
- Asma Vafadar
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Shabaninejad
- Department of Nanotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Fallahi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. of Iran
| | - Mona Taghavipour
- Department of Gynecology and Obstetrics, Ramsar Campus, Mazandaran University of Medical Sciences, Sari, Iran
| | - Younes Ghasemi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Akbari
- Department of Surgery, Kashan University of Medical Sciences, Kashan, Iran
| | - Alimohammad Shafiee
- Division of General Internal Medicine, Toronto General Hospital, Toronto, ON Canada
| | - Sarah Hajighadimi
- Division of General Internal Medicine, Toronto General Hospital, Toronto, ON Canada
| | - Sanaz Moradizarmehri
- Division of General Internal Medicine, Toronto General Hospital, Toronto, ON Canada
| | - Ebrahim Razi
- The Advocate Center for Clinical Research, Ayatollah Yasrebi Hospital, Kashan, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. of Iran
| |
Collapse
|
22
|
Pang B, Xu X, Lu Y, Jin H, Yang R, Jiang C, Shao D, Liu Y, Shi J. Prediction of new targets and mechanisms for quercetin in the treatment of pancreatic cancer, colon cancer, and rectal cancer. Food Funct 2019; 10:5339-5349. [PMID: 31393490 DOI: 10.1039/c9fo01168d] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Quercetin has been widely found to exhibit anticancer activity with low toxicity and prevalence in foods. Quercetin has been reported to inhibit digestive system cancers including pancreatic cancer (PAAD) and colon cancer (COAD), but rectal cancer (READ) has not been reported. The reported mechanisms and targets are divergent. In this study, new targets and mechanisms were predicted for the influence of quercetin on PAAD, COAD, and READ using bioinformatics methods. The results showed that quercetin may target CD36 and reduce the death rate caused by PAAD by enhancing the cell adhesion, mediating the uptake of fatty acids (FAs), regulating thrombospondin-1, and stimulating the immune response. Quercetin may lower the death rate from READ by targeting SLCO1B1 and producing enhanced effects from use of this compound, inhibiting cell growth, and inducing apoptosis in tumor cells. ACADS, ALDH3B2, UGT2A3, AMH, CDKN2A, FOSL1, CD36, CFL2, CYP3A4, and MAF were identified as targets for quercetin to reduce the death rate caused by COAD. Glutathione metabolism was mainly involved in the effect of quercetin on COAD, including the enhancement of the oxidation of fatty acids, the metabolism of anticancer medications, and the stiffness of cells, and the reduction of chemical carcinogenesis, the level of anti-Müllerian hormone, the proliferation of cancer cells and transcriptional misregulation, and mediation of the activity of glutathione transferases. The combined analyses of three databases can be referred to and used to seek medications and targets that can be applied to other diseases.
Collapse
Affiliation(s)
- Bing Pang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Xiaoguang Xu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Yao Lu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Han Jin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Rongrong Yang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Chunmei Jiang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Dongyan Shao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| | - Yanlin Liu
- College of Enology, Northwest A&F University, 28 Xinong Road, Yangling, Shaanxi Province 712100, China
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi Province 710072, China.
| |
Collapse
|
23
|
Sustained DDB-2 and TRX transcriptional response of quercetin-treated lymphocytes exposed to Co-60 radiation. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2019.100416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
24
|
Ye Q, Liu K, Shen Q, Li Q, Hao J, Han F, Jiang RW. Reversal of Multidrug Resistance in Cancer by Multi-Functional Flavonoids. Front Oncol 2019; 9:487. [PMID: 31245292 PMCID: PMC6581719 DOI: 10.3389/fonc.2019.00487] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/23/2019] [Indexed: 12/22/2022] Open
Abstract
Multidrug resistance (MDR) resulting from different defensive mechanisms in cancer is one of the major obstacles of clinical treatment. To circumvent MDR many reversal agents have been developed, but most of them fail in clinical trials due to severely adverse effects. Recently, certain natural products have been reported to overcome MDR, including flavonoids which are abundant in plants, foods, and herbs. The structure of flavonoids can be abbreviated as C6-C3-C6 (C for carbon), and further categorized into flavonoids, iso-flavonoids and neo-flavonoids, according to their structural backbones. Flavonoids possess multiple bioactivities, and a growing body of research has indicated that both flavonoids and iso-flavonoids can either kill or re-sensitize conventional chemotherapeutics to resistant cancer cells. Here, we summarize the research and discuss the underlying mechanisms, concluding that these flavonoids do not function as specific regulators of target proteins, but rather as multi-functional agents that negatively regulate the key factors contributing to MDR.
Collapse
Affiliation(s)
| | - Kai Liu
- Hainan General Hospital, Haikou, China
| | - Qun Shen
- Hainan General Hospital, Haikou, China
| | | | - Jinghui Hao
- Jiaozuo Second People's Hospital, Jiaozuo, China
| | | | - Ren-Wang Jiang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, Jinan University, Guangzhou, China
| |
Collapse
|
25
|
Nani A, Belarbi M, Murtaza B, Benammar C, Merghoub T, Rialland M, Akhtar Khan N, Hichami A. Polyphenols from Pennisetum glaucum grains induce MAP kinase phosphorylation and cell cycle arrest in human osteosarcoma cells. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.01.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
26
|
Kerimi A, Williamson G. Differential Impact of Flavonoids on Redox Modulation, Bioenergetics, and Cell Signaling in Normal and Tumor Cells: A Comprehensive Review. Antioxid Redox Signal 2018; 29:1633-1659. [PMID: 28826224 PMCID: PMC6207159 DOI: 10.1089/ars.2017.7086] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
SIGNIFICANCE Flavonoids can interact with multiple molecular targets to elicit their cellular effects, leading to changes in signal transduction, gene expression, and/or metabolism, which can, subsequently, affect the entire cell and organism. Immortalized cell lines, derived from tumors, are routinely employed as a surrogate for mechanistic studies, with the results extrapolated to tissues in vivo. Recent Advances: We review the activities of selected flavonoids on cultured tumor cells derived from various tissues in comparison to corresponding primary cells or tissues in vivo, mainly using quercetin and flavanols (epicatechin and (-)-epigallocatechin gallate) as exemplars. Several studies have indicated that flavonoids could retard cancer progression in vivo in animal models as well as in tumor cell models. CRITICAL ISSUES Extrapolation from in vitro and animal models to humans is not straightforward given both the extensive conjugation and complex microbiota-dependent metabolism of flavonoids after consumption, as well as the heterogeneous metabolism of different tumors. FUTURE DIRECTIONS Comparison of data from studies on primary cells or in vivo are essential not only to validate results obtained from cultured cell models, but also to highlight whether any differences may be further exploited in the clinical setting for chemoprevention. Tumor cell models can provide a useful mechanistic tool to study the effects of flavonoids, provided that the limitations of each model are understood and taken into account in interpretation of the data.
Collapse
Affiliation(s)
- Asimina Kerimi
- School of Food Science and Nutrition, University of Leeds , Leeds, United Kingdom
| | - Gary Williamson
- School of Food Science and Nutrition, University of Leeds , Leeds, United Kingdom
| |
Collapse
|
27
|
Lohse I, Wildermuth E, Brothers SP. Naturally occurring compounds as pancreatic cancer therapeutics. Oncotarget 2018; 9:35448-35457. [PMID: 30459936 PMCID: PMC6226042 DOI: 10.18632/oncotarget.26234] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 10/06/2018] [Indexed: 02/06/2023] Open
Abstract
Naturally occurring small molecule compounds have long been in the spotlight of pancreatic cancer research as potential therapeutics to prevent cancer progression and sensitize chemoresistant tumors. The hope is that terminal pancreatic cancer patients receiving aggressive chemotherapy can benefit from an increase in treatment efficacy without adding further toxicity by way of utilizing natural compounds. While preclinical studies on a number of natural compounds, such as resveratrol, curcumin, rapalogs and cannabinoids, show promising preclinical results, little has translated into clinical practice, though a number of other compounds hold clinical potential. Nevertheless, recent advances in compound formulation may increase the clinical utility of these compounds.
Collapse
Affiliation(s)
- Ines Lohse
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation University of Miami Miller School of Medicine, University of Miami, Miami, FL, USA.,Molecular Therapeutics Shared Resource, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Erin Wildermuth
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation University of Miami Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Shaun P Brothers
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation University of Miami Miller School of Medicine, University of Miami, Miami, FL, USA.,Molecular Therapeutics Shared Resource, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
28
|
Turner KA, Manouchehri JM, Kalafatis M. Sensitization of recombinant human tumor necrosis factor-related apoptosis-inducing ligand-resistant malignant melanomas by quercetin. Melanoma Res 2018; 28:277-285. [PMID: 29596115 PMCID: PMC6039425 DOI: 10.1097/cmr.0000000000000447] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 02/20/2018] [Indexed: 12/30/2022]
Abstract
Malignant melanoma is the most commonly diagnosed skin cancer associated with a high rate of metastasis. Low-stage melanoma is easily treated, but metastatic malignant melanoma is an extremely treatment-resistant malignancy with low survival rates. The application of recombinant human tumor necrosis factor-related apoptosis-inducing ligand (rhTRAIL) for the treatment of metastatic malignant melanoma holds considerable promise because of its selective proapoptotic activity towards cancer cells and not nontransformed cells. Unfortunately, the clinical utilization of rhTRAIL has been terminated due to the resistance of many cancer cells to undergo apoptosis in response to rhTRAIL. However, rhTRAIL-resistance can be abrogated through the cotreatment with compounds derived from 'Mother Nature' such as quercetin that can modulate cellular components responsible for rhTRAIL-resistance. Here, we show that rhTRAIL-resistant malignant melanomas are sensitized by quercetin. Quercetin action is manifested by the upregulation of rhTRAIL-binding receptors DR4 and DR5 on the surface of cancer cells and by increased rate of the proteasome-mediated degradation of the antiapoptotic protein FLIP. Our data provide for a new efficient and nontoxic treatment of malignant melanoma.
Collapse
Affiliation(s)
- Katherine A. Turner
- Department of Chemistry, Cleveland State University
- Center for Gene Regulation in Health and Disease (GRHD)
| | - Jasmine M. Manouchehri
- Department of Chemistry, Cleveland State University
- Center for Gene Regulation in Health and Disease (GRHD)
| | - Michael Kalafatis
- Department of Chemistry, Cleveland State University
- Center for Gene Regulation in Health and Disease (GRHD)
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|