1
|
Pouliquen DL, Trošelj KG, Anto RJ. Curcuminoids as Anticancer Drugs: Pleiotropic Effects, Potential for Metabolic Reprogramming and Prospects for the Future. Pharmaceutics 2023; 15:1612. [PMID: 37376060 DOI: 10.3390/pharmaceutics15061612] [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: 04/28/2023] [Revised: 05/21/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The number of published studies on curcuminoids in cancer research, including its lead molecule curcumin and synthetic analogs, has been increasing substantially during the past two decades. Insights on the diversity of inhibitory effects they have produced on a multitude of pathways involved in carcinogenesis and tumor progression have been provided. As this wealth of data was obtained in settings of various experimental and clinical data, this review first aimed at presenting a chronology of discoveries and an update on their complex in vivo effects. Secondly, there are many interesting questions linked to their pleiotropic effects. One of them, a growing research topic, relates to their ability to modulate metabolic reprogramming. This review will also cover the use of curcuminoids as chemosensitizing molecules that can be combined with several anticancer drugs to reverse the phenomenon of multidrug resistance. Finally, current investigations in these three complementary research fields raise several important questions that will be put among the prospects for the future research related to the importance of these molecules in cancer research.
Collapse
Affiliation(s)
- Daniel L Pouliquen
- Université d'Angers, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Koraljka Gall Trošelj
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Ruby John Anto
- Molecular Bioassay Laboratory, Institute of Advanced Virology, Thiruvananthapuram 695317, India
| |
Collapse
|
2
|
Nguyen T, Maniyar A, Sarkar M, Sarkar TR, Neelgund GM. The Cytotoxicity of Carbon Nanotubes and Hydroxyapatite, and Graphene and Hydroxyapatite Nanocomposites against Breast Cancer Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:556. [PMID: 36770518 PMCID: PMC9919526 DOI: 10.3390/nano13030556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
Cancer is a current dreadful disease and the leading cause of death. Next to cardiovascular diseases, cancer is the most severe threat to human life and health. Breast cancer is the most common invasive cancer diagnosed in women. Each year about 2.3 million women are diagnosed with breast cancer. In consideration of the severity of breast cancer, herein we designed the biocompatible nanomaterials, CNTs-HAP and GR-HAP, through grafting of hydroxyapatite (HAP) to carbon nanotubes (CNTs) and graphene (GR) nanosheets. CNTs-HAP and GR-HAP have been tested for their cytotoxicity, growth and motility inhibitory effects, and their effects on the mesenchymal markers. All these demonstrated significant dose-dependent and time-dependent in vitro cytotoxicity against SUM-159 and MCF-7 breast cancer cell lines. The cell viability assay showed that the CNTs-HAP was more effective over SUM-159 cells than MCF-7 cells. It found that the increase in the concentration of GR-HAP has inhibited the clonogenic ability of breast cancer cells. The GR-HAP exhibited a substantial inhibitory effect on the cell motility of SUM-159 cell lines. It was investigated that the expression of vimentin (mesenchymal marker) was majorly reduced in SUM-159 cells by GR-HAP.
Collapse
Affiliation(s)
- Tristan Nguyen
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Anuj Maniyar
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | - Mrinmoy Sarkar
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| | | | - Gururaj M. Neelgund
- Department of Chemistry, Prairie View A&M University, Prairie View, TX 77446, USA
| |
Collapse
|
3
|
Chiu CC, Chen YC, Bow YD, Chen JYF, Liu W, Huang JL, Shu ED, Teng YN, Wu CY, Chang WT. diTFPP, a Phenoxyphenol, Sensitizes Hepatocellular Carcinoma Cells to C2-Ceramide-Induced Autophagic Stress by Increasing Oxidative Stress and ER Stress Accompanied by LAMP2 Hypoglycosylation. Cancers (Basel) 2022; 14:cancers14102528. [PMID: 35626132 PMCID: PMC9139631 DOI: 10.3390/cancers14102528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/14/2022] [Accepted: 05/16/2022] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Chemotherapy is the major treatment modality for advanced or unresectable hepatocellular carcinoma (HCC). Unfortunately, chemoresistance carries a poor prognosis in HCC patients. Exogenous ceramide, a sphingolipid, has been well documented to exert anticancer effects; however, recent reports showed ceramide resistance, which limits the development of the ceramide-based cancer treatment diTFPP, a novel phenoxyphenol compound that has been shown to sensitize HCC cells to ceramide treatment. Here, we further clarified the mechanism underlying diTFPP-mediated sensitization of HCC to C2-ceramide-induced stresses, including oxidative stress, ER stress, and autophagic stress, especially the modulation of LAMP2 glycosylation, the lysosomal membrane protein that is crucial for autophagic fusion. This study may shed light on the mechanism of ceramide resistance and help in the development of adjuvants for ceramide-based cancer therapeutics. Abstract Hepatocellular carcinoma (HCC), the most common type of liver cancer, is the leading cause of cancer-related mortality worldwide. Chemotherapy is the major treatment modality for advanced or unresectable HCC; unfortunately, chemoresistance results in a poor prognosis for HCC patients. Exogenous ceramide, a sphingolipid, has been well documented to exert anticancer effects. However, recent reports suggest that sphingolipid metabolism in ceramide-resistant cancer cells favors the conversion of exogenous ceramides to prosurvival sphingolipids, conferring ceramide resistance to cancer cells. However, the mechanism underlying ceramide resistance remains unclear. We previously demonstrated that diTFPP, a novel phenoxyphenol compound, enhances the anti-HCC effect of C2-ceramide. Here, we further clarified that treatment with C2-ceramide alone increases the protein level of CERS2, which modulates sphingolipid metabolism to favor the conversion of C2-ceramide to prosurvival sphingolipids in HCC cells, thus activating the unfolded protein response (UPR), which further initiates autophagy and the reversible senescence-like phenotype (SLP), ultimately contributing to C2-ceramide resistance in these cells. However, cotreatment with diTFPP and ceramide downregulated the protein level of CERS2 and increased oxidative and endoplasmic reticulum (ER) stress. Furthermore, insufficient LAMP2 glycosylation induced by diTFPP/ceramide cotreatment may cause the failure of autophagosome–lysosome fusion, eventually lowering the threshold for triggering cell death in response to C2-ceramide. Our study may shed light on the mechanism of ceramide resistance and help in the development of adjuvants for ceramide-based cancer therapeutics.
Collapse
Affiliation(s)
- Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- The Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yen-Chun Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
| | - Yung-Ding Bow
- Ph.D. Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Jeff Yi-Fu Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
| | - Jau-Ling Huang
- Department of Bioscience Technology, College of Health Science, Chang Jung Christian University, Tainan 711, Taiwan;
| | - En-De Shu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan;
| | - Chang-Yi Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Wen-Tsan Chang
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7-312-1101 (ext. 7651); Fax: +886-7-312-6992
| |
Collapse
|
4
|
Sharmin S, Rahaman MM, Martorell M, Sastre-Serra J, Sharifi-Rad J, Butnariu M, Bagiu IC, Bagiu RV, Islam MT. Cytotoxicity of synthetic derivatives against breast cancer and multi-drug resistant breast cancer cell lines: a literature-based perspective study. Cancer Cell Int 2021; 21:612. [PMID: 34801046 PMCID: PMC8606078 DOI: 10.1186/s12935-021-02309-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/31/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is the second most killer worldwide causing millions of people to lose their lives every year. In the case of women, breast cancer takes away the highest proportion of mortality rate than other cancers. Due to the mutation and resistance-building capacity of different breast cancer cell lines against conventional therapies, this death rate is on the verge of growth. New effective therapeutic compounds and treatment method is the best way to look out for in this critical time. For instance, new synthetic derivatives/ analogues synthesized from different compounds can be a ray of hope. Numerous synthetic compounds have been seen enhancing the apoptosis and autophagic pathway that directly exerts cytotoxicity towards different breast cancer cell lines. To cease the ever-growing resistance of multi-drug resistant cells against anti-breast cancer drugs (Doxorubicin, verapamil, tamoxifen) synthetic compounds may play a vital role by increasing effectivity, showing synergistic action. Many recent and previous studies have reported that synthetic derivatives hold potentials as an effective anti-breast cancer agent as they show great cytotoxicity towards cancer cells, thus can be used even vastly in the future in the field of breast cancer treatment. This review aims to identify the anti-breast cancer properties of several synthetic derivatives against different breast cancer and multi-drug-resistant breast cancer cell lines with their reported mechanism of action and effectivity.
Collapse
Affiliation(s)
- Shabnam Sharmin
- Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj (Dhaka), 8100, Bangladesh
| | - Md Mizanur Rahaman
- Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj (Dhaka), 8100, Bangladesh
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepción, 4070386, Concepción, Chile
| | - Jorge Sastre-Serra
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d'Investigació en Ciències de La Salut (IUNICS), Universitat de Les Illes Balears, Palma de Mallorca, Illes Balears, Spain.,Instituto de Investigación Sanitaria de Las Islas Baleares (IdISBa), Hospital Universitario Son Espases, Edificio S, 07120, Palma de Mallorca, Illes Balears, Spain.,Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029, Madrid, Spain
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Monica Butnariu
- Banat's University of Agricultural Sciences and Veterinary Medicine "King Michael I of Romania" From Timisoara, Timisoara, Romania.
| | - Iulia Cristina Bagiu
- Department of Microbiology, Victor Babes University of Medicine and Pharmacy of Timisoara, Timisoara, Romania.,Multidisciplinary Research Center On Antimicrobial Resistance, Timisoara, Romania
| | - Radu Vasile Bagiu
- Department of Microbiology, Victor Babes University of Medicine and Pharmacy of Timisoara, Timisoara, Romania.,Preventive Medicine Study Center, Timisoara, Romania
| | - Mohammad Torequl Islam
- Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj (Dhaka), 8100, Bangladesh
| |
Collapse
|
5
|
Chavda VP, Ertas YN, Walhekar V, Modh D, Doshi A, Shah N, Anand K, Chhabria M. Advanced Computational Methodologies Used in the Discovery of New Natural Anticancer Compounds. Front Pharmacol 2021; 12:702611. [PMID: 34483905 PMCID: PMC8416109 DOI: 10.3389/fphar.2021.702611] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Natural chemical compounds have been widely investigated for their programmed necrosis causing characteristics. One of the conventional methods for screening such compounds is the use of concentrated plant extracts without isolation of active moieties for understanding pharmacological activity. For the last two decades, modern medicine has relied mainly on the isolation and purification of one or two complicated active and isomeric compounds. The idea of multi-target drugs has advanced rapidly and impressively from an innovative model when first proposed in the early 2000s to one of the popular trends for drug development in 2021. Alternatively, fragment-based drug discovery is also explored in identifying target-based drug discovery for potent natural anticancer agents which is based on well-defined fragments opposite to use of naturally occurring mixtures. This review summarizes the current key advancements in natural anticancer compounds; computer-assisted/fragment-based structural elucidation and a multi-target approach for the exploration of natural compounds.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad, India
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey.,ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Vinayak Walhekar
- Department of Medicinal Chemistry, Bharati Vidyapeeth's Poona College of Pharmacy, Pune, India
| | - Dharti Modh
- Department of Medicinal Chemistry, Bharati Vidyapeeth's Poona College of Pharmacy, Pune, India
| | - Avani Doshi
- Department of Chemistry, SAL Institute of Pharmacy, Ahmedabad, India
| | - Nirav Shah
- Department of Pharmaceutics, SAL Institute of Pharmacy, Ahmedabad, India
| | - Krishna Anand
- Faculty of Health Sciences and National Health Laboratory Service, Department of Chemical Pathology, School of Pathology, University of the Free State, Bloemfontein, South Africa
| | - Mahesh Chhabria
- Department of Pharmaceutical Chemistry, L.M. College of Pharmacy, Ahmedabad, India
| |
Collapse
|
6
|
In vivo estrogenicity of p-phenoxyphenol and p-pentyloxyphenol. Sci Rep 2020; 10:17305. [PMID: 33057140 PMCID: PMC7560878 DOI: 10.1038/s41598-020-73271-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 09/11/2020] [Indexed: 11/20/2022] Open
Abstract
p-Alkoxyphenols (AOPs) are a class of ethers that are widely used in industrial and agricultural productions and daily necessities. p-Phenoxyphenol (PhOP) and p-pentyloxyphenol (PeOP) belong to this class and have been reported to be estrogenic in vitro. However, their in vivo estrogenic activities have rarely been of concern. In this study, we performed an immature mouse uterotrophic assay and studied the estrogenic effects of these two compounds in mice. The results revealed that the uterine weights of the animals treated with PhOP significantly increased at doses of 30 and 300 mg kg-1 bw day-1 for 3 days (P < 0.05), while no significant uterotrophic effects were observed in the mice treated with PeOP. Using next-generation transcriptome sequencing (RNA-seq), we also analyzed the gene expression in the uterine tissue of mice treated with PhOP and PeOP. The observed gene regulation patterns of the PhOP- and PeOP-treated specimens were similar to those of the 17β-estradiol (E2)-treated specimens. In particular, some estrogen-responsive genes, such as the Sprr2 gene family, Apoa1, Prap1, and Ahsg, displayed a regulation trend similar to that of E2. In addition, molecule docking analysis revealed that both PhOP and PeOP could be well docked into the active site of hERα, with potential of mean force (PMF) values of − 58.68 and − 52.67 kcal mol-1 for PhOP and PeOP, respectively. The results of this study indicate that PhOP exhibits relatively strong in vivo estrogenic activity, which could be of future concern.
Collapse
|
7
|
The Phenoxyphenol Compound 4-HPPP Selectively Induces Antiproliferation Effects and Apoptosis in Human Lung Cancer Cells through Aneupolyploidization and ATR DNA Repair Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5167292. [PMID: 32089770 PMCID: PMC7024103 DOI: 10.1155/2020/5167292] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023]
Abstract
Lung cancer is a leading cause of cancer death worldwide, and non-small-cell lung cancer (NSCLC) accounts for 85% of lung cancer, which is highly metastatic, leading to the poor survival rate of patients. We recently reported that 4-[4-(4-hydroxyphenoxy)phenoxy]phenol (4-HPPP), a phenoxyphenol, exerts antihepatoma effects by inducing apoptosis and autophagy. In this study, we further examined the effect of 4-HPPP and its analogs on NSCLC cells. Colony formation assays showed that 4-HPPP exerts selective cytotoxicity against NSCLC H1299 cells; furthermore, the inhibitory effect of 4-HPPP on the proliferation and migration of NSCLC cells was validated using an in vivo zebrafish-based tumor xenograft assay. The flow cytometry-based dichlorofluorescein diacetate (DCF-DA) assays indicated that 4-HPPP caused an increase in reactive oxygen species (ROS) in NSCLC cells, and Western blot assays showed that the major ROS scavenging enzymes superoxide dismutases- (SODs-) 1/2 were upregulated, whereas peroxidase (PRX) was downregulated. Furthermore, 4-HPPP caused both aneuploidization and the accumulation of γH2AX, a sensor of DNA damage, as well as the activation of double-strand break (DSB) markers, especially Ataxia-telangiectasia-mutated and Rad3-related (ATR) in NSCLC cells. Our present work suggests that the antiproliferative effects of 4-HPPP on lung cancer cells could be due to its phenoxyphenol structure, and 4-HPPP could be a candidate molecule for treating NSCLC by modulating ROS levels and lowering the threshold of polyploidy-specific cell death in the future.
Collapse
|
8
|
Umme Hani, Kandagalla S, Sharath BS, Jyothsna K, Manjunatha H. Network Pharmacology Approach Uncovering Pathways Involved in Targeting Hsp90 Through Curcumin and Epigallocatechin to Control Inflammation. Curr Drug Discov Technol 2019; 18:127-138. [PMID: 31820701 DOI: 10.2174/1570163816666191210145652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/07/2019] [Accepted: 10/15/2019] [Indexed: 11/22/2022]
Abstract
AIMS To fetch pathways involved in targetting Hsp90 through Curcumin and Epigallocatechin through Network pharmacological approach. BACKGROUND Hsp90 is a molecular chaperone involved in stabilizing inflammatory protein which may lead to chronic diseases. The herbal compounds Curcumin and Epigallocatechin processing antiinflammatory properties are known to follow a common pathway and control the expression of Hsp90. OBJECTIVE To collect the gene targets of Hsp90, Curcumin and Epigallocatechin in order to understand protein-protein interactions of gene targets by constructing the interactome to identify the hub proteins. Hub proteins docking was performed with curcumin and epigallocatechin. Finally, hub proteins involvement with various human diseases were identified. METHODS The gene targets of Hsp90, Curcumin and Epigallocatechin were obtained from there respective databases. Protein-protein interactions of Pkcδ-Nrf2 and Tlr4 pathway gene targets were collected from String database. Protein interaction network was constructed and merged to get intercession network in cytoscape and Cluego was used to predict the disease related target genes. Docking of ligands to target proteins was carried out using Autodock vina tool. RESULT The main key regulators of Curcumin and Epigallocatechin were identified particularly from Pkcδ-Nrf2 and Tlr4 pathway. CONCLUSION The combined action of Curcumin and Epigallocatechin can reduce the expression of Hsp90 eventually controlling the inflammation.
Collapse
Affiliation(s)
- Umme Hani
- Department of Biotechnology, Janana Sahyadri, Kuvempu University, Shankaraghatta, Shivamogga, Karnataka 577451, India
| | - Shivananda Kandagalla
- Department of Biotechnology, Janana Sahyadri, Kuvempu University, Shankaraghatta, Shivamogga, Karnataka 577451, India
| | - B S Sharath
- Department of Biotechnology, Janana Sahyadri, Kuvempu University, Shankaraghatta, Shivamogga, Karnataka 577451, India
| | - K Jyothsna
- Department of Biotechnology, Janana Sahyadri, Kuvempu University, Shankaraghatta, Shivamogga, Karnataka 577451, India
| | - Hanumanthappa Manjunatha
- Department of Biotechnology, Janana Sahyadri, Kuvempu University, Shankaraghatta, Shivamogga, Karnataka 577451, India
| |
Collapse
|
9
|
Curcumin and Its Derivatives as Potential Therapeutic Agents in Prostate, Colon and Breast Cancers. Molecules 2019; 24:molecules24234386. [PMID: 31801262 PMCID: PMC6930580 DOI: 10.3390/molecules24234386] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/27/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
Cancer is a life-threatening disease and is the second leading cause of death around the world. The increasing threats of drug-resistant cancers indicate that there is an urgent need for the improvement or development of more effective anticancer agents. Curcumin, a phenolic compound originally derived from turmeric plant (Curcuma longa L. (Zingiberaceae family)) widely known as a spice and a coloring agent for food have been reported to possess notable anticancer activity by inhibiting the proliferation and metastasis, and enhancing cell cycle arrest or apoptosis in various cancer cells. In spite of all these benefits, the therapeutic application of curcumin in clinical medicine and its bioavailability are still limited due to its poor absorption and rapid metabolism. Structural modification of curcumin through the synthesis of curcumin-based derivatives is a potential approach to overcome the above limitations. Curcumin derivatives can overcome the disadvantages of curcumin while enhancing the overall efficacy and hindering drug resistance. This article reports a review of published curcumin derivatives and their enhanced anticancer activities.
Collapse
|
10
|
Rodríguez Castaño P, Parween S, Pandey AV. Bioactivity of Curcumin on the Cytochrome P450 Enzymes of the Steroidogenic Pathway. Int J Mol Sci 2019; 20:ijms20184606. [PMID: 31533365 PMCID: PMC6770025 DOI: 10.3390/ijms20184606] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 09/11/2019] [Accepted: 09/16/2019] [Indexed: 11/16/2022] Open
Abstract
Turmeric, a popular ingredient in the cuisine of many Asian countries, comes from the roots of the Curcuma longa and is known for its use in Chinese and Ayurvedic medicine. Turmeric is rich in curcuminoids, including curcumin, demethoxycurcumin, and bisdemethoxycurcumin. Curcuminoids have potent wound healing, anti-inflammatory, and anti-carcinogenic activities. While curcuminoids have been studied for many years, not much is known about their effects on steroid metabolism. Since many anti-cancer drugs target enzymes from the steroidogenic pathway, we tested the effect of curcuminoids on cytochrome P450 CYP17A1, CYP21A2, and CYP19A1 enzyme activities. When using 10 µg/mL of curcuminoids, both the 17α-hydroxylase as well as 17,20 lyase activities of CYP17A1 were reduced significantly. On the other hand, only a mild reduction in CYP21A2 activity was observed. Furthermore, CYP19A1 activity was also reduced up to ~20% of control when using 1–100 µg/mL of curcuminoids in a dose-dependent manner. Molecular docking studies confirmed that curcumin could dock onto the active sites of CYP17A1, CYP19A1, as well as CYP21A2. In CYP17A1 and CYP19A1, curcumin docked within 2.5 Å of central heme while in CYP21A2 the distance from heme was 3.4 Å, which is still in the same range or lower than distances of bound steroid substrates. These studies suggest that curcuminoids may cause inhibition of steroid metabolism, especially at higher dosages. Also, the recent popularity of turmeric powder as a dilatory supplement needs further evaluation for the effect of curcuminoids on steroid metabolism. The molecular structure of curcuminoids could be modified to generate better lead compounds with inhibitory effects on CYP17A1 and CYP19A1 for potential drugs against prostate cancer and breast cancer.
Collapse
Affiliation(s)
- Patricia Rodríguez Castaño
- Pediatric Endocrinology, Diabetology, and Metabolism, University Children's Hospital Bern, 3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Shaheena Parween
- Pediatric Endocrinology, Diabetology, and Metabolism, University Children's Hospital Bern, 3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland
| | - Amit V Pandey
- Pediatric Endocrinology, Diabetology, and Metabolism, University Children's Hospital Bern, 3010 Bern, Switzerland.
- Department of Biomedical Research, University of Bern, 3010 Bern, Switzerland.
| |
Collapse
|
11
|
Soares CT, Fachin LRV, Trombone APF, Rosa PS, Ghidella CC, Belone AFF. Potential of AKR1B10 as a Biomarker and Therapeutic Target in Type 2 Leprosy Reaction. Front Med (Lausanne) 2018; 5:263. [PMID: 30320113 PMCID: PMC6166685 DOI: 10.3389/fmed.2018.00263] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
The AKR1B10 (aldo-keto reductase family 1 member B10) gene has important functions in carcinogen-induced neoplasia. AKR1B10 is also expressed in type 2 reaction leprosy patients (R2). We measured the expression of AKR1B10 in the skin lesions of patients with leprosy by immunohistochemistry from biopsies that encompassed the spectrum of types of leprosy, based on the Ridley and Jopling classification [10 samples each of tuberculoid (TT), borderline tuberculoid (BT), mid-borderline (BB), and borderline lepromatous (BL) lesions; four samples of lepromatous lesions (LL)], reactional leprosy [14 samples of type 1 Reaction (R1) and 10 samples of type 2 Reaction (R2)], and biopsies from 9 healthy control (HC) subjects. In addition, 46 lepromatous lesions (BL and LL), 45 lepromatous lesions in regression, and 115 R2 lesions were included. Eight of 10 R2 samples (80%), 3 of 46 active BL and LL samples (6%), 23 of 45 BL and LL samples in regression (51%), and 107 of 115 R2 samples (93%) were positive for AKR1B10, differing significantly between all groups (p < 0.05). AKR1B10 expression was highest in the cytoplasm of macrophages. Thus, AKR1B10 is overexpressed on the lepromatous side (BL and LL) in samples that are in regression, especially type 2 reaction-associated lesions, rendering it a potential marker of type 2 reactional episodes of leprosy and a target of drugs against reactional episodes.
Collapse
Affiliation(s)
- Cleverson T Soares
- Department of Anatomic Pathology, Instituto Lauro de Souza Lima, Bauru, Brazil
| | - Luciana R V Fachin
- Department of Anatomic Pathology, Instituto Lauro de Souza Lima, Bauru, Brazil
| | - Ana P F Trombone
- Department of Health Science, Universidade do Sagrado Coração, Bauru, Brazil
| | - Patricia S Rosa
- Division of Research and Education, Instituto Lauro de Souza Lima, Bauru, Brazil
| | - Cássio C Ghidella
- Ambulatory of Leprosy, Jardim Guanabara Health Center, Rondonópolis, Brazil
| | - Andrea F F Belone
- Department of Anatomic Pathology, Instituto Lauro de Souza Lima, Bauru, Brazil
| |
Collapse
|
12
|
Gad HA, Bouzabata A. Application of chemometrics in quality control of Turmeric (Curcuma longa) based on Ultra-violet, Fourier transform-infrared and 1H NMR spectroscopy. Food Chem 2017; 237:857-864. [PMID: 28764078 DOI: 10.1016/j.foodchem.2017.06.022] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/02/2017] [Accepted: 06/05/2017] [Indexed: 11/19/2022]
Abstract
Turmeric (Curcuma longa L.) belongs to the family Zingiberaceae that is widely used as a spice in food preparations in addition to its biological activities. UV, FT-IR, 1H NMR in addition to HPLC were applied to construct a metabolic fingerprint for Turmeric in an attempt to assess its quality. 30 samples were analyzed, and then principal component analysis (PCA) and hierarchical clustering analysis (HCA) were utilized to assess the differences and similarities between collected samples. PCA score plot based on both HPLC and UV spectroscopy showed the same discriminatory pattern, where the samples were segregated into four main groups depending on their total curcuminoids content. The results revealed that UV could be utilized as a simple and rapid alternative for HPLC. However, FT-IR failed to discriminate between the same species. By applying 1H NMR, the metabolic variability between samples was more evident in the essential oils/fatty acid region.
Collapse
Affiliation(s)
- Haidy A Gad
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, 11566 Abbassia, Cairo, Egypt.
| | - Amel Bouzabata
- Laboratoire de Synthèse Organique, Modélisation et Optimisation des Procèdes chimiques, Badji-Mokhtar Université, BP 23000 Annaba, Algeria.
| |
Collapse
|
13
|
Chang WT, Liu W, Chiu YH, Chen BH, Chuang SC, Chen YC, Hsu YT, Lu MJ, Chiou SJ, Chou CK, Chiu CC. A 4-Phenoxyphenol Derivative Exerts Inhibitory Effects on Human Hepatocellular Carcinoma Cells through Regulating Autophagy and Apoptosis Accompanied by Downregulating α-Tubulin Expression. Molecules 2017; 22:molecules22050854. [PMID: 28531143 PMCID: PMC6154338 DOI: 10.3390/molecules22050854] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cancer worldwide. Advanced HCCs are usually resistant to anticancer drugs, causing unsatisfactory chemotherapy outcomes. In this study, we showed that a 4-phenoxyphenol derivative, 4-[4-(4-hydroxyphenoxy)phenoxy]phenol (4-HPPP), exerts an inhibitory activity against two HCC cell lines, Huh7 and Ha22T. We further investigated the anti-HCC activities of 4-HPPP, including anti-proliferation and induction of apoptosis. Our results showed that higher dosage of 4-HPPP downregulates the expression of α-tubulin and causes nuclear enlargement in both the Huh-7 and Ha22T cell lines. Interestingly, the colony formation results showed a discrepancy in the inhibitory effect of 4-HPPP on HCC and rat liver epithelial Clone 9 cells, suggesting the selective cytotoxicity of 4-HPPP toward HCC cells. Furthermore, the cell proliferation and apoptosis assay results illustrated the differences between the two HCC cell lines. The results of cellular proliferation assays, including trypan blue exclusion and colony formation, revealed that 4-HPPP inhibits the growth of Huh7 cells, but exerts less cytotoxicity in Ha22T cells. Furthermore, the annexin V assay performed for detecting the apoptosis showed similar results. Western blotting results showed 4-HPPP caused the increase of pro-apoptotic factors including cleaved caspase-3, Bid and Bax in HCC cells, especially in Huh-7. Furthermore, an increase of autophagy-associated protein microtubule-associated protein-1 light chain-3B (LC3B)-II and the decrease of Beclin-1 and p62/SQSTM1 were observed following 4-HPPP treatment. Additionally, the level of γH2A histone family, member X (γH2AX), an endogenous DNA damage biomarker, was dramatically increased in Huh7 cells after 4-HPPP treatment, suggesting the involvement of DNA damage pathway in 4-HPPP-induced apoptosis. On the contrary, the western blotting results showed that treatment up-regulates pro-survival proteins, including the phosphorylation of protein kinase B (Akt) and the level of survivin on Ha22T cells, which may confer a resistance toward 4-HPPP. Notably, the blockade of extracellular signal-regulated kinases (ERK), but not Akt, enhanced the cytotoxicity of 4-HPPP against Ha22T cells, indicating the pro-survival role of ERK in 4-HPPP-induced anti-HCC effect. Our present work suggests that selective anti-HCC activity of 4-HPPP acts through induction of DNA damage. Accordingly, the combination of ERK inhibitor may significantly enhance the anti-cancer effect of 4-HPPP for those HCC cells which overexpress ERK in the future.
Collapse
Affiliation(s)
- Wen-Tsan Chang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yi-Han Chiu
- Department of Nursing, St. Mary's Junior College of Medicine, Nursing and Management, Yi-Lan 266, Taiwan.
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- The Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| | - Shih-Chang Chuang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Transplantation Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Yen-Chun Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yun-Tzh Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Mei-Jei Lu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Shean-Jaw Chiou
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chon-Kit Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Translational Research Center, Cancer Center, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
14
|
Ali NM, Yeap SK, Abu N, Lim KL, Ky H, Pauzi AZM, Ho WY, Tan SW, Alan-Ong HK, Zareen S, Alitheen NB, Akhtar MN. Synthetic curcumin derivative DK1 possessed G2/M arrest and induced apoptosis through accumulation of intracellular ROS in MCF-7 breast cancer cells. Cancer Cell Int 2017; 17:30. [PMID: 28239299 PMCID: PMC5320730 DOI: 10.1186/s12935-017-0400-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 02/09/2017] [Indexed: 12/31/2022] Open
Abstract
Aims Curcumin is a lead compound of the rhizomes of Curcuma longa and possess a broad range of pharmacological activities. Chemically, curcumin is 1,3-dicarbonyl class of compound, which exhibits keto-enol tautomerism. Despite of its strong biological properties, curcumin has yet been recommended as a therapeutic agent because of its poor bioavailability. Main methods A curcumin derivative (Z)-3-hydroxy-1-(2-hydroxyphenyl)-3-phenylprop-2-en-1-one (DK1) was synthesized and its cytotoxicity was tested on breast cancer cell MCF-7 and normal cell MCF-10A using MTT assay. Meanwhile, cell cycle regulation and apoptosis on MCF-7 cell were evaluated using flow cytometry. Regulation of cell cycle and apoptosis related genes expression was investigated by quantitative real time polymerase chain reaction (qRT-PCR), western blot and caspases activity analyses. Activation of oxidative stress on MCF-7 were evaluated by measuring ROS and GSH levels. Key findings DK1 was found to possess selective cytotoxicity on breast cancer MCF-7 cell than normal MCF-10A cell. Flow cytometry cell cycle and AnnexinV/PI analyses reported that DK1 effectively arrested MCF-7 at G2/M phase and induced apoptosis after 72 h of incubation than curcumin. Upregulation of p53, p21 and downregulation of PLK-1 subsequently promote phosphorylation of CDC2 which were found contributed to the arrest of G2/M phase. Moreover, increased of reactive oxygen species and reduced of antioxidant glutathione level correlate with apoptosis observed with raised of cytochrome c and active caspase 9. Significance DK1 was found to be more effective in inducing cell cycle arrest and apoptosis against MCF-7 cell with much higher selectivity index of MCF-10A/MCF-7 than curcumin, which might be contributed by the overexpression of p53 protein.
Collapse
Affiliation(s)
- Norlaily Mohd Ali
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Lot PT 21144, Jalan Sungai Long, Bandar Sungai Long, Cheras, 43000 Kajang, Selangor Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Jalan Sunsuria, Bandar Sunsuria, 43900 Sepang, Selangor Malaysia
| | - Nadiah Abu
- UKM Molecular Biology Institute (UMBI), UKM Medical Centre, Jalan Yaa'cob Latiff, Bandar Tun Razak, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Kian Lam Lim
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Lot PT 21144, Jalan Sungai Long, Bandar Sungai Long, Cheras, 43000 Kajang, Selangor Malaysia
| | - Huynh Ky
- Department of Agriculture Genetics and Breeding, College of Agriculture and Applied Biology, Cantho University, 3/2 Street, CanTho City, Vietnam
| | - Ahmad Zaim Mat Pauzi
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Wan Yong Ho
- School of Biomedical Sciences, The University of Nottingham Malaysia Campus, JalanBroga, 43500 Semenyih, Selangor Malaysia
| | - Sheau Wei Tan
- Institute of Bioscience, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Han Kiat Alan-Ong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Lot PT 21144, Jalan Sungai Long, Bandar Sungai Long, Cheras, 43000 Kajang, Selangor Malaysia
| | - Seema Zareen
- Faculty of Industrial Sciences & Technology, Universiti Malaysia Pahang, Lebuhraya Tun Razak, 26300 Kuantan Pahang, Malaysia
| | - Noorjahan Banu Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - M Nadeem Akhtar
- Faculty of Industrial Sciences & Technology, Universiti Malaysia Pahang, Lebuhraya Tun Razak, 26300 Kuantan Pahang, Malaysia
| |
Collapse
|
15
|
Huang L, He R, Luo W, Zhu YS, Li J, Tan T, Zhang X, Hu Z, Luo D. Aldo-Keto Reductase Family 1 Member B10 Inhibitors: Potential Drugs for Cancer Treatment. Recent Pat Anticancer Drug Discov 2017; 11:184-96. [PMID: 26844556 PMCID: PMC5403964 DOI: 10.2174/1574892811888160304113346] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/01/2016] [Accepted: 02/01/2016] [Indexed: 01/11/2023]
Abstract
Cytosolic NADPH-dependent reductase AKR1B10 is a member of the aldo-keto reductase (AKR) superfamily. This enzyme is normally expressed in the gastrointestinal tract. However, it is overexpressed in many solid tumors, such as hepatocarcinoma, lung cancer and breast cancer. AKR1B10 may play a role in the formation and development of carcinomas through multiple mechanisms including detoxification of cytotoxic carbonyls, modulation of retinoic acid level, and regulation of cellular fatty acid synthesis and lipid metabolism. Studies have suggested that AKR1B10 may be a useful biomarker for cancer diagnosis and a potential target for cancer treatment. Over the last decade, a number of AKR1B10 inhibitors including aldose reductase inhibitors (ARIs), endogenous substances, natural-based derivatives and synthetic compounds have been developed, which could be novel anticancer drugs. This review provides an overview on related articles and patents about AKR1B10 inhibitors, with a focus on their inhibition selectivity and mechanism of function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zheng Hu
- Translational Medicine Institute, National & Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Postdoctoral Mobile Stations of Central South University, Affiliated the First Peoples Hospital of Chenzhou of University of South China, Chenzhou 432000, P.R.China.
| | | |
Collapse
|
16
|
Allegra A, Innao V, Russo S, Gerace D, Alonci A, Musolino C. Anticancer Activity of Curcumin and Its Analogues: Preclinical and Clinical Studies. Cancer Invest 2016; 35:1-22. [PMID: 27996308 DOI: 10.1080/07357907.2016.1247166] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Curcumin has been shown to have a wide variety of therapeutic effects, ranging from anti-inflammatory, chemopreventive, anti-proliferative, and anti-metastatic. This review provides an overview of the recent research conducted to overcome the problems with the bioavailability of curcumin, and of the preclinical and clinical studies that have reported success in combinatorial strategies coupling curcumin with other treatments. Research on the signaling pathways that curcumin treatment targets shows that it potently acts on major intracellular components involved in key processes such as genomic modulations, cell invasion and cell death pathways. Curcumin is a promising molecule for the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Alessandro Allegra
- a Division of Hematology, Department of General Surgery, Oncology and Pathological Anatomy , University of Messina , Messina , Italy
| | - Vanessa Innao
- a Division of Hematology, Department of General Surgery, Oncology and Pathological Anatomy , University of Messina , Messina , Italy
| | - Sabina Russo
- a Division of Hematology, Department of General Surgery, Oncology and Pathological Anatomy , University of Messina , Messina , Italy
| | - Demetrio Gerace
- a Division of Hematology, Department of General Surgery, Oncology and Pathological Anatomy , University of Messina , Messina , Italy
| | - Andrea Alonci
- a Division of Hematology, Department of General Surgery, Oncology and Pathological Anatomy , University of Messina , Messina , Italy
| | - Caterina Musolino
- a Division of Hematology, Department of General Surgery, Oncology and Pathological Anatomy , University of Messina , Messina , Italy
| |
Collapse
|
17
|
Dietary polyphenols in prevention and treatment of prostate cancer. Int J Mol Sci 2015; 16:3350-76. [PMID: 25654230 PMCID: PMC4346900 DOI: 10.3390/ijms16023350] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 01/21/2015] [Accepted: 01/26/2015] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer is the most prevalent disease affecting males in many Western countries, with an estimated 29,480 deaths in 2014 in the US alone. Incidence rates for prostate cancer deaths have been decreasing since the early 1990s in men of all races/ethnicities, though they remain about 60% higher in African Americans than in any other group. The relationship between dietary polyphenols and the prevention of prostate cancer has been examined previously. Although results are sometimes inconsistent and variable, there is a general agreement that polyphenols hold great promise for the future management of prostate cancer. Various dietary components, including polyphenols, have been shown to possess anti-cancer properties. Generally considered as non-toxic, dietary polyphenols act as key modulators of signaling pathways and are therefore considered ideal chemopreventive agents. Besides possessing various anti-tumor properties, dietary polyphenols also contribute to epigenetic changes associated with the fate of cancer cells and have emerged as potential drugs for therapeutic intervention. Polyphenols have also been shown to affect post-translational modifications and microRNA expressions. This article provides a systematic review of the health benefits of selected dietary polyphenols in prostate cancer, especially focusing on the subclasses of polyphenols, which have a great effect on disease prevention and treatment.
Collapse
|