51
|
Pal D, Tyagi A, Chandrasekaran B, Alattasi H, Ankem MK, Sharma AK, Damodaran C. Suppression of Notch1 and AKT mediated epithelial to mesenchymal transition by Verrucarin J in metastatic colon cancer. Cell Death Dis 2018; 9:798. [PMID: 30038258 PMCID: PMC6056562 DOI: 10.1038/s41419-018-0810-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 01/10/2023]
Abstract
Epithelial to mesenchymal transition (EMT) in colorectal cancer (CRC) has been attributed to activation of AKT and Notch1 signaling pathways. As EMT corresponds to increased aggressiveness of CRC, approaches that prevent metastasis by targeting AKT/Notch1 pathways are at the forefront of current research paradigms. This study examined the anti-metastatic potential of Verrucarin J (VJ), a small molecule, in CRC cells overexpressing AKT and Notch1. VJ significantly inhibited AKT/HCT 116 cell growth by acting on the AKT/NFκB/Bcl-2 signaling axis and initiated apoptotic signaling as was evident from increased expression of pro-apoptotic markers such as cleaved PARP, cleaved caspase 3, and cleaved caspase 9. Also, VJ inhibited the cell growth in AKT/Notch1-overexpressing CRC cells and abrogated EMT. The down-regulation of AKT and Notch1 signaling was apparent in immunoblot analysis and corresponded with down-regulation of mesenchymal markers including Snail, and β-catenin. Intraperitoneal administration of VJ in control (pCMV/HCT 116) and AKT/HCT 116 mice significantly suppressed AKT-induced tumor growth in a xenograft model. In addition, down-regulation of prosurvival markers as well as AKT and Notch1 was observed in the immunohistochemical analysis of the xenografted tumors. In conclusion, our study substantiates the role of AKT and Notch1 in cell proliferation, angiogenesis, and EMT of CRC cells and demonstrates that VJ may be a viable therapeutic option to counter AKT-induced cell proliferation and tumor outgrowth in CRC.
Collapse
Affiliation(s)
- Deeksha Pal
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Ashish Tyagi
- Department of Urology, University of Louisville, Louisville, KY, USA
| | | | - Houda Alattasi
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - Murali K Ankem
- Department of Urology, University of Louisville, Louisville, KY, USA
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Chendil Damodaran
- Department of Urology, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
52
|
Hemati M, Haghiralsadat F, Yazdian F, Jafari F, Moradi A, Malekpour-Dehkordi Z. Development and characterization of a novel cationic PEGylated niosome-encapsulated forms of doxorubicin, quercetin and siRNA for the treatment of cancer by using combination therapy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 47:1295-1311. [DOI: 10.1080/21691401.2018.1489271] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Mahdie Hemati
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Haghiralsadat
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Fatemeh Yazdian
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Farzaneh Jafari
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Moradi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zahra Malekpour-Dehkordi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
53
|
Minas A, Najafi G, Jalali AS, Razi M. Fennel induces cytotoxic effects against testicular germ cells in mice; evidences for suppressed pre-implantation embryo development. ENVIRONMENTAL TOXICOLOGY 2018; 33:841-850. [PMID: 29761655 DOI: 10.1002/tox.22570] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 06/08/2023]
Abstract
Foeniculum vulgare (FVE; fennel) is an aromatic plant belonging to Umbelliferae family, which is widely used in traditional societies because of its different pharmaceutical properties. To uncover the fennel-derived essential oil (FVEO)-induced effects on male reproductive potential, 24 mature male albino mice were divided into, control, 0.37, 0.75, and 1.5 mg kg-1 FVEO-received groups. Following 35 days, the animals were euthanized and the testicular tissue and sperm samples were collected. The histological alterations, tubular differentiation (TDI), spermiogenesis (SPI) indices, apoptosis ratio, and RNA damage of germinal cells were analyzed. Moreover, the sperm count, motility, viability, chromatin condensation, and DNA fragmentation were assessed. Finally, the pre-implantation embryo development including; the percentage of zygote, 2-cell embryos and blastocysts were assessed. Observations showed that the FVEO, dose dependently, increased histological damages, resulted in germ cells dissociation, depletion, nuclear shrinkage and significantly (P < .05) decreased tubular differentiation and spermiogenesis ratios. Moreover, the FVEO-received animals (more significantly in 1.5 mg kg-1 -received group) exhibited decreased sperm count, viability, and motility and represented enhanced percentage of sperms with decondensed chromatin and DNA fragmentation. Finally, the animals in FVEO-received group showed diminished zygote formation and represented decreased pre-implantation embryo development compared to control animals. In conclusion, our data showed that, FVEO albeit at higher doses, is able to adversely affect cellular DNA and RNA contents, which in turn is able to negatively affect the sperm count and morphology. All these impairments are able to negatively affect the fertilization potential as well as pre-implantation embryo development.
Collapse
Affiliation(s)
- Aram Minas
- Department of Basic Science, Faculty of Veterinary, Urmia University, Urmia, Iran
| | - Gholamreza Najafi
- Department of Basic Science, Faculty of Veterinary, Urmia University, Urmia, Iran
| | - Ali Shalizar Jalali
- Department of Basic Science, Faculty of Veterinary, Urmia University, Urmia, Iran
| | - Mazdak Razi
- Department of Basic Science, Faculty of Veterinary, Urmia University, Urmia, Iran
| |
Collapse
|
54
|
Darband SG, Kaviani M, Yousefi B, Sadighparvar S, Pakdel FG, Attari JA, Mohebbi I, Naderi S, Majidinia M. Quercetin: A functional dietary flavonoid with potential chemo-preventive properties in colorectal cancer. J Cell Physiol 2018; 233:6544-6560. [PMID: 29663361 DOI: 10.1002/jcp.26595] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/12/2018] [Indexed: 02/06/2023]
Abstract
Recently, an intense attention has been paid to the application of natural compounds as a novel therapeutic strategy for cancer treatment. Quercetin, a natural flavonol present in many commonly consumed food items, is widely demonstrated to exert inhibitory effects on cancer progression through various mechanisms. Since there is a strong association with diets containing abundant vegetables, fruits, and grains, and significant decline in the risk of colon cancer, accumulation studies have focused on the anticancer potential of quercetin in colorectal cancer. Cell cycle arrest, increase in apoptosis, antioxidant replication, modulation of estrogen receptors, regulation of signaling pathways, inhibition of and metastasis and angiogenesis are among various mechanisms underlying the chemo-preventive effects of quercetin in colorectal cancer. This review covers various therapeutic interactions of Quercetin as to how targets cellular involved in cancer treatment.
Collapse
Affiliation(s)
- Saber G Darband
- Danesh Pey Hadi Co., Health Technology, Development Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Mojtaba Kaviani
- School of Nutrition and Dietetics, Acadia University, Wolfville, Nova Scotia, Canada
| | - Bahman Yousefi
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Sadighparvar
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Firouz G Pakdel
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Javad A Attari
- Department of Neurosurgery, Urmia University of Medical Sciences, Urmia, Iran
| | - Iraj Mohebbi
- Social Determinants of Health Center, Occupational Medicine Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Somayeh Naderi
- Danesh Pey Hadi Co., Health Technology, Development Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
55
|
Li M, Qian X, Zhu M, Li A, Fang M, Zhu Y, Zhang J. miR‑1273g‑3p promotes proliferation, migration and invasion of LoVo cells via cannabinoid receptor 1 through activation of ERBB4/PIK3R3/mTOR/S6K2 signaling pathway. Mol Med Rep 2018; 17:4619-4626. [PMID: 29328379 DOI: 10.3892/mmr.2018.8397] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/24/2017] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miR) are important in various crucial cell processes including proliferation, migration and invasion. Dysregulation of miRNAs have been increasingly reported to contribute to colorectal cancer. However, the detailed biological function and potential mechanisms of miR‑1273g‑3p in colorectal cancer remain poorly understood. The expression levels of miR‑1273g‑3p in human colorectal cancer LoVo cell lines were detected via reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). The target genes of miR‑1273g‑3p were predicted by bioinformatics and verified by a luciferase reporter assay, RT‑qPCR and western blotting. The MTT, wound‑healing and Transwell assays were used to examine the biological functions of miR‑1273g‑3p in LoVo cells. The potential molecular mechanisms of miR‑1273g‑3p on LoVo cell proliferation, migration and invasion was detected by western blotting. The results of the present study demonstrated that miR‑1273g‑3p expression was extensively upregulated in LoVo cells compared with the normal colon epithelial NCM460 cell line. Further studies indicated that miR‑1273g‑3p inhibitor significantly suppressed LoVo cell proliferation, migration and invasion compared with inhibitor control. Following this, the cannabinoid receptor 1 (CNR1) was identified as a direct target gene of miR‑1273g‑3p. Knockdown of CNR1 restored the phenotypes of LoVo cells transfected with miR‑1273g‑3p inhibitor. Furthermore, the potential molecular mechanism of miR‑1273g‑3p on LoVo cell proliferation, migration and invasion may be mediated by activating the Erb‑B2 receptor tyrosine kinase 4 (ERBB4)/phosphoinositide‑3‑kinase regulatory subunit 3 (PIK3R3)/mechanistic target of rapamycin (mTOR)/S6 kinase 2 (S6K2) signaling pathway. These observations indicated that miR‑1273g‑3p promoted the proliferation, migration and invasion of LoVo cells via CNR1, and this may have occurred through activation of the ERBB4/PIK3R3/mTOR/S6K2 signaling pathway, suggesting that miR‑1273g‑3p may serve as a novel therapeutic target for the effective treatment of colorectal cancer.
Collapse
Affiliation(s)
- Min Li
- Department of Oncology, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210000, P.R. China
| | - Xiaoping Qian
- Department of The Comprehensive Cancer Center, Affiliated Nanjing Drum Tower Hospital, Nanjing University, Nanjing, Jiangsu 210008, P.R. China
| | - Mingzhen Zhu
- The Department of Tumor‑Chemotherapy, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu 222023, P.R. China
| | - Aiyi Li
- The Department of Tumor‑Chemotherapy, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu 222023, P.R. China
| | - Mingzhi Fang
- Department of Oncology, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210000, P.R. China
| | - Yong Zhu
- National Medical Centre of Colorectal Disease, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210000, P.R. China
| | - Jingyu Zhang
- The Department of Tumor‑Chemotherapy, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu 222023, P.R. China
| |
Collapse
|
56
|
Suman S, Das TP, Sirimulla S, Alatassi H, Ankem MK, Damodaran C. Withaferin-A suppress AKT induced tumor growth in colorectal cancer cells. Oncotarget 2017; 7:13854-64. [PMID: 26883103 PMCID: PMC4924683 DOI: 10.18632/oncotarget.7351] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/29/2016] [Indexed: 12/12/2022] Open
Abstract
The oncogenic activation of AKT gene has emerged as a key determinant of the aggressiveness of colorectal cancer (CRC); hence, research has focused on targeting AKT signaling for the treatment of advanced stages of CRC. In this study, we explored the anti-tumorigenic effects of withaferin A (WA) on CRC cells overexpressing AKT in preclinical (in vitro and in vivo) models. Our results indicated that WA, a natural compound, resulted in significant inhibition of AKT activity and led to the inhibition of cell proliferation, migration and invasion by downregulating the epithelial to mesenchymal transition (EMT) markers in CRC cells overexpressing AKT. The oral administration of WA significantly suppressed AKT-induced aggressive tumor growth in a xenograft model. Molecular analysis revealed that the decreased expression of AKT and its downstream pro-survival signaling molecules may be responsible for tumor inhibition. Further, significant inhibition of some important EMT markers, i.e., Snail, Slug, β-catenin and vimentin, was observed in WA-treated human CRC cells overexpressing AKT. Significant inhibition of micro-vessel formation and the length of vessels were evident in WA-treated tumors, which correlated with a low expression of the angiogenic marker RETIC. In conclusion, the present study emphasizes the crucial role of AKT activation in inducing cell proliferation, angiogenesis and EMT in CRC cells and suggests that WA may overcome AKT-induced cell proliferation and tumor growth in CRC.
Collapse
Affiliation(s)
- Suman Suman
- Department of Urology, University of Louisville, Louisville, KY 40202, USA
| | - Trinath P Das
- Department of Urology, University of Louisville, Louisville, KY 40202, USA
| | - Suman Sirimulla
- Department of Basic Sciences, St. Louis College of Pharmacy, St. Louis, MO 63110, USA
| | - Houda Alatassi
- Department of Pathology, University of Louisville, Louisville, KY 40202, USA
| | - Murali K Ankem
- Department of Urology, University of Louisville, Louisville, KY 40202, USA
| | - Chendil Damodaran
- Department of Urology, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
57
|
De Santis S, Galleggiante V, Scandiffio L, Liso M, Sommella E, Sobolewski A, Spilotro V, Pinto A, Campiglia P, Serino G, Santino A, Notarnicola M, Chieppa M. Secretory Leukoprotease Inhibitor (Slpi) Expression Is Required for Educating Murine Dendritic Cells Inflammatory Response Following Quercetin Exposure. Nutrients 2017; 9:nu9070706. [PMID: 28684695 PMCID: PMC5537821 DOI: 10.3390/nu9070706] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/29/2017] [Accepted: 07/04/2017] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells' (DCs) ability to present antigens and initiate the adaptive immune response confers them a pivotal role in immunological defense against hostile infection and, at the same time, immunological tolerance towards harmless components of the microbiota. Food products can modulate the inflammatory status of intestinal DCs. Among nutritionally-derived products, we investigated the ability of quercetin to suppress inflammatory cytokines secretion, antigen presentation, and DCs migration towards the draining lymph nodes. We recently identified the Slpi expression as a crucial checkpoint required for the quercetin-induced inflammatory suppression. Here we demonstrate that Slpi-KO DCs secrete a unique panel of cytokines and chemokines following quercetin exposure. In vivo, quercetin-enriched food is able to induce Slpi expression in the ileum, while little effects are detectable in the duodenum. Furthermore, Slpi expressing cells are more frequent at the tip compared to the base of the intestinal villi, suggesting that quercetin exposure could be more efficient for DCs projecting periscopes in the intestinal lumen. These data suggest that quercetin-enriched nutritional regimes may be efficient for suppressing inflammatory syndromes affecting the ileo-colonic tract.
Collapse
Affiliation(s)
- Stefania De Santis
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Via Turi, 27, 70013 Castellana Grotte, Italy.
- Institute of Sciences of Food Production C.N.R., Unit of Lecce, via Monteroni, 73100 Lecce, Italy.
| | - Vanessa Galleggiante
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Via Turi, 27, 70013 Castellana Grotte, Italy.
| | - Letizia Scandiffio
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Via Turi, 27, 70013 Castellana Grotte, Italy.
| | - Marina Liso
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Via Turi, 27, 70013 Castellana Grotte, Italy.
| | - Eduardo Sommella
- Department of Pharmacy, School of Pharmacy, University of Salerno, 84084 Fisciano, Italy.
| | | | - Vito Spilotro
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Via Turi, 27, 70013 Castellana Grotte, Italy.
| | - Aldo Pinto
- Department of Pharmacy, School of Pharmacy, University of Salerno, 84084 Fisciano, Italy.
| | - Pietro Campiglia
- Department of Pharmacy, School of Pharmacy, University of Salerno, 84084 Fisciano, Italy.
- European Biomedical Research Institute of Salerno (EBRIS), Via S. de Renzi, 3, 84125 Salerno, Italy.
| | - Grazia Serino
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Via Turi, 27, 70013 Castellana Grotte, Italy.
| | - Angelo Santino
- Institute of Sciences of Food Production C.N.R., Unit of Lecce, via Monteroni, 73100 Lecce, Italy.
| | - Maria Notarnicola
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Via Turi, 27, 70013 Castellana Grotte, Italy.
| | - Marcello Chieppa
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Via Turi, 27, 70013 Castellana Grotte, Italy.
- Department of Pharmacy, School of Pharmacy, University of Salerno, 84084 Fisciano, Italy.
| |
Collapse
|
58
|
Seidel DV, Azcárate-Peril MA, Chapkin RS, Turner ND. Shaping functional gut microbiota using dietary bioactives to reduce colon cancer risk. Semin Cancer Biol 2017; 46:191-204. [PMID: 28676459 DOI: 10.1016/j.semcancer.2017.06.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/18/2022]
Abstract
Colon cancer is a multifactorial disease associated with a variety of lifestyle factors. Alterations in the gut microbiota and the intestinal metabolome are noted during colon carcinogenesis, implicating them as critical contributors or results of the disease process. Diet is a known determinant of health, and as a modifier of the gut microbiota and its metabolism, a critical element in maintenance of intestinal health. This review summarizes recent evidence demonstrating the role and responses of the intestinal microbiota during colon tumorigenesis and the ability of dietary bioactive compounds and probiotics to impact colon health from the intestinal lumen to the epithelium and systemically. We first describe changes to the intestinal microbiome, metabolome, and epithelium associated with colon carcinogenesis. This is followed by a discussion of recent evidence indicating how specific classes of dietary bioactives, prebiotics, or probiotics affect colon carcinogenesis. Lastly, we briefly address the prospects of using multiple 'omics' techniques to integrate the effects of diet, host, and microbiota on colon tumorigenesis with the goal of more fully appreciating the interconnectedness of these systems and thus, how these approaches can be used to advance personalized nutrition strategies and nutrition research.
Collapse
Affiliation(s)
- Derek V Seidel
- Nutrition and Food Science Department, and Faculty of Genetics, Texas A&M University, College Station, TX 77843-2253, USA.
| | - M Andrea Azcárate-Peril
- Department of Medicine GI Division, University of North Carolina, Chapel Hill, NC 27599-7555, USA.
| | - Robert S Chapkin
- Nutrition and Food Science Department, and Faculty of Genetics, Texas A&M University, College Station, TX 77843-2253, USA.
| | - Nancy D Turner
- Nutrition and Food Science Department, and Faculty of Genetics, Texas A&M University, College Station, TX 77843-2253, USA.
| |
Collapse
|
59
|
Badal S, Smith KN, Rajnarayanan R. Analysis of natural product regulation of cannabinoid receptors in the treatment of human disease. Pharmacol Ther 2017; 180:24-48. [PMID: 28583800 DOI: 10.1016/j.pharmthera.2017.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The organized, tightly regulated signaling relays engaged by the cannabinoid receptors (CBs) and their ligands, G proteins and other effectors, together constitute the endocannabinoid system (ECS). This system governs many biological functions including cell proliferation, regulation of ion transport and neuronal messaging. This review will firstly examine the physiology of the ECS, briefly discussing some anomalies in the relay of the ECS signaling as these are consequently linked to maladies of global concern including neurological disorders, cardiovascular disease and cancer. While endogenous ligands are crucial for dispatching messages through the ECS, there are also commonalities in binding affinities with copious exogenous ligands, both natural and synthetic. Therefore, this review provides a comparative analysis of both types of exogenous ligands with emphasis on natural products given their putative safer efficacy and the role of Δ9-tetrahydrocannabinol (Δ9-THC) in uncovering the ECS. Efficacy is congruent to both types of compounds but noteworthy is the effect of a combination therapy to achieve efficacy without unideal side-effects. An example is Sativex that displayed promise in treating Huntington's disease (HD) in preclinical models allowing for its transition to current clinical investigation. Despite the in vitro and preclinical efficacy of Δ9-THC to treat neurodegenerative ailments, its psychotropic effects limit its clinical applicability to treating feeding disorders. We therefore propose further investigation of other compounds and their combinations such as the triterpene, α,β-amyrin that exhibited greater binding affinity to CB1 than CB2 and was more potent than Δ9-THC and the N-alkylamides that exhibited CB2 selective affinity; the latter can be explored towards peripherally exclusive ECS modulation. The synthetic CB1 antagonist, Rimonabant was pulled from commercial markets for the treatment of diabetes, however its analogue SR144528 maybe an ideal lead molecule towards this end and HU-210 and Org27569 are also promising synthetic small molecules.
Collapse
Affiliation(s)
- S Badal
- Department of Basic Medical Sciences, Faculty of Medical Sciences, University of the West Indies, Mona, Jamaica.
| | - K N Smith
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - R Rajnarayanan
- Jacobs School of Medicine and Biomedical Sciences, Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14228, USA
| |
Collapse
|
60
|
Maryam R, Faegheh S, Majid AS, Kazem NK. Effect of quercetin on secretion and gene expression of leptin in breast cancer. J TRADIT CHIN MED 2017. [DOI: 10.1016/s0254-6272(17)30067-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
61
|
Li QC, Liang Y, Hu GR, Tian Y. Enhanced therapeutic efficacy and amelioration of cisplatin-induced nephrotoxicity by quercetin in 1,2-dimethyl hydrazine-induced colon cancer in rats. Indian J Pharmacol 2017; 48:168-71. [PMID: 27127319 PMCID: PMC4825434 DOI: 10.4103/0253-7613.178834] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Objective: The aim of quercetin treatment in combination of cisplatin (CP)-induced nephrotoxicity as well on 1,2-dimethyl hydrazine (DMH)-induced colon cancer. Materials and Methods: In this study, animals are exposed to DMH hydrochloride to induce colon cancer. In these groups, nephrotoxicity was assessed with the help of blood urea nitrogen, urea, and creatinine. The antitumor activity of quercetin and CP assessed with the help of number of aberrant crypts and foci formation. Tumor size in different treatment group determined with the help of vernier caliper. Results: CP is one of the most widely used antineoplastic drugs against colon cancer, but it has a major dose-limiting drawbacks of causing nephrotoxicity. Therefore, there is a need for a novel therapeutic regimen which will reduce the nephrotoxicity and enhance the anticancer activity of CP. The protective effect of quercetin on CP-induced nephrotoxicity is well-known. Moreover, quercetin is proven to have antitumor activity in colon cancer. Keeping all the facts in view, this study was conceived to evaluate the role of quercetin on therapeutic efficacy and nephrotoxicity of CP in DMH-induced colon cancer in male Sprague–Dawley rats. Treatment of quercetin with CP (7.5 mg/kg) prevents the CP-induced nephrotoxicity along with enhance the anticancer activity confirmed by the reduction of aberrant crypt foci number. Treatment of CP and quercetin alone leads to significant increase in the anticancer activity as compared to control colon tumor rats. Conclusion: In DMH-induced colon cancer model, combination of quercetin and CP ameliorates CP-induced nephrotoxicity as well as enhanced antitumor activity.
Collapse
Affiliation(s)
- Qing-Chun Li
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Yun Liang
- Department of Physical Examination, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Guang-Rui Hu
- Department of Physical Examination, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Yuan Tian
- Department of Physical Examination, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| |
Collapse
|
62
|
Zhou J, Fang L, Liao J, Li L, Yao W, Xiong Z, Zhou X. Investigation of the anti-cancer effect of quercetin on HepG2 cells in vivo. PLoS One 2017; 12:e0172838. [PMID: 28264020 PMCID: PMC5338765 DOI: 10.1371/journal.pone.0172838] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 02/10/2017] [Indexed: 12/18/2022] Open
Abstract
Quercetin, a natural polyphenolic flavonoid compound, can inhibit the growth of several malignant cancers. However, the mechanism still remains unclear. Our previous findings have suggested that quercetin can significantly inhibit HepG2 cell proliferation and induce cell apoptosis in vitro. It can also affect cell cycle distribution and significantly decrease cyclin D1 expression. In this study, we investigated the anti-cancer effect of quercetin on HepG2 tumor-bearing nude mice and its effect on cyclin D1 expression in the tumor tissue. First, the nude murine tumor model was established by subcutaneous inoculation of HepG2 cells, then quercetin was administered intraperitoneally, and the mice injected with saline solution were used as controls. The daily behavior of the tumor-bearing mice was observed and differences in tumor growth and survival rate were monitored. The expression of cyclin D1 in isolated tumor sections was evaluated by immunohistochemistry. We found that HepG2 tumor became palpable in the mice one-week post-inoculation. Tumors in the control group grew rapidly and the daily behavior of the mice changed significantly, including listlessness, poor feeding and ataxia. The mice in quercetin-treated group showed delayed tumor growth, no significant changes in daily behavior, and the survival rate was significantly improved. Finally, we observed increased tumor necrosis and a lighter cyclin D1 staining with reduced staining areas. Our findings thus suggest that quercetin can significantly inhibit HepG2 cell proliferation, and this effect may be achieved through the regulation of cyclin D1 expression.
Collapse
Affiliation(s)
- Jin Zhou
- Department of Chemotherapy, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
- * E-mail:
| | - Li Fang
- Department of Gastroenterology, the First Affiliated Hospital, Chengdu Medical College, Chengdu, Sichuan, P.R. China
| | - Jiaxu Liao
- Department of Radiology, the Sixth Hospital, Chengdu, Sichuan, P.R. China
| | - Lin Li
- Department of Nuclear Medicine, the Second Hospital, Chengdu, Sichuan, P.R. China
| | - Wenxiu Yao
- Department of Chemotherapy, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| | - Zhujuan Xiong
- Department of Chemotherapy, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| | - Xiang Zhou
- Department of Chemotherapy, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, P.R. China
| |
Collapse
|
63
|
Wang Y, DiSalvo M, Gunasekara DB, Dutton J, Proctor A, Lebhar MS, Williamson IA, Speer J, Howard RL, Smiddy NM, Bultman SJ, Sims CE, Magness ST, Allbritton NL. Self-renewing Monolayer of Primary Colonic or Rectal Epithelial Cells. Cell Mol Gastroenterol Hepatol 2017; 4:165-182.e7. [PMID: 29204504 PMCID: PMC5710741 DOI: 10.1016/j.jcmgh.2017.02.011] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/15/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Three-dimensional organoid culture has fundamentally changed the in vitro study of intestinal biology enabling novel assays; however, its use is limited because of an inaccessible luminal compartment and challenges to data gathering in a three-dimensional hydrogel matrix. Long-lived, self-renewing 2-dimensional (2-D) tissue cultured from primary colon cells has not been accomplished. METHODS The surface matrix and chemical factors that sustain 2-D mouse colonic and human rectal epithelial cell monolayers with cell repertoires comparable to that in vivo were identified. RESULTS The monolayers formed organoids or colonoids when placed in standard Matrigel culture. As with the colonoids, the monolayers exhibited compartmentalization of proliferative and differentiated cells, with proliferative cells located near the peripheral edges of growing monolayers and differentiated cells predominated in the central regions. Screening of 77 dietary compounds and metabolites revealed altered proliferation or differentiation of the murine colonic epithelium. When exposed to a subset of the compound library, murine organoids exhibited similar responses to that of the monolayer but with differences that were likely attributable to the inaccessible organoid lumen. The response of the human primary epithelium to a compound subset was distinct from that of both the murine primary epithelium and human tumor cells. CONCLUSIONS This study demonstrates that a self-renewing 2-D murine and human monolayer derived from primary cells can serve as a physiologically relevant assay system for study of stem cell renewal and differentiation and for compound screening. The platform holds transformative potential for personalized and precision medicine and can be applied to emerging areas of disease modeling and microbiome studies.
Collapse
Key Words
- 2-D, two-dimensional
- 3-D, three-dimensional
- ALP, alkaline phosphatase
- CAG, cytomegalovirus enhancer plus chicken actin promoter
- CI, confidence interval
- Colonic Epithelial Cells
- Compound Screening
- ECM, extracellular matrix
- EDU, 5-ethynyl-2′-deoxyuridine
- EGF, epidermal growth factor
- ENR-W, cell medium with [Wnt-3A] of 30 ng/mL
- ENR-w, cell medium with [Wnt-3A] of 10 ng/mL
- HISC, human intestinal stem cell medium
- IACUC, Institutional Animal Care and Use Committee
- ISC, intestinal stem cell
- Monolayer
- Organoids
- PBS, phosphate-buffered saline
- PDMS, polydimethylsiloxane
- RFP, red fluorescent protein
- SEM, scanning electron microscope
- SSMD, strictly standardized mean difference
- UNC, University of North Carolina
- α-ChgA, anti-chromogranin A
- α-Muc2, anti-mucin2
Collapse
Affiliation(s)
- Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Matthew DiSalvo
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Dulan B. Gunasekara
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Johanna Dutton
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Angela Proctor
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Michael S. Lebhar
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Ian A. Williamson
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Jennifer Speer
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Riley L. Howard
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, North Carolina
| | - Nicole M. Smiddy
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Scott J. Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina
| | - Scott T. Magness
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina,Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina,Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, North Carolina,Correspondence Address correspondence to: Nancy L. Allbritton, MD, PhD, Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599. fax: (919) 962-2388.Department of ChemistryUniversity of North CarolinaChapel HillNorth Carolina 27599
| |
Collapse
|
64
|
Notarnicola M, Tutino V, De Nunzio V, Dituri F, Caruso MG, Giannelli G. Dietary ω-3 Polyunsaturated Fatty Acids Inhibit Tumor Growth in Transgenic Apc Min/+ Mice, Correlating with CB1 Receptor Up-Regulation. Int J Mol Sci 2017; 18:ijms18030485. [PMID: 28245562 PMCID: PMC5372501 DOI: 10.3390/ijms18030485] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/17/2017] [Accepted: 02/20/2017] [Indexed: 02/07/2023] Open
Abstract
Mediterranean diet components, such as olive oil and ω-3 polyunsaturated fatty acids (ω-3 PUFAs), can arrest cell growth and promote cell apoptosis. Recently, olive oil has been demonstrated to modulate type-1 cannabinoid (CB1) receptor gene expression in both human colon cancer cells and rat colon. The aim of this study was to investigate a possible link between olive oil and ω-3 PUFAs effects and CB1 receptor expression in both intestinal and adipose tissue of ApcMin/+ mice. To confirm the role for the CB1 receptor as a negative modulator of cell proliferation in human colon cancer, CB1 receptor gene expression was also detected in tumor tissue and in surrounding normal mucosa of patients with colorectal cancer (CRC). Dietary ω-3 PUFAs significantly inhibited intestinal polyp growth in mice, correlating with CB1 receptor gene and protein expression induction. CB1 receptor gene up-regulation was also detected in adipose tissue, suggesting a close communication between cancer cells and the surrounding environment. Tissue CB1 receptor induction was associated with a concurrent inactivation of the Wnt/β-catenin pathway. Moreover, there was a significant reduction in CB1 receptor gene expression levels in cancer tissue compared to normal surrounding mucosa of patients with CRC, confirming that in cancer the “protective” action of the CB1 receptor is lost.
Collapse
Affiliation(s)
- Maria Notarnicola
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy.
| | - Valeria Tutino
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy.
| | - Valentina De Nunzio
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy.
| | - Francesco Dituri
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy.
| | - Maria Gabriella Caruso
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy.
| |
Collapse
|
65
|
Kangawa Y, Yoshida T, Maruyama K, Okamoto M, Kihara T, Nakamura M, Ochiai M, Hippo Y, Hayashi SM, Shibutani M. Cilostazol and enzymatically modified isoquercitrin attenuate experimental colitis and colon cancer in mice by inhibiting cell proliferation and inflammation. Food Chem Toxicol 2017; 100:103-114. [DOI: 10.1016/j.fct.2016.12.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 12/04/2016] [Accepted: 12/14/2016] [Indexed: 12/26/2022]
|
66
|
Zhang L, Zhang J, Qi B, Jiang G, Liu J, Zhang P, Ma Y, Li W. The anti-tumor effect and bioactive phytochemicals of Hedyotis diffusa willd on ovarian cancer cells. JOURNAL OF ETHNOPHARMACOLOGY 2016; 192:132-139. [PMID: 27426510 DOI: 10.1016/j.jep.2016.07.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/06/2016] [Accepted: 07/07/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hedyotis diffusa willd (HDW) is a widely used medicinal herb in China. It processed various medicinal properties including antioxidative, anti-inflamatory and anti-cancer effects. This study aimed to investigate the anti-tumor effects of HDW on ovarian cancer cells and the underlying mechanisms as well as identify the bioactive compounds. MATERIALS AND METHODS Effects of HDW on the viability of ovarian cancer A2780 cells were detected by MTT assay. Apoptosis was detected by cell morphologic observation through DAPI staining and flow cytometry analysis. The migration of ovarian cancer cells which exposed to HDW were detected by wound healing and transwell assays. The protein levels of caspase 3/9, Bcl-2 and MMP-2/9 in human ovarian cancer cells treated with HDW were assessed by western blotting analysis. The potential bioactive compounds were characterized by HPLC-Q-TOF-MS. RESULTS HDW significantly inhibited the growth of A2780 ovarian cancer cells and induced apoptosis. The induction of apoptosis by HDW was associated with down-regulation of anti-apoptotic protein Bcl-2 and the activation of caspase 3/9. Wound healing and transwell chamber assays indicated HDW suppressed the migration of ovarian cancer cells. HDW dramatically decreased MMP-2/9 expression. A HPLC-Q-TOF-MS analysis of HDW indicated the presence of 13 flavonoids compounds and one anthraquinone compound, which may contribute to the anticancer activity of the HDW. CONCLUSIONS HDW effectively restricted the growth of ovarian cancer cells and induced apoptosis through the mitochondria-associated apoptotic pathway. Furthermore, HDW suppressed the migration of ovarian cancer cells through down-regulation of MMP-2 and MMP-9 expression. These results showed that HDW hold potential therapeutic effect for ovarian cancer patients.
Collapse
Affiliation(s)
- Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Jing Zhang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Bing Qi
- Acute Abdomen Department of the First Affiliated Hospital, Dalian medical University, Dalian 116000, China
| | - Guoqiang Jiang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Jia Liu
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Pei Zhang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China
| | - Yuan Ma
- Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
67
|
Kashyap D, Mittal S, Sak K, Singhal P, Tuli HS. Molecular mechanisms of action of quercetin in cancer: recent advances. Tumour Biol 2016; 37:12927-12939. [PMID: 27448306 DOI: 10.1007/s13277-016-5184-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 07/13/2016] [Indexed: 02/06/2023] Open
Abstract
In the last few decades, the scientific community has discovered an immense potential of natural compounds in the treatment of dreadful diseases such as cancer. Besides the availability of a variety of natural bioactive molecules, efficacious cancer therapy still needs to be developed. So, to design an efficacious cancer treatment strategy, it is essential to understand the interactions of natural molecules with their respective cellular targets. Quercetin (Quer) is a naturally occurring flavonol present in many commonly consumed food items. It governs numerous intracellular targets, including the proteins involved in apoptosis, cell cycle, detoxification, antioxidant replication, and angiogenesis. The weight of available synergistic studies vigorously fortifies the utilization of Quer as a chemoprevention drug. This extensive review covers various therapeutic interactions of Quer with their recognized cellular targets involved in cancer treatment.
Collapse
Affiliation(s)
- Dharambir Kashyap
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab, 160012, India
| | - Sonam Mittal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Katrin Sak
- Department of Hematology and Oncology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Paavan Singhal
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana, Ambala, 133203, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana, Ambala, 133203, India.
| |
Collapse
|
68
|
John ASP, Ankem MK, Damodaran C. Oxidative Stress: A Promising Target for Chemoprevention. ACTA ACUST UNITED AC 2016; 2:73-81. [PMID: 27088073 DOI: 10.1007/s40495-016-0052-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cancer is a leading cause of death worldwide, and treating advanced stages of cancer remains clinically challenging. Epidemiological studies have shown that oxidants and free radicals induced DNA damage is one of the predominant causative factors for cancer pathogenesis. Hence, oxidants are attractive targets for chemoprevention as well as therapy. Dietary agents are known to exert an anti-oxidant property which is one of the most efficient preventive strategy in cancer progression. In this article, we highlight dietary agents can potentially target oxidative stress, in turn delaying, preventing, or treating cancer development. Some of these agents are currently in use in basic research, while some have been launched successfully into clinical trials.
Collapse
Affiliation(s)
| | - Murali K Ankem
- Department of Urology, University of Louisville, KY 40202
| | | |
Collapse
|
69
|
Novel Investigations of Flavonoids as Chemopreventive Agents for Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:840542. [PMID: 26858957 PMCID: PMC4695650 DOI: 10.1155/2015/840542] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/19/2015] [Indexed: 12/16/2022]
Abstract
We would like to highlight the application of natural products to hepatocellular carcinoma (HCC). We will focus on the natural products known as flavonoids, which target this disease at different stages of hepatocarcinogenesis. In spite of the use of chemotherapy and radiotherapy in treating HCC, patients with HCC still face poor prognosis because of the nature of multidrug resistance and toxicity derived from chemotherapy and radiotherapy. Flavonoids can be found in many vegetables, fruits, and herbal medicines that exert their different anticancer effects via different intracellular signaling pathways and serve as antioxidants. In this review, we will discuss seven common flavonoids that exert different biological effects against HCC via different pathways.
Collapse
|
70
|
Nath LR, Gorantla JN, Joseph SM, Antony J, Thankachan S, Menon DB, Sankar S, Lankalapalli RS, Anto RJ. Kaempferide, the most active among the four flavonoids isolated and characterized from Chromolaena odorata, induces apoptosis in cervical cancer cells while being pharmacologically safe. RSC Adv 2015. [DOI: 10.1039/c5ra19199h] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We report the isolation and characterization of four compounds from a cytotoxic fraction F-17, isolated from the DCM extract of C. odorata by bioactivity guided fractionation.
Collapse
Affiliation(s)
- Lekshmi R. Nath
- Division of Cancer Research
- Rajiv Gandhi Centre for Biotechnology
- Thiruvananthapuram-695014
- India
| | - Jaggaiah N. Gorantla
- Chemical Sciences and Technology Division
- CSIR-National Institute for Interdisciplinary Science and Technology
- Thiruvananthapuram-695019
- India
| | - Sophia Margaret Joseph
- Division of Cancer Research
- Rajiv Gandhi Centre for Biotechnology
- Thiruvananthapuram-695014
- India
| | - Jayesh Antony
- Division of Cancer Research
- Rajiv Gandhi Centre for Biotechnology
- Thiruvananthapuram-695014
- India
| | - Sanu Thankachan
- Division of Cancer Research
- Rajiv Gandhi Centre for Biotechnology
- Thiruvananthapuram-695014
- India
| | - Darsan B. Menon
- Division of Cancer Research
- Rajiv Gandhi Centre for Biotechnology
- Thiruvananthapuram-695014
- India
| | - S. Sankar
- Department of Pathology
- Government Medical College
- Thiruvananthapuram-695011
- India
| | - Ravi S. Lankalapalli
- Chemical Sciences and Technology Division
- CSIR-National Institute for Interdisciplinary Science and Technology
- Thiruvananthapuram-695019
- India
| | - Ruby John Anto
- Division of Cancer Research
- Rajiv Gandhi Centre for Biotechnology
- Thiruvananthapuram-695014
- India
| |
Collapse
|