1
|
Flaxseed Ethanol Extracts’ Antitumor, Antioxidant, and Anti-Inflammatory Potential. Antioxidants (Basel) 2022; 11:antiox11050892. [PMID: 35624757 PMCID: PMC9137875 DOI: 10.3390/antiox11050892] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
The antitumoral, antioxidant, and anti-inflammatory effects of flaxseed ethanol extract was screened. Phytochemical analysis was performed by measuring the total phenolic content and by HPLC-DAD-ESI MS. In vitro antiproliferative activity was appreciated by MMT test of four adenocarcinomas and two normal cell lines. In vitro, antioxidant activity was evaluated by DPPH, FRAP, H2O2, and NO scavenging tests. The in vivo growth inhibitory activity against Ehrlich ascites carcinoma (EAC) in female BALB/c mice was determined using the trypan blue test. In EAC mice serum and ascites total oxidative status, total antioxidant reactivity, oxidative stress index, malondialdehyde, total thiols, total nitrites, 3-nitrotyrosine, and NFkB were measured. The phytochemical analysis found an significant content of phenols, with lignans having the highest concentration. The extract had an significant in vitro antioxidant effect and different inhibitory effects on different cell lines. After treatment of EAC mice with flaxseeds extract, body weight, ascites volume and viable tumour cell count, serum and ascites oxidative stress, and inflammatory markers decreased significantly. The ethanol flaxseeds extract has potential antiproliferative activity against some ovary and endometrial malignant cells and EAC. This effect can be attributed to the phenols content, and its antioxidant and anti-inflammatory activity.
Collapse
|
2
|
Abstract
The lack of preclinical models of spontaneous ovarian cancer (OVCA), a fatal gynecological malignancy, is a significant barrier to generating information on early changes indicative of OVCA. In contrast to rodents, laying hens develop OVCA spontaneously, with remarkable similarities to OVCA in women regarding tumor histology, OVCA dissemination, immune responses, and risk factors. These important features of OVCA will be useful to develop an early detection test for OVCA, which would significantly reduce mortality rates; preventive strategies; immunotherapeutics; prevention of resistance to chemotherapeutics; and exploration of gene therapies. A transvaginal ultrasound (TVUS) imaging method for imaging of hen ovarian tumors has been developed. Hens can be monitored prospectively by using serum markers, together with TVUS imaging, to detect early-stage OVCA, provided that a panel of serum markers can be established and imaging agents developed. Recent sequencing of the chicken genome will further facilitate the hen model to explore gene therapies against OVCA.
Collapse
Affiliation(s)
- Animesh Barua
- Laboratory of Translational Research on Ovarian Cancer, Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA;
| | - Janice M Bahr
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA;
| |
Collapse
|
3
|
Pal P, Starkweather KN, Hales KH, Hales DB. A Review of Principal Studies on the Development and Treatment of Epithelial Ovarian Cancer in the Laying Hen Gallus gallus. Comp Med 2021; 71:271-284. [PMID: 34325771 DOI: 10.30802/aalas-cm-20-000116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Often referred to as the silent killer, ovarian cancer is the most lethal gynecologic malignancy. This disease rarely shows any physical symptoms until late stages and no known biomarkers are available for early detection. Because ovarian cancer is rarely detected early, the physiology behind the initiation, progression, treatment, and prevention of this disease remains largely unclear. Over the past 2 decades, the laying hen has emerged as a model that naturally develops epithelial ovarian cancer that is both pathologically and histologically similar to that of the human form of the disease. Different molecular signatures found in human ovarian cancer have also been identified in chicken ovarian cancer including increased CA125 and elevated E-cadherin expression, among others. Chemoprevention studies conducted in this model have shown that decreased ovulation and inflammation are associated with decreased incidence of ovarian cancer development. The purpose of this article is to review the major studies performed in laying hen model of ovarian cancer and discuss how these studies shape our current understanding of the pathophysiology, prevention, and treatment of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Purab Pal
- Department of Physiology, Southern Illinois University, Carbondale, Illinois
| | | | - Karen Held Hales
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, Illinois
| | - Dale Buchanan Hales
- Department of Physiology, Southern Illinois University, Carbondale, Illinois; Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Springfield, Illinois;,
| |
Collapse
|
4
|
Ashfaq W, Rehman K, Siddique MI, Khan QAA. Eicosapentaenoic Acid and Docosahexaenoic Acid from Fish Oil and Their Role in Cancer Research. FOOD REVIEWS INTERNATIONAL 2019. [DOI: 10.1080/87559129.2019.1686761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Wardah Ashfaq
- Department of Medicine, Ameer ud Din Medical College, Lahore, Pakistan
| | - Khurram Rehman
- Department of Pharmacy, Forman Christan College (A Chartered University), Lahore, Pakistan
| | - Muhammad Irfan Siddique
- Institute of Pharmaceutical Sciences, University of Veterinary & Animal Sciences, Lahore, Pakistan
| | - Qurrat-Al-Ain Khan
- Institute of Pharmaceutical Sciences, University of Veterinary & Animal Sciences, Lahore, Pakistan
| |
Collapse
|
5
|
Koshiyama M. The Effects of the Dietary and Nutrient Intake on Gynecologic Cancers. Healthcare (Basel) 2019; 7:healthcare7030088. [PMID: 31284691 PMCID: PMC6787610 DOI: 10.3390/healthcare7030088] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/23/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022] Open
Abstract
The contribution of diet to cancer risk has been considered to be higher in advanced countries than in developing countries. In this paper, I review the current issues (a review of the relevant literature), and the effects of the dietary and nutrient intake on three types of gynecologic cancer (cervical, endometrial and ovarian cancers). In cervical cancer, the most important roles of diet/nutrition in relation to cancer are prophylaxis and countermeasures against human papillomavirus (HPV) infection. The main preventive and reductive factors of cervical cancer are antioxidants, such as vitamin A, C, D and E, carotenoids, vegetables and fruits. These antioxidants may have different abilities to intervene in the natural history of diseases associated with HPV infection. For endometrial cancer, the increase in peripheral estrogens as a result of the aromatization of androgens to estrogens in adipose tissue in obese women and insulin resistance are risk factors. Thus, we must mainly take care to avoid the continuous intake of fat energy and sugar. In ovarian cancer, the etiology has not been fully understood. To the best of our knowledge, the long-term consumption of pro-inflammatory foods, including saturated fat, carbohydrates and animal proteins is a risk factor. The intake of acrylamide is also a risk factor for both endometrial and ovarian cancer. Most papers have been epidemiological studies. Thus, further research using in vitro and in vivo approaches is needed to clarify the effects of the dietary and nutrient intake in detail.
Collapse
Affiliation(s)
- Masafumi Koshiyama
- Department of Women's Health, Graduate School of Human Nursing, The University of Shiga Prefecture, Shiga 522-8533, Japan.
| |
Collapse
|
6
|
Khadge S, Thiele GM, Sharp JG, McGuire TR, Klassen LW, Black PN, DiRusso CC, Cook L, Talmadge JE. Long-chain omega-3 polyunsaturated fatty acids decrease mammary tumor growth, multiorgan metastasis and enhance survival. Clin Exp Metastasis 2018; 35:797-818. [PMID: 30327985 DOI: 10.1007/s10585-018-9941-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023]
Abstract
Epidemiological studies show a reduced risk of breast cancer (BC) in women consuming high levels of long-chain (LC) omega-3 (ω-3) fatty acids (FAs) compared with women who consumed low levels. However, the regulatory and mechanistic roles of dietary ω-6 and LC-ω-3 FAs on tumor progression, metastasis and survival are poorly understood. Female BALB/c mice (10-week old) were pair-fed with a diet containing ω-3 or an isocaloric, isolipidic ω-6 diet for 16 weeks prior to the orthotopic implantation of 4T1 mammary tumor cells. Major outcomes studied included: mammary tumor growth, survival analysis, and metastases analyses in multiple organs including pulmonary, hepatic, bone, cardiac, renal, ovarian, and contralateral MG (CMG). The dietary regulation of the tumor microenvironment was evaluated in mice autopsied on day-35 post tumor injection. In mice fed the ω-3 containing diet, there was a significant delay in tumor initiation and prolonged survival relative to the ω-6 diet-fed group. The tumor size on day 35 post tumor injection in the ω-3 group was 50% smaller and the frequencies of pulmonary and bone metastases were significantly lower relative to the ω-6 group. Similarly, the incidence/frequencies and/or size of cardiac, renal, ovarian metastases were significantly lower in mice fed the ω-3 diet. The analyses of the tumor microenvironment showed that tumors in the ω-3 group had significantly lower numbers of proliferating tumor cells (Ki67+)/high power field (HPF), and higher numbers of apoptotic tumor cells (TUNEL+)/HPF, lower neo-vascularization (CD31+ vessels/HPF), infiltration by neutrophil elastase+ cells, and macrophages (F4/80+) relative to the tumors from the ω-6 group. Further, in tumors from the ω-3 diet-fed mice, T-cell infiltration was 102% higher resulting in a neutrophil to T-lymphocyte ratio (NLR) that was 76% lower (p < 0.05). Direct correlations were observed between NLR with tumor size and T-cell infiltration with the number of apoptotic tumor cells. qRT-PCR analysis revealed that tumor IL10 mRNA levels were significantly higher (six-fold) in the tumors from mice fed the ω-3 diet and inversely correlated with the tumor size. Our data suggest that dietary LC-ω-3FAs modulates the mammary tumor microenvironment slowing tumor growth, and reducing metastases to both common and less preferential organs resulting in prolonged survival. The surrogate analyses undertaken support a mechanism of action by dietary LC-ω-3FAs that includes, but is not limited to decreased infiltration by myeloid cells (neutrophils and macrophages), an increase in CD3+ lymphocyte infiltration and IL10 associated anti-inflammatory activity.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA
| | - Geoffrey M Thiele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA.,Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - John Graham Sharp
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Timothy R McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lynell W Klassen
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA.,Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Paul N Black
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Concetta C DiRusso
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Leah Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA
| | - James E Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA. .,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA.
| |
Collapse
|
7
|
Shahnazi M, Mohammadi M, Mohaddes G, Latifi Z, Ghasemnejad T, Nouri M, Fattahi A. Dietary omega-3 and -6 fatty acids affect the expression of prostaglandin E2 synthesis enzymes and receptors in mice uteri during the window of pre-implantation. Biochem Biophys Res Commun 2018; 503:1754-1760. [DOI: 10.1016/j.bbrc.2018.07.109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 07/22/2018] [Indexed: 02/06/2023]
|
8
|
Preclinical Models of Ovarian Cancer: Pathogenesis, Problems, and Implications for Prevention. Clin Obstet Gynecol 2018; 60:789-800. [PMID: 28719396 DOI: 10.1097/grf.0000000000000312] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Preclinical models are relatively underutilized and underfunded resources for modeling the pathogenesis and prevention of ovarian cancers. Several reviews have detailed the numerous published models of ovarian cancer. In this review, we will provide an overview of experimental model systems, their strengths and limitations, and use selected models to illustrate how they can be used to address specific issues about ovarian cancer pathogenesis. We will then highlight some of the preclinical prevention studies performed to date and discuss experiments needed to address important unanswered questions about ovarian cancer prevention strategies.
Collapse
|
9
|
Elhamouly M, Isobe N, Yoshimura Y. Expression and localization of cyclooxygenases in the oviduct of laying hens during the ovulatory cycle. Theriogenology 2017; 101:1-7. [DOI: 10.1016/j.theriogenology.2017.06.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/06/2017] [Accepted: 06/10/2017] [Indexed: 11/28/2022]
|
10
|
Zhao J, Li Y, Gao J, De Y. Hesperidin inhibits ovarian cancer cell viability through endoplasmic reticulum stress signaling pathways. Oncol Lett 2017; 14:5569-5574. [PMID: 29142606 DOI: 10.3892/ol.2017.6873] [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] [Received: 05/22/2016] [Accepted: 03/30/2017] [Indexed: 12/14/2022] Open
Abstract
Hesperidin is a vitamin P flavonoid compound primarily present in citrus fruits. The aim of the present study was to investigate whether hesperidin inhibits ovarian cancer cell viability via endoplasmic reticulum stress signaling pathways. A2780 cells were treated with various doses of hesperidin for 6, 12 or 24 h, and the viability of A2780 cells was assessed using the MTT assay. Hesperidin decreased the viability of A2780 cells and increased cytotoxicity in a dose- and time-dependent manner. In addition, hesperidin induced apoptosis and increased cleaved caspase-3 protein expression levels in A2780 cells. Furthermore, hesperidin markedly increased the protein expression of anti-growth arrest- and DNA damage-inducible gene 153, anti-CCAAT'enhancer-binding protein homologous protein, glucose-regulated protein 78 and cytochrome c in A2780 cells. The results of the present study indicated that hesperidin inhibits cell viability and induces apoptosis in ovarian cancer cells via endoplasmic reticulum stress signaling pathways. Thus, hesperidin may offer a novel therapeutic tool for ovarian carcinoma.
Collapse
Affiliation(s)
- Jun Zhao
- Department for Gynaecology and Obstetrics, General Hospital of People's Liberation Army, Beijing 100053, P.R. China
| | - Yali Li
- Department for Gynaecology and Obstetrics, General Hospital of People's Liberation Army, Beijing 100053, P.R. China
| | - Jinfang Gao
- Department for Gynaecology and Obstetrics, Navy General Hospital, Beijing 100048, P.R. China
| | - Yinshan De
- Department for Gynaecology and Obstetrics, Navy General Hospital, Beijing 100048, P.R. China
| |
Collapse
|
11
|
Dikshit A, Hales K, Hales DB. Whole flaxseed diet alters estrogen metabolism to promote 2-methoxtestradiol-induced apoptosis in hen ovarian cancer. J Nutr Biochem 2017; 42:117-125. [PMID: 28178600 PMCID: PMC5360509 DOI: 10.1016/j.jnutbio.2017.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 01/12/2017] [Accepted: 01/14/2017] [Indexed: 12/12/2022]
Abstract
The study reported here demonstrates that a flaxseed-supplemented diet causes ovarian tumors in the laying hen to undergo apoptosis, resulting in a reduction of tumor burden, reducing the frequency and severity of ovarian cancer. We have previously shown in normal ovaries that flaxseed and its components down-regulate ERalpha and alter the expression of enzymes that metabolize estrogen. In this study, we analyzed the effects of the two main components of whole flaxseed, ligan and omega 3 fatty acids on estrogen metabolism and the estrogen receptor in ovarian tumors. ER alpha expression was up-regulated in the ovarian tumors and was not affected by diet. Liver CYP1A1 expression was significantly increased by the whole flaxseed diet with a corresponding increase in 2-methoxyestradiol plasma levels. We also observed increased p38 and ERK 1/2 MAPK activation in the ovary as well as an increase in apoptosis in the tumor epithelium. SMAD 7, a factor involved in the 2-methoxyestradiol-mediated apoptosis pathway was also up-regulated in tumors from the whole flaxseed diet group. 2-methoxyestradiol-induced antitumor effects were further validated by in human ovarian cancer cells. This study details the effect of flaxseed diet on estrogen metabolism and demonstrates the antiovarian cancer effects of 2-methoxyestradiol.
Collapse
Affiliation(s)
- Anushka Dikshit
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, 62901, USA
| | - Karen Hales
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, 62901, USA
| | - Dale Buchanan Hales
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL, 62901, USA.
| |
Collapse
|
12
|
Abstract
This article is a combination of an autobiography and a review of outstanding research done by over 70 graduate students, postdoctoral fellows, and visiting scientists along with excellent collaborators during my over-40-year career as a professor of reproductive physiology at the University of Illinois, Urbana-Champaign. I have also shared thoughts on mentoring, how research has changed over the years, and the future of reproductive physiology. I provide the reader with a snapshot of the challenges faced by a woman eager to obtain a PhD under the guidance of renowned professors in the early 1970s and to be hired as the first woman, and the only permanent female faculty member, for more than 20 years on a faculty of 40 men. As a comparative reproductive physiologist, I describe the various animal models used because they were the best models to answer specific questions in reproduction. Also, my graduate students and postdoctoral fellows were given the freedom to identify their research topics, articulate hypotheses to be tested, and select appropriate animal models. This approach caused students to take ownership of their research, resulting in the development of independent and creative scientists and over 170 publications, excluding chapters in top-tier journals. Finally, I am so grateful for a truly rich life mentoring graduate students and postdoctoral fellows who have become my lifelong friends.
Collapse
Affiliation(s)
- Janice M Bahr
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, Illinois 61801.,Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Illinois 61801;
| |
Collapse
|
13
|
Dikshit A, Gao C, Small C, Hales K, Hales DB. Flaxseed and its components differentially affect estrogen targets in pre-neoplastic hen ovaries. J Steroid Biochem Mol Biol 2016; 159:73-85. [PMID: 26925929 PMCID: PMC4821676 DOI: 10.1016/j.jsbmb.2016.02.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/12/2016] [Accepted: 02/24/2016] [Indexed: 02/07/2023]
Abstract
Flaxseed has been studied for decades for its health benefits that include anti-cancer, cardio-protective, anti-diabetic, anti-inflammatory properties. The biologically active components that mediate these effects are the omega-3 fatty acids and the lignan, secoisolariciresinol diglucoside. We have previously shown that whole flaxseed supplemented diet decreases the severity and incidence of ovarian cancer while a 15% dose of flaxseed is most protective against inflammation and estrogen-induced chemical and genotoxicity. The objective of this study was to dissect the independent effects of the two flaxseed components on estrogen signaling and metabolism. Two and half year old hens were fed either a control diet, 15% whole flaxseed diet, defatted flax meal diet or 5% flax oil diet for 3 months after which the animals were sacrificed and blood and tissues were harvested. Whole flaxseed diet caused a decrease in expression of ERα. ERα target gene expression was assessed using RT(2) profiler PCR array. Some targets involved in the IGF/insulin signaling pathway (IRS1, IGFBP4, IGFBP5) were downregulated by flaxseed and its components. Flaxseed diet also downregulated AKT expression. A number of targets related to NF-kB signaling were altered by flaxseed diet including a series of targets implicated in cancer. Whole flaxseed diet also affected E2 metabolism by increasing CYP1A1 expression with a corresponding increase in the onco-protective E2 metabolite, 2-methoxyestradiol. The weak anti-estrogens, enterolactone, enterodiol and 2-methoxyestradiol, might be working synergistically to generate a protective effect on the ovaries from hens on whole flaxseed diet by altering the estrogen signaling and metabolism.
Collapse
Affiliation(s)
- Anushka Dikshit
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA
| | - Chunqi Gao
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA
| | - Carrie Small
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA
| | - Karen Hales
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Carbondale, Illinois, USA
| | - Dale Buchanan Hales
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA.
| |
Collapse
|
14
|
Dikshit A, Filho MAG, Eilati E, McGee S, Small C, Gao C, Klug T, Hales DB. Flaxseed reduces the pro-carcinogenic micro-environment in the ovaries of normal hens by altering the PG and oestrogen pathways in a dose-dependent manner. Br J Nutr 2015; 113:1384-95. [PMID: 25850566 PMCID: PMC4445837 DOI: 10.1017/s000711451500029x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The objective of the present study was to find the optimum dose of flaxseed that would decrease PG and alter oestrogen pathway endpoints implicated in ovarian cancer. In the study, four groups of fifty 1.5-year-old chickens were fed different amounts of flaxseed (0, 5, 10 or 15% of their total diet) for 4 months and were then killed to collect blood and tissues. Levels of flaxseed lignan metabolites, Enterolactone (EL) and Enterodiol (ED) were measured in the serum, liver and ovaries by liquid chromatography-MS/MS, and n-3 and n-6 fatty acid (FA) levels were measured by GC. The effects of the varied flaxseed doses were assessed by measuring levels of PGE2 and oestrogen metabolites (16-hydroxyestrone (16-OHE1) and 2-hydroxyestrone (2-OHE1)) as well as by analysing the expression of the oestradiol metabolising enzymes CYP3A4 (cytochrome p450, family 3, subfamily A, polypeptide 4), CYP1B1 (cytochrome p450, family 1, subfamily B, polypeptide 1) and CYP1A1 (cytochrome p450, family 1, subfamily A, polypeptide 1) and that of oestrogen receptor α (ERα) in the ovaries. The ratio of n-3:n-FA increased with an increase in flaxseed supplementation and corresponded to a dose-dependent decrease in cyclo-oxygenase-2 protein and PGE2 levels. EL and ED increased in the serum, liver and ovaries with increased concentrations of flaxseed. Flaxseed decreased the expression of ERα in the ovaries. The ratio of 2-OHE1:16-OHE1 in the serum increased significantly in the 15% flaxseed diet, and there was a corresponding increase in CYP1A1 in the liver and decrease in CYP3A4 in the ovaries. CYP1B1 mRNA also decreased with flaxseed diet in the ovaries. The 15% flaxseed-supplemented diet significantly decreased inflammatory PGE2, ERα, CYP3A4, CYP1B1 and 16-OHE1, but it increased CYP1A1 and 2-OHE1, which thus reduced the inflammatory and pro-carcinogenic micro-environment of the ovaries.
Collapse
Affiliation(s)
- Anushka Dikshit
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL 62901, USA
| | | | - Erfan Eilati
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL 62901, USA
| | - Stacey McGee
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL 62901, USA
| | - Carrie Small
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL 62901, USA
| | - Chunqi Gao
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL 62901, USA
| | | | - Dale Buchanan Hales
- Department of Physiology, Southern Illinois School of Medicine, Carbondale, IL 62901, USA
| |
Collapse
|
15
|
Abstract
The complexity and heterogeneity of ovarian cancer cases are difficult to reproduce in in vitro studies, which cannot adequately elucidate the molecular events involved in tumor initiation and disease metastasis. It has now become clear that, although the multiple histological subtypes of ovarian cancer are being treated with similar surgical and therapeutic approaches, they are in fact characterized by distinct phenotypes, cell of origin, and underlying key genetic and genomic alterations. Consequently, the development of more personalized treatment methodologies, which are aimed at improving patient care and prognosis, will greatly benefit from a better understanding of the key differences between various subtypes. To accomplish this, animal models of all histotypes need to be generated in order to provide accurate in vivo platforms for research and the testing of targeted treatments and immune therapies. Both genetically engineered mouse models (GEMMs) and xenograft models have the ability to further our understanding of key mechanisms facilitating tumorigenesis, and at the same time offer insight into enhanced imaging and treatment modalities. While genetic models may be better suited to examine oncogenic functions and interactions during tumorigenesis, patient-derived xenografts (PDXs) are likely a superior model to assess drug efficacy, especially in concurrent clinical trials, due to their similarity to the tumors from which they are derived. Genetic and avatar models possess great clinical utility and have both benefits and limitations. Additionally, the laying hen model, which spontaneously develops ovarian tumors, has inherent advantages for the study of epithelial ovarian cancer (EOC) and recent work champions this model especially when assessing chemoprevention strategies. While high-grade ovarian serous tumors are the most prevalent form of EOC, rarer ovarian cancer variants, such as small cell ovarian carcinoma of the hypercalcemic type and transitional cell carcinoma, or non-epithelial tumors, including germ cell tumors, will also benefit from the generation of improved models to advance our understanding of tumorigenic mechanisms and the development of selective therapeutic options.
Collapse
Affiliation(s)
- Noor Hasan
- Department of Pathology, Division of Women's and Perinatal Pathology, Eugene Braunwald Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anders W Ohman
- Department of Pathology, Division of Women's and Perinatal Pathology, Eugene Braunwald Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Daniela M Dinulescu
- Department of Pathology, Division of Women's and Perinatal Pathology, Eugene Braunwald Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
16
|
Eilati E, Small CC, McGee SR, Kurrey NK, Hales DB. Anti-inflammatory effects of fish oil in ovaries of laying hens target prostaglandin pathways. Lipids Health Dis 2013; 12:152. [PMID: 24156238 PMCID: PMC3874764 DOI: 10.1186/1476-511x-12-152] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 10/18/2013] [Indexed: 12/17/2022] Open
Abstract
Background An effective way to control cancer is by prevention. Ovarian cancer is the most lethal gynecological malignancy. Progress in the treatment and prevention of ovarian cancer has been hampered due to the lack of an appropriate animal model and absence of effective chemo-prevention strategies. The domestic hens spontaneously develop ovarian adenocarcinomas that share similar histological appearance and symptoms such as ascites and metastasis with humans. There is a link between chronic inflammation and cancer. Prostaglandin E2 (PGE2) is the most pro-inflammatory ecoisanoid and one of the downstream products of two isoforms of cyclooxygenase (COX) enzymes: COX-1 and COX-2. PGE2 exerts its effects on target cells by coupling to four subtypes of receptors which have been classified as EP1-4. Fish oil is a source of omega-3 fatty acids (OM-3FAs) which may be effective in prevention of ovarian cancer. Our objective was to assess the potential impact of fish oil on expression of COX enzymes, PGE2 concentration, apoptosis and proliferation in ovaries of laying hens. Methods 48 white Leghorn hens were fed 50, 100, 175, 375 and 700 mg/kg fish oil for 21 days. The OM3-FAs and omega-6 fatty acids contents of egg yolks were determined by Gas Chromatography. Proliferation, apoptosis, COX-1, COX-2 and prostaglandin receptor subtype 4 (EP4) protein and mRNA expression and PGE2 concentration in ovaries were measured by PCNA, TUNEL, Western blot, quantitative real-time qPCR and ELISA, respectively. Results Consumption of fish oil increased the incorporation of OM-3FAs into yolks and decreased both COX-1 and COX-2 protein and mRNA expression. In correlation with COXs down-regulation, fish oil significantly reduced the concentrations of PGE2 in ovaries. EP4 protein and mRNA expression in ovaries of hens was not affected by fish oil treatment. A lower dose of fish oil increased the egg laying frequency. 175 and 700 mg/kg fish oil reduced proliferation and 700 mg/kg increased apoptosis in hen ovaries. Conclusions Our findings suggest that the lower doses of fish oil reduce inflammatory PG and may be an effective approach in preventing ovarian carcinogenesis. These findings may provide the basis for clinical trials utilizing fish oil as a dietary intervention targeting prostaglandin biosynthesis for the prevention and treatment of ovarian cancer.
Collapse
Affiliation(s)
| | | | | | | | - Dale Buchanan Hales
- Department of Physiology, Southern Illinois University Carbondale, School of Medicine, Life Science II, Room 250 (M/C 6512), 1125 Lincoln Drive, Carbondale, IL 62901, USA.
| |
Collapse
|