1
|
Karmokar PF, Moniri NH. Oncogenic signaling of the free-fatty acid receptors FFA1 and FFA4 in human breast carcinoma cells. Biochem Pharmacol 2022; 206:115328. [PMID: 36309079 DOI: 10.1016/j.bcp.2022.115328] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022]
Abstract
Globally, breast cancer is the most frequent type of cancer in women, and most breast cancer-associated deaths are due to metastasis and recurrence of the disease. Dietary habits, specifically dietary fat intake is a crucial risk factor involved in breast cancer development and progression. Decades of research has revealed that free-fatty acids (FFA) modulate carcinogenic processes through fatty acid metabolism and lipid peroxidation. The ground-breaking discovery of free-fatty acid receptors, which are members of the G-protein coupled receptor (GPCR) superfamily, has led to the realization that FFA can also act via these receptors to modulate carcinogenic effects. The long-chain free-fatty acid receptors FFA1 (previously termed GPR40) and FFA4 (previously termed GPR120) are activated by mono- and polyunsaturated fatty acids including ω-3, 6, and 9 fatty acids. Initial enthusiasm towards the study of these receptors focused on their insulin secretagogue and sensitization effects, and the downstream associated metabolic regulation. However, recent studies have demonstrated that abnormal expression and/or aberrant FFA1/FFA4 signaling are evident in human breast carcinomas, suggesting that FFA receptors could be a promising target in the treatment of breast cancer. The current review discusses the diverse roles of FFA1 and FFA4 in the regulation of cell proliferation, migration, invasion, and chemotherapy resistance in human breast carcinoma cells and tissue.
Collapse
Affiliation(s)
- Priyanka F Karmokar
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, GA 30341, USA
| | - Nader H Moniri
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, GA 30341, USA; Department of Biomedical Sciences, School of Medicine, Mercer University Health Sciences Center, Mercer University, Macon, GA 31207, USA.
| |
Collapse
|
2
|
Wajszczyk B, Charzewska J, Godlewski D, Zemła B, Nowakowska E, Kozaczka M, Chilimoniuk M, Pathak DR. Consumption of Dairy Products and the Risk of Developing Breast Cancer in Polish Women. Nutrients 2021; 13:nu13124420. [PMID: 34959971 PMCID: PMC8703752 DOI: 10.3390/nu13124420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Lack of consistency in the relationship between dairy products consumption and breast cancer (BC) risk motivated us to evaluate this association in a case-control study of BC among Polish women. The study includes 1699 women 26–79 years of age, 823 BC cases identified in Cancer Registries and 876 randomly selected controls from the national population registry. Using a validated, semiquantitative food frequency questionnaire (FFQ), the consumption of dairy products was collected for a time period of 10–15 years prior to BC diagnosis. We used logistic regression, adjusting for potential confounders, to assess the relationship between total dairy consumption as well as individual dairy groups of milk, cottage cheese and hard cheese and BC risk for premenopausal and postmenopausal women. For total consumption, a significant decrease in BC risk was observed with increased consumption of one serving/week, OR trend = 0.98, 2% decrease in risk, for premenopausal women only. For milk, a significant decrease in BC risk was observed for an increase in consumption of one glass/week, OR trend = 0.95, 5% decrease, in both strata of menopause. In contrast, for hard cheese, a significant increase in the risk of 10% was observed only in premenopausal women, OR trend = 1.10. Cottage cheese consumption significantly reduced BC risk by 20%, OR trend = 0.80, for an increase in one serving/week for postmenopausal women only. Our results show that individual dairy products have a statistically significant but bi-directional relationship with BC risk, which differs for premenopausal and postmenopausal women.
Collapse
Affiliation(s)
- Bożena Wajszczyk
- Department of Nutrition and Nutritional Value of Food, National Institute of Public Health NIH-National Research Institute, 00-791 Warszawa, Poland
- Correspondence: (B.W.); (J.C.); (D.R.P.)
| | - Jadwiga Charzewska
- Department of Nutrition and Nutritional Value of Food, National Institute of Public Health NIH-National Research Institute, 00-791 Warszawa, Poland
- Correspondence: (B.W.); (J.C.); (D.R.P.)
| | - Dariusz Godlewski
- Center of Cancer Prevention and Epidemiology OPEN, 61-863 Poznań, Poland;
| | | | | | - Maciej Kozaczka
- II Clinic of Radiology and Chemiotherapy, National Institute of Oncology, 44-102 Gliwice, Poland;
| | | | - Dorothy R. Pathak
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
- Correspondence: (B.W.); (J.C.); (D.R.P.)
| |
Collapse
|
3
|
Monk JM, Liddle DM, Hutchinson AL, Burns JL, Wellings H, Cartwright NM, Muller WJ, Power KA, Robinson LE, Ma DWL. Fish oil supplementation increases expression of mammary tumor apoptosis mediators and reduces inflammation in an obesity-associated HER-2 breast cancer model. J Nutr Biochem 2021; 95:108763. [PMID: 33965532 DOI: 10.1016/j.jnutbio.2021.108763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 03/10/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022]
Abstract
Obesity is associated with inflammation and has been shown to increase breast cancer severity. The objective of this study was to examine the effect of fish oil (FO) supplementation in obesity-associated mammary tumorigenesis in the MMTV-neu(ndl)-YD5 mouse model of human epidermal growth factor receptor-2 positive BC. Female mice were fed one of three diets for 16 weeks: i) high fat diet [HF, % kacl: 41.2% lard, 18.7% corn oil (CO)], ii) an isocaloric HF plus menhaden FO diet (HF+FO, % kcal: 41.2 lard, 13.4% CO, 5.3% FO), iii) low fat diet (LF, % kcal: 4.7% lard, 6% CO). HF mice had increased body weight, visceral adipose weight and serum hormone concentrations (increased leptin and resistin; decreased adiponectin) versus LF, which was attenuated in the HF+FO group versus HF (P<.05). Compared to HF, tumor onset was delayed in HF+FO and LF mice (P<0.05). Compared to HF, HF+FO reduced mammary tumor multiplicity (-27%), tumor weight (-46%) and total tumor volume (-50%) (P<0.05). Additionally, HF+FO reduced mammary tumor multiplicity (-33%), tumor weight (-39%) and total tumor volume (-60%) versus LF. HF+FO improved mammary tumor apoptosis status with increased expression of pro-apoptotic Bad and decreased expression of anti-apoptotic Bcl-xLmediators versus HF (P<0.05). Additionally, HF+FO decreased tumor protein expression of activated Akt, NFκB p65 and STAT3, versus HF (P<0.05). Tumor mRNA expression of inflammatory mediators TNFα, IL-6 and leptin were reduced in HF+FO, whereas IL-10 expression was increased compared to HF (P<0.05). Collectively these results demonstrate the efficacy of FO supplementation for improving obesity-associated breast cancer outcomes.
Collapse
Affiliation(s)
- Jennifer M Monk
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1.
| | - Danyelle M Liddle
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - Amber L Hutchinson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - Jessie L Burns
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - Hannah Wellings
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - Nadia M Cartwright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - William J Muller
- Department of Biochemistry, McGill University, Rosalind and Morris Goodman Cancer Research, Montreal, QC, Canada
| | - Krista A Power
- School of Nutrition Sciences, University of Ottawa, Ottawa ON, Canada, K1H 8L1
| | - Lindsay E Robinson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1
| | - David W L Ma
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON, Canada, N1G 2W1.
| |
Collapse
|
4
|
Wilkin AM, Sullivan R, Trinh T, Edson M, Kozlowski B, Meckling KA. Differential effects of the 1,25D3-MARRS receptor (ERp57/PDIA3) on murine mammary gland development depend on the vitamin D3 dose. Steroids 2020; 158:108621. [PMID: 32119872 DOI: 10.1016/j.steroids.2020.108621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/23/2020] [Accepted: 02/26/2020] [Indexed: 12/19/2022]
Abstract
1,25 dihydroxyvitamin D3 (1,25D3) is the most potent biologically active form of vitamin D3. Its actions on the mammary gland include cell growth inhibition and anti-cancer effects. This study's purpose was to explore the role of the 1,25D3-membrane associated rapid response steroid (MARRS) receptor in the mammary gland using a tissue-specific knockout mouse model and a vitamin D3 dietary intervention. Three genotype groups were created using the Cre/loxp system to knock-down (+/-) and knockout (-/-) the MARRS receptor in epithelial cells of mammary glands (MG). Abdominal MGs were collected from 6-week old female mice (n = 94) on diets of 10,000 IU/kg (excess), 1,000 IU/kg (sufficient) or 0 IU/kg (deficient) of D3. There was a significant interaction between genotype and diet regarding number of terminal end buds (TEBs) (p = 0.001) and ductal coverage of the fat pad (p = 0.03). MARRS -/- mice on the sufficient diet had significantly fewer TEBs (p = 0.001) compared to MARRS +/+ on the same diet, but the opposite effect was seen in mice on the excess diet. There were no effects of genotype on TEBs when animals were vitamin D3 deficient. These results suggest that there is an effect of MARRS on mammary gland development that is dependent on 25(OH)D status, specifically, altering the number of highly proliferative TEBs. Increased numbers of TEBs have been correlated with increased breast cancer risk later in life. Therefore the results of this study warrant further examination of 25(OH)D status and recommendations in adolescent humans to reduce dietary effects on future breast cancer risk.
Collapse
Affiliation(s)
- Allison M Wilkin
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| | - Robert Sullivan
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| | - Thao Trinh
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| | - Michael Edson
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| | - Benjamin Kozlowski
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| | - Kelly A Meckling
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd. E. Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
5
|
Krisanits B, Randise JF, Burton CE, Findlay VJ, Turner DP. Pubertal mammary development as a "susceptibility window" for breast cancer disparity. Adv Cancer Res 2020; 146:57-82. [PMID: 32241392 PMCID: PMC10084741 DOI: 10.1016/bs.acr.2020.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Factors such as socioeconomic status, age at menarche and childbearing patterns are components that have been shown to influence mammary gland development and establish breast cancer disparity. Pubertal mammary gland development is selected as the focus of this review, as it is identified as a "window of susceptibility" for breast cancer risk and disparity. Here we recognize non-Hispanic White, African American, and Asian American women as the focus of breast cancer disparity, in conjunction with diets associated with changes in breast cancer risk. Diets consisting of high fat, N-3 polyunsaturated fatty acids, N-6 polyunsaturated fatty acids, as well as obesity and the Western diet have shown to lead to changes in pubertal mammary gland development in mammalian models, therefore increasing the risk of breast cancer and breast cancer disparity. While limited intervention strategies are offered to adolescents to mitigate development changes and breast cancer risk, the prominent solution to closing the disparity among the selected population is to foster lifestyle changes that avoid the deleterious effects of unhealthy diets.
Collapse
Affiliation(s)
- Bradley Krisanits
- Department of Pathology & Laboratory Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Jaime F Randise
- Department of Pathology & Laboratory Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Clare E Burton
- Department of Pathology & Laboratory Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Victoria J Findlay
- Department of Pathology & Laboratory Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - David P Turner
- Department of Pathology & Laboratory Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
6
|
Lifelong n-3 Polyunsaturated Fatty Acid Exposure Modulates Size of Mammary Epithelial Cell Populations and Expression of Caveolae Resident Proteins in Fat-1 Mice. Nutrients 2019; 11:nu11102477. [PMID: 31619022 PMCID: PMC6835466 DOI: 10.3390/nu11102477] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/30/2019] [Accepted: 10/05/2019] [Indexed: 01/26/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFA) have been associated with reduced breast cancer risk; however, the exact mechanism remains elusive. Female wildtype (WT) and fat-1 mice were fed a 10% safflower diet until 6 weeks of age. Mammary gland epithelial cells (EC) were isolated and EC populations were determined by CD24 surface expression. Fat-1 mice expressed 65%, 20%, and 15% while WT mice expressed 65%, 26% and 9% for non-, myo- and luminal ECs, respectively. The luminal EC population was significantly greater in fat-1 mice (p ≤ 0.05), while the total number of mammary ECs were similar between groups (p = 0.79). Caveolae was isolated from ECs and Her-2/neu, ER-α and cav-1 protein expression was determined by Western blotting. Fat-1 mice had a two-fold greater ER-α (p ≤ 0.05) and a 1.5-fold greater cav-1 (p ≤ 0.05) expression than WT with a similar amount of Her-2/neu protein (p = 0.990) between groups. Overall, this study provides novel mechanistic evidence by which n-3 PUFA modifies early mammary gland development that may potentially reduce breast cancer risk later in life.
Collapse
|
7
|
Li DD, Ling SC, Wu K, Luo Z. Identification of Five Key Genes Involved in Intrinsic Apoptotic Pathway From Yellow Catfish Pelteobagrus fulvidraco and Their Transcriptional Responses to High Fat Diet (HFD). Front Physiol 2019; 10:921. [PMID: 31427980 PMCID: PMC6687843 DOI: 10.3389/fphys.2019.00921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/08/2019] [Indexed: 11/28/2022] Open
Abstract
The hypothesis of the present study is that apoptosis through an intrinsic mitochondrial pathway may mediate high fat diet (HFD)-induced changes in the metabolism of Pelteobagrus fulvidraco. To this end, we cloned the full-length cDNA sequences of Cycs, Apaf1, Casp9, Casp3a, and Casp3b involved in the mitochondria apoptotic pathway, and explored their mRNA tissue expressions and transcriptional responses to HFD. All of these members shared similar domains to their orthologous vertebrate genes. They were constitutively expressed in all analyzed tissues but varied from tissue to tissue. Compared to the control, HFD up-regulated the mRNA expression of partial genes among these five key genes (Cycs, Apaf1, Casp9, Casp3a, and Casp3b) in mesenteric fat, intestine, ovary and the kidney, indicating the induction of apoptosis in these tissues; in contrast, HFD down-regulated mRNA levels of partial genes among the five key genes (Cycs, Apaf1, Casp9, Casp3a, and Casp3b) in the heart, spleen and gill tissues, indicating the inhibition of apoptosis in these tissues. The present study will facilitate further exploration into the functions of these genes at the molecular level and disclose the critical involvement of these genes against nutrient changes, indicating that processes of apoptosis in various tissues may differentially be modified by HFD.
Collapse
Affiliation(s)
- Dan-Dan Li
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Shi-Cheng Ling
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Kun Wu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China
| | - Zhi Luo
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
8
|
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
|
9
|
Grassi TF, Bidinotto LT, Lopes GAD, Zapaterini JR, Rodrigues MAM, Barbisan LF. Maternal western-style diet enhances the effects of chemically-induced mammary tumors in female rat offspring through transcriptome changes. Nutr Res 2018; 61:41-52. [PMID: 30683438 DOI: 10.1016/j.nutres.2018.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 09/06/2018] [Accepted: 09/27/2018] [Indexed: 01/09/2023]
Abstract
Previous studies have shown that early life intake of high-fat diet or western-style diet (WD) enhances the development of mammary tumors in adult female rats. Thus, we hypothesized that maternal WD throughout pregnancy and the lactation period could speed up the development of MNU-induced mammary tumors and alter their gene expression. For this, the present study investigated the gene expression profile of chemically-induced mammary tumors in female rat offspring from dams fed a WD or a control diet. Pregnant female Sprague-Dawley rats received a WD (high-fat, low-fiber and oligoelements) or a control diet from gestational day 12 until post-natal day (PND) 21. At PND 21, female offspring received a single dose of N-Methyl-N-Nitrosourea (MNU, 50 mg/kg body weight) and were fed a control diet for 13 weeks. Tumor incidence, multiplicity, and latency were recorded and mammary gland samples were collected for histopathology and gene expression analysis. Tumor multiplicity and histological grade were significantly higher and tumor latency was lower in WD offspring compared to control offspring. Transcriptome profiling identified 57 differentially expressed genes in tumors from WD offspring as compared to control offspring. There was also an increase in mRNA expression of genes such as Emp3, Ccl7, Ets1, Abcc5, and Cyr61, indicative of more aggressive disease detected in tumors from WD offspring. Thus, maternal WD diet increased MNU-induced mammary carcinogenesis in adult female offspring through transcriptome changes that resulted in a more aggressive disease.
Collapse
Affiliation(s)
- Tony F Grassi
- UNESP - Univ. Estadual Paulista, Botucatu Medical School, Department of Pathology, Botucatu, 18610-307, SP, Brazil; UNESP - Univ. Estadual Paulista, Institute of Biosciences of Botucatu, Department of Morphology, Botucatu 18618-689, SP, Brazil
| | - Lucas T Bidinotto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, SP, Brazil; Barretos School of Health Sciences, Dr. Paulo Prata -FACISB, Barretos 14785-002, SP, Brazil
| | - Gisele A D Lopes
- UNESP - Univ. Estadual Paulista, Botucatu Medical School, Department of Pathology, Botucatu, 18610-307, SP, Brazil
| | - Joyce R Zapaterini
- UNESP - Univ. Estadual Paulista, Botucatu Medical School, Department of Pathology, Botucatu, 18610-307, SP, Brazil; UNESP - Univ. Estadual Paulista, Institute of Biosciences of Botucatu, Department of Morphology, Botucatu 18618-689, SP, Brazil
| | - Maria A M Rodrigues
- UNESP - Univ. Estadual Paulista, Botucatu Medical School, Department of Pathology, Botucatu, 18610-307, SP, Brazil
| | - Luís F Barbisan
- UNESP - Univ. Estadual Paulista, Institute of Biosciences of Botucatu, Department of Morphology, Botucatu 18618-689, SP, Brazil.
| |
Collapse
|
10
|
Strategies to reduce non-communicable diseases in the offspring: negative and positive in utero programming. J Dev Orig Health Dis 2018; 9:642-652. [PMID: 30111388 DOI: 10.1017/s2040174418000569] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Non-communicable diseases (NCDs) are a major problem as they are the leading cause of death and represent a substantial economic cost. The 'Developmental Origins of Health and Disease Hypothesis' proposes that adverse stimuli at different life stages can increase the predisposition to these diseases. In fact, adverse in utero programming is a major origin of these diseases due to the high malleability of embryonic development. This review provides a comprehensive analysis of the scientific literature on in utero programming and NCDs highlighting potential medical strategies to prevent these diseases based upon this programming. We fully address the concept and mechanisms involved in this programming (anatomical disruptions, epigenetic modifications and microbiota alterations). We also examine the negative role of in utero programming on the increased predisposition of NCDs in the offspring, which introduces the passive medical approach that consists of avoiding adverse stimuli including an unhealthy diet and environmental chemicals. Finally, we extensively discuss active medical approaches that target the causes of NCDs and have the potential to significantly and rapidly reduce the incidence of NCDs. These approaches can be classified as direct in utero programming modifications and personalized lifestyle pregnancy programs; they could potentially provide transgenerational NCDs protection. Active strategies against NCDs constitute a promising tool for the reduction in NCDs.
Collapse
|
11
|
Wilkin AM, Harnett A, Underschultz M, Cragg C, Meckling KA. Role of the ERp57 protein (1,25D3-MARRS receptor) in murine mammary gland growth and development. Steroids 2018; 135:63-68. [PMID: 29477346 DOI: 10.1016/j.steroids.2018.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/09/2018] [Accepted: 02/19/2018] [Indexed: 12/13/2022]
Abstract
The protein disulfide isomerase ERp57 (GRp58/PDIA3/1,25D3-MARRS) has been implicated in a multitude of signaling pathways throughout the entire body. Most thoroughly studied for its protein-folding role, ERp57 has also been found to have multiple binding partners, and have significant effects on cellular growth. ERp57 has been studied n the context of several neurodegenerative disorders, metabolic conditions, and can be used as a prognosis marker in certain cancers. One role, as an alternate vitamin D binding receptor, has prompted research in tissues with known vitamin D activity, such as the intestine and bone. Vitamin D has been studied in relation to mammary gland growth and development, but it is not yet known if ERp57 plays an independent role in this tissue. In this study, ERp57 was knocked out in murine mammary gland epithelial cells of 30 4-week old mice. Several markers of mammary gland growth were measured, including number of terminal end buds (TEB), ductal coverage of the fat pad, and ductal extension. It was found the knockout animals had decreased numbers of TEBs (p = 0.019), and decreased ductal extension (p = 0.018) compared to wildtype animals, with no differences in gross body weight. Immunohistochemistry analysis of mammary glands showed ERp57 localized to the apical side of alveolar branches, and on leading edges of TEBs. These results provide further evidence for ERp57 functioning separately to the VDR, and further insights into the roles of ERp57.
Collapse
Affiliation(s)
- Allison M Wilkin
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada.
| | - Amber Harnett
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada.
| | - Michael Underschultz
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada
| | - Cheryl Cragg
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada.
| | - Kelly A Meckling
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Rd E, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
12
|
Khadge S, Thiele GM, Sharp JG, McGuire TR, Klassen LW, Black PN, DiRusso CC, Talmadge JE. Long-Chain Omega-3 Polyunsaturated Fatty Acids Modulate Mammary Gland Composition and Inflammation. J Mammary Gland Biol Neoplasia 2018; 23:43-58. [PMID: 29574638 DOI: 10.1007/s10911-018-9391-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/19/2018] [Indexed: 01/07/2023] Open
Abstract
Studies in rodents have shown that dietary modifications as mammary glands (MG) develop, regulates susceptibility to mammary tumor initiation. However, the effects of dietary PUFA composition on MGs in adult life, remains poorly understood. This study investigated morphological alterations and inflammatory microenvironments in the MGs of adult mice fed isocaloric and isolipidic liquid diets with varying compositions of omega (ω)-6 and long-chain (Lc)-ω3FA that were pair-fed. Despite similar consumption levels of the diets, mice fed the ω-3 diet had significantly lower body-weight gains, and abdominal-fat and mammary fat pad (MFP) weights. Fatty acid analysis showed significantly higher levels of Lc-ω-3FAs in the MFPs of mice on the ω-3 diet, while in the MFPs from the ω-6 group, Lc-ω-3FAs were undetectable. Our study revealed that MGs from ω-3 group had a significantly lower ductal end-point density, branching density, an absence of ductal sprouts, a thinner ductal stroma, fewer proliferating epithelial cells and a lower transcription levels of estrogen receptor 1 and amphiregulin. An analysis of the MFP and abdominal-fat showed significantly smaller adipocytes in the ω-3 group, which was accompanied by lower transcription levels of leptin, IGF1, and IGF1R. Further, MFPs from the ω-3 group had significantly decreased numbers and sizes of crown-like-structures (CLS), F4/80+ macrophages and decreased expression of proinflammatory mediators including Ptgs2, IL6, CCL2, TNFα, NFκB, and IFNγ. Together, these results support dietary Lc-ω-3FA regulation of MG structure and density and adipose tissue inflammation with the potential for dietary Lc-ω-3FA to decrease the risk of mammary gland tumor formation.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 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, 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
| | - James E Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
13
|
Zanoaga O, Jurj A, Raduly L, Cojocneanu-Petric R, Fuentes-Mattei E, Wu O, Braicu C, Gherman CD, Berindan-Neagoe I. Implications of dietary ω-3 and ω-6 polyunsaturated fatty acids in breast cancer. Exp Ther Med 2017; 15:1167-1176. [PMID: 29434704 PMCID: PMC5776638 DOI: 10.3892/etm.2017.5515] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/20/2017] [Indexed: 12/20/2022] Open
Abstract
Breast cancer represents one of the most common forms of cancer in women worldwide, with an increase in the number of newly diagnosed patients in the last decade. The role of fatty acids, particularly of a diet rich in ω-3 and ω-6 polyunsaturated fatty acids (PUFAs), in breast cancer development is not fully understood and remains controversial due to their complex mechanism of action. However, a large number of animal models and cell culture studies have demonstrated that high levels of ω-3 PUFAs have an inhibitory role in the development and progression of breast cancer, compared to ω-6 PUFAs. The present review focused on recent studies regarding the correlation between dietary PUFAs and breast cancer development, and aimed to emphasize the main molecular mechanisms involved in the modification of cell membrane structure and function, modulation of signal transduction pathways, gene expression regulation, and antiangiogenic and antimetastatic effects. Furthermore, the anticancer role of ω-3 PUFAs through the modulation of microRNA expression levels was also reviewed.
Collapse
Affiliation(s)
- Oana Zanoaga
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.,Department of Physiopathology, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Roxana Cojocneanu-Petric
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Enrique Fuentes-Mattei
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Oscar Wu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Texas Tech University Honors College, McClellan Hall, Lubbock, TX 79409, USA
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Claudia Diana Gherman
- Surgical Clinic II Hospital, 400006 Cluj-Napoca, Romania.,Department of Surgery, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.,MEDFUTURE-Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, 400349 Cluj-Napoca, Romania.,Department of Functional Genomics, Proteomics and Experimental Pathology, Prof Dr Ion Chiricuta Oncology Institute, 400015 Cluj-Napoca, Romania
| |
Collapse
|
14
|
Fasano E, Serini S, Cittadini A, Calviello G. Long-chain n-3 PUFA against breast and prostate cancer: Which are the appropriate doses for intervention studies in animals and humans? Crit Rev Food Sci Nutr 2017; 57:2245-2262. [PMID: 25897862 DOI: 10.1080/10408398.2013.850060] [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] [Indexed: 01/05/2023]
Abstract
The potential antineoplastic effect of the long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) eicosapentaenoic (EPA) and docosahexaenoic acid (DHA) remains a highly controversial issue. Numerous animal studies have supported the anticancer role of these dietary fatty acids, whereas conflicting results have been obtained in population studies, and only a few intervention human trials have been so far performed. In view of the possibility that the anticancer effects may be maximally observed within a defined range of EPA and DHA doses, herein we critically review the results and doses used in both animal studies and human clinical trials focusing on the possible n-3 PUFA protective effects against breast and prostate cancer. Our main aim is to identify the EPA and/or DHA ranges of doses needed to obtain clear anticancer effects. This may be of great help in designing future animal studies, and also in understanding the most appropriate dose for further human intervention studies. Moreover, since the healthy effects of these fatty acids have been strictly related to their increased incorporation in plasma and tissue lipids, we also examine and discuss the incorporation changes following the administration of the effective anticancer EPA and/or DHA doses in animals and humans.
Collapse
Affiliation(s)
- Elena Fasano
- a Institute of General Pathology, Università Cattolica S. Cuore , Rome , Italy
| | - Simona Serini
- a Institute of General Pathology, Università Cattolica S. Cuore , Rome , Italy
| | - Achille Cittadini
- a Institute of General Pathology, Università Cattolica S. Cuore , Rome , Italy
| | - Gabriella Calviello
- a Institute of General Pathology, Università Cattolica S. Cuore , Rome , Italy
| |
Collapse
|
15
|
Mutagenicity of ω-3 fatty acid peroxidation products in the Ames test. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2017. [PMID: 28622825 DOI: 10.1016/j.mrgentox.2017.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Polyunsaturated fatty acids (PUFA) represent one of the main building blocks of cellular membranes and their varying composition impacts lifespan as well as susceptibility to cancer and other degenerative diseases. Increased intake of ω-3 PUFA is taught to compensate for the abundance of ω-6 PUFA in modern human diet and prevent cardiocirculatory diseases. However, highly unsaturated PUFA of marine and seed origin easily oxidize to aldehydic products which form DNA adducts. With increased PUFA consumption it is prudent to re-evaluate ω-3 PUFA safety and the genotoxic hazards of their metabolites. We have used the standard Ames test to examine the mutagenicity of 2 hexenals derived from lipid peroxidation of the common ω-3 PUFA in human diet and tissues. Both 4-hydroxyhexenal and 2-hexenal derived from the ω-3 docosahexaenoic and α-linolenic acid, respectively, induced base substitutions in the TA104 and TA100 Ames strains in a dose dependent manner. Their mutagenicity was dependent on the Y-family DNA polymerase RI and they did not induce other types of mutations such as the -2 and -1 frameshifts in the TA98 and TA97 strains. Our results expand previous findings about the mutagenicity of related ω-3 peroxidation product 4-oxohexenal and raise alert that overuse of ω-3 rich oils may have adverse effect on genome stability.
Collapse
|
16
|
De Luca P, Dalton GN, Scalise GD, Moiola CP, Porretti J, Massillo C, Kordon E, Gardner K, Zalazar F, Flumian C, Todaro L, Vazquez ES, Meiss R, De Siervi A. CtBP1 associates metabolic syndrome and breast carcinogenesis targeting multiple miRNAs. Oncotarget 2017; 7:18798-811. [PMID: 26933806 PMCID: PMC4951330 DOI: 10.18632/oncotarget.7711] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/11/2016] [Indexed: 12/22/2022] Open
Abstract
Metabolic syndrome (MeS) has been identified as a risk factor for breast cancer. C-terminal binding protein 1 (CtBP1) is a co-repressor of tumor suppressor genes that is activated by low NAD+/NADH ratio. High fat diet (HFD) increases intracellular NADH. We investigated the effect of CtBP1 hyperactivation by HFD intake on mouse breast carcinogenesis. We generated a MeS-like disease in female mice by chronically feeding animals with HFD. MeS increased postnatal mammary gland development and generated prominent duct patterns with markedly increased CtBP1 and Cyclin D1 expression. CtBP1 induced breast cancer cells proliferation. Serum from animals with MeS enriched the stem-like/progenitor cell population from breast cancer cells. CtBP1 increased breast tumor growth in MeS mice modulating multiple genes and miRNA expression implicated in cell proliferation, progenitor cells phenotype, epithelial to mesenchymal transition, mammary development and cell communication in the xenografts. These results define a novel function for CtBP1 in breast carcinogenesis.
Collapse
Affiliation(s)
- Paola De Luca
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Guillermo N Dalton
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Georgina D Scalise
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Cristian P Moiola
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Juliana Porretti
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Cintia Massillo
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Edith Kordon
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET, Buenos Aires, Argentina
| | - Kevin Gardner
- National Cancer Institute and National Institute of Minority Health and Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Florencia Zalazar
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Carolina Flumian
- Área de Investigación del Instituto de Oncología A.H. Roffo, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Laura Todaro
- Área de Investigación del Instituto de Oncología A.H. Roffo, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Elba S Vazquez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN - CONICET, Buenos Aires, Argentina
| | - Roberto Meiss
- Departamento de Patología, Instituto de Estudios Oncológicos, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Adriana De Siervi
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| |
Collapse
|
17
|
Abdelmagid SA, MacKinnon JL, Janssen SM, Ma DWL. Role of n-3 Polyunsaturated Fatty Acids and Exercise in Breast Cancer Prevention: Identifying Common Targets. Nutr Metab Insights 2016; 9:71-84. [PMID: 27812288 PMCID: PMC5089819 DOI: 10.4137/nmi.s39043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/18/2016] [Accepted: 08/23/2016] [Indexed: 12/21/2022] Open
Abstract
Diet and exercise are recognized as important lifestyle factors that significantly influence breast cancer risk. In particular, dietary n-3 polyunsaturated fatty acids (PUFAs) have been shown to play an important role in breast cancer prevention. Growing evidence also demonstrates a role for exercise in cancer and chronic disease prevention. However, the potential synergistic effect of n-3 PUFA intake and exercise is yet to be determined. This review explores targets for breast cancer prevention that are common between n-3 PUFA intake and exercise and that may be important study outcomes for future research investigating the combined effect of n-3 PUFA intake and exercise. These lines of evidence highlight potential new avenues for research and strategies for breast cancer prevention.
Collapse
Affiliation(s)
- Salma A Abdelmagid
- Department of Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, Ontario, Canada
| | - Jessica L MacKinnon
- Department of Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, Ontario, Canada
| | - Sarah M Janssen
- Department of Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, Ontario, Canada
| | - David W L Ma
- Department of Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
18
|
Giles K, Guan C, Jagoe TR, Mazurak V. Diet composition as a source of variation in experimental animal models of cancer cachexia. J Cachexia Sarcopenia Muscle 2016; 7:110-25. [PMID: 27493865 PMCID: PMC4863732 DOI: 10.1002/jcsm.12058] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/24/2015] [Accepted: 06/16/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND A variety of experimental animal models are used extensively to study mechanisms underlying cancer cachexia, and to identify potential treatments. The important potential confounding effect of dietary composition and intake used in many preclinical studies of cancer cachexia is frequently overlooked. Dietary designs applied in experimental studies should maximize the applicability to human cancer cachexia, meeting the essential requirements of the species used in the study, matched between treatment and control groups as well as also being generally similar to human consumption. METHODS A literature review of scientific studies using animal models of cancer and cancer cachexia with dietary interventions was performed. Studies that investigated interventions using lipid sources were selected as the focus of discussion. RESULTS The search revealed a number of nutrient intervention studies (n = 44), with the majority including n-3 fatty acids (n = 16), mainly eicosapentaenoic acid and/or docosahexaenoic acid. A review of the literature revealed that the majority of studies do not provide information about dietary design; food intake or pair-feeding is rarely reported. Further, there is a lack of standardization in dietary design, content, source, and overall composition in animal models of cancer cachexia. A model is proposed with the intent of guiding dietary design in preclinical studies to enable comparisons of dietary treatments within the same study, translation across different study designs, as well as application to human nutrient intakes. CONCLUSION The potential for experimental endpoints to be affected by variations in food intake, macronutrient content, and diet composition is likely. Diet content and composition should be reported, and food intake assessed. Minimum standards for diet definition in cachexia studies would improve reproducibility of pre-clinical studies and aid the interpretation and translation of results to humans with cancer.
Collapse
Affiliation(s)
- Kaitlin Giles
- Department of Agricultural, Food, and Nutritional Science University of Alberta Edmonton Canada
| | - Chen Guan
- McGill Cancer Nutrition Rehabilitation Program Jewish General Hospital Montreal Canada
| | - Thomas R Jagoe
- McGill Cancer Nutrition Rehabilitation Program Jewish General Hospital Montreal Canada; Department of Medicine McGill University Montreal Canada
| | - Vera Mazurak
- Department of Agricultural, Food, and Nutritional Science University of Alberta Edmonton Canada
| |
Collapse
|
19
|
Dietary supplementation with n-3 fatty acids from weaning limits brain biochemistry and behavioural changes elicited by prenatal exposure to maternal inflammation in the mouse model. Transl Psychiatry 2015; 5:e641. [PMID: 26393487 PMCID: PMC5068805 DOI: 10.1038/tp.2015.126] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/02/2015] [Accepted: 07/22/2015] [Indexed: 12/15/2022] Open
Abstract
Prenatal exposure to maternal immune activation (MIA) increases the risk of schizophrenia and autism in the offspring. The MIA rodent model provides a valuable tool to directly test the postnatal consequences of exposure to an early inflammatory insult; and examine novel preventative strategies. Here we tested the hypotheses that behavioural differences in the MIA mouse model are accompanied by in vivo and ex vivo alterations in brain biochemistry; and that these can be prevented by a post-weaning diet enriched with n-3 polyunsaturated fatty acid (PUFA). The viral analogue PolyI:C (POL) or saline (SAL) was administered to pregnant mice on gestation day 9. Half the resulting male offspring (POL=21; SAL=17) were weaned onto a conventional lab diet (n-6 PUFA); half were weaned onto n-3 PUFA-enriched diet. In vivo magnetic resonance spectroscopy measures were acquired prior to behavioural tests; glutamic acid decarboxylase 67 (GAD67) and tyrosine hydroxylase protein levels were measured ex vivo. The main findings were: (i) Adult MIA-exposed mice fed a standard diet had greater N-acetylaspartate/creatine (Cr) and lower myo-inositol/Cr levels in the cingulate cortex in vivo. (ii) The extent of these metabolite differences was correlated with impairment in prepulse inhibition. (iii) MIA-exposed mice on the control diet also had higher levels of anxiety and altered levels of GAD67 ex vivo. (iv) An n-3 PUFA diet prevented all the in vivo and ex vivo effects of MIA observed. Thus, n-3 PUFA dietary enrichment from early life may offer a relatively safe and non-toxic approach to limit the otherwise persistent behavioural and biochemical consequences of prenatal exposure to inflammation. This result may have translational importance.
Collapse
|
20
|
Andrade FDO, de Assis S, Jin L, Fontelles CC, Barbisan LF, Purgatto E, Hilakivi-Clarke L, Ong TP. Lipidomic fatty acid profile and global gene expression pattern in mammary gland of rats that were exposed to lard-based high fat diet during fetal and lactation periods associated to breast cancer risk in adulthood. Chem Biol Interact 2015; 239:118-28. [PMID: 26115784 DOI: 10.1016/j.cbi.2015.06.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 06/05/2015] [Accepted: 06/23/2015] [Indexed: 02/07/2023]
Abstract
The persistent effects of animal fat consumption during pregnancy and nursing on the programming of breast cancer risk among female offspring were studied here. We have previously found that female offspring of rat dams that consumed a lard-based high-fat (HF) diet (60% fat-derived energy) during pregnancy, or during pregnancy and lactation, were at a reduced risk of developing mammary cancer. To better understand the unexpected protective effects of early life lard exposure, we have applied lipidomics and nutrigenomics approaches to investigate the fatty acid profile and global gene expression patterns in the mammary tissue of the female offspring. Consumption of this HF diet during gestation had few effects on the mammary tissue fatty acids profile of young adult offspring, while exposure from gestation throughout nursing promoted significant alterations in the fatty acids profile. Major differences were related to decreases in saturated fatty acids (SFA) and increases in omega-6 polyunsaturated fatty acids (PUFAs), monounsaturated fatty acids (MUFAs) and conjugated linolenic acid (CLA) concentrations. In addition several differences in gene expression patterns by microarray analysis between the control and in utero or in utero and during lactation HF exposed offspring were identified. Differential dependency network (DDN) analysis indicated that many of the genes exhibited unique connections to other genes only in the HF offspring. These unique connections included Hrh1-Ythdf1 and Repin1-Elavl2 in the in utero HF offspring, and Rnf213-Htr3b and Klf5-Chrna4 in the in utero and lactation HF offspring, compared with the control offspring. We conclude that an exposure to a lard-based HF diet during early life changes the fatty acid profile and transcriptional network in mammary gland in young adult rats, and these changes appear to be consistent with reduced mammary cancer risk observed in our previous study.
Collapse
Affiliation(s)
- Fábia de Oliveira Andrade
- Departament of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 580, Bloco 14, 05508-900 São Paulo, SP, Brazil
| | - Sonia de Assis
- Department of Oncology, Georgetown University Lombardi Comprehensive Cancer Center, Research Building, Room E407, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20007, USA
| | - Lu Jin
- Department of Oncology, Georgetown University Lombardi Comprehensive Cancer Center, Research Building, Room E407, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20007, USA
| | - Camile Castilho Fontelles
- Departament of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 580, Bloco 14, 05508-900 São Paulo, SP, Brazil
| | - Luís Fernando Barbisan
- Department of Morphology, Botucatu Bioscience Institute, State University of São Paulo Botucatu, Distrito de Rubião Júnior, s/n. Rubião Júnior, 18618000 Botucatu, SP, Brazil
| | - Eduardo Purgatto
- Departament of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 580, Bloco 14, 05508-900 São Paulo, SP, Brazil
| | - Leena Hilakivi-Clarke
- Department of Oncology, Georgetown University Lombardi Comprehensive Cancer Center, Research Building, Room E407, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20007, USA
| | - Thomas Prates Ong
- Departament of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 580, Bloco 14, 05508-900 São Paulo, SP, Brazil; Food Research Center (NAPAN), Av. Prof. Lineu Prestes 580, Bloco 14, 05508-900 São Paulo, SP, Brazil.
| |
Collapse
|
21
|
HU ZHIMEI, QI HAIXIA, ZHANG RUIXUE, ZHANG KUN, SHI ZHEMIN, CHANG YANAN, CHEN LINFENG, ESMAEILI MOHSEN, BANIAHMAD ARIA, HONG WEI. Docosahexaenoic acid inhibits the growth of hormone-dependent prostate cancer cells by promoting the degradation of the androgen receptor. Mol Med Rep 2015; 12:3769-3774. [DOI: 10.3892/mmr.2015.3813] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 03/19/2015] [Indexed: 11/05/2022] Open
|
22
|
Witte TR, Hardman WE. The effects of omega-3 polyunsaturated Fatty Acid consumption on mammary carcinogenesis. Lipids 2015; 50:437-46. [PMID: 25860692 DOI: 10.1007/s11745-015-4011-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 03/23/2015] [Indexed: 12/21/2022]
Abstract
The consumption of omega-3 polyunsaturated fatty acids (n-3 PUFA) is associated with a reduced risk of breast cancer. Studies in animals and in vitro have demonstrated mechanisms that could explain this apparent effect, but clinical and epidemiological studies have returned conflicting results on the practical benefits of dietary n-3 PUFA for prevention of breast cancer. Effects are often only significant within a population when comparing the highest n-3 PUFA consumption group to the lowest n-3 group or highest n-6 group. The beneficial effects of n-3 PUFA eicosapentaenoic and docosahexaenoic on the risk of breast cancer are dose dependent and are negatively affected by total n-6 consumption. The majority of the world population, including the most highly developed regions, consumes insufficient n-3 PUFA to significantly reduce breast cancer risk. This review discusses the physiological and dietary context in which reduction of breast cancer risk may occur, some proposed mechanisms of action and meaningful recommendations for consumption of n-3 PUFA in the diet of developed regions.
Collapse
Affiliation(s)
- Theodore R Witte
- Department of Biochemistry and Microbiology, Marshall University School of Medicine, 1600 Medical Center Drive, Huntington, WV, 25701, USA
| | | |
Collapse
|
23
|
Ma Z, Blackwelder AJ, Lee H, Zhao M, Yang X. In Utero exposure to low-dose alcohol induces reprogramming of mammary development and tumor risk in MMTV-erbB-2 transgenic mice. Int J Mol Sci 2015; 16:7655-71. [PMID: 25853264 PMCID: PMC4425041 DOI: 10.3390/ijms16047655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 03/30/2015] [Accepted: 03/30/2015] [Indexed: 12/20/2022] Open
Abstract
There is increasing evidence that prenatal exposure to environmental factors may modify breast cancer risk later in life. This study aimed to investigate the effects of in utero exposure to low-dose alcohol on mammary development and tumor risk. Pregnant MMTV-erbB-2 mice were exposed to alcohol (6 g/kg/day) between day 13 and day 19 of gestation, and the female offspring were examined for tumor risk. Whole mount analysis indicated that in utero exposure to low-dose alcohol induced significant increases in ductal extension at 10 weeks of age. Molecular analysis showed that in utero alcohol exposure induced upregulation of ERα signaling and activation of Akt and Erk1/2 in pubertal mammary glands. However, enhanced signaling in the EGFR/erbB-2 pathway appeared to be more prominent in 10-week-old glands than did signaling in the other pathways. Interestingly, tumor development in mice with in utero exposure to low-dose alcohol was slightly delayed compared to control mice, but tumor multiplicity was increased. The results indicate that in utero exposure to low-dose alcohol induces the reprogramming of mammary development by mechanisms that include altered signaling in the estrogen receptor (ER) and erbB-2 pathways. The intriguing tumor development pattern might be related to alcohol dose and exposure conditions, and warrants further investigation.
Collapse
MESH Headings
- Animals
- Cell Transformation, Viral/genetics
- Estrogen Receptor alpha/metabolism
- Ethanol/pharmacology
- Ethanol/toxicity
- Female
- Fetus/drug effects
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/chemically induced
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/virology
- Mammary Tumor Virus, Mouse/pathogenicity
- Mice
- Mice, Transgenic
- Pregnancy
- Prenatal Exposure Delayed Effects/chemically induced
- Prenatal Exposure Delayed Effects/pathology
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Signal Transduction/drug effects
- Up-Regulation
Collapse
Affiliation(s)
- Zhikun Ma
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Kannapolis, NC 28081, USA.
| | - Amanda J Blackwelder
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Kannapolis, NC 28081, USA.
| | - Harry Lee
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Kannapolis, NC 28081, USA.
| | - Ming Zhao
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Kannapolis, NC 28081, USA.
| | - Xiaohe Yang
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Kannapolis, NC 28081, USA.
| |
Collapse
|
24
|
|
25
|
Liu J, Ma DWL. The role of n-3 polyunsaturated fatty acids in the prevention and treatment of breast cancer. Nutrients 2014; 6:5184-223. [PMID: 25412153 PMCID: PMC4245586 DOI: 10.3390/nu6115184] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/28/2014] [Accepted: 11/04/2014] [Indexed: 02/07/2023] Open
Abstract
Breast cancer (BC) is the most common cancer among women worldwide. Dietary fatty acids, especially n-3 polyunsaturated fatty acids (PUFA), are believed to play a role in reducing BC risk. Evidence has shown that fish consumption or intake of long-chain n-3 PUFA, such as eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are beneficial for inhibiting mammary carcinogenesis. The evidence regarding α-linolenic acid (ALA), however, remains equivocal. It is essential to clarify the relation between ALA and cancer since ALA is the principal source of n-3 PUFA in the Western diet and the conversion of ALA to EPA and DHA is not efficient in humans. In addition, the specific anticancer roles of individual n-3 PUFA, alone, have not yet been identified. Therefore, the present review evaluates ALA, EPA and DHA consumed individually as well as in n-3 PUFA mixtures. Also, their role in the prevention of BC and potential anticancer mechanisms of action are examined. Overall, this review suggests that each n-3 PUFA has promising anticancer effects and warrants further research.
Collapse
Affiliation(s)
- Jiajie Liu
- Department of Human Health & Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - David W L Ma
- Department of Human Health & Nutritional Sciences, College of Biological Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
26
|
Lopes GAD, Fan WYC, Ciol H, Bidinotto LT, Rodrigues MAM, Barbisan LF. Maternal western style diet increases susceptibility to chemically-induced mammary carcinogenesis in female rats offspring. Nutr Cancer 2014; 66:1293-303. [PMID: 25333700 DOI: 10.1080/01635581.2014.956256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The present study investigated whether maternal exposure to western style diet (WD) increases susceptibility to mammary carcinogenesis induced by N-methyl-N-nitrosourea (MNU) in female offspring. Pregnant female Sprague-Dawley rats received WD diet or control diet from gestational day 12 until postnatal day (PND) 21. At PND 21, female offspring received a single dose of MNU (50 mg/kg body weight) and were fed chow diet until PND 110. Mammary gland structures were assessed on whole-mount preparations in the offspring at PND 21, and tumor morphology was examined at PND 110. Immunohistochemical analysis for cell proliferation (PCNA), apoptosis (cleaved caspase-3) and estrogen receptor alpha (ER-α) was performed in mammary terminal end buds (TEBs) at PND 21, and PCNA, ER-α, and p63 analysis in mammary tumors at PND 110. Maternal WD intake induced a significant increase in the number of TEBs (P = 0.024) and in PCNA labeling index (P < 0.020) in the mammary glands at PND 21. Tumor multiplicity, tumor weight, and PCNA labeling indexes were significantly higher in the WD offspring than that of the control offspring (P < 0.05). These findings indicate that maternal western style diet potentially enhanced the development of mammary tumors induced by MNU in female offspring, possibly by affecting the mammary gland differentiation.
Collapse
Affiliation(s)
- Gisele A D Lopes
- a Department of Pathology, Botucatu Medical School , São Paulo State University , São Paulo , Brazil
| | | | | | | | | | | |
Collapse
|
27
|
Zheng H, Tang H, Liu M, He M, Lai P, Dong H, Lin J, Jia C, Zhong M, Dai Y, Bai X, Wang L. Inhibition of Endometrial Cancer by n-3 Polyunsaturated Fatty Acids in Preclinical Models. Cancer Prev Res (Phila) 2014; 7:824-34. [DOI: 10.1158/1940-6207.capr-13-0378-t] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Olivo-Marston SE, Hursting SD, Perkins SN, Schetter A, Khan M, Croce C, Harris CC, Lavigne J. Effects of calorie restriction and diet-induced obesity on murine colon carcinogenesis, growth and inflammatory factors, and microRNA expression. PLoS One 2014; 9:e94765. [PMID: 24732966 PMCID: PMC3986228 DOI: 10.1371/journal.pone.0094765] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/28/2013] [Indexed: 12/17/2022] Open
Abstract
Obesity is an established colon cancer risk factor, while preventing or reversing obesity via a calorie restriction (CR) diet regimen decreases colon cancer risk. Unfortunately, the biological mechanisms underlying these associations are poorly understood, hampering development of mechanism-based approaches for preventing obesity-related colon cancer. We tested the hypotheses that diet-induced obesity (DIO) would increase (and CR would decrease) colon tumorigenesis in the mouse azoxymethane (AOM) model. In addition, we established that changes in inflammatory cytokines, growth factors, and microRNAs are associated with these energy balance-colon cancer links, and thus represent mechanism-based targets for colon cancer prevention. Mice were injected with AOM once a week for 5 weeks and randomized to: 1) control diet; 2) 30% CR diet; or 3) DIO diet. Mice were euthanized at week 5 (n = 12/group), 10 (n = 12/group), and 20 (n = 20/group) after the last AOM injection. Colon tumors were counted, and cytokines, insulin-like growth factor 1 (IGF-1), IGF binding protein 3 (IGFBP-3), adipokines, proliferation, apoptosis, and expression of microRNAs (miRs) were measured. The DIO diet regimen induced an obese phenotype (∼36% body fat), while CR induced a lean phenotype (∼14% body fat); controls were intermediate (∼26% body fat). Relative to controls, DIO increased (and CR decreased) the number of colon tumors (p = 0.01), cytokines (p<0.001), IGF-1 (p = 0.01), and proliferation (p<0.001). DIO decreased (and CR increased) IGFBP-3 and apoptosis (p<0.001). miRs including mir-425, mir-196, mir-155, mir-150, mir-351, mir-16, let-7, mir34, and mir-138 were differentially expressed between the dietary groups. We conclude that the enhancing effects of DIO and suppressive effects of CR on colon carcinogenesis are associated with alterations in several biological pathways, including inflammation, IGF-1, and microRNAs.
Collapse
Affiliation(s)
- Susan E. Olivo-Marston
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, United States of America
- Division of Epidemioogy, The Ohio State University College of Public Health, Columbus, Ohio, United States of America
- * E-mail:
| | - Stephen D. Hursting
- Department of Nutritional Sciences, University of Texas-Austin, Austin, Texas, United States of America
- Department of Molecular Carcinogenesis, University of Texas-MD Anderson Cancer Center, Smithville, Texas, United States of America
| | - Susan N. Perkins
- Center for Cancer Training, The National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Aaron Schetter
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mohammed Khan
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Carlo Croce
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
| | - Curtis C. Harris
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jackie Lavigne
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| |
Collapse
|
29
|
Anderson BM, MacLennan MB, Hillyer LM, Ma DWL. Lifelong exposure to n-3 PUFA affects pubertal mammary gland development. Appl Physiol Nutr Metab 2014; 39:699-706. [PMID: 24758708 DOI: 10.1139/apnm-2013-0365] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is growing evidence that early developmental periods may importantly influence future breast cancer risk. Also, there is great interest in the role of dietary fat in breast cancer risk, but the role of dietary fat during pubertal mammary gland development remains poorly understood. This study investigated the effect of n-3 polyunsaturated fatty acids (PUFA) using complementary dietary and genetic approaches to examine the effect of lifelong exposure of n-3 PUFA or n-6 PUFA (control) on mammary gland development and fatty acid composition. n-3 PUFA from both diet and genetics were enriched in mammary glands as early as 3 weeks of age. Parameters related to mammary gland development, including number of terminal end buds (TEB), percent coverage of ductal tree, and infiltration of TEB, were influenced by n-3 PUFA at 3 and 4 weeks of age. Overall, findings suggest that n-3 PUFA incorporation into the mammary gland early in life plays a role in the morphological development of the mammary gland during puberty.
Collapse
Affiliation(s)
- Breanne M Anderson
- Department of Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Animal Science/Nutrition Building, Rm 342, 491 Gordon Street, Guelph, ON N1G 2W1, Canada
| | | | | | | |
Collapse
|
30
|
de Oliveira Andrade F, Fontelles CC, Rosim MP, de Oliveira TF, de Melo Loureiro AP, Mancini-Filho J, Rogero MM, Moreno FS, de Assis S, Barbisan LF, Hilakivi-Clarke L, Ong TP. Exposure to lard-based high-fat diet during fetal and lactation periods modifies breast cancer susceptibility in adulthood in rats. J Nutr Biochem 2014; 25:613-22. [PMID: 24746835 DOI: 10.1016/j.jnutbio.2014.02.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/27/2014] [Accepted: 02/04/2014] [Indexed: 12/16/2022]
Abstract
The present study investigated whether early life exposure to high levels of animal fat increases breast cancer risk in adulthood in rats. Dams consumed a lard-based high-fat (HF) diet (60% fat-derived energy) or an AIN93G control diet (16% fat-derived energy) during gestation or gestation and lactation. Their 7-week-old female offspring were exposed to 7,12-dimethyl-benzo[a]anthracene to induce mammary tumors. Pregnant dams consuming an HF diet had higher circulating leptin levels than pregnant control dams. However, compared to the control offspring, significantly lower susceptibility to mammary cancer development was observed in the offspring of dams fed an HF diet during pregnancy (lower tumor incidence, multiplicity and weight), or pregnancy and lactation (lower tumor multiplicity only). Mammary epithelial elongation, cell proliferation (Ki67) and expression of NFκB p65 were significantly lower and p21 expression and global H3K9me3 levels were higher in the mammary glands of rats exposed to an HF lard diet in utero. They also tended to have lower Rank/Rankl ratios (P=.09) and serum progesterone levels (P=.07) than control offspring. In the mammary glands of offspring of dams consuming an HF diet during both pregnancy and lactation, the number of terminal end buds, epithelial elongation and the BCL-2/BAX ratio were significantly lower and serum leptin levels were higher than in the controls. Our data confirm that the breast cancer risk of offspring can be programmed by maternal dietary intake. However, contrary to our expectation, exposure to high levels of lard during early life decreased later susceptibility to breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fernando Salvador Moreno
- Faculty of Pharmaceutical Sciences; Food and Nutrition Research Center (NAPAN), São Paulo, Brazil
| | - Sonia de Assis
- Georgetown University Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | | | | | - Thomas Prates Ong
- Faculty of Pharmaceutical Sciences; Food and Nutrition Research Center (NAPAN), São Paulo, Brazil.
| |
Collapse
|
31
|
Abstract
Epigenetic processes play a central role in regulating the tissue-specific expression of genes. Alterations in these processes can lead to profound changes in phenotype and have been implicated in the pathogenesis of many human diseases including human cancer. There is growing evidence that the environment, particularly variations in diet, during specific developmental periods can induce changes in the epigenome, which are then stably maintained throughout life influencing susceptibility to cancer in later life. This chapter will review the evidence that alterations in early life nutritional exposure can affect breast cancer risk through the altered epigenetic regulation of genes and discuss how detection of such altered epigenetic marks in early life may provide biomarkers to detect individuals at increased risk of disease.
Collapse
|
32
|
Chen Z, Zhang Y, Jia C, Wang Y, Lai P, Zhou X, Wang Y, Song Q, Lin J, Ren Z, Gao Q, Zhao Z, Zheng H, Wan Z, Gao T, Zhao A, Dai Y, Bai X. mTORC1/2 targeted by n-3 polyunsaturated fatty acids in the prevention of mammary tumorigenesis and tumor progression. Oncogene 2013; 33:4548-57. [PMID: 24096482 DOI: 10.1038/onc.2013.402] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 07/21/2013] [Accepted: 08/23/2013] [Indexed: 12/17/2022]
Abstract
Although epidemiological and preclinical studies have shown the preventative effects of n-3 polyunsaturated fatty acids (PUFAs) on breast cancer, inconsistencies still remain in the data and the underlying mechanisms remain unclear. In this study, we identified mammalian target of rapamycin (mTOR) signaling, which plays an essential role in cell proliferation and breast tumorigenesis, as a target of n-3 PUFAs. In breast cancer cell lines, n-3 PUFAs rapidly and efficiently suppress both mTOR complex 1 (mTORC1) and mTORC2 and their downstream signaling, and subsequently inhibit cell proliferation and angiogenesis while promoting apoptosis. Further study indicates that stabilization of the mTOR-raptor complex by n-3 PUFAs may contribute to their inhibitory effect on mTORC1. Importantly, four complementary and well-controlled animal models were utilized to identify the role and molecular target of n-3 PUFAs in the prevention of breast carcinogenesis and progression, namely: (1) chemically induced mammary tumor rats with a high dietary intake of n-3 PUFAs; (2) nude mice implanted with mammary tumor cell lines stably expressing fat-1, a desaturase that catalyzes the conversion of n-6 to n-3 PUFAs and produces n-3 PUFAs endogenously; (3) fat-1 transgenic severe combined immune deficiency mice implanted with breast tumor cells; and (4) the fat-1 transgenic mouse mammary tumor virus-polyoma virus middle T oncogene double-hybrid mice, a model of aggressive breast cancer. In summary, dietary and endogenous n-3 PUFAs abrogate the activity of mTORC1/2 pathways in vitro and in vivo and prevent breast carcinogenesis, tumor growth and metastasis. Taken together, our findings convincingly clarify the causal relationship between n-3 PUFAs and breast cancer prevention and establish mTORC1/2 as a target of n-3 PUFAs.
Collapse
Affiliation(s)
- Z Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Y Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - C Jia
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Y Wang
- State Key Laboratory of Reproductive Medicine and Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - P Lai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - X Zhou
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Y Wang
- State Key Laboratory of Reproductive Medicine and Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Q Song
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Lin
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Z Ren
- State Key Laboratory of Reproductive Medicine and Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Q Gao
- State Key Laboratory of Reproductive Medicine and Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Z Zhao
- College of Life Sciences, Nankai University, Tianjin, China
| | - H Zheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Z Wan
- State Key Laboratory of Reproductive Medicine and Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - T Gao
- Department of Anatomy and Neurobiology, Southern Medical University, Guangzhou, China
| | - A Zhao
- State Key Laboratory of Reproductive Medicine and Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Y Dai
- State Key Laboratory of Reproductive Medicine and Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - X Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
33
|
Zou Z, Bidu C, Bellenger S, Narce M, Bellenger J. n-3 polyunsaturated fatty acids and HER2-positive breast cancer: interest of the fat-1 transgenic mouse model over conventional dietary supplementation. Biochimie 2013; 96:22-7. [PMID: 24012777 DOI: 10.1016/j.biochi.2013.08.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 08/23/2013] [Indexed: 01/10/2023]
Abstract
Overexpression of the tyrosine kinase receptor ErbB2/HER2/Neu, occurs in 25%-30% of invasive breast cancer (BC) with poor patient prognosis. Even if numerous studies have shown prevention of breast cancer by n-3 fatty acid intake, the experimental conditions under which n-3 fatty acids exert their protective effect have been variable from study to study, preventing unifying conclusions. Due to confounding factors, inconsistencies still remain regarding protective effects of n-3 polyunsaturated fatty acids (PUFA) on BC. When animals are fed with dietary supplementation in n-3 fatty acids (the traditional approach to modify tissue content and decrease the n-6/n-3 ratio) complex dietary interactions can occur among dietary lipids (antioxidants, vitamins…) that can modulate the activity of n-3 fatty acids. So, what are the specific roles of these n-3 PUFA in reducing breast cancer risk and particularly preventing HER2-positive breast cancer? In this review, we discuss crucial points that may account for discrepancies of results and provide a highly effective genetic approach that can eliminate confounding factors of diet for evaluating the molecular mechanisms of n-3 PUFA in HER2 signaling pathway regulation. The fat-1 transgenic mouse model is capable of converting n-6 to n-3 fatty acids leading to an increase in n-3 fatty acid content with a balanced n-6/n-3 fatty acid ratio in all tissues. The fat-1 mouse model allows well-controlled studies in HER2-positive breast cancer prevention to be performed, without the conflict of potential confounding factors of diet.
Collapse
Affiliation(s)
- Zuquan Zou
- Université de Bourgogne, UFR Sciences de la Vie, de la Terre et de l'Environnement, 6 Boulevard Gabriel, 21000 Dijon, France; INSERM UMR U866 Lipides Nutrition Cancer, Université de Bourgogne, 6 Boulevard Gabriel, 21000 Dijon, France
| | | | | | | | | |
Collapse
|
34
|
Luijten M, Singh AV, Bastian CA, Westerman A, Pisano MM, Pennings JLA, Verhoef A, Green ML, Piersma AH, de Vries A, Knudsen TB. Lasting effects on body weight and mammary gland gene expression in female mice upon early life exposure to n-3 but not n-6 high-fat diets. PLoS One 2013; 8:e55603. [PMID: 23409006 PMCID: PMC3567116 DOI: 10.1371/journal.pone.0055603] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 01/03/2013] [Indexed: 01/21/2023] Open
Abstract
Exposure to an imbalance of nutrients prior to conception and during critical developmental periods can have lasting consequences on physiological processes resulting in chronic diseases later in life. Developmental programming has been shown to involve structural and functional changes in important tissues. The aim of the present study was to investigate whether early life diet has a programming effect on the mammary gland. Wild-type mice were exposed from 2 weeks prior to conception to 6 weeks of age to a regular low-fat diet, or to high-fat diets based on either corn oil or flaxseed oil. At 6 weeks of age, all mice were shifted to the regular low-fat diet until termination at 10 weeks of age. Early life exposure to a high-fat diet, either high in n-6 (corn oil) or in n-3 (flaxseed oil) polyunsaturated fatty acids, did not affect birth weight, but resulted in an increased body weight at 10 weeks of age. Transcriptome analyses of the fourth abdominal mammary gland revealed differentially expressed genes between the different treatment groups. Exposure to high-fat diet based on flaxseed oil, but not on corn oil, resulted in regulation of pathways involved in energy metabolism, immune response and inflammation. Our findings suggest that diet during early life indeed has a lasting effect on the mammary gland and significantly influences postnatal body weight gain, metabolic status, and signaling networks in the mammary gland of female offspring.
Collapse
Affiliation(s)
- Mirjam Luijten
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Amar V. Singh
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville Birth Defects Center, Louisville, Kentucky, United States of America
| | - Caleb A. Bastian
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville Birth Defects Center, Louisville, Kentucky, United States of America
| | - Anja Westerman
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - M. Michele Pisano
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville Birth Defects Center, Louisville, Kentucky, United States of America
| | - Jeroen L. A. Pennings
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Aart Verhoef
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Maia L. Green
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville Birth Defects Center, Louisville, Kentucky, United States of America
| | - Aldert H. Piersma
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Annemieke de Vries
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Thomas B. Knudsen
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville Birth Defects Center, Louisville, Kentucky, United States of America
- National Center for Computational Toxicology, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
35
|
de Assis S, Warri A, Cruz MI, Laja O, Tian Y, Zhang B, Wang Y, Huang THM, Hilakivi-Clarke L. High-fat or ethinyl-oestradiol intake during pregnancy increases mammary cancer risk in several generations of offspring. Nat Commun 2013; 3:1053. [PMID: 22968699 PMCID: PMC3570979 DOI: 10.1038/ncomms2058] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 08/10/2012] [Indexed: 01/05/2023] Open
Abstract
Maternal exposures to environmental factors during pregnancy influence the risk of many chronic adult-onset diseases in the offspring. Here we investigate whether feeding pregnant rats a high-fat (HF)- or ethinyl-oestradiol (EE2)-supplemented diet affects carcinogen-induced mammary cancer risk in daughters, granddaughters and great-granddaughters. We show that mammary tumourigenesis is higher in daughters and granddaughters of HF rat dams and in daughters and great-granddaughters of EE2 rat dams. Outcross experiments suggest that the increase in mammary cancer risk is transmitted to HF granddaughters equally through the female or male germ lines, but it is only transmitted to EE2 granddaughters through the female germ line. The effects of maternal EE2 exposure on offspring's mammary cancer risk are associated with changes in the DNA methylation machinery and methylation patterns in mammary tissue of all three EE2 generations. We conclude that dietary and oestrogenic exposures in pregnancy increase breast cancer risk in multiple generations of offspring, possibly through epigenetic means. Environmental factors can influence one's susceptibility to cancer, but it is not clear whether such an influence extends beyond the directly exposed generations. Here, feeding pregnant rats with a high-fat diet or a hormone derivative, the authors observe increased breast cancer risk in up to three subsequent generations.
Collapse
Affiliation(s)
- Sonia de Assis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3970 Reservoir Road NW, The Research Building, Room E407, Washington, District of Columbia 20057, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Mammary tumor development is directly inhibited by lifelong n-3 polyunsaturated fatty acids. J Nutr Biochem 2012; 24:388-95. [PMID: 23026490 DOI: 10.1016/j.jnutbio.2012.08.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 07/19/2012] [Accepted: 08/01/2012] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Despite the advocacy that diet may be a significant contributor to cancer prevention, there is a lack of direct evidence from epidemiological and experimental studies to substantiate such claims. Experimental studies suggest that n-3 polyunsaturated fatty acids (n-3 PUFA) from marine oils may reduce breast cancer risk, however, findings are equivocal. Thus, in this study, novel transgenic mouse models were employed to provide, for the first time, direct evidence for an anti-cancer role of n-3 PUFA in mammary tumorigenesis. METHODS fat-1 Mice, which are capable of endogenous n-3 PUFA synthesis, were bred with mouse mammary tumor virus (MMTV)-neu(ndl)-YD5 mice, an aggressive breast cancer model. The resultant offspring, including novel hybrid progeny, were assessed for tumor onset, size and multiplicity as well as n-3 PUFA composition in mammary gland and tumor tissue. A complementary group of MMTV-neu(ndl)-YD5 mice were fed n-3 PUFA in the diet. RESULTS Mice expressing MMTV-neu(ndl)-YD5 and fat-1 displayed significant (P<.05) reductions in tumor volume (~30%) and multiplicity (~33%), as well as reduced n-6 PUFA and enriched n-3 PUFA in tumor phospholipids relative to MMTV-neu(ndl)-YD5 control mice. The effect observed in hybrid progeny was similarly observed in n-3 PUFA diet fed mice. CONCLUSION Using complementary genetic and conventional dietary approaches we provide, for the first time, unequivocal experimental evidence that n-3 PUFA is causally linked to tumor prevention.
Collapse
|
37
|
Inhibition of neuroblastoma cell proliferation with omega-3 fatty acids and treatment of a murine model of human neuroblastoma using a diet enriched with omega-3 fatty acids in combination with sunitinib. Pediatr Res 2012; 71:168-78. [PMID: 22258128 DOI: 10.1038/pr.2011.28] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION We investigated the use of dietary omega-3 (ω-3) polyunsaturated fatty acids (PUFAs) in the treatment of neuroblastoma both as a sole agent and in combination with sunitinib, a broad-spectrum tyrosine kinase receptor inhibitor. RESULTS Substitution of all dietary fat with menhaden oil (ω-3 PUFA rich) resulted in a 40-70% inhibition of tumor growth and a statistically significant difference in the levels of several PUFAs (18:2 ω-6, 20:4 ω-6, 22:4 ω-6, 20:5 ω-3) as compared with a control diet. Furthermore, tumors from animals on the ω-3 fatty acid (FA)-enriched diet had an elevated triene/tetraene ratio suggestive of a change in local eicosanoid metabolism in these tissues similar to that seen with essential fatty acid deficiency. The ω-3 FA-enriched diet also decreased tumor-associated inflammatory cells and induced mitochondrial changes suggestive of mitochondrial damage. Combination treatment with sunitinib resulted in further reduction in tumor proliferation and microvessel density. DISCUSSION These findings suggest a potential role for ω-3 PUFAs in the combination treatment of neuroblastoma. METHODS We used a murine model of orthotopic and subcutaneous human neuroblastoma and diets that differ in the FA content to define the optimal dietary ω-3/omega-6 (ω-6) FA ratio required for the inhibition of these tumors.
Collapse
|
38
|
Bidinotto LT, López de Cicco R, Russo J. Omega-3 fatty acids: a potential booster for tamoxifen therapy? Expert Rev Anticancer Ther 2012; 11:1151-3. [PMID: 21916567 DOI: 10.1586/era.11.106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
The effects of Tamoxifen and fish oil on mammary carcinogenesis in polyoma middle T transgenic mice. Discov Oncol 2011; 2:249-59. [PMID: 21769696 DOI: 10.1007/s12672-011-0078-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In these experiments, we tested the hypothesis that inhibition of the estrogen receptor (ER) with Tamoxifen and activation of PPARγ with fish oil (FO) rich in omega-3 (n-3; known PPAR agonists) inhibit the development of hormone-independent breast cancer in view of the known crosstalk between the ER and PPARγ pathways. We selected the polyoma middle T transgenic mouse model, since in this system the development of ER- tumors is preceded by ER positive preneoplastic lesions. Tamoxifen admixed with a 20% corn oil (CO) modified AIN-76A diet delayed mammary carcinogenesis and inhibited tumor multiplicity, volume, and weight in a dose-dependent (1, 10, and 100 ppm) fashion. Administration of increasing concentrations of FO in the diet (5%, 10%, and 17%) did not affect any of the tumor parameters. Combined administration of different doses of Tamoxifen and FO delayed carcinogenesis and suppressed tumor multiplicity and volume to the same extent as Tamoxifen alone. Mice fed 10% FO exhibited the expected increase in n-3/n-6 ratio in plasma and tumor based on diet analysis. Further increase in the n-3/n-6 ratio was not observed in mice fed the 17% FO diet. FO reduced tissue levels of arachidonic acid and its metabolite PGF-2α. Our results support the role of ER expression by preneoplastic lesions in the development of hormone-independent tumors and consequently the importance of including ER targeting in combination with mechanistically based novel chemopreventive agents.
Collapse
|
40
|
MacLennan MB, Anderson BM, Ma DWL. Differential mammary gland development in FVB and C57Bl/6 mice: implications for breast cancer research. Nutrients 2011; 3:929-36. [PMID: 22254086 PMCID: PMC3257723 DOI: 10.3390/nu3110929] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 10/05/2011] [Accepted: 10/11/2011] [Indexed: 12/31/2022] Open
Abstract
A growing body of research suggests a linkage between pubertal mammary gland development and environmental factors such as diet as modifiers of long term breast cancer risk. Much of this research is dependent upon mouse models, which may vary between studies. However, effects may be strain dependent and further modified by diet, which has not been previously examined. Therefore, the objective of the present study was to determine whether mammary gland development differs between FVB and C57Bl/6 strains on diets containing either n-6 or n-3 polyunsaturated fats. Developmental measures related to onset of puberty and mammary gland development differed between strains. Mice fed the n-3 polyunsaturated fatty acids (PUFA) diet were shown to have lower numbers of terminal end buds, a marker of mammary gland development. This study helps to further clarify differences in development and dietary response between FVB and C57Bl/6 mice in order to more appropriately relate mammary gland research to human populations.
Collapse
Affiliation(s)
- Mira B MacLennan
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| | | | | |
Collapse
|
41
|
Signori C, El-Bayoumy K, Russo J, Thompson HJ, Richie JP, Hartman TJ, Manni A. Chemoprevention of breast cancer by fish oil in preclinical models: trials and tribulations. Cancer Res 2011; 71:6091-6. [PMID: 21933885 DOI: 10.1158/0008-5472.can-11-0977] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite the perception that omega-3 fatty acids (n-3 FA) protect against breast cancer, epidemiologic studies have yielded inconsistent results. Although preclinical data have been, in general, more supportive of a protective effect of n-3 FA on breast cancer, inconsistencies still remain, which preclude definite conclusions or in-depth mechanistic investigations despite 30 years of research in this area. In this review, we discuss key variables that may account for inconsistencies of results across preclinical studies and provide recommendations for future experiments testing the chemopreventive effect of n-3 FAs in breast cancer, as part of a multiagent approach under rigorously controlled conditions.
Collapse
Affiliation(s)
- Carina Signori
- Department of Medicine, Penn State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Xue H, Sawyer MB, Wischmeyer PE, Baracos VE. Nutrition modulation of gastrointestinal toxicity related to cancer chemotherapy: from preclinical findings to clinical strategy. JPEN J Parenter Enteral Nutr 2011; 35:74-90. [PMID: 21224434 DOI: 10.1177/0148607110377338] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chemotherapy-induced gut toxicity is a major dose-limiting toxicity for many anticancer drugs. Gastrointestinal (GI) complications compromise the efficacy of chemotherapy, promote overall malnutrition, aggravate cancer cachexia, and may contribute to worsened prognosis. The GI tract is an attractive target for nutrition modulation, owing to its direct exposure to the diet, participation in uptake and metabolism of nutrients, high rate of cell turnover, and plasticity to nutrition stimuli. Glutamine, ω-3 polyunsaturated fatty acids, and probiotics/prebiotics are therapeutic factors that potentially modulate GI toxicity related to cancer treatments. Preclinical and clinical evidence are reviewed to critically define plausible benefits of these factors and their potential development into adjuncts to cancer chemotherapy. Mechanisms underlying the action of these nutrients are being unraveled in the laboratory. Optimal strategies to translate these findings into clinical care still remain to be elucidated. Key questions that remain to be answered include the following: which nutrient or combination of nutrients is selected for which patient and chemotherapy regimen? What mechanisms are responsible for modulation, and how are nutrient(s) administered in a clinically optimal manner? Research exploring interactions between different nutrients in GI protection is ongoing and demands further understanding. How nutrition preparations given to chemotherapy-treated patients are formulated in terms of component selection and dose optimization should be carefully studied and justified.
Collapse
Affiliation(s)
- Hongyu Xue
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.
| | | | | | | |
Collapse
|
43
|
Nielsen TS, Khan G, Davis J, Michels KB, Hilakivi-Clarke L. Prepubertal exposure to cow's milk reduces susceptibility to carcinogen-induced mammary tumorigenesis in rats. Int J Cancer 2010; 128:12-20. [PMID: 20232392 DOI: 10.1002/ijc.25313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cow's milk contains high levels of estrogens, progesterone and insulin-like growth factor 1 (IGF-1), all of which are associated with breast cancer. We investigated whether prepubertal milk exposure affects mammary gland development and carcinogenesis in rats. Sprague-Dawley rats were given either whole milk or tap water to drink from postnatal day (PND) 14 to PND 35, and thereafter normal tap water. Mammary tumorigenesis was induced by administering 7,12-dimethylbenz[a]anthracene on PND 50. Milk exposure increased circulating E2 levels on PND 25 by 10-fold (p < 0.001) and accelerated vaginal opening, which marks puberty onset, by 2.5 days (p < 0.001). However, rats exposed to milk before puberty exhibited reduced carcinogen-induced mammary carcinogenesis; that is, their tumor latency was longer (p < 0.03) and incidence was lower (p < 0.05) than in the controls. On PND 25 and 50, mammary glands of the milk-exposed rats had significantly less terminal end buds (TEBs) than the tap water-exposed controls (p < 0.019). ER-α protein levels were elevated in the TEBs and lobules of milk rats, compared to rats given tap water (p < 0.019), but no changes in cyclin D1 expression, cell proliferation or apoptosis were seen. IGF-1 mRNA levels were reduced on PND 50 in the mammary glands of rats exposed to milk at puberty. Our results suggest that drinking milk before puberty reduces later risk of developing mammary cancer in rats. This might be mediated by a reduction in the number of TEBs and lower expression of IGF-1 mRNA in the mammary glands of milk-exposed animals.
Collapse
Affiliation(s)
- Tina S Nielsen
- Department of Animal Health and Bioscience, Welfare and Nutrition, Faculty of Agricultural Sciences, University of Aarhus, Aarhus, Denmark
| | | | | | | | | |
Collapse
|
44
|
Ly A, Lee H, Chen J, Sie KKY, Renlund R, Medline A, Sohn KJ, Croxford R, Thompson LU, Kim YI. Effect of Maternal and Postweaning Folic Acid Supplementation on Mammary Tumor Risk in the Offspring. Cancer Res 2010; 71:988-97. [DOI: 10.1158/0008-5472.can-10-2379] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Abstract
Breast cancer is the most common cancer among women worldwide. Estimates suggest up to 35% of cases may be preventable through diet and lifestyle modification. Growing research on the role of fats in human health suggests that early exposure in life to specific fatty acids, when tissues are particularly sensitive to their environment, can have long-term health impacts. The present review examines the role of dietary fat in mammary gland development and breast cancer throughout the lifecycle. Overall, n-3 polyunsaturated fatty acids have promising cancer-preventive effects when introduced early in life, and warrant further research to elucidate the mechanisms of action.
Collapse
|
46
|
Olivo-Marston SE, Hursting SD, Lavigne J, Perkins SN, Maarouf RS, Yakar S, Harris CC. Genetic reduction of circulating insulin-like growth factor-1 inhibits azoxymethane-induced colon tumorigenesis in mice. Mol Carcinog 2009; 48:1071-6. [PMID: 19760669 DOI: 10.1002/mc.20577] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
High levels of insulin-like growth factor-1 (IGF-1) have been associated with a significant increase in colon cancer risk. Additionally, IGF-1 inhibits apoptosis and stimulates proliferation of colonic epithelial cells in vitro. Unfortunately, IGF-1 knockout mice have severe developmental abnormalities and most do not survive, making it difficult to study how genetic ablation of IGF-1 affects colon tumorigenesis. To test the hypothesis that inhibition of IGF-1 prevents colon tumorigenesis, we utilized a preexisting mouse model containing a deletion of the igf1 gene in the liver through a Cre/loxP system. These liver-specific IGF-1 deficient (LID) mice display a 50-75% reduction in circulating IGF-1 levels. We conducted a pilot study to assess the impact of liver-specific IGF-1 deficiency on azoxymethane (AOM)-induced colon tumors. LID mice had a significant inhibition of colon tumor multiplicity in the proximal area of the colon compared to their wild-type littermates. We examined markers of proliferation and apoptosis in the colons of the LID and wild-type mice to see if these were consistent with tumorigenesis. We observed a decrease in proliferation in the colons of the LID mice and an increase in apoptosis. Finally, we examined cytokine levels to determine whether IGF-1 interacts with inflammatory pathways to affect colon tumorigenesis. We observed a significant reduction in the levels of 7 out of 10 cytokines that were measured in the LID mice as compared to wild-type littermates. Results from this pilot study support the hypothesis that reductions in circulating IGF-1 levels may prevent colon tumorigenesis and affect both proliferation and apoptosis. Future experiments will investigate downstream genes of the IGF-1 receptor.
Collapse
Affiliation(s)
- Susan E Olivo-Marston
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, NIH, Bethesda, Maryland 20892-4258, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Su HM, Hsieh PH, Chen HF. A maternal high n-6 fat diet with fish oil supplementation during pregnancy and lactation in rats decreases breast cancer risk in the female offspring. J Nutr Biochem 2009; 21:1033-7. [PMID: 19954943 DOI: 10.1016/j.jnutbio.2009.08.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 08/14/2009] [Accepted: 08/20/2009] [Indexed: 10/20/2022]
Abstract
The timing of dietary fat intake may modify breast cancer risk. In addition, n-3 fatty acids reduce, and n-6 fatty acids increase, the risk of breast cancer and a maternal high n-6 fat diet results in a greater risk of breast cancer in the female offspring. We hypothesized that the timing of n-3 fatty acid-enriched fish oil supplementation would be important for reducing the risk of breast cancer. Female rats were fed to a high n-6 fat diet containing 20% of the sunflower oil by weight during pregnancy and lactation, and the female offspring were exposed to fish oil by oral gavage either during the perinatal period via maternal intake or during puberty or adulthood. Exposure during the perinatal period to a maternal high n-6 fat diet with fish oil supplementation significantly reduced the incidence of carcinogen-induced mammary tumors in the female offspring compared to a maternal high n-6 fat diet with no fish oil supplementation or fish oil supplementation later in life (P=.0228 by Cox proportional hazards model). We found that a maternal high n-6 fat diet during pregnancy is more important in increasing the risk of mammary tumors in the female offspring than a maternal high n-6 fat diet during lactation. This study suggests that fish oil supplementation during the perinatal period decreases the effect of a maternal high n-6 fat diet on subsequent carcinogen-induced mammary tumor risk, whereas fish oil supplementation during puberty or adulthood does not.
Collapse
Affiliation(s)
- Hui-Min Su
- Department of Physiology, National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | | | | |
Collapse
|
48
|
Medvedovic M, Gear R, Freudenberg JM, Schneider J, Bornschein R, Yan M, Mistry MJ, Hendrix H, Karyala S, Halbleib D, Heffelfinger S, Clegg DJ, Anderson MW. Influence of fatty acid diets on gene expression in rat mammary epithelial cells. Physiol Genomics 2009; 38:80-8. [PMID: 19351911 PMCID: PMC2696152 DOI: 10.1152/physiolgenomics.00007.2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Accepted: 04/01/2009] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND This study examines the impact of dietary fatty acids on regulation of gene expression in mammary epithelial cells before and during puberty. METHODS Diets primarily consisted of n-9 monounsaturated fatty acids (olive oil), n-6 polyunsaturated fatty acids (safflower), saturated acids (butter), and the reference AIN-93G diet (soy oil). The dietary regimen mimics the repetitive nature of fatty acid exposure in Western diets. Diet-induced changes in gene expression were examined in laser capture microdissected mammary ductal epithelial cells at day of weaning and end of puberty. PCNA immunohistochemistry analysis compared proliferation rates between diets. RESULTS Genes differentially expressed between each test diets and the reference diet were significantly enriched by cell cycle genes. Some of these genes were involved in activation of the cell cycle pathway or the G2/M check point pathway. Although there were some differences in the level of differential expression, all diets showed qualitatively the same pattern of differential expression compared to the reference diet. Cluster analysis identified an expanded set of cell cycle as well as immunity and sterol metabolism related clusters of differentially expressed genes. CONCLUSION Fatty acid-enriched diets significantly upregulated proliferation above normal physiological levels during puberty. Higher cellular proliferation during puberty caused by enriched fatty acid diets poses a potential increase risk of mammary cancer in later life. The human homologs of 27 of 62 cell cycle rat genes are included in a human breast cancer cluster of 45 cell cycle genes, further emphasizing the importance of our findings in the rat model.
Collapse
Affiliation(s)
- M Medvedovic
- Department of Cancer and Cell Biology, University of Cincinnati, Cincinnati, Ohio 45267-0521, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Olivo-Marston SE, Zhu Y, Lee RY, Cabanes A, Khan G, Zwart A, Wang Y, Clarke R, Hilakivi-Clarke L. Gene signaling pathways mediating the opposite effects of prepubertal low-fat and high-fat n-3 polyunsaturated fatty acid diets on mammary cancer risk. Cancer Prev Res (Phila) 2009; 1:532-45. [PMID: 19139003 DOI: 10.1158/1940-6207.capr-08-0030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In rats, prepubertal exposure to low-fat diet containing n-3 polyunsaturated fatty acids (PUFA) reduces mammary cell proliferation, increases apoptosis, and lowers risk of mammary tumors in adulthood, whereas prepubertal high-fat n-3 PUFA exposure has opposite effects. To identify signaling pathways mediating these effects, we performed gene microarray analyses and determined protein levels of genes related to mammary epithelial cell proliferation. Nursing female rats and rat pups were fed low-fat (16% energy from fat) or high-fat (39% energy from fat) n-3 or n-6 PUFA diets between postnatal days 5 and 24. cDNA gene expression microarrays were used to identify global changes in the mammary glands of 50-day-old rats. Differences in gene expression were confirmed by real-time quantitative PCR, and immunohistochemistry was used to assess changes in the peroxisome proliferator-activated receptor gamma and cyclin D1 levels. DNA damage was determined by 8-hydroxy-2'-deoxyguanosine assay. Expressions of the antioxidant genes thioredoxin, heme oxygenase, NADP-dependent isocitrate dehydrogenase, and metallothionein III, as well as peroxisome proliferator-activated receptor gamma protein, were increased in the mammary glands of 50-day-old rats prepubertally fed the low-fat n-3 PUFA diet. Prepubertal exposure to the high-fat n-3 PUFA diet increased DNA damage and cyclin D1 protein and reduced expression of BRCA1 and cardiotrophin-1. Reduction in mammary tumorigenesis among rats prepubertally fed a low-fat n-3 PUFA diet was associated with an up-regulation of antioxidant genes, whereas the increase in mammary tumorigenesis in the high-fat n-3 PUFA fed rats was linked to up-regulation of genes that induce cell proliferation and down-regulation of genes that repair DNA damage and induce apoptosis.
Collapse
Affiliation(s)
- Susan E Olivo-Marston
- Cancer Prevention Fellowship Program, Office of Preventive Oncology, Division of Cancer Prevention, National Cancer Institute/NIH, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Comparison of biochemical effects of statins and fish oil in brain: the battle of the titans. ACTA ACUST UNITED AC 2007; 56:443-71. [PMID: 17959252 DOI: 10.1016/j.brainresrev.2007.09.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2007] [Revised: 09/16/2007] [Accepted: 09/17/2007] [Indexed: 11/20/2022]
Abstract
Neural membranes are composed of glycerophospholipids, sphingolipids, cholesterol and proteins. The distribution of these lipids within the neural membrane is not random but organized. Neural membranes contain lipid rafts or microdomains that are enriched in sphingolipids and cholesterol. These rafts act as platforms for the generation of glycerophospholipid-, sphingolipid-, and cholesterol-derived second messengers, lipid mediators that are necessary for normal cellular function. Glycerophospholipid-derived lipid mediators include eicosanoids, docosanoids, lipoxins, and platelet-activating factor. Sphingolipid-derived lipid mediators include ceramides, ceramide 1-phosphates, and sphingosine 1-phosphate. Cholesterol-derived lipid mediators include 24-hydroxycholesterol, 25-hydroxycholesterol, and 7-ketocholesterol. Abnormal signal transduction processes and enhanced production of lipid mediators cause oxidative stress and inflammation. These processes are closely associated with the pathogenesis of acute neural trauma (stroke, spinal cord injury, and head injury) and neurodegenerative diseases such as Alzheimer disease. Statins, the HMG-CoA reductase inhibitors, are effective lipid lowering agents that significantly reduce risk for cardiovascular and cerebrovascular diseases. Beneficial effects of statins in neurological diseases are due to their anti-excitotoxic, antioxidant, and anti-inflammatory properties. Fish oil omega-3 fatty acids, eicosapentaenoic acid and docosahexaenoic acid, have similar anti-excitotoxic, antioxidant and anti-inflammatory effects in brain tissue. Thus the lipid mediators, resolvins, protectins, and neuroprotectins, derived from eicosapentaenoic acid and docosahexaenoic acid retard neuroinflammation, oxidative stress, and apoptotic cell death in brain tissue. Like statins, ingredients of fish oil inhibit generation of beta-amyloid and provide protection from oxidative stress and inflammatory processes. Collective evidence suggests that antioxidant, anti-inflammatory, and anti-apoptotic properties of statins and fish oil contribute to the clinical efficacy of treating neurological disorders with statins and fish oil. We speculate that there is an overlap between neurochemical events associated with neural cell injury in stroke and neurodegenerative diseases. This commentary compares the neurochemical effects of statins with those of fish oil.
Collapse
|