1
|
Joyce R, Visvader JE. Cells-of-Origin of Breast Cancer and Intertumoral Heterogeneity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:151-165. [PMID: 39821025 DOI: 10.1007/978-3-031-70875-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Both intrinsic and extrinsic mechanisms underpin the profound intertumoral heterogeneity in breast cancer. Increasing evidence suggests that the intrinsic characteristics of breast epithelial precursor cells may influence tumour phenotype. These "cells-of-origin" of cancer preside in normal breast tissue and are uniquely susceptible to mutagenesis upon exposure to distinct oncogenic stimuli. Notably, molecular profiling studies have revealed strong concordance between the gene expression profiles of breast cancer subtypes and discrete cell types within the normal breast epithelium. Further characterisation of cells-of-origin of breast cancer requires comprehensive delineation of the normal mammary stem cell hierarchy. To this end, mouse models have provided valuable tools for exploring stem and progenitor cell function and identifying potential targets of neoplastic transformation via in vivo lineage-tracing studies. Nonetheless, the murine mammary differentiation hierarchy does not fully recapitulate human biology, and complementary studies using patient-direct breast tissue are critical. There is also accumulating evidence that extrinsic factors such as the microenvironment of premalignant cells can influence tumour initiation, highlighting opportunities for targeting cancer cells-of-origin via deconvolution of the premalignant epithelial niche. Pertinently, the identification of premalignant clones and targetable molecular perturbations responsible for driving their oncogenic transformation has critical implications for disease management and prevention.
Collapse
Affiliation(s)
- Rachel Joyce
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Wurundjeri Country, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Wurundjeri Country, Melbourne, Australia
| | - Jane E Visvader
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Wurundjeri Country, Melbourne, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Wurundjeri Country, Melbourne, Australia.
| |
Collapse
|
2
|
Juarez MN, McDermott A, Wade MG, Plante I. Exposure to brominated flame retardants in utero and through lactation delays the development of DMBA-induced mammary cancer: potential effects on subtypes? Front Endocrinol (Lausanne) 2024; 15:1429142. [PMID: 39610845 PMCID: PMC11602300 DOI: 10.3389/fendo.2024.1429142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/21/2024] [Indexed: 11/30/2024] Open
Abstract
Introduction Brominated flame retardants (BFRs) are chemical compounds used to reduce the flammability of various products; some BFRs exhibit endocrine-disrupting properties and can leach into the environment leading to human and wildlife exposure. The mammary gland has specific vulnerability windows during which it is more sensitive to the effects of endocrine disrupting compounds (EDCs), such as the in utero life, puberty and pregnancy. Our previous studies revealed precocious mammary gland development, disruptions in junctional proteins, and altered proliferation-apoptosis balance during puberty in rats exposed to BFRs in utero and through lactation. Such effects have been associated with increased mammary cancer risk. Objective The current study aimed to determine if in utero and lactational exposure to BFRs renders the mammary gland more susceptible to 7,12-dimethylbenz[a]anthracene (DMBA)-induced mammary cancer. Methods Dams were exposed to a BFRs mixture (0. 0.06 or 60 mg/kg/day), and mammary cancer was induced in pups using DMBA at post-natal day 46. Tumors onset and growth were monitored, and tumors were characterized using histology and molecular biology. Results Although BFRs exposure did not significantly affect mammary tumor number or burden, it showed significant delay in mammary tumor onset and growth in BFR-exposed animal. These effects could potentially be due to BFRs' impact on cellular responses, DMBA metabolism, or mammary gland shift of the sensitivity window. Molecular analysis of mammary tumors showed a shift in the ratio of luminal A, luminal B, and (HER2)-enriched tumors, and an increase in triple-negative breast cancer (TNBC) subtypes in BFR-exposed animals. Additionally, BFRs exposure showed lung lesions indicative of inflammation, independent of mammary cancer development. Conclusion Our study highlights the complex relationship between BFRs exposure and mammary cancer risk, emphasizing the need for further investigation into underlying mechanisms and long-term effects of BFRs on mammary gland development and carcinogenesis.
Collapse
MESH Headings
- Animals
- Female
- Flame Retardants/toxicity
- 9,10-Dimethyl-1,2-benzanthracene/toxicity
- Pregnancy
- Lactation
- Rats
- Prenatal Exposure Delayed Effects/chemically induced
- Prenatal Exposure Delayed Effects/pathology
- Mammary Neoplasms, Experimental/chemically induced
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/metabolism
- Rats, Sprague-Dawley
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/pathology
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/metabolism
- Endocrine Disruptors/toxicity
- Carcinogens/toxicity
- Halogenated Diphenyl Ethers/toxicity
- Maternal Exposure/adverse effects
Collapse
Affiliation(s)
- Melany N. Juarez
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Alec McDermott
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Michael G. Wade
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, Canada
| | - Isabelle Plante
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| |
Collapse
|
3
|
Rakoczy K, Kaczor J, Sołtyk A, Jonderko L, Sędzik M, Lizon J, Lewandowska A, Saczko M, Kulbacka J. Pregnancy, abortion, and birth control methods' complicity with breast cancer occurrence. Mol Cell Endocrinol 2024; 590:112264. [PMID: 38705365 DOI: 10.1016/j.mce.2024.112264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/05/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Reproductive factors play significantly important roles in determining the breast cancer (BC) risk. The impact of pregnancy, abortion, and birth control methods on tumor development remains unclear. It has been found that early full-term pregnancies in young women can lower their lifetime risk of developing the type of cancer in question. However, having a first full-term pregnancy at an older age can increase this risk. The relationship between pregnancy and breast cancer (BC) is, however, much more complicated. Both induced and spontaneous abortions lead to sudden changes in hormonal balance, which could cause different effects on sensitive breast epithelial cells, making abortion a potential risk factor for breast cancer. The influence of hormonal contraception on carcinogenesis is not comprehensively understood, and therefore, more exhaustive analysis of existing data and further investigation is needed. This review explores how the mentioned reproductive factors affect the risk of breast cancer (BC), focusing on the molecular mechanisms that contribute to its complexity. By comprehending this intricate network of relationships, we can develop new strategies for predicting and treating the disease.
Collapse
Affiliation(s)
- Katarzyna Rakoczy
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Justyna Kaczor
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Adam Sołtyk
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Laura Jonderko
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Mikołaj Sędzik
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Julia Lizon
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Anna Lewandowska
- Faculty of Medicine, Wroclaw Medical University, J. Mikulicza-Radeckiego 5, 50-345, Wroclaw, Poland
| | - Małgorzata Saczko
- A. Falkiewicz Specialist Hospital in Wroclaw, Warszawska 2, 52-114 Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland; Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Santariškių 5, 08410, Vilnius, Lithuania.
| |
Collapse
|
4
|
Mustafa M, Sarfraz S, Saleem G, Khan TA, Shahid D, Taj S, Amir N. Beyond Milk and Nurture: Breastfeeding's Powerful Impact on Breast Cancer. Geburtshilfe Frauenheilkd 2024; 84:541-554. [PMID: 38884025 PMCID: PMC11175834 DOI: 10.1055/a-2313-0637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/21/2024] [Indexed: 06/18/2024] Open
Abstract
Breast cancer (BC) stands as a global concern, given its high incidence and impact on women's mortality. This complex disease has roots in various risk factors, some modifiable and others not. Understanding and identifying these factors can be instrumental in both preventing BC and improving survival rates. Remarkably, women's reproductive behaviors have emerged as critical determinants of BC susceptibility. Numerous studies have shed light on how aspects including age of menarche, first pregnancy and menopause along with number of pregnancies, hormone replacement therapies, can influence one's risk of developing BC. Furthermore, the act of breastfeeding and its duration have shown an inverse relationship with BC risk. This review delves into the biological and molecular mechanisms associated with breastfeeding that contribute to BC protection. It highlights the role of endocrine processes triggered by suckling stimulation, the gradual onset of lactational amenorrhea, delayed weaning, reduced lifetime menstrual cycles, chromosomal repair mechanisms, and immunological events throughout the lactation cycle. These insights provide a potential explanation for the protective effects conferred by breastfeeding against breast carcinomas.
Collapse
Affiliation(s)
- Muhammad Mustafa
- Kauser Abdulla Malik School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Sadaf Sarfraz
- Kauser Abdulla Malik School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Gullelalah Saleem
- Kauser Abdulla Malik School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Touqeer Ahmad Khan
- Kauser Abdulla Malik School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Damiya Shahid
- Kauser Abdulla Malik School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Saba Taj
- Kauser Abdulla Malik School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Noor Amir
- Kauser Abdulla Malik School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan
| |
Collapse
|
5
|
Silva J, Faustino-Rocha AI, Duarte JA, Oliveira PA. Realistic aspects behind the application of the rat model of chemically-induced mammary cancer: Practical guidelines to obtain the best results. Vet World 2023; 16:1222-1230. [PMID: 37577198 PMCID: PMC10421542 DOI: 10.14202/vetworld.2023.1222-1230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/04/2023] [Indexed: 08/15/2023] Open
Abstract
Cancer is one of the most important public health problems worldwide. Despite the great contribution of in-vitro studies for biomedical research, animals are essential to study diseases' biopathology and diagnosis, and searching for new preventive and therapeutic strategies. Breast cancer is currently the most common cancer globally, accounting for 12.5% of all new annual cancer cases worldwide. Although the rat model of mammary cancer chemically-induced is widely used to study this disease, there is a lack of standardization in procedures for cancer induction, sample collection, and analysis. Therefore, it is important to provide a practical guide for researchers aiming to work with this model to make the analysis of results more uniform. Thus, in this review, we provide the researchers with a detailed step-by-step guide to implement a rat model of mammary cancer, based on our wide experience in this field, to obtain the best results, maximum throughput of each experiment, and easy comparison among researches.
Collapse
Affiliation(s)
- Jéssica Silva
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-food Production (Inov4Agro), Vila Real, Portugal
| | - Ana I. Faustino-Rocha
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-food Production (Inov4Agro), Vila Real, Portugal
- Department of Zootechnics, School of Sciences and Technology, University of Évora, Portugal
- Comprehensive Health Research Center, University of Évora, Évora, Portugal
| | - José Alberto Duarte
- Research Center for Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, Porto, Portugal
- Toxicology Research Unit (TOXRUN), Advanced Polytechnic and University Cooperative (CESPU), Gandra, Portugal
| | - Paula A. Oliveira
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-food Production (Inov4Agro), Vila Real, Portugal
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| |
Collapse
|
6
|
Ferreira T, Gama A, Seixas F, Faustino-Rocha AI, Lopes C, Gaspar VM, Mano JF, Medeiros R, Oliveira PA. Mammary Glands of Women, Female Dogs and Female Rats: Similarities and Differences to Be Considered in Breast Cancer Research. Vet Sci 2023; 10:379. [PMID: 37368765 DOI: 10.3390/vetsci10060379] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Breast cancer is one of the most common and well-known types of cancer among women worldwide and is the most frequent neoplasm in intact female dogs. Female dogs are considered attractive models or studying spontaneous breast cancer, whereas female rats are currently the most widely used animal models for breast cancer research in the laboratory context. Both female dogs and female rats have contributed to the advancement of scientific knowledge in this field, and, in a "One Health" approach, they have allowed broad understanding of specific biopathological pathways, influence of environmental factors and screening/discovery of candidate therapies. This review aims to clearly showcase the similarities and differences among woman, female dog and female rat concerning to anatomical, physiological and histological features of the mammary gland and breast/mammary cancer epidemiology, in order to better portray breast tumorigenesis, and to ensure appropriate conclusions and extrapolation of results among species. We also discuss the major aspects that stand out in these species. The mammary glands of female dogs and women share structural similarities, especially with respect to the lactiferous ducts and lymphatic drainage. In contrast, female rats have only one lactiferous duct per nipple. A comprehensive comparison between humans and dogs is given a special focus, as these species share several aspects in terms of breast/mammary cancer epidemiology, such as age of onset, hormonal etiology, risk factors, and the clinical course of the disease. Holistically, it is clear that each species has advantages and limitations that researchers must consider during the development of experimental designs and data analysis.
Collapse
Affiliation(s)
- Tiago Ferreira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Adelina Gama
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Fernanda Seixas
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Ana I Faustino-Rocha
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Department of Zootechnics, School of Sciences and Technology, University of Évora, 7004-516 Évora, Portugal
- Comprehensive Health Research Center, 7004-516 Évora, Portugal
| | - Carlos Lopes
- Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto.CCC), 4200-072 Porto, Portugal
- Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- Research Department of the Portuguese League against Cancer-Regional Nucleus of the North (Liga Portuguesa Contra o Cancro-Núcleo Regional do Norte), 4200-177 Porto, Portugal
- Virology Service, Portuguese Institute of Oncology (IPO), 4200-072 Porto, Portugal
- Biomedical Research Center (CEBIMED), Faculty of Health Sciences of the Fernando Pessoa University, 4249-004 Porto, Portugal
| | - Paula A Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| |
Collapse
|
7
|
Pederson HJ, Batur P. Use of exogenous hormones in those at increased risk for breast cancer: contraceptive and menopausal hormones in gene carriers and other high-risk patients. Menopause 2023; 30:341-347. [PMID: 36626703 DOI: 10.1097/gme.0000000000002136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
IMPORTANCE AND OBJECTIVE Addressing the hormonal needs of individuals at increased risk of breast cancer (BC) can be a challenge. Observational, prospective, and case-control data support the safety of hormonal contraception in women, often with the added benefits of ovarian and endometrial cancer risk reduction. The majority of data on menopausal hormone therapy (HT) in the highest-risk patients comes from studies of patients with pathogenic variants in BRCA1 and BRCA2 who undergo early surgical menopause. The benefits of risk-reducing salpingo-oophorectomy are not minimized by HT, whereas its use mitigates accelerated osteoporosis and cardiovascular disease. In other patients at increased risk, such as with family history, studies have shown little risk with significant benefit. METHODS We review evidence to help women's health practitioners aid patients in making choices. The paper is divided into four parts: 1, contraception in the very high-risk patient (ie, with a highly penetrant BC predisposition gene); 2, contraception in other patients at increased risk; 3, menopausal HT in the gene carrier; and 4, HT in other high-risk patients. DISCUSSION AND CONCLUSION Women at increased risk for BC both early and later in life should be offered reassurance around the use of premenopausal and postmenopausal hormone therapies. The absolute risks associated with these therapies are low, even in the very high-risk patient, and the benefits are often substantial. Shared decision making is key in presenting options, and knowledge of the data in this area is fundamental to these discussions.
Collapse
Affiliation(s)
- Holly J Pederson
- From the Department of General Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic; and the
| | - Pelin Batur
- Cleveland Clinic Ob/Gyn and Women's Health Institute, Department of Subspecialty Women's Health, Cleveland OH
| |
Collapse
|
8
|
Su Y, Dang NM, Depypere H, Santucci-Pereira J, Gutiérrez-Díez PJ, Kanefsky J, Janssens JP, Russo J. Recombinant human chorionic gonadotropin induces signaling pathways towards cancer prevention in the breast of BRCA1/2 mutation carriers. Eur J Cancer Prev 2023; 32:126-138. [PMID: 35881946 PMCID: PMC9800649 DOI: 10.1097/cej.0000000000000763] [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: 02/04/2023]
Abstract
BACKGROUND Strategies for breast cancer prevention in women with germline BRCA1/2 mutations are limited. We previously showed that recombinant human chorionic gonadotropin (r-hCG) induces mammary gland differentiation and inhibits mammary tumorigenesis in rats. The present study investigated hCG-induced signaling pathways in the breast of young nulliparous women carrying germline BRCA1/2 mutations. METHODS We performed RNA-sequencing on breast tissues from 25 BRCA1/2 mutation carriers who received r-hCG treatment for 3 months in a phase II clinical trial, we analyzed the biological processes, reactome pathways, canonical pathways, and upstream regulators associated with genes differentially expressed after r-hCG treatment, and validated genes of interest. RESULTS We observed that r-hCG induces remarkable transcriptomic changes in the breast of BRCA1/2 carriers, especially in genes related to cell development, cell differentiation, cell cycle, apoptosis, DNA repair, chromatin remodeling, and G protein-coupled receptor signaling. We revealed that r-hCG inhibits Wnt/β-catenin signaling, MYC, HMGA1 , and HOTAIR , whereas activates TGFB/TGFBR-SMAD2/3/4, BRCA1, TP53, and upregulates BRCA1 protein. CONCLUSION Our data suggest that the use of r-hCG at young age may reduce the risk of breast cancer in BRCA1/2 carriers by inhibiting pathways associated with stem/progenitor cell maintenance and neoplastic transformation, whereas activating genes crucial for breast epithelial differentiation and lineage commitment, and DNA repair.
Collapse
Affiliation(s)
- Yanrong Su
- The Irma H Russo, MD, Breast Cancer Research Laboratory at the Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA
- These authors contributed equally: Yanrong Su, Nhi M. Dang, and Herman Depypere
| | - Nhi M. Dang
- The Irma H Russo, MD, Breast Cancer Research Laboratory at the Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA
- These authors contributed equally: Yanrong Su, Nhi M. Dang, and Herman Depypere
| | - Herman Depypere
- Department of Gynecology, Breast and Menopause clinic, University Hospital of Ghent, Corneel Heymanslaan 10, 9000 Ghent, Belgium
- These authors contributed equally: Yanrong Su, Nhi M. Dang, and Herman Depypere
| | - Julia Santucci-Pereira
- The Irma H Russo, MD, Breast Cancer Research Laboratory at the Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | | | - Joice Kanefsky
- The Irma H Russo, MD, Breast Cancer Research Laboratory at the Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Jaak Ph. Janssens
- European Cancer Prevention Organization, University of Hasselt, Klein Hilststraat 5, 3500 Hasselt, Belgium
| | - Jose Russo
- The Irma H Russo, MD, Breast Cancer Research Laboratory at the Fox Chase Cancer Center-Temple Health, 333 Cottman Avenue, Philadelphia, PA 19111, USA
- Dr. Russo conceived the study and supervised the work. Dr. Russo passed away on September 24, 2021
| |
Collapse
|
9
|
Long-term impact of hypothyroidism during gestation and lactation on the mammary gland. J Dev Orig Health Dis 2023; 14:122-131. [PMID: 35670520 DOI: 10.1017/s2040174422000320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The functional differentiation of the mammary gland (MG) is fundamental for the prevention of mammary pathologies. This process occurs throughout pregnancy and lactation, making these stages key events for the study of pathologies associated with development and differentiation. Many studies have investigated the link between mammary pathologies and thyroid diseases, but most have ignored the role of thyroid hormone (TH) in the functional differentiation of the MG. In this work, we show the long-term impact of hypothyroidism in an animal model whose lactogenic differentiation occurred at low TH levels. We evaluated the ability of the MG to respond to hormonal control and regulate cell cycle progression. We found that a deficit in TH throughout pregnancy and lactation induces a long-term decrease in Rb phosphorylation, increases p53, p21, Cyclin D1 and Ki67 expression, reduces progesterone receptor expression, and induces nonmalignant lesions in mammary tissue. This paper shows the importance of TH level control during mammary differentiation and its long-term impact on mammary function.
Collapse
|
10
|
In Vitro and In Vivo Antiproliferative Actions of Solanum gilo Raddi (Solanaceae) Fruit Extract on Breast Tissues. Adv Pharmacol Pharm Sci 2022; 2022:6834626. [PMID: 36199376 PMCID: PMC9529474 DOI: 10.1155/2022/6834626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/05/2022] [Indexed: 12/24/2022] Open
Abstract
Background Menopause is a normal event characterized by a drop in estrogen's production, leading to numerous symptoms. To face these later, women rely on hormone replacement therapy (HRT), which alleviates numerous menopausal symptoms. Unfortunately, long-term exposure to estrogens is associated with an increase in endometrial and breast cancers. This study dealt with the evaluation of in vitro and in vivo antiproliferative effects of Solanum gilo Raddi, a plant used in folk medicine to treat tumors in Cameroon. Materials and Methods The in vitro antiproliferative effect of S. gilo fruit extract was investigated through the well-characterized MTT assay in one normal and three cancerous breast cells. For the in vivo study, one normal group (NOR) of rats received distilled water (vehicle), and five other groups (n = 6) were treated either with tamoxifen (3.3 mg/kg BW) as standard or with the vehicle (negative control) or S. gilo fruit hydroethanolic extract (125, 250, and 500 mg/kg BW). The treatments were administered concomitantly with the E2V to induce breast hyperplasia for 16 weeks, and the endpoints were the histopathology of the mammary glands and some biochemical parameters. Results The S. gilo extract significantly inhibited human (MCF-7 and MDA-MB-231) and rodent (4T1) breast carcinoma cell growth. Rats exposed only to E2V presented atypical mammary hyperplasia compared to the normal parenchyma observed in normal rats. While rats treated with S. gilo extract at the dose of 125 mg/kg BW showed a microarchitecture of mammary glands with moderate hyperplasia, the higher doses (250 and 500 mg/kg) inhibited mammary gland hyperplasia compared to the E2V group. Conclusion S. gilo fruit extract has antiproliferative constituents that could help to fight against estrogen-dependent breast cancer, thanks to their ability to scavenge free radicals, as exhibited in this study.
Collapse
|
11
|
Miller JL, Bartlett AP, Harman RM, Majhi PD, Jerry DJ, Van de Walle GR. Induced mammary cancer in rat models: pathogenesis, genetics, and relevance to female breast cancer. J Mammary Gland Biol Neoplasia 2022; 27:185-210. [PMID: 35904679 DOI: 10.1007/s10911-022-09522-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 10/16/2022] Open
Abstract
Mammary cancer, or breast cancer in women, is a polygenic disease with a complex etiopathogenesis. While much remains elusive regarding its origin, it is well established that chemical carcinogens and endogenous estrogens contribute significantly to the initiation and progression of this disease. Rats have been useful models to study induced mammary cancer. They develop mammary tumors with comparable histopathology to humans and exhibit differences in resistance or susceptibility to mammary cancer depending on strain. While some rat strains (e.g., Sprague-Dawley) readily form mammary tumors following treatment with the chemical carcinogen, 7,12-dimethylbenz[a]-anthracene (DMBA), other strains (e.g., Copenhagen) are resistant to DMBA-induced mammary carcinogenesis. Genetic linkage in inbred strains has identified strain-specific quantitative trait loci (QTLs) affecting mammary tumors, via mechanisms that act together to promote or attenuate, and include 24 QTLs controlling the outcome of chemical induction, 10 QTLs controlling the outcome of estrogen induction, and 4 QTLs controlling the outcome of irradiation induction. Moreover, and based on shared factors affecting mammary cancer etiopathogenesis between rats and humans, including orthologous risk regions between both species, rats have served as useful models for identifying methods for breast cancer prediction and treatment. These studies in rats, combined with alternative animal models that more closely mimic advanced stages of breast cancer and/or human lifestyles, will further improve our understanding of this complex disease.
Collapse
Affiliation(s)
- James L Miller
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 14853, Ithaca, NY, USA
| | - Arianna P Bartlett
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 14853, Ithaca, NY, USA
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 14853, Ithaca, NY, USA
| | - Prabin Dhangada Majhi
- Department of Veterinary & Animal Sciences, University of Massachusetts, 01003, Amherst, MA, USA
| | - D Joseph Jerry
- Department of Veterinary & Animal Sciences, University of Massachusetts, 01003, Amherst, MA, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 14853, Ithaca, NY, USA.
| |
Collapse
|
12
|
Kumar R, Abreu C, Toi M, Saini S, Casimiro S, Arora A, Paul AM, Velaga R, Rameshwar P, Lipton A, Gupta S, Costa L. Oncobiology and treatment of breast cancer in young women. Cancer Metastasis Rev 2022; 41:749-770. [PMID: 35488982 DOI: 10.1007/s10555-022-10034-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/14/2022] [Indexed: 12/20/2022]
Abstract
Female breast cancer emerged as the leading cancer type in terms of incidence globally in 2020. Although mortality due to breast cancer has improved during the past three decades in many countries, this trend has reversed in women less than 40 years since the past decade. From the biological standpoint, there is consensus among experts regarding the clinically relevant definition of breast cancer in young women (BCYW), with an age cut-off of 40 years. The idea that breast cancer is an aging disease has apparently broken in the case of BCYW due to the young onset and an overall poor outcome of BCYW patients. In general, younger patients exhibit a worse prognosis than older pre- and postmenopausal patients due to the aggressive nature of cancer subtypes, a high percentage of cases with advanced stages at diagnosis, and a high risk of relapse and death in younger patients. Because of clinically and biologically unique features of BCYW, it is suspected to represent a distinct biologic entity. It is unclear why BCYW is more aggressive and has an inferior prognosis with factors that contribute to increased incidence. However, unique developmental features, adiposity and immune components of the mammary gland, hormonal interplay and crosstalk with growth factors, and a host of intrinsic and extrinsic risk factors and cellular regulatory interactions are considered to be the major contributing factors. In the present article, we discuss the status of BCYW oncobiology, therapeutic interventions and considerations, current limitations in fully understanding the basis and underlying cause(s) of BCYW, understudied areas of BCYW research, and postulated advances in the coming years for the field.
Collapse
Affiliation(s)
- Rakesh Kumar
- Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India. .,Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India. .,Department of Medicine, Division of Hematology and Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA. .,Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.
| | - Catarina Abreu
- Department of Medical Oncology, Hospital de Santa Maria- Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Masakazu Toi
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sunil Saini
- Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India
| | - Sandra Casimiro
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Anshika Arora
- Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India
| | - Aswathy Mary Paul
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Ravi Velaga
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Pranela Rameshwar
- Department of Medicine, Division of Hematology and Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Allan Lipton
- Hematology-Oncology, Department of Medicine, Penn State University School of Medicine, Hershey, PA, USA
| | - Sudeep Gupta
- Department of Medical Oncology, Tata Memorial Centre and Homi Bhabha National Institute, Mumbai, India
| | - Luis Costa
- Department of Medical Oncology, Hospital de Santa Maria- Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal.,Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
13
|
Wang K, Ge M, Liu L, Lv H, Wang S, Jia F, Sun J. Birth weight and the risk of overall breast cancer, premenopausal and postmenopausal breast cancer in adulthood: a dose-response meta-analysis of observational studies. Menopause 2021; 29:114-124. [PMID: 34905748 DOI: 10.1097/gme.0000000000001885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
IMPORTANCE The effect of birth weight on breast cancer across different menopausal states remains unknown. OBJECTIVE The aim of this study was to systematically evaluate the association of birth weight with the risk of overall breast cancer (OBC) and premenopausal and postmenopausal breast cancer during adulthood. In parallel, the dose-response analyses were performed. EVIDENCE REVIEW Relevant studies were systematically searched from the PubMed, Embase, and the Cochrane Library databases from the inception to May 25, 2021, without language restrictions. All the results were pooled according to risk ratios (RRs). FINDINGS In total, 21 cohort studies comprising 1,139,032 participants were included. An increase in the birth weight was not associated with the risk of OBC and premenopausal and postmenopausal breast cancer. Compared with women having normal weight at birth, those with a high birth weight are likely to have an increased risk of invasive breast cancer (RR: 1.19, 95% confidence intervals: 1.03-1.38; I2: 28.6%). The dose-response analyses showed that the risk of premenopausal breast cancer increased significantly in unknown singleton status with birth weight over 2850 g (RR: 1.14 [1.02-1.30]). Similarly, postmenopausal breast cancer risk was increased in singleton births with birth weight over 3750 g (RR: 1.21 [1.00-1.47]). CONCLUSIONS AND RELEVANCE High weight at birth might be not significantly associated with the risk of OBC, premenopausal and postmenopausal breast cancer and ER+ and ER- breast cancer but is positively associated with the risk of invasive breast cancer, regardless of parity. Furthermore, with an increase in birth weight, the risk of postmenopausal breast cancer is likely to increase in the singleton births, whereas the risk of premenopausal breast cancer is likely to increase in unknown singleton status.
Collapse
Affiliation(s)
- Kang Wang
- Department of Oncology, Zaozhuang Municipal Hospital, Zaozhuang, China
| | - Mingxiu Ge
- Department of Obstetrics, Zaozhuang Municipal Hospital, Zaozhuang, China
| | - Lulu Liu
- Department of Obstetrics, Maternity and Child Health Care of Zaozhuang, Zaozhuang, China
| | - Haihong Lv
- Department of Obstetrics, Maternity and Child Health Care of Zaozhuang, Zaozhuang, China
| | - Shujuan Wang
- Department of Surgery, Maternity and Child Health Care of Zaozhuang, Zaozhuang, China
| | - Fei Jia
- Department of Internal Medicine, Maternity and Child Health Care of Zaozhuang, Zaozhuang, China
| | - Jinjun Sun
- Department of Oncology, Zaozhuang Municipal Hospital, Zaozhuang, China
| |
Collapse
|
14
|
Watson KL, Yi R, Moorehead RA. Transgenic overexpression of the miR-200b/200a/429 cluster inhibits mammary tumor initiation. Transl Oncol 2021; 14:101228. [PMID: 34562686 PMCID: PMC8473771 DOI: 10.1016/j.tranon.2021.101228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 12/24/2022] Open
Abstract
The miR-200 family consists of five members expressed as two clusters: miR-200c/141 cluster and miR-200b/200a/429 cluster. In the mammary gland, miR-200s maintain epithelial identity by decreasing the expression of mesenchymal markers leading to high expression of epithelial markers. While the loss of miR-200s is associated with breast cancer growth and metastasis the impact of miR-200 expression on mammary tumor initiation has not been investigated. Using mammary specific expression of the miR-200b/200a/429 cluster in transgenic mice, we found that elevated expression miR-200s could almost completely prevent mammary tumor development. Only 1 of 16 MTB-IGFIRba429 transgenic mice (expressing both the IGF-IR and miR-200b/200a/429 transgenes) developed a mammary tumor while 100% of MTB-IGFIR transgenic mice (expressing only the IGF-IR transgene) developed mammary tumors. RNA sequencing, qRT-PCR, and immunohistochemistry of mammary tissue from 55-day old mice found Spp1, Saa1, and Saa2 to be elevated in mammary tumors and inhibited by miR-200b/200a/429 overexpression. This study suggests that miR-200s could be used as a preventative strategy to protect women from developing breast cancer. One concern with this approach is the potential negative impact miR-200 overexpression may have on mammary function. However, transgenic overexpression of miR-200s, on their own, did not significantly impact mammary ductal development indicating the miR-200 overexpression should not significantly impact mammary function. Thus, this study provides the initial foundation for using miR-200s for breast cancer prevention and additional studies should be performed to identify strategies for increasing mammary miR-200 expression and determine whether miR-200s can prevent mammary tumor initiation by other genetic alterations.
Collapse
Affiliation(s)
- Katrina L Watson
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Rui Yi
- Department of Pathology, Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Roger A Moorehead
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
15
|
Kim H, Moon WK. Histological Findings of Mammary Gland Development and Risk of Breast Cancer in BRCA1 Mutant Mouse Models. J Breast Cancer 2021; 24:455-462. [PMID: 34652081 PMCID: PMC8561134 DOI: 10.4048/jbc.2021.24.e44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/30/2021] [Accepted: 09/27/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose The breast cancer susceptibility gene, BRCA1, is involved in normal development and carcinogenesis of mammary glands. Here, we aimed to evaluate the relationship between histological findings of mammary gland development and breast cancer risk in BRCA1 mutant mice. Methods Five BRCA1 mutant mice and five non-mutant FVB/NJ mice were used for each group of 1-month-old (pubertal), 3-month-old (fertile), and 8-month-old (menopausal) mice. In another experiment, 15 BRCA1 mutant mice were followed up to 8 months after birth and classified into tumor-bearing (11 mice) and tumor-free (4 mice) groups. Excised mammary gland tissues were stained with Carmine Alum, and the number of terminal end buds (or alveolar buds), branching density, and duct elongation were measured using image analysis programs. Differences between the two groups were assessed using paired t-test. Results One-month-old BRCA1 mutant mice showed a higher number of terminal end buds (23.8 ± 1.0 vs. 15.6 ± 0.8, p = 0.0002), branching density (11.7 ± 0.4 vs. 9.6 ± 0.5%, p = 0.0082), and duct elongation (9.7 ± 0.7 vs. 7.3 ± 0.4 mm, p = 0.0186) than controls. However, there was no difference between the 3- and 8-month-old groups. In BRCA1 mutant mice, the tumor-bearing group showed a significantly higher number of alveolar buds (142.7 ± 5.5 vs. 105.5 ± 5.4, p = 0.0008) and branching density (30.0 ± 1.0 vs. 24.1 ± 1.1%, p = 0.008) than the tumor-free group; however, duct elongation was not different (23.9 ± 0.6 vs. 23.6 ± 0.6 mm, p = 0.8099) between the groups. Conclusion BRCA1 mutant mice exhibited early pubertal mammary gland development and delayed age-related mammary gland involution was associated with breast cancer. Our results may have clinical implications for predicting breast cancer risk and developing prevention strategies for BRCA1 mutation carriers.
Collapse
Affiliation(s)
- Hyelim Kim
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Woo Kyung Moon
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Department of Radiology, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
16
|
Yusuf H, Kamarlis RK, Yusni Y, Fahriani M. The anticancer activity of ethanol extract of Chromolaena odorata leaves in 7,12-Dimethylbenz[a]anthracene in (DMBA) induced breast cancer Wistar rats (Rattus novergicus). PHARMACIA 2021. [DOI: 10.3897/pharmacia.68.e63956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background: Breast cancer chemotherapy with standard drugs such as doxorubicin will induce cardiotoxicity. Therefore, this study aims to evaluate the anticancer activity of C. odorata leaves extract in DMBA induced breast cancer on rats.
Methods: Seven groups of Rattus novergicus were used: Four treatment groups of C. odorata extract (500, 1000, 2000, and 4000 mg/kg BW), normal control, breast cancer control, and doxorubicin treatment group. The number, volume, and weight of the nodule and the rats’ body weight were compared among groups. Data was analyzed using paired t-test or one-way ANOVA with post hoc analysis as appropriate.
Results: Significant decline of the number, volume, and weight of cancer nodules was observed in the treatment group (p < 0.001). The weight of the cancer nodule at week 16th was also significantly reduced in GCo2000 compared to Gdoxo (p < 0.0001). A significant increase in body weight was also dose-dependent, especially at week 11th (p < 0.05 in all comparisons) and week 16th (p < 0.001 in all comparisons).
Conclusion: This study suggested that the ethanol extract of C. odorata leaves has anticancer and antiproliferative activity.
Collapse
|
17
|
Pubertal Growth, IGF-1, and Windows of Susceptibility: Puberty and Future Breast Cancer Risk. J Adolesc Health 2021; 68:517-522. [PMID: 32888770 PMCID: PMC7902462 DOI: 10.1016/j.jadohealth.2020.07.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Risk markers for breast cancer include earlier onset of menarche (age at menarche [AAM]) and peak height velocity (PHV). Insulin-like growth factor-1 (IGF-1) is associated with pubertal milestones, as well as cancer risk. This study examined the relationships between pubertal milestones associated with breast cancer risk and hormone changes in puberty. METHODS This is a longitudinal study of pubertal maturation in 183 girls, recruited at ages 6-7, followed up between 2004 and 2018. Measures included age at onset of puberty, and adult height attained; PHV; AAM; adult height, and serum IGF-1, and estrone-to-androstenedione (E:A) ratio. RESULTS PHV was greatest in early, and least in late maturing girls; length of the pubertal growth spurt was longest in early, and shortest in late maturing girls. Earlier AAM was related to greater PHV. IGF-1 concentrations tracked significantly during puberty; higher IGF-1 was related to earlier age of PHV, earlier AAM, greater PHV, and taller adult height. Greater E:A ratio was associated with earlier AAM. CONCLUSIONS Factors driving the association of earlier menarche and pubertal growth with breast cancer risk may be explained through a unifying concept relating higher IGF-1 concentrations, greater lifelong estrogen exposure, and longer pubertal growth period, with an expanded pubertal window of susceptibility.
Collapse
|
18
|
Ni Y, Chen Q, Cai J, Xiao L, Zhang J. Three lactation-related hormones: Regulation of hypothalamus-pituitary axis and function on lactation. Mol Cell Endocrinol 2021; 520:111084. [PMID: 33232781 DOI: 10.1016/j.mce.2020.111084] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/08/2020] [Accepted: 11/11/2020] [Indexed: 01/10/2023]
Abstract
The endocrine system plays a central role in many aspects of lactation, including mammogenesis (mammary gland development), lactogenesis (onset of lactation), and galactopoiesis (maintenance of milk secretion). Many hormones of the endocrine system directly or indirectly regulate lactation process. The secretion of prolactin (PRL), one of the most important lactation-related hormones, is inhibited by hypothalamus-pituitary dopaminergic system and stimulated by hypothalamus-pituitary oxytocinergic system. This hormone is essential in all stages of lactation. The growth hormone (GH) regulates metabolism and the distribution of nutrients between tissues mammary glands, and stimulates the production of IGF-I from the liver which binds to IGF-IR of mammary epithelial cells (MECs) to indirectly promote lactation. The synthesis and secretion of estrogen (E) are affected by the hypothalamus-pituitary axis. The hormone regulates duct morphogenesis and MECs proliferation. It also modulates the synthesis and secretion of PRL and GH, which together regulate the lactation in female animals. In this article, we reviewed the three main lactation-related hormones (PRL, GH, and E), summarize their regulation by the hypothalamus-pituitary axis and how they influence lactation.
Collapse
Affiliation(s)
- Yifan Ni
- College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Qiangqiang Chen
- College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Jianfeng Cai
- College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Lixia Xiao
- College of Animal Science, Zhejiang University, Hangzhou, 310058, China
| | - Jinzhi Zhang
- College of Animal Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
19
|
Berger E, Maitre N, Romana Mancini F, Baglietto L, Perduca V, Colineaux H, Sieri S, Panico S, Sacerdote C, Tumino R, Vineis P, Boutron-Ruault MC, Severi G, Castagné R, Delpierre C. The impact of lifecourse socio-economic position and individual social mobility on breast cancer risk. BMC Cancer 2020; 20:1138. [PMID: 33228587 PMCID: PMC7684912 DOI: 10.1186/s12885-020-07648-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/17/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Women with an advantaged socioeconomic position (SEP) have a higher risk of developing breast cancer (BC). The reasons for this association do not seem to be limited to reproductive factors and remain to be understood. We aimed to investigate the impact of lifecourse SEP from childhood and social mobility on the risk of BC considering a broad set of potential mediators. METHODS We used a discovery-replication strategy in two European prospective cohorts, E3N (N = 83,436) and EPIC-Italy (N = 20,530). In E3N, 7877 women were diagnosed with BC during a median 24.4 years of follow-up, while in EPIC-Italy, 893 BC cases were diagnosed within 15.1 years. Hazard ratios (HR) were estimated using Cox proportional hazard models on imputed data. RESULTS In E3N, women with higher education had a higher risk of BC (HR [95%CI] = 1.21 [1.12, 1.30]). This association was attenuated by adjusting for reproductive factors, in particular age at first childbirth (HR[95%CI] = 1.13 [1.04, 1.22]). Health behaviours, anthropometric variables, and BC screening had a weaker effect on the association. Women who remained in a stable advantaged SEP had a higher risk of BC (HR [95%CI] = 1.24 [1.07; 1.43]) attenuated after adjustment for potential mediators (HR [95%CI] = 1.13 [0.98; 1.31]). These results were replicated in EPIC-Italy. CONCLUSIONS These results confirm the important role of reproductive factors in the social gradient in BC risk, which does not appear to be fully explained by the large set of potential mediators, including cancer screening, suggesting that further research is needed to identify additional mechanisms.
Collapse
Affiliation(s)
- Eloïse Berger
- UMR LEASP, Université de Toulouse III, UPS, Inserm, Toulouse, France.
| | - Noële Maitre
- CESP, Faculté de Médecine, Université Paris-Saclay, UVSQ, INSERM, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Francesca Romana Mancini
- CESP, Faculté de Médecine, Université Paris-Saclay, UVSQ, INSERM, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Laura Baglietto
- CESP, Faculté de Médecine, Université Paris-Saclay, UVSQ, INSERM, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Vittorio Perduca
- CESP, Faculté de Médecine, Université Paris-Saclay, UVSQ, INSERM, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
- Université de Paris, CNRS, MAP5 UMR 8145, F-75006, Paris, France
| | - Hélène Colineaux
- UMR LEASP, Université de Toulouse III, UPS, Inserm, Toulouse, France
- Epidemiology Department, Toulouse Teaching Hospital, Toulouse, France
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Salvatore Panico
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Department, Provicial Health Authority (ASP) Ragusa, Ragusa, Italy
| | - Paolo Vineis
- Department of Epidemiology and Biostatistics, Imperial College London, MRC-PHE Centre for Environment and Health, School of Public Health, London, UK
- Italian Institute for Genomic Medicine, Torino, Italy
| | - Marie-Christine Boutron-Ruault
- CESP, Faculté de Médecine, Université Paris-Saclay, UVSQ, INSERM, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
| | - Gianluca Severi
- CESP, Faculté de Médecine, Université Paris-Saclay, UVSQ, INSERM, Villejuif, France
- Gustave Roussy, F-94805, Villejuif, France
- Department of Statistics, Computer Science and Applications "G. Parenti" (DISIA), University of Florence, Florence, Italy
| | - Raphaële Castagné
- UMR LEASP, Université de Toulouse III, UPS, Inserm, Toulouse, France
| | - Cyrille Delpierre
- UMR LEASP, Université de Toulouse III, UPS, Inserm, Toulouse, France
| |
Collapse
|
20
|
Cytotoxic and cancer chemopreventive potentials of the Anthonotha macrophylla P. Beauv aqueous extract on 7,12-dimethylbenz[a]anthracene-induced breast cancer in rats. Biologia (Bratisl) 2020. [DOI: 10.2478/s11756-020-00607-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
21
|
Nguedia MY, Tueche AB, Yaya AJG, Yadji V, Ndinteh DT, Njamen D, Zingue S. Daucosterol from Crateva adansonii DC (Capparaceae) reduces 7,12-dimethylbenz(a)anthracene-induced mammary tumors in Wistar rats. ENVIRONMENTAL TOXICOLOGY 2020; 35:1125-1136. [PMID: 32449848 DOI: 10.1002/tox.22948] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/29/2020] [Accepted: 05/08/2020] [Indexed: 06/11/2023]
Abstract
This study aimed to evaluate the in vivo anticancer effects of daucosterol which was earlier reported to possess in vitro anticancer effects. Breast tumor was induced in 30 rats using the environmental carcinogen 7,12-dimethylbenz(a)anthracene (DMBA) while 6 control rats received olive oil (NOR). Animals with palpable tumors were randomized into five groups (n = 6) each as follows: negative control group treated with the vehicle (DMBA); positive control group treated with 5 mg/kg BW doxorubicin (DOXO + DMBA); three groups treated with daucosterol at doses of 2.5, 5, and 10 mg/kg BW (DAU + DMBA). Treatment lasted 28 days afterward, tumor (mass, volume, cancer antigen [CA] 15-3 level and histoarchitecture), hematological and toxicological parameters were examined. The tumor volume gradually increased in the DMBA group during the 28 days, with a tumor volume gain of ∼390 cm3 . Daucosterol at all doses reduced tumor volume (∼133.7 cm3 at 10 mg/kg) as well as protein, malondialdehyde (MDA), and CA 15-3 levels compared to DMBA rats. Tumor sections in daucosterol-treated rats showed a lower proliferation of mammary ducts with mild (5 and 10 mg/kg) to moderate (2.5 mg/kg) inflammatory responses. Moreover, it exhibited an antioxidant effect, evidenced by a significant and dose-dependent decreased in MDA levels, as well as an increase in catalase activity compared to the DMBA group. Daucosterol showed for the first time in vivo antitumor effects that corroborate its previous in vitro effects.
Collapse
MESH Headings
- 9,10-Dimethyl-1,2-benzanthracene/toxicity
- Animals
- Antineoplastic Agents, Phytogenic/isolation & purification
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/therapeutic use
- Antioxidants/metabolism
- Capparaceae/chemistry
- Carcinogens/toxicity
- Dose-Response Relationship, Drug
- Female
- Mammary Neoplasms, Experimental/chemically induced
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Molecular Structure
- Plant Bark/chemistry
- Rats
- Rats, Wistar
- Sitosterols/isolation & purification
- Sitosterols/pharmacology
- Sitosterols/therapeutic use
Collapse
Affiliation(s)
- Merline Ymele Nguedia
- Laboratory of Physiology and Natural Products Research, Department of Life and Earth Sciences, Higher Teachers' Training College, University of Maroua, Maroua, Cameroon
| | - Alain Brice Tueche
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé, Yaounde, Cameroon
| | - Abel Joël Gbaweng Yaya
- Department of Chemistry, Faculty of Science, University of Ngaoundere, Ngaoundere, Cameroon
| | - Vincent Yadji
- Laboratory of Physiology and Natural Products Research, Department of Life and Earth Sciences, Higher Teachers' Training College, University of Maroua, Maroua, Cameroon
| | - Derek Tantoh Ndinteh
- Department of Chemical Sciences, Faculty of Sciences, University of Johannesburg, Doornfontein, South Africa
| | - Dieudonné Njamen
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaoundé, Yaounde, Cameroon
- Department of Chemical Sciences, Faculty of Sciences, University of Johannesburg, Doornfontein, South Africa
| | - Stéphane Zingue
- Laboratory of Physiology and Natural Products Research, Department of Life and Earth Sciences, Higher Teachers' Training College, University of Maroua, Maroua, Cameroon
- Department of Chemical Sciences, Faculty of Sciences, University of Johannesburg, Doornfontein, South Africa
| |
Collapse
|
22
|
Abstract
Despite decades of laboratory, epidemiological and clinical research, breast cancer incidence continues to rise. Breast cancer remains the leading cancer-related cause of disease burden for women, affecting one in 20 globally and as many as one in eight in high-income countries. Reducing breast cancer incidence will likely require both a population-based approach of reducing exposure to modifiable risk factors and a precision-prevention approach of identifying women at increased risk and targeting them for specific interventions, such as risk-reducing medication. We already have the capacity to estimate an individual woman's breast cancer risk using validated risk assessment models, and the accuracy of these models is likely to continue to improve over time, particularly with inclusion of newer risk factors, such as polygenic risk and mammographic density. Evidence-based risk-reducing medications are cheap, widely available and recommended by professional health bodies; however, widespread implementation of these has proven challenging. The barriers to uptake of, and adherence to, current medications will need to be considered as we deepen our understanding of breast cancer initiation and begin developing and testing novel preventives.
Collapse
Affiliation(s)
- Kara L Britt
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia.
| | - Jack Cuzick
- Centre for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Kelly-Anne Phillips
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
23
|
Parity reduces mammary repopulating activity but does not affect mammary stem cells defined as CD24 + CD29/CD49fhi in mice. Breast Cancer Res Treat 2020; 183:565-575. [PMID: 32696317 DOI: 10.1007/s10549-020-05804-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 07/11/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Breast cancer (BCa) mortality is decreasing with early detection and improvement in therapies. The incidence of BCa, however, continues to increase, particularly estrogen-receptor-positive (ER +) subtypes. One of the greatest modifiers of ER + BCa risk is childbearing (parity), with BCa risk halved in young multiparous mothers. Despite convincing epidemiological data, the biology that underpins this protection remains unclear. Parity-induced protection has been postulated to be due to a decrease in mammary stem cells (MaSCs); however, reports to date have provided conflicting data. METHODS We have completed rigorous functional testing of repopulating activity in parous mice using unfractionated and MaSC (CD24midCD49fhi)-enriched populations. We also developed a novel serial transplant method to enable us to assess self-renewal of MaSC following pregnancy. Lastly, as each pregnancy confers additional BCa protection, we subjected mice to multiple rounds of pregnancy to assess whether additional pregnancies impact MaSC activity. RESULTS Here, we report that while repopulating activity in the mammary gland is reduced by parity in the unfractionated gland, it is not due to a loss in the classically defined MaSC (CD24+CD49fhi) numbers or function. Self-renewal was unaffected by parity and additional rounds of pregnancy also did not lead to a decrease in MaSC activity. CONCLUSIONS Our data show instead that parity impacts on the stem-like activity of cells outside the MaSC population.
Collapse
|
24
|
Atashgaran V, Dasari P, Hodson LJ, Evdokiou A, Barry SC, Ingman WV. Foxp3 heterozygosity does not overtly affect mammary gland development during puberty or the oestrous cycle in mice. Reprod Fertil Dev 2020; 32:774-782. [PMID: 32389178 DOI: 10.1071/rd19378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/20/2020] [Indexed: 11/23/2022] Open
Abstract
Female mice heterozygous for a genetic mutation in transcription factor forkhead box p3 (Foxp3) spontaneously develop mammary cancers; however, the underlying mechanism is not well understood. We hypothesised that increased cancer susceptibility is associated with an underlying perturbation in mammary gland development. The role of Foxp3 in mammary ductal morphogenesis was investigated in heterozygous Foxp3Sf/+ and wildtype Foxp3+/+ mice during puberty and at specific stages of the oestrous cycle. No differences in mammary ductal branching morphogenesis, terminal end bud formation or ductal elongation were observed in pubertal Foxp3Sf/+ mice compared with Foxp3+/+ mice. During adulthood, all mice underwent normal regular oestrous cycles. No differences in epithelial branching morphology were detected in mammary glands from mice at the oestrus, metoestrus, dioestrus and pro-oestrus stages of the cycle. Furthermore, abundance of Foxp3 mRNA and protein in the mammary gland and lymph nodes was not altered in Foxp3Sf/+ mice compared with Foxp3+/+ mice. These studies suggest that Foxp3 heterozygosity does not overtly affect mammary gland development during puberty or the oestrous cycle. Further studies are required to dissect the underlying mechanisms of increased mammary cancer susceptibility in Foxp3Sf/+ heterozygous mice and the function of this transcription factor in normal mammary gland development.
Collapse
Affiliation(s)
- Vahid Atashgaran
- Discipline of Surgical Specialties, Adelaide Medical School, The Queen Elizabeth Hospital, University of Adelaide, Woodville, SA 5011, Australia; and Robinson Research Institute, University of Adelaide, SA 5005, Australia
| | - Pallave Dasari
- Discipline of Surgical Specialties, Adelaide Medical School, The Queen Elizabeth Hospital, University of Adelaide, Woodville, SA 5011, Australia; and Robinson Research Institute, University of Adelaide, SA 5005, Australia
| | - Leigh J Hodson
- Discipline of Surgical Specialties, Adelaide Medical School, The Queen Elizabeth Hospital, University of Adelaide, Woodville, SA 5011, Australia; and Robinson Research Institute, University of Adelaide, SA 5005, Australia
| | - Andreas Evdokiou
- Discipline of Surgical Specialties, Adelaide Medical School, The Queen Elizabeth Hospital, University of Adelaide, Woodville, SA 5011, Australia
| | - Simon C Barry
- Robinson Research Institute, University of Adelaide, SA 5005, Australia; and Molecular Immunology Laboratory, Discipline of Paediatrics, Adelaide Medical School, University of Adelaide, North Adelaide, SA 5006, Australia
| | - Wendy V Ingman
- Discipline of Surgical Specialties, Adelaide Medical School, The Queen Elizabeth Hospital, University of Adelaide, Woodville, SA 5011, Australia; and Robinson Research Institute, University of Adelaide, SA 5005, Australia; and Corresponding author.
| |
Collapse
|
25
|
Li H, Terry MB, Antoniou AC, Phillips KA, Kast K, Mooij TM, Engel C, Noguès C, Stoppa-Lyonnet D, Lasset C, Berthet P, Mari V, Caron O, Barrowdale D, Frost D, Brewer C, Evans DG, Izatt L, Side L, Walker L, Tischkowitz M, Rogers MT, Porteous ME, Snape K, Meijers-Heijboer HEJ, Gille JJP, Blok MJ, Hoogerbrugge N, Daly MB, Andrulis IL, Buys SS, John EM, McLachlan SA, Friedlander M, Tan YY, Osorio A, Caldes T, Jakubowska A, Simard J, Singer CF, Olah E, Navratilova M, Foretova L, Gerdes AM, Roos-Blom MJ, Arver B, Olsson H, Schmutzler RK, Hopper JL, Milne RL, Easton DF, Van Leeuwen FE, Rookus MA, Andrieu N, Goldgar DE. Alcohol Consumption, Cigarette Smoking, and Risk of Breast Cancer for BRCA1 and BRCA2 Mutation Carriers: Results from The BRCA1 and BRCA2 Cohort Consortium. Cancer Epidemiol Biomarkers Prev 2020; 29:368-378. [PMID: 31792088 PMCID: PMC7611162 DOI: 10.1158/1055-9965.epi-19-0546] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/08/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Tobacco smoking and alcohol consumption have been intensively studied in the general population to assess their effects on the risk of breast cancer, but very few studies have examined these effects in BRCA1 and BRCA2 mutation carriers. Given the high breast cancer risk for mutation carriers and the importance of BRCA1 and BRCA2 in DNA repair, better evidence on the associations of these lifestyle factors with breast cancer risk is essential. METHODS Using a large international pooled cohort of BRCA1 and BRCA2 mutation carriers, we conducted retrospective (5,707 BRCA1 mutation carriers and 3,525 BRCA2 mutation carriers) and prospective (2,276 BRCA1 mutation carriers and 1,610 BRCA2 mutation carriers) analyses of alcohol and tobacco consumption using Cox proportional hazards models. RESULTS For both BRCA1 and BRCA2 mutation carriers, none of the smoking-related variables was associated with breast cancer risk, except smoking for more than 5 years before a first full-term pregnancy (FFTP) when compared with parous women who never smoked. For BRCA1 mutation carriers, the HR from retrospective analysis (HRR) was 1.19 [95% confidence interval (CI), 1.02-1.39] and the HR from prospective analysis (HRP) was 1.36 (95% CI, 0.99-1.87). For BRCA2 mutation carriers, smoking for more than 5 years before an FFTP showed an association of a similar magnitude, but the confidence limits were wider (HRR = 1.25; 95% CI, 1.01-1.55 and HRP = 1.30; 95% CI, 0.83-2.01). For both carrier groups, alcohol consumption was not associated with breast cancer risk. CONCLUSIONS The finding that smoking during the prereproductive years increases breast cancer risk for mutation carriers warrants further investigation. IMPACT This is the largest prospective study of BRCA mutation carriers to assess these important risk factors.
Collapse
Affiliation(s)
- Hongyan Li
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University, New York, New York
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Strangeways Research Laboratory, Worts Causeway, University of Cambridge, Cambridge, United Kingdom
| | - Kelly-Anne Phillips
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Victoria, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Karin Kast
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- German Cancer Consortium (DKTK), Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thea M Mooij
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Germany
| | - Catherine Noguès
- Institut Paoli-Calmettes, Département d'Anticipation et de Suivi des Cancers, Oncogénétique Clinique and Aix Marseille Univ, INSERM, IRD, SESSTIM, Marseille, France
| | - Dominique Stoppa-Lyonnet
- Institut Curie, Service de Génétique Médicale, Paris, France
- Inserm, U830, Université Paris Descartes, Paris, France
| | - Christine Lasset
- Unité de prévention et Epidémiologie Génétique, Centre Léon Bérard - Lyon/UMR CNRS 5558, Université de Lyon - Lyon, France
| | - Pascaline Berthet
- Département de biopathologie, Oncogénétique clinique, Centre François Baclesse - Caen, France
| | - Veronique Mari
- CLCC Antoine Lacassagne, Département d'Hématologie - Oncologie médicale, Nice, France
| | - Olivier Caron
- Département de Médecine, Gustave Roussy Hôpital Universitaire - Villejuif, France
| | - Daniel Barrowdale
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Strangeways Research Laboratory, Worts Causeway, University of Cambridge, Cambridge, United Kingdom
| | - Debra Frost
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Strangeways Research Laboratory, Worts Causeway, University of Cambridge, Cambridge, United Kingdom
| | - Carole Brewer
- Department of Clinical Genetics, Royal Devon & Exeter Hospital, Exeter, United Kingdom
| | - D Gareth Evans
- Genomic Medicine, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Sciences Centre, Division of Evolution and Genomic Sciences, Manchester University, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Louise Izatt
- Clinical Genetics, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
| | - Lucy Side
- Wessex Clinical Genetics Service, The Princess Anne Hospital, Southampton, United Kingdom
| | - Lisa Walker
- Oxford Regional Genetics Service, Churchill Hospital, Oxford, United Kingdom
| | - Marc Tischkowitz
- University of Cambridge Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, and Cancer Research UK Cambridge Center, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Mark T Rogers
- All Wales Medical Genetics Services, University Hospital of Wales, Cardiff, United Kingdom
| | - Mary E Porteous
- South East of Scotland Regional Genetics Service, Western General Hospital, Edinburgh, United Kingdom
| | - Katie Snape
- Medical Genetics Unit, St. George's, University of London, London, United Kingdom
| | | | - Johan J P Gille
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Marinus J Blok
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mary B Daly
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Irene L Andrulis
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Saundra S Buys
- Department of Medicine, University of Utah Health Sciences Center, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Esther M John
- Stanford University School of Medicine, Department of Medicine, Division of Oncology, and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Sue-Anne McLachlan
- Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Parkville, Victoria, Australia
- Department of Medical Oncology, St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Michael Friedlander
- Department of Medical Oncology, Prince of Wales Hospital, Randwick, New South Wales, Australia
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Yen Y Tan
- Department of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Ana Osorio
- Human Genetics Group, Spanish National Cancer Centre (CNIO) and Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Trinidad Caldes
- Molecular Oncology Laboratory, Hospital Clinico San Carlos, IdISSC, CIBERONC (ISCIII), Madrid, Spain
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, Poland
- Independent Laboratory of Molecular Biology and Genetic Diagnostics, Pomeranian Medical University, Szczecin, Poland
| | - Jacques Simard
- Genomics Center, Centre Hospitalier Universitaire de Québec - Université Laval Research Center, Quebec City, Quebec, Canada
| | - Christian F Singer
- Department of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Edith Olah
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
| | - Marie Navratilova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, Czech Republic
| | - Lenka Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, Czech Republic
| | - Anne-Marie Gerdes
- The Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Marie-José Roos-Blom
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Brita Arver
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - Håkan Olsson
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - Rita K Schmutzler
- Center for Familial Breast and Ovarian Cancer, Center for Integrated Oncology (CIO), Medical Faculty, University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Victoria, Australia
| | - Roger L Milne
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Victoria, Australia
- Division of Cancer Epidemiology and Intelligence, Cancer Council Victoria, Melbourne, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Strangeways Research Laboratory, Worts Causeway, University of Cambridge, Cambridge, United Kingdom
- Centre for Cancer Genetic Epidemiology, Department of Oncology, Strangeways Research Laboratory, Worts Causeway, University of Cambridge, Cambridge, United Kingdom
| | - Flora E Van Leeuwen
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Matti A Rookus
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nadine Andrieu
- INSERM, U900, Paris, France.
- Institut Curie, Paris, France
- Mines Paris Tech, Fontainebleau, France
- PSL Research University, Paris, France
| | - David E Goldgar
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah.
- Department of Dermatology, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
26
|
Environmental Exposures during Puberty: Window of Breast Cancer Risk and Epigenetic Damage. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17020493. [PMID: 31941024 PMCID: PMC7013753 DOI: 10.3390/ijerph17020493] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022]
Abstract
During puberty, a woman’s breasts are vulnerable to environmental damage (“window of vulnerability”). Early exposure to environmental carcinogens, endocrine disruptors, and unhealthy foods (refined sugar, processed fats, food additives) are hypothesized to promote molecular damage that increases breast cancer risk. However, prospective human studies are difficult to perform and effective interventions to prevent these early exposures are lacking. It is difficult to prevent environmental exposures during puberty. Specifically, young women are repeatedly exposed to media messaging that promotes unhealthy foods. Young women living in disadvantaged neighborhoods experience additional challenges including a lack of access to healthy food and exposure to contaminated air, water, and soil. The purpose of this review is to gather information on potential exposures during puberty. In future directions, this information will be used to help elementary/middle-school girls to identify and quantitate environmental exposures and develop cost-effective strategies to reduce exposures.
Collapse
|
27
|
Moradinazar M, Marzbani B, Shahebrahimi K, Shahabadi S, Marzbani B, Moradinazar Z. Hormone Therapy and Factors Affecting Fertility of Women Under 50-Year-Old with Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2019; 11:309-319. [PMID: 31920374 PMCID: PMC6938198 DOI: 10.2147/bctt.s218394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 12/10/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Although the use of contraceptive hormones is a risk factor for development of breast cancer, level of risk is unknown; thus the current research was conducted to investigate the effect of factors related to fertility and hormone use on risk of breast cancer in women aged under 50 years old in the west of Iran. METHODS In this case-control study, all incidence cases of breast cancer aged between 25-49 years old (n=212) were selected. Twice as many as the case group, the individuals referred to other outpatient sections of the same hospital at the time of study and up to 2 years after the follow-up not diagnosed with breast or other cancers were selected as a control group. The data were collected using healthy fertility program and middle-aged periodical care forms developed by Iran's Ministry of Health (MOH). RESULTS After controlling for confounding variables, history of hormonal use for contraception (OR=2.02, 95% CI=1.2-3.3) and hormone therapies (OR=1.9, 95% CI=1.2-3.04) were identified as factors increasing the risk of breast cancer. Dose-response relationships between breast cancer and the use of hormones for contraception and hormone therapy indicated that these factors increased the risk of breast cancer. The risk was found to be higher in women who had been under hormone therapy for more than 120 months. With an increase in the age of the first menstruation, risk of breast cancer increased linearly, but with an increase in the age of the first pregnancy, risk of breast cancer increased exponentially. After 20 years of age, the risk increased with a steeper slope. CONCLUSION Considering the effect of hormone therapy and fertility factors on breast cancer and changeability of listed risk factors, the researchers suggest planning for sensitizing, increasing the awareness, and educating women and professionals regarding the influence of fertility and hormonal factors including pregnancy at lower ages, minimizing the use of hormones for contraception, and hormone therapy.
Collapse
Affiliation(s)
- Mehdi Moradinazar
- Research Center for Environmental Determinants of Health, School of Public Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Behjat Marzbani
- Health Education and Promotion Group, Vice Chancellor for Health Affairs, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Karoon Shahebrahimi
- Department of Internal Medicine, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sara Shahabadi
- Health Education and Promotion Group, Vice Chancellor for Health Affairs, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Behnaz Marzbani
- Health Education and Promotion Group, Health Network of Kermanshah, Vice Chancellor for Health Affairs, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeinab Moradinazar
- School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
28
|
Cheng TYD, Yao S, Omilian AR, Khoury T, Buas MF, Payne-Ondracek R, Sribenja S, Bshara W, Hong CC, Bandera EV, Davis W, Higgins MJ, Ambrosone CB. FOXA1 Protein Expression in ER + and ER - Breast Cancer in Relation to Parity and Breastfeeding in Black and White Women. Cancer Epidemiol Biomarkers Prev 2019; 29:379-385. [PMID: 31871111 DOI: 10.1158/1055-9965.epi-19-0787] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 10/28/2019] [Accepted: 12/12/2019] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Forkhead box protein A1 (FOXA1) promotes luminal differentiation, and hypermethylation of the gene can be a mechanism of developing estrogen receptor-negative (ER-) breast cancer. We examined FOXA1 in breast tumor and adjacent normal tissue in relation to reproductive factors, particularly higher parity and no breastfeeding, that are associated with ER- tumors. METHODS We performed IHC for FOXA1 in breast tumors (n = 1,329) and adjacent normal tissues (n = 298) in the Women's Circle of Health Study (949 Blacks and 380 Whites). Protein expression levels were summarized by histology (H) scores. Generalized linear models were used to assess FOXA1 protein expression in relation to reproductive factors by ER status. RESULTS ER-positive (ER+) versus ER- tumors had higher FOXA1 protein expression (P < 0.001). FOXA1 expression was higher in tumor versus paired adjacent normal tissue in women with ER+ or non-triple-negative cancer (both P < 0.001), but not in those with ER- or triple-negative cancer. Higher number of births (1, 2, and 3+) was associated with lower FOXA1 protein expression in ER+ tumors [differences in H score, or β = -8.5; 95% confidence interval (CI), -15.1 to -2.0], particularly among parous women who never breastfed (β = -10.4; 95% CI, -19.7 to -1.0), but not among those who breastfed (β = -7.5; 95% CI, -16.9 to 1.8). The associations for ER- tumors were similar, although they were not statistically significant. CONCLUSIONS In this tumor-based study, higher parity was associated with lower FOXA1 expression in ER+ tumors, and breastfeeding may ameliorate the influence. IMPACT These findings contribute to our understanding of FOXA1 methylation and breast cancer etiology.
Collapse
Affiliation(s)
- Ting-Yuan David Cheng
- Department of Epidemiology, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida. .,Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Song Yao
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Angela R Omilian
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Thaer Khoury
- Department of Pathology & Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Matthew F Buas
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Rochelle Payne-Ondracek
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Sirinapa Sribenja
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Wiam Bshara
- Department of Pathology & Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Chi-Chen Hong
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Elisa V Bandera
- Cancer Prevention and Control Program, Rutgers Cancer Institute of New Jersey, The State University of New Jersey, New Brunswick, New Jersey
| | - Warren Davis
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Michael J Higgins
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Christine B Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
29
|
The lncRNA SOX2OT rs9839776 C>T Polymorphism Indicates Recurrent Miscarriage Susceptibility in a Southern Chinese Population. Mediators Inflamm 2019; 2019:9684703. [PMID: 31827385 PMCID: PMC6885167 DOI: 10.1155/2019/9684703] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 09/25/2019] [Accepted: 10/22/2019] [Indexed: 12/26/2022] Open
Abstract
Genetic susceptibility may be involved in the onset of recurrent miscarriage. Previous studies have shown that some genetic polymorphisms that regulate cell migration are associated with susceptibility to recurrent miscarriage. The SOX2 overlapping transcript (SOX2OT) may regulate the migration and invasion of multiple tumor cells and is related to susceptibility to various diseases. However, whether lncRNA SOX2OT polymorphisms are related to recurrent miscarriage susceptibility is unclear. Therefore, we investigated the relationship between the lncRNA SOX2OT rs9839776 C>T polymorphism and recurrent miscarriage susceptibility. We recruited 570 subjects with recurrent miscarriage and 578 healthy control subjects from a population in southern China and used the TaqMan method for genotyping. We found a significant association between the rs9839776 CT genotype in the SOX2OT gene and an increased risk for recurrent miscarriage (CT vs CC: adjusted OR = 1.357, 95%CI = 1.065 - 1.728, P = 0.0134). However, we did not observe any significant associations between the recurrent miscarriage risk and the number of miscarriages in different age groups. In conclusion, our study indicated that the rs9839776 CT genotype may contribute to an increased risk of recurrent miscarriage in the southern Chinese population and that rs9839776 may act as a prognostic biomarker in recurrent miscarriage patients. However, an experiment-based study with a larger sample size should be performed to confirm these results.
Collapse
|
30
|
Terry MB, Michels KB, Brody JG, Byrne C, Chen S, Jerry DJ, Malecki KMC, Martin MB, Miller RL, Neuhausen SL, Silk K, Trentham-Dietz A. Environmental exposures during windows of susceptibility for breast cancer: a framework for prevention research. Breast Cancer Res 2019; 21:96. [PMID: 31429809 PMCID: PMC6701090 DOI: 10.1186/s13058-019-1168-2] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background The long time from exposure to potentially harmful chemicals until breast cancer occurrence poses challenges for designing etiologic studies and for implementing successful prevention programs. Growing evidence from animal and human studies indicates that distinct time periods of heightened susceptibility to endocrine disruptors exist throughout the life course. The influence of environmental chemicals on breast cancer risk may be greater during several windows of susceptibility (WOS) in a woman’s life, including prenatal development, puberty, pregnancy, and the menopausal transition. These time windows are considered as specific periods of susceptibility for breast cancer because significant structural and functional changes occur in the mammary gland, as well as alterations in the mammary micro-environment and hormone signaling that may influence risk. Breast cancer research focused on these breast cancer WOS will accelerate understanding of disease etiology and prevention. Main text Despite the plausible heightened mechanistic influences of environmental chemicals on breast cancer risk during time periods of change in the mammary gland’s structure and function, most human studies of environmental chemicals are not focused on specific WOS. This article reviews studies conducted over the past few decades that have specifically addressed the effect of environmental chemicals and metals on breast cancer risk during at least one of these WOS. In addition to summarizing the broader evidence-base specific to WOS, we include discussion of the NIH-funded Breast Cancer and the Environment Research Program (BCERP) which included population-based and basic science research focused on specific WOS to evaluate associations between breast cancer risk and particular classes of endocrine-disrupting chemicals—including polycyclic aromatic hydrocarbons, perfluorinated compounds, polybrominated diphenyl ethers, and phenols—and metals. We outline ways in which ongoing transdisciplinary BCERP projects incorporate animal research and human epidemiologic studies in close partnership with community organizations and communication scientists to identify research priorities and effectively translate evidence-based findings to the public and policy makers. Conclusions An integrative model of breast cancer research is needed to determine the impact and mechanisms of action of endocrine disruptors at different WOS. By focusing on environmental chemical exposure during specific WOS, scientists and their community partners may identify when prevention efforts are likely to be most effective.
Collapse
Affiliation(s)
- Mary Beth Terry
- Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168th Street, Room 1611, New York, NY, 10032, USA
| | - Karin B Michels
- Department of Epidemiology, Fielding School of Public Health, University of California, 650 Charles E. Young Drive South, CHS 71-254, Los Angeles, CA, 90095, USA
| | | | - Celia Byrne
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road A-1039F, Bethesda, MD, 20814, USA
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of City of Hope, 1450 E. Duarte Road, Duarte, CA, 91010, USA
| | - D Joseph Jerry
- Pioneer Valley Life Sciences Institute and Department of Veterinary & Animal Sciences, University of Massachusetts Amherst, 661 North Pleasant St., Amherst, MA, 01003, USA
| | - Kristen M C Malecki
- Department of Population Health Sciences and the Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, 610 Walnut St., WARF Room 605, Madison, WI, 53726, USA
| | - Mary Beth Martin
- Departments of Oncology and Biochemistry & Molecular Biology, Georgetown University Medical Center, E411 New Research Building, Washington, DC, 20057, USA
| | - Rachel L Miller
- Departments of Medicine, Pediatrics, Environmental Health Sciences; Vagelos College of Physicians and Surgeons, Mailman School of Public Health, Columbia University, PH8E-101B, 630 W. 168th St, New York, NY, 10032, USA
| | - Susan L Neuhausen
- Department of Population Sciences, Beckman Research Institute of City of Hope, 1450 E. Duarte Road, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Kami Silk
- Department of Communication, University of Delaware, 250 Pearson Hall, 125 Academy St, Newark, DE, 19716, USA
| | - Amy Trentham-Dietz
- Department of Population Health Sciences and Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, 610 Walnut St., WARF Room 307, Madison, WI, 53726, USA.
| | | |
Collapse
|
31
|
Santucci-Pereira J, Zeleniuch-Jacquotte A, Afanasyeva Y, Zhong H, Slifker M, Peri S, Ross EA, López de Cicco R, Zhai Y, Nguyen T, Sheriff F, Russo IH, Su Y, Arslan AA, Bordas P, Lenner P, Åhman J, Landström Eriksson AS, Johansson R, Hallmans G, Toniolo P, Russo J. Genomic signature of parity in the breast of premenopausal women. Breast Cancer Res 2019; 21:46. [PMID: 30922380 PMCID: PMC6438043 DOI: 10.1186/s13058-019-1128-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/14/2019] [Indexed: 12/17/2022] Open
Abstract
Background Full-term pregnancy (FTP) at an early age confers long-term protection against breast cancer. Previously, we reported that a FTP imprints a specific gene expression profile in the breast of postmenopausal women. Herein, we evaluated gene expression changes induced by parity in the breast of premenopausal women. Methods Gene expression profiling of normal breast tissue from 30 nulliparous (NP) and 79 parous (P) premenopausal volunteers was performed using Affymetrix microarrays. In addition to a discovery/validation analysis, we conducted an analysis of gene expression differences in P vs. NP women as a function of time since last FTP. Finally, a laser capture microdissection substudy was performed to compare the gene expression profile in the whole breast biopsy with that in the epithelial and stromal tissues. Results Discovery/validation analysis identified 43 differentially expressed genes in P vs. NP breast. Analysis of expression as a function of time since FTP revealed 286 differentially expressed genes (238 up- and 48 downregulated) comparing all P vs. all NP, and/or P women whose last FTP was less than 5 years before biopsy vs. all NP women. The upregulated genes showed three expression patterns: (1) transient: genes upregulated after FTP but whose expression levels returned to NP levels. These genes were mainly related to immune response, specifically activation of T cells. (2) Long-term changing: genes upregulated following FTP, whose expression levels decreased with increasing time since FTP but did not return to NP levels. These were related to immune response and development. (3) Long-term constant: genes that remained upregulated in parous compared to nulliparous breast, independently of time since FTP. These were mainly involved in development/cell differentiation processes, and also chromatin remodeling. Lastly, we found that the gene expression in whole tissue was a weighted average of the expression in epithelial and stromal tissues. Conclusions Genes transiently activated by FTP may have a role in protecting the mammary gland against neoplastically transformed cells through activation of T cells. Furthermore, chromatin remodeling and cell differentiation, represented by the genes that are maintained upregulated long after the FTP, may be responsible for the lasting preventive effect against breast cancer. Electronic supplementary material The online version of this article (10.1186/s13058-019-1128-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Santucci-Pereira
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA.
| | - Anne Zeleniuch-Jacquotte
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA.,New York University Perlmutter Cancer Center, New York, NY, 10016, USA
| | - Yelena Afanasyeva
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA
| | - Hua Zhong
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA
| | - Michael Slifker
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center - Temple University Health System, Philadelphia, PA, 19111, USA
| | - Suraj Peri
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center - Temple University Health System, Philadelphia, PA, 19111, USA
| | - Eric A Ross
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center - Temple University Health System, Philadelphia, PA, 19111, USA
| | - Ricardo López de Cicco
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Yubo Zhai
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Theresa Nguyen
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Fathima Sheriff
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Irma H Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Yanrong Su
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| | - Alan A Arslan
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY, 10016, USA.,Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY, 10016, USA
| | - Pal Bordas
- Sunderby Hospital, Luleå and the Norrbotten Mammography Screening Program, Luleå, Sweden.,Departments of Radiation Sciences and Oncology, Umeå University, Umeå, Sweden
| | - Per Lenner
- Departments of Radiation Sciences and Oncology, Umeå University, Umeå, Sweden
| | - Janet Åhman
- Sunderby Hospital, Luleå and the Norrbotten Mammography Screening Program, Luleå, Sweden
| | | | | | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Paolo Toniolo
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY, 10016, USA
| | - Jose Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, 333 Cottman Ave, P2051, Philadelphia, PA, 19111, USA
| |
Collapse
|
32
|
Katuwal S, Tapanainen JS, Pukkala E, Kauppila A. The effect of length of birth interval on the risk of breast cancer by subtype in grand multiparous women. BMC Cancer 2019; 19:199. [PMID: 30832620 PMCID: PMC6399864 DOI: 10.1186/s12885-019-5404-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 02/25/2019] [Indexed: 12/19/2022] Open
Abstract
Background The length of interval between successive childbirths (birth interval) might influence the incidence of breast cancer, either by stimulating or by inhibiting the factors that are responsible for the initiation of breast cancer or its early development. Methods This is a case-control study nested in a cohort of 47,479 Finnish grand-multiparous (GM) women born after 1934, and registered as having had at least five births before 2013. The 1354 women with breast cancer diagnosis were compared with controls (1:5) matched by parity and date of birth of the mother. Conditional logistic regression was used to estimate odds ratios of the risk of ductal and lobular breast cancer subtypes associated with each of the intervals between the 1st and 5th birth, stratified by age at diagnosis. Age at first and last birth before index date were used as covariates. Results Increased intervals between the 1st and 5th births were associated with an increased risk of lobular breast cancer. In contrast, regarding ductal cancer, premenopausal women with shorter length of interval (< 2 years) between the 1st and 2nd birth had greater risk and longer intervals (3+ years) were associated with reduced risk. Spacing between the 2nd and 5th birth did not influence the risk of ductal breast cancer. Conclusion The findings of our study suggest that the effect of the length of birth interval on breast cancer depends on the age and histology. The protective effect of shorter birth intervals on lobular breast among postmenopausal women and the opposite effect on ductal cancer in premenopausal women may reflect distinct differentiation and functional roles of lobular and ductal cells, and possibly also different response to hormonal exposure.
Collapse
Affiliation(s)
- Sushmita Katuwal
- Faculty of Social Sciences, University of Tampere, Kauppi Campus, Arvo Ylpön Street 34, 33520, Tampere, Finland.
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki University Hospital, Helsinki, Finland.,Department of Obstetrics and Gynecology, Oulu University Hospital, University of Oulu, Medical Research Center Oulu, Oulu, Finland.,Department of Obstetrics and Gynecology, University of Oulu and Oulu University Hospital, Medical Research Center, PEDEGO Research Unit, Oulu, Finland
| | - Eero Pukkala
- Faculty of Social Sciences, University of Tampere, Kauppi Campus, Arvo Ylpön Street 34, 33520, Tampere, Finland.,Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| | - Antti Kauppila
- Department of Obstetrics and Gynecology, Oulu University Hospital, University of Oulu, Medical Research Center Oulu, Oulu, Finland
| |
Collapse
|
33
|
Nguyen B, Venet D, Lambertini M, Desmedt C, Salgado R, Horlings HM, Rothé F, Sotiriou C. Imprint of parity and age at first pregnancy on the genomic landscape of subsequent breast cancer. Breast Cancer Res 2019; 21:25. [PMID: 30770770 PMCID: PMC6377756 DOI: 10.1186/s13058-019-1111-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Background Although parity and age at first pregnancy are among the most known extrinsic factors that modulate breast cancer risk, their impact on the biology of subsequent breast cancer has never been explored in depth. Recent data suggest that pregnancy-induced tumor protection is different according to breast cancer subtypes, with parity and young age at first pregnancy being associated with a marked reduction in the risk of developing luminal subtype but not triple negative breast cancer. In this study, we investigated the imprint of parity and age at first pregnancy on the pattern of somatic mutations, somatic copy number alterations, transcriptomic profiles, and tumor immune microenvironment by assessing tumor-infiltrating lymphocytes (TILs) levels of subsequent breast cancer. Methods A total of 313 patients with primary breast cancer with available whole genome, RNA sequencing, and TILs data were included in this study. We used a multivariate analysis adjusted for age at diagnosis, pathological stage, molecular subtypes, and histological subtypes. We compared nulliparous vs. parous, late parous vs. early parous, and nulliparous vs. pregnancy-associated breast cancer (PABC) patients. Late and early parous patients were grouped by using the median age at first pregnancy. PABC was defined as patients diagnosed up to 10 years postpartum. Results Genomic alterations of breast cancer were associated with age at first pregnancy but not with parity status alone. Independently of clinicopathological features, early parous patients developed tumors characterized by a higher number of Indels (Padj = 0.002), a lower frequency of CDH1 mutations (1.2% vs. 12.7%; Padj = 0.013), a higher frequency of TP53 mutations (50% vs. 22.5%; Padj = 0.010), and MYC amplification (28% vs. 7%; Padj = 0.008). PABC were associated with increased TILs infiltration (Padj = 0.0495). Conclusions These findings highlight an unprecedented link between reproductive history and the genomic landscape of subsequent breast cancer. We further hypothesize that TP53-mutant premalignant lesions could be less susceptible to the protective effect of an early parity, which might explain the difference of parity-induced protection according to breast cancer subtypes. This work also advocates that reproductive history should be routinely collected in future large-scale genomic studies addressing the biology of female cancers. Electronic supplementary material The online version of this article (10.1186/s13058-019-1111-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bastien Nguyen
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium. .,Present Address: Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, USA.
| | - David Venet
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Matteo Lambertini
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Department of Medical Oncology, Clinica di Oncologia Medica, Ospedale Policlinico San Martino, & Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Christine Desmedt
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Roberto Salgado
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Department of Pathology, GZA-ZNA, Antwerp, Belgium
| | - Hugo Mark Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Françoise Rothé
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory J.-C. Heuson, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
34
|
Terry MB, Liao Y, Kast K, Antoniou AC, McDonald JA, Mooij TM, Engel C, Nogues C, Buecher B, Mari V, Moretta-Serra J, Gladieff L, Luporsi E, Barrowdale D, Frost D, Henderson A, Brewer C, Evans DG, Eccles D, Cook J, Ong KR, Izatt L, Ahmed M, Morrison PJ, Dommering CJ, Oosterwijk JC, Ausems MGEM, Kriege M, Buys SS, Andrulis IL, John EM, Daly M, Friedlander M, McLachlan SA, Osorio A, Caldes T, Jakubowska A, Simard J, Singer CF, Tan Y, Olah E, Navratilova M, Foretova L, Gerdes AM, Roos-Blom MJ, Arver B, Olsson H, Schmutzler RK, Hopper JL, van Leeuwen FE, Goldgar D, Milne RL, Easton DF, Rookus MA, Andrieu N. The Influence of Number and Timing of Pregnancies on Breast Cancer Risk for Women With BRCA1 or BRCA2 Mutations. JNCI Cancer Spectr 2018; 2:pky078. [PMID: 30873510 PMCID: PMC6405439 DOI: 10.1093/jncics/pky078] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/08/2018] [Accepted: 12/08/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Full-term pregnancy (FTP) is associated with a reduced breast cancer (BC) risk over time, but women are at increased BC risk in the immediate years following an FTP. No large prospective studies, however, have examined whether the number and timing of pregnancies are associated with BC risk for BRCA1 and BRCA2 mutation carriers. METHODS Using weighted and time-varying Cox proportional hazards models, we investigated whether reproductive events are associated with BC risk for mutation carriers using a retrospective cohort (5707 BRCA1 and 3525 BRCA2 mutation carriers) and a prospective cohort (2276 BRCA1 and 1610 BRCA2 mutation carriers), separately for each cohort and the combined prospective and retrospective cohort. RESULTS For BRCA1 mutation carriers, there was no overall association with parity compared with nulliparity (combined hazard ratio [HRc] = 0.99, 95% confidence interval [CI] = 0.83 to 1.18). Relative to being uniparous, an increased number of FTPs was associated with decreased BC risk (HRc = 0.79, 95% CI = 0.69 to 0.91; HRc = 0.70, 95% CI = 0.59 to 0.82; HRc = 0.50, 95% CI = 0.40 to 0.63, for 2, 3, and ≥4 FTPs, respectively, P trend < .0001) and increasing duration of breastfeeding was associated with decreased BC risk (combined cohort P trend = .0003). Relative to being nulliparous, uniparous BRCA1 mutation carriers were at increased BC risk in the prospective analysis (prospective hazard ration [HRp] = 1.69, 95% CI = 1.09 to 2.62). For BRCA2 mutation carriers, being parous was associated with a 30% increase in BC risk (HRc = 1.33, 95% CI = 1.05 to 1.69), and there was no apparent decrease in risk associated with multiparity except for having at least 4 FTPs vs. 1 FTP (HRc = 0.72, 95% CI = 0.54 to 0.98). CONCLUSIONS These findings suggest differential associations with parity between BRCA1 and BRCA2 mutation carriers with higher risk for uniparous BRCA1 carriers and parous BRCA2 carriers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Nadine Andrieu
- Correspondence to: Nadine Andrieu, PhD, Cancer Genetic Epidemiology Team, INSERM Unit 900, Institut Curie, 26 rue d’Ulm, 75005 Paris, France (e-mail: )
| | | |
Collapse
|
35
|
Swerdlow AJ, Wright LB, Schoemaker MJ, Jones ME. Maternal breast cancer risk in relation to birthweight and gestation of her offspring. Breast Cancer Res 2018; 20:110. [PMID: 30286782 PMCID: PMC6172803 DOI: 10.1186/s13058-018-1035-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/01/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Parity and age at first pregnancy are well-established risk factors for breast cancer, but the effects of other characteristics of pregnancies are uncertain and the literature is inconsistent. METHODS In a cohort of 83,451 parous women from the general population of the UK, which collected detailed information on each pregnancy and a wide range of potential confounders, we investigated the associations of length of gestation and birthweight of offspring in a woman's pregnancies with her breast cancer risk, adjusting for a full range of non-reproductive as well as reproductive risk factors unlike in previous large studies. RESULTS Gestation of the first-born offspring was significantly inversely related to the risk of pre-menopausal breast cancer (p trend = 0.03; hazard ratio (HR) for 26-31 compared with 40-41 weeks, the baseline group, = 2.38, 95% confidence interval (CI) 1.26-4.49), and was borderline significantly related to risk of breast cancer overall (p trend = 0.05). Risk was significantly raised in mothers of high birthweight first-born (HR for breast cancer overall = 1.53, 95% CI 1.06-2.21 for ≥ 4500 g compared with 3000-3499 g, the baseline group). For gestation and birthweight of most recent birth, there were no clear effects. Analyses without adjustment for confounders (other than age) gave similar results. CONCLUSIONS Our data add to evidence that short gestation pregnancies may increase the risk of breast cancer, at least pre-menopausally, perhaps by hormonal stimulation and breast proliferation early in pregnancy without the opportunity for the differentiation that occurs in late pregnancy. High birthweight first pregnancies may increase breast cancer risk, possibly through the association of birthweight with oestrogen and insulin-like growth factor 1 levels.
Collapse
Affiliation(s)
- Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sir Richard Doll Building, London, SM2 5NG, UK. .,Division of Breast Cancer Research, The Institute of Cancer Research, London, SW3 6JB, UK.
| | - Lauren B Wright
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sir Richard Doll Building, London, SM2 5NG, UK
| | - Minouk J Schoemaker
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sir Richard Doll Building, London, SM2 5NG, UK
| | - Michael E Jones
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sir Richard Doll Building, London, SM2 5NG, UK
| |
Collapse
|
36
|
Abstract
The human betaretrovirus and the closely related mouse mammary tumor virus have been linked with the development of cholangitis and mitochondrial antibody production in patients with primary biliary cholangitis (PBC) and mouse models of autoimmune biliary disease, respectively. In vitro, betaretroviruses have been found to stimulate the expression of mitochondrial autoantigens on the cell surface of biliary epithelial cells. In vivo, both mitochondrial autoantigens and viral proteins have been shown to be co-expressed in biliary epithelium and lymphoid tissue. Notably, both mice and humans make poor antibody responses to betaretrovirus infection, whereas proinflammatory responses to viral proteins have been observed in T lymphocyte studies. Furthermore, proviral integration studies have confirmed the presence of human betaretrovirus in biliary epithelium of patients with PBC. Preliminary proof of principal studies using combination antiretroviral therapy have shown that suppression of viral expression is associated with sustained biochemical response. As the previous regimen used was poorly tolerated, further randomized controlled trials are planned to determine whether betaretrovirus infection plays an important role in the development of PBC.
Collapse
Affiliation(s)
- Andrew L Mason
- Department of Medicine, University of Alberta, Edmonton, AB, T6G 2E1, Canada.
| |
Collapse
|
37
|
Dall GV, Hawthorne S, Seyed-Razavi Y, Vieusseux J, Wu W, Gustafsson JA, Byrne D, Murphy L, Risbridger GP, Britt KL. Estrogen receptor subtypes dictate the proliferative nature of the mammary gland. J Endocrinol 2018; 237:323-336. [PMID: 29636363 DOI: 10.1530/joe-17-0582] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022]
Abstract
Estrogen induces proliferation of breast epithelial cells and is responsible for breast development at puberty. This tightly regulated control is lost in estrogen-receptor-positive (ER+) breast cancers, which comprise over 70% of all breast cancers. Currently, breast cancer diagnosis and treatment considers only the α isoform of ER; however, there is a second ER, ERβ. Whilst ERα mediates estrogen-driven proliferation of the normal breast in puberty and breast cancers, ERβ has been shown to exert an anti-proliferative effect on the normal breast. It is not known how the expression of each ER (alone or in combination) correlates with the ability of estrogen to induce proliferation in the breast. We assessed the levels of each ER in normal mouse mammary glands subdivided into proliferative and non-proliferative regions. ERα was most abundant in the proliferative regions of younger mice, with ERβ expressed most abundantly in old mice. We correlated this expression profile with function by showing that the ability of estrogen to induce proliferation was reduced in older mice. To show that the ER profile associated with breast cancer risk, we assessed ER expression in parous mice which are known to have a reduced risk of developing ERα breast cancer. ERα expression was significantly decreased yet co-localization analysis revealed ERβ expression increased with parity. Parous mice had less unopposed nuclear ERα expression and increased levels of ERβ. These changes suggest that the nuclear expression of ERs dictates the proliferative nature of the breast and may explain the decreased breast cancer risk with parity.
Collapse
Affiliation(s)
| | - Samuel Hawthorne
- Department of Anatomy and Developmental BiologyMonash University, Melbourne, Australia
| | - Yashar Seyed-Razavi
- Department of Anatomy and Developmental BiologyMonash University, Melbourne, Australia
| | | | - Wanfu Wu
- Department of Biology and BiochemistryUniversity of Houston, Houston, Texas, USA
| | - Jan-Ake Gustafsson
- Department of Biology and BiochemistryUniversity of Houston, Houston, Texas, USA
| | - David Byrne
- Department of PathologyPeter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Gail P Risbridger
- Peter MacCallum Cancer CentreMelbourne, Australia
- Department of Anatomy and Developmental BiologyMonash University, Melbourne, Australia
| | - Kara L Britt
- Peter MacCallum Cancer CentreMelbourne, Australia
- The Sir Peter MacCallumDepartment of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
38
|
Brantley KD, Hartman TJ, Patel AV, Gapstur SM, Flanders WD, McCullough ML. Test-Retest Reproducibility of Adult-Reported High School Diet Varies among Racially and Ethnically Diverse US Men and Women. J Nutr 2018; 148:599-606. [PMID: 29659953 DOI: 10.1093/jn/nxy001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/28/2017] [Indexed: 01/11/2023] Open
Abstract
Background Adolescent diet is thought to play an important role in future chronic disease risk. However, few studies have examined the reproducibility of adult-reported adolescent diet, and evidence for possible differences in reproducibility by demographic characteristics is limited. Objective We assessed the ability of adults to consistently report past high school diet over a 1-y period and examined differences in reproducibility by selected demographic characteristics. Methods By using age-adjusted partial Spearman (ρ) or Pearson (r) correlations, we assessed 1-y reproducibility for 33 line items, 20 food groups, and 2 dietary patterns of high school diet reported in adulthood via a questionnaire completed by 742 participants in the Cancer Prevention Study 3 (CPS-3) Diet Substudy. Results Participants' median age was 53 y (range: 31-70 y), 65.2% were women, 59.8% were non-Hispanic white, 24.8% were non-Hispanic black, and 15.4% were Hispanic. The mean Spearman correlation assessing reproducibility across the 33 line items was 0.60 and ranged from 0.44 to 0.72, with no differences in mean correlations by age, sex, race/ethnicity, education, or body mass index (BMI). Reproducibility was similar across food groups (ρ = 0.62; range: 0.44-0.68), with differences by sex, ethnicity, age, or BMI observed for some food groups (e.g., sugar-sweetened beverages). Pearson correlations for the reproducibility of 2 major eating patterns, "fast food" and "whole food," were 0.73 and 0.72, respectively. Conclusion These results show good 1-y reproducibility of assessed high school diet, as reported from memory in adulthood, by line item, food group, and dietary pattern, with noted differences by demographic characteristics.
Collapse
Affiliation(s)
- Kristen D Brantley
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Terryl J Hartman
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA.,Winship Cancer Institute, Atlanta, GA
| | - Alpa V Patel
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | - Susan M Gapstur
- Epidemiology Research Program, American Cancer Society, Atlanta, GA
| | - W D Flanders
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | | |
Collapse
|
39
|
Zingue S, Njuh AN, Tueche AB, Tamsa J, Tchoupang EN, Kakene SD, Sipping MTK, Njamen D. In Vitro Cytotoxicity and In Vivo Antimammary Tumor Effects of the Hydroethanolic Extract of Acacia seyal (Mimosaceae) Stem Bark. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2024602. [PMID: 29770327 PMCID: PMC5889903 DOI: 10.1155/2018/2024602] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/11/2018] [Accepted: 02/19/2018] [Indexed: 12/21/2022]
Abstract
The present study was designed to evaluate the in vitro and in vivo antitumor effects of A. seyal hydroethanolic extract on breast cancer. The cytotoxicity of A. seyal extract was evaluated using resazurin reduction assay in 9 cell lines. Further, the protective effect of the hydroethanolic extract of A. seyal stem barks was evaluated on 7,12-dimethylbenz(a)anthracene- (DMBA-) induced breast cancer rat model. Incidence, burden, volume, and histological analysis of mammary tumors were measured. The Acacia seyal extract exhibited CC50 of 100 in MCF-7 cells after 24 h. In vivo, no tumors were detected in rats from the control group, while 11 rats out of 12 (91.66%) developed mammary tumors in the DMBA-exposed group receiving only the vehicle. Acacia seyal extract significantly (p < 0.01) and in the dose-dependent manner reduced tumor incidence (3 rats out of 12 at the dose of 300 mg/kg), burden [62.1% (150 mg/kg) and 65.8% (300 mg/kg)], and mass. It protected rats against DMBA-induced breast hyperplasia, with an optimal effect at the dose of 300 mg/kg. Taken altogether, these results suggest that the hydroethanolic extract of Acacia seyal might contain phytoconstituents endowed with antitumoral properties, which could protect against the breast cancer induced in rats.
Collapse
Affiliation(s)
- Stephane Zingue
- Department of Life and Earth Sciences, Higher Teachers' Training College, University of Maroua, P.O. Box 55, Maroua, Cameroon
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde 1, P.O. Box 812, Yaounde, Cameroon
| | - Amstrong Nang Njuh
- Department of Biological Sciences, Faculty of Science, University of Maroua, P.O. Box 814, Maroua, Cameroon
| | - Alain Brice Tueche
- Department of Biological Sciences, Faculty of Science, University of Maroua, P.O. Box 814, Maroua, Cameroon
| | - Jeremie Tamsa
- Department of Life and Earth Sciences, Higher Teachers' Training College, University of Maroua, P.O. Box 55, Maroua, Cameroon
| | - Edwige Nana Tchoupang
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde 1, P.O. Box 812, Yaounde, Cameroon
| | - Stanislas Djaouli Kakene
- Department of Life and Earth Sciences, Higher Teachers' Training College, University of Maroua, P.O. Box 55, Maroua, Cameroon
| | | | - Dieudonné Njamen
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde 1, P.O. Box 812, Yaounde, Cameroon
| |
Collapse
|
40
|
Martins FC, Botelho MF, Cabrita AM, de Oliveira CF. Influence of Normal Mammary Epithelium on Breast Cancer Progression: The Protective Role of Early Pregnancy. TUMORI JOURNAL 2018. [DOI: 10.1177/548.6522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Filipe Correia Martins
- Department of Gynecology, University Hospital of Coimbra
- CIMAGO (Oncobiology, Genetics and Environment Research Center), Coimbra Medical School
- Biophysics and Biomathematics Institute, IBILI, Coimbra Medical School
- Experimental Pathology Institute, Coimbra Medical School, Coimbra, Portugal
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA; Gulbenkian Program for Advanced Medical Education
| | - Maria Filomena Botelho
- CIMAGO (Oncobiology, Genetics and Environment Research Center), Coimbra Medical School
- Biophysics and Biomathematics Institute, IBILI, Coimbra Medical School
| | - António Manuel Cabrita
- CIMAGO (Oncobiology, Genetics and Environment Research Center), Coimbra Medical School
- Experimental Pathology Institute, Coimbra Medical School, Coimbra, Portugal
| | - Carlos Freire de Oliveira
- Department of Gynecology, University Hospital of Coimbra
- CIMAGO (Oncobiology, Genetics and Environment Research Center), Coimbra Medical School
| |
Collapse
|
41
|
Martins FC, Teixeira F, Reis I, Geraldes N, Cabrita AMS, Dias MF. Increased Transglutaminase 2 and GLUT-1 Expression in Breast Tumors not Susceptible to Chemoprevention with Antioxidants. TUMORI JOURNAL 2018; 95:227-32. [DOI: 10.1177/030089160909500215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Goals Expression of GLUT-1 and transglutaminase 2 is increased in aggressive breast cancer, whereas claudin-1, which is expressed in normal tissues, is absent in such tumors. This experimental study was undertaken to establish the aggressiveness and prognosis of DMBA-induced mammary tumors in female Wistar rats based on the assessment of these markers. Materials and methods The rats were divided into two groups, a control group (n = 70) and a chemoprevention group (n = 70). Breast tumors were induced in both groups by administration of 7,12-dimethylbenz[a]anthracene (DMBA). The chemoprevention group also received alpha-tocopherol and a solution of micronutrients containing ascorbic acid and selenium. Neoplastic lesions of both groups were randomly selected for immunohistochemical assessment of the expression of GLUT-1, transglutaminase 2 and claudin-1. Results A higher proportion of mammary tumors expressed GLUT-1 and transglutaminase 2 in the chemoprevention group. Claudin-1 expression was absent in all tumors of both groups. Conclusions These results are suggestive of increased aggressiveness of tumors not susceptible to chemoprevention by the agents used in this study.
Collapse
Affiliation(s)
- Filipe C Martins
- Gynecology Department, University Hospital of Coimbra, Coimbra
- Institute of Experimental Pathology, Coimbra Faculty of Medicine, Coimbra
- CIMAGO, Coimbra Faculty of Medicine, Coimbra, Portugal
| | - Filipa Teixeira
- Institute of Experimental Pathology, Coimbra Faculty of Medicine, Coimbra
- CIMAGO, Coimbra Faculty of Medicine, Coimbra, Portugal
| | - Ines Reis
- Institute of Experimental Pathology, Coimbra Faculty of Medicine, Coimbra
- CIMAGO, Coimbra Faculty of Medicine, Coimbra, Portugal
| | - Nuno Geraldes
- Institute of Experimental Pathology, Coimbra Faculty of Medicine, Coimbra
- CIMAGO, Coimbra Faculty of Medicine, Coimbra, Portugal
| | - AM Silvério Cabrita
- Institute of Experimental Pathology, Coimbra Faculty of Medicine, Coimbra
- CIMAGO, Coimbra Faculty of Medicine, Coimbra, Portugal
| | - Margarida F Dias
- Gynecology Department, University Hospital of Coimbra, Coimbra
- Institute of Experimental Pathology, Coimbra Faculty of Medicine, Coimbra
- CIMAGO, Coimbra Faculty of Medicine, Coimbra, Portugal
| |
Collapse
|
42
|
Ruiz R, Herrero C, Strasser-Weippl K, Touya D, St. Louis J, Bukowski A, Goss PE. Epidemiology and pathophysiology of pregnancy-associated breast cancer: A review. Breast 2017; 35:136-141. [DOI: 10.1016/j.breast.2017.07.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/10/2017] [Accepted: 07/12/2017] [Indexed: 11/24/2022] Open
|
43
|
Bertrand KA, Gerlovin H, Bethea TN, Palmer JR. Pubertal growth and adult height in relation to breast cancer risk in African American women. Int J Cancer 2017; 141:2462-2470. [PMID: 28845597 DOI: 10.1002/ijc.31019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/31/2017] [Accepted: 08/16/2017] [Indexed: 01/09/2023]
Abstract
Adult height has been positively associated with breast cancer risk. The timing of pubertal growth-as measured by age at menarche and age at attained height-may also influence risk. We evaluated associations of adult height, age at attained height, and age at menarche with incidence of invasive breast cancer in 55,687 African American women in the prospective Black Women's Health Study. Over 20 years, 1,826 invasive breast cancers [1,015 estrogen receptor (ER) positive; 542 ER negative] accrued. We used multivariable Cox proportional hazards regression to estimate hazards ratios (HRs) and 95% confidence intervals (CIs) for associations with breast cancer overall and by ER status, mutually adjusted for the three factors of interest. Adult height was associated with increased risk of ER+ breast cancer (HR for ≥70 inches vs ≤63 inches: 1.44; 95% CI: 1.09, 1.89) but not ER- (corresponding HR: 1.16; 95% CI: 0.78, 1.71) (p heterogeneity = 0.34). HRs for attained height before age 13 versus age >17 were 1.30 (95% CI: 0.96, 1.76) for ER+ and 1.25 (95% CI: 0.80, 1.96) for ER- breast cancer. Results for age at menarche (≤11 vs ≥14 years) were similar for ER+ and ER- breast cancer (HR for breast cancer overall: 1.30; 95% CI: 1.12, 1.50). We confirmed height as a strong risk factor for ER+ breast cancer in African American women and identified early age at attained height as a risk factor for both ER+ and ER- breast cancer, albeit without statistical significance of the latter associations. While adult height and timing of pubertal growth are inter-related, our findings suggest that they may be independent risk factors for breast cancer.
Collapse
Affiliation(s)
| | - Hanna Gerlovin
- Slone Epidemiology Center at Boston University, Boston, MA.,Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Traci N Bethea
- Slone Epidemiology Center at Boston University, Boston, MA
| | - Julie R Palmer
- Slone Epidemiology Center at Boston University, Boston, MA
| |
Collapse
|
44
|
Harris HR, Willett WC, Vaidya RL, Michels KB. An Adolescent and Early Adulthood Dietary Pattern Associated with Inflammation and the Incidence of Breast Cancer. Cancer Res 2017; 77:1179-1187. [PMID: 28249935 DOI: 10.1158/0008-5472.can-16-2273] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/17/2016] [Accepted: 11/23/2016] [Indexed: 11/16/2022]
Abstract
Adolescence is a highly susceptible period for mammary carcinogenesis, but few prospective studies have examined the role of adolescent diet in breast cancer risk. Reduced rank regression has previously been used to identify a dietary pattern associated with markers of inflammation (C-reactive protein, IL6, and TNFα receptor 2). We investigated whether an adolescent and early adulthood inflammatory dietary pattern was associated with breast cancer among 45,204 women in the Nurses' Health Study II using reduced rank regression. Participants completed a food frequency questionnaire in 1998 about their high school diet (HS-FFQ) and a FFQ in 1991 when they were ages 27-44 years. Among women who completed the HS-FFQ, 1,477 cases of breast cancer were diagnosed during 22 years of follow-up. An adolescent and early adulthood dietary pattern characterized by inflammation was associated with an increased incidence of premenopausal but not postmenopausal breast cancer. Women in the fifth quintile of the inflammatory pattern score had multivariable adjusted HRs for premenopausal breast cancer of 1.35 for adolescent diet [95% confidence interval (95% CI), 1.06-1.73; Ptrend = 0.002] and 1.41 for early adulthood diet (95% CI, 1.11-1.78; Ptrend = 0.006) compared with women in the first quintile. The corresponding RRs for postmenopausal breast cancer were 0.84 (95% CI, 0.60-1.17) for adolescent and 0.76 (95% CI, 0.54-1.06) for adult intake. Overall, our findings support the notion that an adolescent and early adulthood diet characterized by high intake of sugar-sweetened and diet soft drinks, refined grains, red and processed meat, and margarine, and low intake of green leafy vegetables, cruciferous vegetables, and coffee may increase the incidence of premenopausal breast cancer. Cancer Res; 77(5); 1179-87. ©2017 AACR.
Collapse
Affiliation(s)
- Holly R Harris
- Department of Obstetrics, Gynecology and Reproductive Biology, Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Division of Nutritional Epidemiology, The National Institute for Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Walter C Willett
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts
| | - Rita L Vaidya
- Department of Obstetrics, Gynecology and Reproductive Biology, Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Karin B Michels
- Department of Obstetrics, Gynecology and Reproductive Biology, Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. .,Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
45
|
Imaoka T, Nishimura M, Daino K, Hosoki A, Takabatake M, Kokubo T, Doi K, Showler K, Nishimura Y, Moriyama H, Morioka T, Shimada Y, Kakinuma S. Age Modifies the Effect of 2-MeV Fast Neutrons on Rat Mammary Carcinogenesis. Radiat Res 2017; 188:419-425. [DOI: 10.1667/rr14829.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Tatsuhiko Imaoka
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Mayumi Nishimura
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Kazuhiro Daino
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Ayaka Hosoki
- Radiation Effect Accumulation and Prevention Project, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo 116-8551, Japan
| | - Masaru Takabatake
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Toshiaki Kokubo
- Department of Engineering and Safety, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Kazutaka Doi
- Center for Radiation Protection Knowledge, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Kaye Showler
- Radiobiology for Children's Health Program, NIRS, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo 116-8551, Japan
| | - Yukiko Nishimura
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Hitomi Moriyama
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | - Takamitsu Morioka
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| | | | - Shizuko Kakinuma
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba 263-8555, Japan
| |
Collapse
|
46
|
Dall G, Risbridger G, Britt K. Mammary stem cells and parity-induced breast cancer protection- new insights. J Steroid Biochem Mol Biol 2017; 170:54-60. [PMID: 26907964 DOI: 10.1016/j.jsbmb.2016.02.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/09/2016] [Accepted: 02/18/2016] [Indexed: 11/26/2022]
Abstract
Parity (childbearing) significantly decreases a woman's risk of breast cancer and the protective effect is greater if the woman is younger and has more children. The mechanism/s of parity-induced protection are not known. Although several factors are postulated to play a role, we discuss how a reduction in the number of mammary stem cells (MaSCs) may lead to a reduction in breast cancer risk in parous women. Firstly we review the epidemiology linking childbearing to reduced breast cancer risk and discuss how additional births, a young age at first full term birth, and breastfeeding impact the protection. We then detail the mouse and human studies implicating MaSC in parity induced protection and the in-vivo work being performed in mice to directly investigate the effect of parity on MaSC. Finally we discuss the transplant and lineage tracing experiments assessing MaSC activity according to parity and the need to define if MaSC are indeed more carcinogen sensitive than mature mammary epithelial cells. Continuing and future studies attempting to define the parity induced mechanisms will aid in the development of preventative therapies.
Collapse
Affiliation(s)
- Genevieve Dall
- Metastasis Research Laboratory, Peter MacCallum Cancer Centre, 7 St Andrews Place, East Melbourne 3002, Australia; Department of Anatomy and Developmental Biology, Monash University Clayton, Wellington Rd 3800, Australia
| | - Gail Risbridger
- Department of Anatomy and Developmental Biology, Monash University Clayton, Wellington Rd 3800, Australia
| | - Kara Britt
- Metastasis Research Laboratory, Peter MacCallum Cancer Centre, 7 St Andrews Place, East Melbourne 3002, Australia; Department of Anatomy and Developmental Biology, Monash University Clayton, Wellington Rd 3800, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia.
| |
Collapse
|
47
|
White AJ, D'Aloisio AA, Nichols HB, DeRoo LA, Sandler DP. Breast cancer and exposure to tobacco smoke during potential windows of susceptibility. Cancer Causes Control 2017; 28:667-675. [PMID: 28523418 DOI: 10.1007/s10552-017-0903-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/03/2017] [Indexed: 11/30/2022]
Abstract
PURPOSE An association between smoking and breast cancer is unresolved, although a higher risk from exposure during windows of susceptibility has been proposed. The objective of this prospective study was to evaluate the association between tobacco smoke and breast cancer with a focus on timing of exposure, especially during early life. METHODS Sister study participants (n = 50,884) aged 35-74 were enrolled from 2003 to 2009. Women in the United States and Puerto Rico were eligible if they were breast cancer-free but had a sister with breast cancer. Participants completed questionnaires on smoking and environmental tobacco smoke (ETS) exposure. Cox regression was used to estimate adjusted hazard ratios (HRs) and 95% confidence intervals (95% CIs) for breast cancer risk. RESULTS During follow-up (mean = 6.4 years), 1,843 invasive breast cancers were diagnosed. Neither active smoking nor adult ETS was associated with breast cancer risk. However, never smoking women exposed to ETS throughout their childhood had a 17% higher risk of breast cancer (95% CI 1.00-1.36) relative to those with no exposure. In utero ETS exposure was also associated with breast cancer (HR = 1.16, 95% CI 1.01-1.32) and the HR was most elevated for women born in earlier birth cohorts (<1940, HR = 1.44, 95% CI 1.02-2.02; 1940-1949, HR = 1.28, 95% CI 1.01-1.62). CONCLUSION In utero ETS and ETS exposure during childhood and adolescence were associated with increased risk of breast cancer and associations varied by birth cohort.
Collapse
Affiliation(s)
- Alexandra J White
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, 27709-2233, USA.
| | - Aimee A D'Aloisio
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, 27709-2233, USA.,Social & Scientific Systems, Inc., Durham, NC, 27703, USA
| | - Hazel B Nichols
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Lisa A DeRoo
- Department of Global Public Health & Primary Care, University of Bergen, Bergen, Norway
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, 27709-2233, USA
| |
Collapse
|
48
|
Muenst S, Mechera R, Däster S, Piscuoglio S, Ng CK, Meier-Abt F, Weber WP, Soysal SD. Pregnancy at early age is associated with a reduction of progesterone-responsive cells and epithelial Wnt signaling in human breast tissue. Oncotarget 2017; 8:22353-22360. [PMID: 28423605 PMCID: PMC5410228 DOI: 10.18632/oncotarget.16023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/22/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Pregnancy at early age is the most significant modifiable factor which consistently decreases lifetime breast cancer risk. However, the underlying mechanisms haven't been conclusively identified. Studies in mice suggest a reduction in progesterone-receptor (PR) sensitive epithelial cells as well as a downregulation of the Wnt signaling pathway as being one of the main mechanisms for the protective effect of early pregnancy. The aim of our study was to validate these findings in humans. METHODS We collected benign breast tissue of 125 women who had been stratified according to age at first pregnancy and the occurrence of subsequent breast cancer, and performed immunohistochemistry for PR, Wnt4 and the Wnt-target Versican. RESULTS The number of PR positive epithelial cells was significantly lower in the group of women with early pregnancy and no subsequent breast cancer compared to the group of nulliparous women with subsequent invasive breast cancer (p = 0.0135). In women with early pregnancy, expression of Versican and Wnt4 was significantly lower compared to nulliparous women (p = 0.0036 and p = 0.0241 respectively), and Versican expression was also significant lower compared to women with late pregnancy (p < 0.0001). DISCUSSION Our results confirm prior observations in mice and suggest a role of downregulation of epithelial Wnt signaling in the protective effect of early pregnancy in humans. This results in a decreased proliferation of stem/progenitor cells; therefore, the Wnt signaling pathway may represent a potential target for breast cancer prevention in humans.
Collapse
Affiliation(s)
- Simone Muenst
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Robert Mechera
- Department of Surgery, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Silvio Däster
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | | | - Charlotte K.Y. Ng
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Fabienne Meier-Abt
- Institute of Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Walter P. Weber
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Savas D. Soysal
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
49
|
Alvarado A, Faustino-Rocha AI, Colaço B, Oliveira PA. Experimental mammary carcinogenesis - Rat models. Life Sci 2017; 173:116-134. [PMID: 28188729 DOI: 10.1016/j.lfs.2017.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/26/2017] [Accepted: 02/06/2017] [Indexed: 12/22/2022]
Abstract
Mammary cancer is one of the most common cancers, victimizing more than half a million of women worldwide every year. Despite all the studies in this field, the current therapeutic approaches are not effective and have several devastating effects for patients. In this way, the need to better understand the mammary cancer biopathology and find effective therapies led to the development of several rodent models over years. With this review, the authors intended to provide the readers with an overview of the rat models used to study mammary carcinogenesis, with a special emphasis on chemically-induced models.
Collapse
Affiliation(s)
- Antonieta Alvarado
- Área de Patología, Decanato de Ciencias Veterinarias, Universidad Centroccidental "Lisandro Alvarado", UCLA, Lara, Venezuela; Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Ana I Faustino-Rocha
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, UTAD, Vila Real, Portugal
| | - Bruno Colaço
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Department of Zootechnics, School of Agrarian and Veterinary Sciences, UTAD, Vila Real, Portugal
| | - Paula A Oliveira
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, UTAD, Vila Real, Portugal.
| |
Collapse
|
50
|
SCA-1 Labels a Subset of Estrogen-Responsive Bipotential Repopulating Cells within the CD24 + CD49f hi Mammary Stem Cell-Enriched Compartment. Stem Cell Reports 2017; 8:417-431. [PMID: 28132885 PMCID: PMC5312257 DOI: 10.1016/j.stemcr.2016.12.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 12/26/2022] Open
Abstract
Estrogen stimulates breast development during puberty and mammary tumors in adulthood through estrogen receptor-α (ERα). These effects are proposed to occur via ERα+ luminal cells and not the mammary stem cells (MaSCs) that are ERαneg. Since ERα+ luminal cells express stem cell antigen-1 (SCA-1), we sought to determine if SCA-1 could define an ERα+ subset of EpCAM+/CD24+/CD49fhi MaSCs. We show that the MaSC population has a distinct SCA-1+ population that is abundant in pre-pubertal mammary glands. The SCA-1+ MaSCs have less stem cell markers and less in vivo repopulating activity than their SCA-1neg counterparts. However, they express ERα and specifically enter the cell cycle at puberty. Using estrogen-deficient aromatase knockouts (ArKO), we showed that the SCA-1+ MaSC could be directly modulated by estrogen supplementation. Thus, SCA-1 enriches for an ERα+, estrogen-sensitive subpopulation within the CD24+/CD49fhi MaSC population that may be responsible for the hormonal sensitivity of the developing mammary gland. SCA-1+ delineates ER-positive cells in the CD24+ CD49fhi mammary stem population SCA-1+ cells have lower repopulation activity SCA-1+ cells are estrogen responsive
Collapse
|