1
|
García-Sancha N, Corchado-Cobos R, Pérez-Losada J. Understanding Susceptibility to Breast Cancer: From Risk Factors to Prevention Strategies. Int J Mol Sci 2025; 26:2993. [PMID: 40243654 PMCID: PMC11988588 DOI: 10.3390/ijms26072993] [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] [Received: 02/05/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Breast cancer is the most common malignancy among women globally, with incidence rates continuing to rise. A comprehensive understanding of its risk factors and the underlying biological mechanisms that drive tumor initiation is essential for developing effective prevention strategies. This review examines key non-modifiable risk factors, such as genetic predisposition, demographic characteristics, family history, mammographic density, and reproductive milestones, as well as modifiable risk factors like exogenous hormone exposure, obesity, diet, and physical inactivity. Importantly, reproductive history plays a dual role, providing long-term protection while temporarily increasing breast cancer risk shortly after pregnancy. Current chemoprevention strategies primarily depend on selective estrogen receptor modulators (SERMs), including tamoxifen and raloxifene, which have demonstrated efficacy in reducing the incidence of estrogen receptor-positive breast cancer but remain underutilized due to adverse effects. Emerging approaches such as aromatase inhibitors, RANKL inhibitors, progesterone antagonists, PI3K inhibitors, and immunoprevention strategies show promise for expanding preventive options. Understanding the interactions between risk factors, hormonal influences, and tumorigenesis is critical for optimizing breast cancer prevention and advancing safer, more targeted chemopreventive interventions.
Collapse
Affiliation(s)
- Natalia García-Sancha
- Institute of Molecular and Cellular Biology of Cancer (IBMCC-CIC), CSIC-University of Salamanca, 37007 Salamanca, Spain; (R.C.-C.); (J.P.-L.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Roberto Corchado-Cobos
- Institute of Molecular and Cellular Biology of Cancer (IBMCC-CIC), CSIC-University of Salamanca, 37007 Salamanca, Spain; (R.C.-C.); (J.P.-L.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Jesús Pérez-Losada
- Institute of Molecular and Cellular Biology of Cancer (IBMCC-CIC), CSIC-University of Salamanca, 37007 Salamanca, Spain; (R.C.-C.); (J.P.-L.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
2
|
Vandeloo MJ, Kellen E, Fang CY, Ross EA, Vancoillie L, Bruckers LM, Neven KY, Bijnens EM, Nawrot TS, Van Ongeval C. Age at First Full-term Pregnancy and Other Reproductive Factors Are Associated with Mammographic Breast Density in Postmenopausal Women: A Study in Flanders, Belgium. CANCER RESEARCH COMMUNICATIONS 2025; 5:267-276. [PMID: 39835404 PMCID: PMC11803437 DOI: 10.1158/2767-9764.crc-24-0561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/30/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
SIGNIFICANCE We consider our findings to be highly novel, and to the best of our knowledge, our study is one of the first to investigate associations between age at FFTP and three markers of MBD (GLAND, VBD, and BI-RADS), uncovering that MBD is significantly higher when FFTP occurs after 25.7 years.
Collapse
Affiliation(s)
- Magda J. Vandeloo
- Department of Imaging & Pathology, University of Leuven, Leuven, Belgium
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
- Cancer Prevention and Control, Fox Chase Cancer Center-Temple Health, Philadelphia, Pennsylvania
| | - Eliane Kellen
- LUCK, Campus St Rafael, University Hospital Leuven, Leuven, Belgium
- Centre of Cancer Detection, Bruges, Belgium
| | - Carolyn Y. Fang
- Cancer Prevention and Control, Fox Chase Cancer Center-Temple Health, Philadelphia, Pennsylvania
| | - Eric A. Ross
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center-Temple Health, Philadelphia, Pennsylvania
| | | | | | - Kristof Y. Neven
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Esmée M. Bijnens
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
- Faculty of Science/Department of Environmental Sciences, Open University Heerlen, Heerlen, the Netherlands
| | - Tim S. Nawrot
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
- Department of Public Health and Primary Care, University of Leuven, Leuven, Belgium
| | - Chantal Van Ongeval
- Department of Imaging & Pathology, University of Leuven, Leuven, Belgium
- Department of Radiology, University Hospital Leuven, Leuven, Belgium
| |
Collapse
|
3
|
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
|
4
|
Expression and functions of long non-coding RNA NEAT1 and isoforms in breast cancer. Br J Cancer 2022; 126:551-561. [PMID: 34671127 PMCID: PMC8854383 DOI: 10.1038/s41416-021-01588-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/08/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023] Open
Abstract
NEAT1 is a highly abundant nuclear architectural long non-coding RNA. There are two overlapping NEAT1 isoforms, NEAT1_1 and NEAT1_2, of which the latter is an essential scaffold for the assembly of a class of nuclear ribonucleoprotein bodies called paraspeckles. Paraspeckle formation is elevated by a wide variety of cellular stressors and in certain developmental processes, either through transcriptional upregulation of the NEAT1 gene or through a switch from NEAT1_1 to NEAT1_2 isoform production. In such conditions, paraspeckles modulate cellular processes by sequestering proteins or RNA molecules. NEAT1 is abnormally expressed in many cancers and a growing body of evidence suggests that, in many cases, high NEAT1 levels are associated with therapy resistance and poor clinical outcome. Here we review the current knowledge of NEAT1 expression and functions in breast cancer, highlighting its established role in postnatal mammary gland development. We will discuss possible isoform-specific roles of NEAT1_1 and NEAT1_2 in different breast cancer subtypes, which critically needs to be considered when studying NEAT1 and breast cancer.
Collapse
|
5
|
Slepicka PF, Somasundara AVH, Dos Santos CO. The molecular basis of mammary gland development and epithelial differentiation. Semin Cell Dev Biol 2021; 114:93-112. [PMID: 33082117 PMCID: PMC8052380 DOI: 10.1016/j.semcdb.2020.09.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
Our understanding of the molecular events underpinning the development of mammalian organ systems has been increasing rapidly in recent years. With the advent of new and improved next-generation sequencing methods, we are now able to dig deeper than ever before into the genomic and epigenomic events that play critical roles in determining the fates of stem and progenitor cells during the development of an embryo into an adult. In this review, we detail and discuss the genes and pathways that are involved in mammary gland development, from embryogenesis, through maturation into an adult gland, to the role of pregnancy signals in directing the terminal maturation of the mammary gland into a milk producing organ that can nurture the offspring. We also provide an overview of the latest research in the single-cell genomics of mammary gland development, which may help us to understand the lineage commitment of mammary stem cells (MaSCs) into luminal or basal epithelial cells that constitute the mammary gland. Finally, we summarize the use of 3D organoid cultures as a model system to study the molecular events during mammary gland development. Our increased investigation of the molecular requirements for normal mammary gland development will advance the discovery of targets to predict breast cancer risk and the development of new breast cancer therapies.
Collapse
Affiliation(s)
- Priscila Ferreira Slepicka
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | | | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
6
|
Depypere H, Su Y, Dang N, Poppe B, Stanczyk F, Janssens J, Russo J. Prolonged recombinant pregnancy hormone use in BRCA1 and BRCA2 mutation carriers. Eur J Cancer Prev 2021; 30:195-203. [PMID: 33720054 PMCID: PMC8011504 DOI: 10.1097/cej.0000000000000664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 12/03/2022]
Abstract
BACKGROUND An early first full-time pregnancy substantially reduces the risk of developing breast cancer later in life. Extensive studies indicate that this protective effect is mediated by the pregnancy hormone human chorionic gonadotrophin (hCG). METHODS In this proof-of-concept study 33 women with a BRCA mutation received recombinant-hCG (r-hCG). A 4-mm breast biopsy was obtained before (T1) and after 12 weeks of r-hCG injections (T2), as well as 6 months later (T3). The tissue was examined using RNA-sequencing methodology to determine if the 'high-risk' transcriptomic signature was converted to a 'low-risk' signature as in an early first full-time pregnancy. A stringent clinical safety monitoring was performed. RESULTS The r-hCG administration was well tolerated in all participants. No clinically relevant changes were observed. In 25 women, the RNA quality was good for RNA sequencing in all three breast tissue biopsies. In response to the r-hCG, we observed 1907 differentially expressed genes (DEGs) (1032 up, 875 down) at T2 vs. T1 and 1065 DEGs (897 up, 168 down) at T3 vs. T1 in the group of women (n = 11) not using any hormonal contraceptives during the study. There was no response at T2 vs. T1 and a small number of DEGs, 260 (214 up, 46 down) at T3 vs. T1 in the group of 14 women using contraceptives. CONCLUSIONS In summary, r-hCG has a remarkable effect on the gene expression profile of breast tissues from BRCA1/2 carriers who did not use any contraception. This opens an opportunity for a novel preventive strategy to reduce the incidence of breast cancer.
Collapse
Affiliation(s)
- Herman Depypere
- Department of Gynecology, Breast and Menopause Clinic, University Hospital, Ghent, Belgium
| | - Yanrong Su
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, Philadelphia, Pennsylvania, USA
| | - Nhi Dang
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, Philadelphia, Pennsylvania, USA
| | - Bruce Poppe
- Department of Clinical Genetics, University Hospital, Ghent, Belgium
| | - Frank Stanczyk
- Departments of Obstetrics and Gynecology and Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jaak Janssens
- European Cancer Prevention Organization, University of Hasselt, Hasselt, Belgium
| | - Jose Russo
- The Irma H Russo, MD-Breast Cancer Research Laboratory, Fox Chase Cancer Center-Temple Health, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
García Solá ME, Stedile M, Beckerman I, Kordon EC. An Integrative Single-cell Transcriptomic Atlas of the Post-natal Mouse Mammary Gland Allows Discovery of New Developmental Trajectories in the Luminal Compartment. J Mammary Gland Biol Neoplasia 2021; 26:29-42. [PMID: 33913090 DOI: 10.1007/s10911-021-09488-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
The mammary gland is a highly dynamic organ which undergoes periods of expansion, differentiation and cell death in each reproductive cycle. Partly because of the dynamic nature of the gland, mammary epithelial cells (MECs) are extraordinarily heterogeneous. Single cell RNA-seq (scRNA-seq) analyses have contributed to understand the cellular and transcriptional heterogeneity of this complex tissue. Here, we integrate scRNA-seq data from three foundational reports that have explored the mammary gland cell populations throughout development at single-cell level using 10× Chromium Drop-Seq. We center our analysis on post-natal development of the mammary gland, from puberty to post-involution. The new integrated study corresponds to RNA sequences from 53,686 individual cells, which greatly outnumbers the three initial data sets. The large volume of information provides new insights, as a better resolution of the previously detected Procr+ stem-like cell subpopulation or the identification of a novel group of MECs expressing immune-like markers. Moreover, here we present new pseudo-temporal trajectories of MEC populations at two resolution levels, that is either considering all mammary cell subtypes or focusing specifically on the luminal lineages. Interestingly, the luminal-restricted analysis reveals distinct expression patterns of various genes that encode milk proteins, suggesting specific and non-redundant roles for each of them. In summary, our data show that the application of bioinformatic tools to integrate multiple scRNA-seq data-sets helps to describe and interpret the high level of plasticity involved in gene expression regulation throughout mammary gland post-natal development.
Collapse
Affiliation(s)
- Martín E García Solá
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIByNE), CONICET, Departamento de Fisiología y Biología Molecular y Celular. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Micaela Stedile
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIByNE), CONICET, Departamento de Fisiología y Biología Molecular y Celular. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Inés Beckerman
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIByNE), CONICET, Departamento de Fisiología y Biología Molecular y Celular. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Edith C Kordon
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIByNE), CONICET, Departamento de Fisiología y Biología Molecular y Celular. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
- Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Ivanova E, Le Guillou S, Hue-Beauvais C, Le Provost F. Epigenetics: New Insights into Mammary Gland Biology. Genes (Basel) 2021; 12:genes12020231. [PMID: 33562534 PMCID: PMC7914701 DOI: 10.3390/genes12020231] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/23/2021] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
The mammary gland undergoes important anatomical and physiological changes from embryogenesis through puberty, pregnancy, lactation and involution. These steps are under the control of a complex network of molecular factors, in which epigenetic mechanisms play a role that is increasingly well described. Recently, studies investigating epigenetic modifications and their impacts on gene expression in the mammary gland have been performed at different physiological stages and in different mammary cell types. This has led to the establishment of a role for epigenetic marks in milk component biosynthesis. This review aims to summarize the available knowledge regarding the involvement of the four main molecular mechanisms in epigenetics: DNA methylation, histone modifications, polycomb protein activity and non-coding RNA functions.
Collapse
|
9
|
Boyles AL, Beverly BE, Fenton SE, Jackson CL, Jukic AMZ, Sutherland VL, Baird DD, Collman GW, Dixon D, Ferguson KK, Hall JE, Martin EM, Schug TT, White AJ, Chandler KJ. Environmental Factors Involved in Maternal Morbidity and Mortality. J Womens Health (Larchmt) 2021; 30:245-252. [PMID: 33211615 PMCID: PMC7891208 DOI: 10.1089/jwh.2020.8855] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nongenetic, environmental factors contribute to maternal morbidity and mortality through chemical exposures via air, water, soil, food, and consumer products. Pregnancy represents a particularly sensitive window of susceptibility during which physiological changes to every major organ system increase sensitivity to chemicals that can impact a woman's long-term health. Nonchemical stressors, such as low socioeconomic status, may exacerbate the effects of chemical exposures on maternal health. Racial/ethnic minorities are exposed disproportionately to both chemicals and nonchemical stressors, which likely contribute to the observed health disparities for maternal morbidities and mortality. Epidemiological studies linking exposures to adverse maternal health outcomes underscore the importance of environmental health impacts, and mechanistic studies in model systems reveal how chemicals perturb biological pathways and processes. Environmental stressors are associated with a variety of immediate maternal health impacts, including hypertensive disorders of pregnancy, fibroids, and infertility, as well as long-term maternal health impacts, such as higher risk of breast cancer and metabolic disorders. Identifying and reducing a pregnant woman's environmental exposures is not only beneficial to her offspring but also important to preserve her short- and long-term health.
Collapse
Affiliation(s)
- Abee L. Boyles
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Brandiese E. Beverly
- National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Suzanne E. Fenton
- National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Chandra L. Jackson
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
- Intramural Program, National Institute on Minority Health and Health Disparities, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, USA
| | - Anne Marie Z. Jukic
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Vicki L. Sutherland
- National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Donna D. Baird
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Gwen W. Collman
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Darlene Dixon
- National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Kelly K. Ferguson
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Janet E. Hall
- Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Elizabeth M. Martin
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
- Postdoctoral Research Associate Training Program, National Institute of General Medical Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, USA
| | - Thaddeus T. Schug
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Alexandra J. White
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| | - Kelly J. Chandler
- Office of Policy, Planning, and Evaluation, National Institute of Environmental Health Sciences, National Institutes of Health, U.S. Department of Health and Human Services, Durham, North Carolina, USA
| |
Collapse
|
10
|
Gutiérrez-Díez PJ, Gomez-Pilar J, Hornero R, Martínez-Rodríguez J, López-Marcos MA, Russo J. The role of gene to gene interaction in the breast's genomic signature of pregnancy. Sci Rep 2021; 11:2643. [PMID: 33514799 PMCID: PMC7846553 DOI: 10.1038/s41598-021-81704-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Full-term pregnancy at an early age confers long-term protection against breast cancer. Published data shows a specific transcriptomic profile controlling chromatin remodeling that could play a relevant role in the pregnancy-induced protection. This process of chromatin remodeling, induced by the breast differentiation caused by the first full-term pregnancy, has mainly been measured by the expression level of genes individually considered. However, genes equally expressed during the process of chromatin remodeling may behave differently in their interaction with other genes. These changes at the gene cluster level could constitute an additional dimension of chromatin remodeling and therefore of the pregnancy-induced protection. In this research, we apply Information and Graph Theories, Differential Co-expression Network Analysis, and Multiple Regression Analysis, specially designed to examine structural and informational aspects of data sets, to analyze this question. Our findings demonstrate that, independently of the changes in the gene expression at the individual level, there are significant changes in gene-gene interactions and gene cluster behaviors. These changes indicate that the parous breast, through the process of early full-term pregnancy, generates more modules in the networks, with higher density, and a genomic structure performing additional and more complex functions than those found in the nulliparous breast.
Collapse
Affiliation(s)
- Pedro J Gutiérrez-Díez
- IMUVA Mathematical Institute, University of Valladolid, Valladolid, Spain
- Faculty of Economics, University of Valladolid, Valladolid, Spain
| | - Javier Gomez-Pilar
- Biomedical Engineering Group, University of Valladolid, Paseo de Belén, 15, 47011, Valladolid, Spain.
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Valladolid, Spain.
| | - Roberto Hornero
- IMUVA Mathematical Institute, University of Valladolid, Valladolid, Spain
- Biomedical Engineering Group, University of Valladolid, Paseo de Belén, 15, 47011, Valladolid, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales Y Nanomedicina (CIBER-BBN), Valladolid, Spain
| | - Julia Martínez-Rodríguez
- IMUVA Mathematical Institute, University of Valladolid, Valladolid, Spain
- Faculty of Economics, University of Valladolid, Valladolid, Spain
| | - Miguel A López-Marcos
- IMUVA Mathematical Institute, University of Valladolid, Valladolid, Spain
- Faculty of Science, University of Valladolid, Valladolid, Spain
| | - Jose Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center - Temple University Health System, Philadelphia, USA
| |
Collapse
|
11
|
Daraei A, Izadi P, Khorasani G, Nafissi N, Naghizadeh MM, Meysamie A, Mansoori Y, Nariman-Saleh-Fam Z, Bastami M, Saadatian Z, Roshan SJ, Bayani N, Tavakkoly-Bazzaz J. A methylation signature at the CpG island promoter of estrogen receptor beta (ER-β) in breasts of women may be an early footmark of lack of breastfeeding and nulliparity. Pathol Res Pract 2020; 218:153328. [PMID: 33422777 DOI: 10.1016/j.prp.2020.153328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/17/2020] [Accepted: 12/23/2020] [Indexed: 11/24/2022]
Abstract
Although little is known regarding the mechanisms behind the onset of breast cancer (BC) through reproductive risk factors, new researches have highlighted some early tumor-related methylation footmarks in the breast tissue of apparently clinically healthy women as their potential epigenetic mechanism. Previous evidence supports that the estrogen receptor beta (ER-β), whose anti-cancer roles had already been revealed in BC, is downregulated in the breasts of healthy nulliparous women. Nevertheless, data on such a link about its methylation alterations have not been reported. The goal of current study was to determine possible methylation alterations at CpG island promoter of the ER-β gene, including promoter 0 N and exon 0 N, in relation to aspects of reproductive history in the healthy breasts. The DNA was extracted from the breasts of 120 subjects undergoing cosmetic mammoplasty. Thereafter, the methylation levels of targeted regions in ER-β gene were determined by using MeDIP-qPCR assay. The results revealed that ER-β exon 0 N had no methylation in 84.2 % of the women, whereas the rest, comprising 2.5 % and 13.3 % of the samples, showed a lower and higher of its methylation, respectively. Interestingly, nulliparous women were found to have an elevated methylation level of the ER-β exon 0 N than parous women (P = 0.036). Moreover, we observed a high methylation of the ER-β exon 0 N in the breasts of non-breastfeeding women compared to breastfeeding subgroup (P = 0.048). Likewise, the non-breastfeeding subgroup showed exon 0N high methylation in comparison to women with breastfeeding >24 months (P = 0.023). Finally, although we found that 6.67 % of the samples had a high methylation level at the promoter 0N, no any relationship was found between its methylation and reproductive history. These results may provide key clues to revealing the epigenetic mechanism through which the nulliparity and lack of breastfeeding influencing the risk factor of BC as well as introducing the potential new early prediction and prevention strategies. Although further investigations need to be done in order to gain a better understanding the roles of these epigenetic signatures.
Collapse
Affiliation(s)
- Abdolreza Daraei
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Pantea Izadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasemali Khorasani
- Division of Plastic and Reconstructive Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Nahid Nafissi
- Surgical Department, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alipasha Meysamie
- Community and Preventive Medicine Department, Medical Faculty, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Ziba Nariman-Saleh-Fam
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Bastami
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Zahra Saadatian
- Department of Physiology, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Samaneh Jafari Roshan
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Niloofar Bayani
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| | - Javad Tavakkoly-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Feigman MJ, Moss MA, Chen C, Cyrill SL, Ciccone MF, Trousdell MC, Yang ST, Frey WD, Wilkinson JE, Dos Santos CO. Pregnancy reprograms the epigenome of mammary epithelial cells and blocks the development of premalignant lesions. Nat Commun 2020; 11:2649. [PMID: 32461571 PMCID: PMC7253414 DOI: 10.1038/s41467-020-16479-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Pregnancy causes a series of cellular and molecular changes in mammary epithelial cells (MECs) of female adults. In addition, pregnancy can also modify the predisposition of rodent and human MECs to initiate oncogenesis. Here, we investigate how pregnancy reprograms enhancer chromatin in the mammary epithelium of mice and influences the transcriptional output of the oncogenic transcription factor cMYC. We find that pregnancy induces an expansion of the active cis-regulatory landscape of MECs, which influences the activation of pregnancy-related programs during re-exposure to pregnancy hormones in vivo and in vitro. Using inducible cMYC overexpression, we demonstrate that post-pregnancy MECs are resistant to the downstream molecular programs induced by cMYC, a response that blunts carcinoma initiation, but does not perturb the normal pregnancy-induced epigenomic landscape. cMYC overexpression drives post-pregnancy MECs into a senescence-like state, and perturbations of this state increase malignant phenotypic changes. Taken together, our findings provide further insight into the cell-autonomous signals in post-pregnancy MECs that underpin the regulation of gene expression, cellular activation, and resistance to malignant development. Mammary epithelial cells are epigenetically modified during pregnancy, these changes can influence the pre-disposition to cancer. Here, the authors examine the epigenetic landscape of mammary epithelial cells pre and post pregnancy and identify changes to the epigenetic landscape, which can protect mice from Myc induced cancer.
Collapse
Affiliation(s)
- Mary J Feigman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Matthew A Moss
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, 11549, USA
| | - Chen Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Samantha L Cyrill
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Michael F Ciccone
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | | | - Shih-Ting Yang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Wesley D Frey
- School of Medicine, Tulane University, New Orleans, LA, 70118, USA
| | - John E Wilkinson
- Department of Comparative Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA.
| |
Collapse
|
13
|
The Basal Level of Gene Expression Associated with Chromatin Loosening Shapes Waddington Landscapes and Controls Cell Differentiation. J Mol Biol 2020; 432:2253-2270. [PMID: 32105732 DOI: 10.1016/j.jmb.2020.02.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 01/01/2023]
Abstract
The baseline level of transcription, which is variable and difficult to quantify, seriously complicates the normalization of comparative transcriptomic data, but its biological importance remains unappreciated. We show that this currently neglected ingredient is essential for controlling gene network multistability and therefore cellular differentiation. Basal expression is correlated to the degree of chromatin loosening measured by DNA accessibility and systematically leads to cellular dedifferentiation as assessed by transcriptomic signatures, irrespective of the molecular and cellular tools used. Modeling gene network motifs formally involved in developmental bifurcations reveals that the epigenetic landscapes of Waddington are restructured by the level of nonspecific expression, such that the attractors of progenitor and differentiated cells can be mutually exclusive. This mechanism is universal and holds beyond the particular nature of the genes involved, provided the multistable circuits are correctly described with autonomous basal expression. These results explain the relationships long established between gene expression noise, chromatin decondensation and cellular dedifferentiation, and highlight how heterochromatin maintenance is essential for preventing pathological cellular reprogramming, age-related diseases, and cancer.
Collapse
|
14
|
Salama EA, Adbeltawab RE, El Tayebi HM. XIST and TSIX: Novel Cancer Immune Biomarkers in PD-L1-Overexpressing Breast Cancer Patients. Front Oncol 2020; 9:1459. [PMID: 31998636 PMCID: PMC6966712 DOI: 10.3389/fonc.2019.01459] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
Escaping antitumor immunity is a hallmark in cancer progression. Programmed cell death protein 1 (PD-1) is an immune checkpoint receptor responsible for the maintenance of immune tolerance; PD-1 ligand (PD-L1) is overexpressed in tumor cells, simplifying their escape from the immune system through T-cell function suppression. Notwithstanding that cancer antigen (CA)125, carcinoembryonic antigen (CEA), CA15-3, and alpha-fetoprotein (AFP) are among conventional breast cancer diagnostic biomarkers, their lack of sensitivity and specificity resides among their major limitations. Furthermore, human epidermal growth factor receptor (HER)2 and interleukin (IL)-6-demonstrated as breast cancer immune biomarkers-still possess limitations, for instance, technical detection problems and stability problems, which necessitate the discovery of novel, stable non-invasive cancer immune biomarkers. XIST and TSIX are two long non-coding (lnc)RNAs possessing a role in X chromosome inactivation (XCI) as well as in breast cancer (BC). In the present study, they were investigated as stable non-invasive breast cancer immune biomarkers. The study demonstrated that PD-L1 was overexpressed in the different molecular subtypes of breast cancer patients as well as in MDA-MB-231 cells. Furthermore, lncRNAs XIST and TSIX were markedly increased in the tissues, lymph nodes, and different body fluids of breast cancer patients compared to controls. In addition, XIST and TSIX were differentially expressed in subtypes of BC patients, and their levels were correlated to PD-L1 expression level. In conclusion, this correlative study has shed light on the role of both lncRNAs XIST and TSIX as potential non-invasive BC immune biomarkers reflecting the evaded immune system of the patient and overcoming the instability problem of common BC biomarkers.
Collapse
Affiliation(s)
- Esraa A. Salama
- Molecular Pharmacology Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Reda E. Adbeltawab
- Department of Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hend M. El Tayebi
- Molecular Pharmacology Research Group, Department of Pharmacology and Toxicology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| |
Collapse
|
15
|
Barton M, Santucci-Pereira J, Vaccaro OG, Nguyen T, Su Y, Russo J. BC200 overexpression contributes to luminal and triple negative breast cancer pathogenesis. BMC Cancer 2019; 19:994. [PMID: 31646972 PMCID: PMC6813071 DOI: 10.1186/s12885-019-6179-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 09/20/2019] [Indexed: 01/04/2023] Open
Abstract
Background Long non coding RNAs (lncRNAs) are RNA molecules longer than 200 nucleotides that are not translated into proteins, but regulate the transcription of genes involved in different cellular processes, including cancer. Epidemiological analyses have demonstrated that parous women have a decreased risk of developing breast cancer in postmenopausal years if they went through a full term pregnancy in their early twenties. We here provide evidence of the role of BC200 in breast cancer and, potentially, in pregnancy’s preventive effect in reducing the lifetime risk of developing breast cancer. Methods Transcriptome analysis of normal breast of parous and nulliparous postmenopausal women revealed that several lncRNAs are differentially expressed in the parous breast. RNA sequencing of healthy postmenopausal breast tissue biopsies from eight parous and eight nulliparous women showed that there are 42 novel lncRNAs differentially expressed between these two groups. Screening of several of these 42 lncRNAs by RT-qPCR in different breast cancer cell lines, provided evidence that one in particular, lncEPCAM (more commonly known as BC200), was a strong candidate involved in cancer progression. Proliferation, migration, invasion and xerograph studies confirmed this hypothesis. Results The poorly studied oncogenic BC200 was selected to be tested in vitro and in vivo to determine its relevance in breast cancer and also to provide us with an understanding of its role in the increased susceptibility of the nulliparous women to cancer. Our results show that BC200 is upregulated in nulliparous women, and breast cancer cells and tissue. The role of BC200 is not completely understood in any of the breast cancer subtypes. We here provide evidence that BC200 has a role in luminal breast cancer as well as in the triple negative breast cancer subtype. Conclusion When overexpressed in luminal and triple negative breast cancer cell lines, BC200 shows increased proliferation, migration, and invasion in vitro. In vivo, overexpression of BC200 increased tumor size. Although treatment for cancer using lncRNAs as targets is in its infancy, the advancement in knowledge and technology to study their relevance in disease could lead to the development of novel treatment and preventive strategies for breast cancer.
Collapse
Affiliation(s)
- Maria Barton
- Biochemistry Department, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA. .,The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, 19111, USA.
| | - Julia Santucci-Pereira
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, 19111, USA
| | - Olivia G Vaccaro
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, 19111, USA
| | - Theresa Nguyen
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, 19111, USA
| | - Yanrong Su
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, 19111, USA
| | - Jose Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, 19111, USA
| |
Collapse
|
16
|
Slepicka PF, Cyrill SL, Dos Santos CO. Pregnancy and Breast Cancer: Pathways to Understand Risk and Prevention. Trends Mol Med 2019; 25:866-881. [PMID: 31383623 DOI: 10.1016/j.molmed.2019.06.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022]
Abstract
Several studies have made strong efforts to understand how age and parity modulate the risk of breast cancer. A holistic understanding of the dynamic regulation of the morphological, cellular, and molecular milieu of the mammary gland offers insights into the drivers of breast cancer development as well as into potential prophylactic interventions, the latter being a longstanding ambition of the research and clinical community aspiring to eradicate the disease. In this review we discuss mechanisms that react to pregnancy signals, and we delineate the nuances of pregnancy-associated dynamism that contribute towards either breast cancer development or prevention. Further definition of the molecular basis of parity and breast cancer risk may allow the elaboration of tools to predict and survey those who are at risk of breast cancer development.
Collapse
Affiliation(s)
- Priscila F Slepicka
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Samantha L Cyrill
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Camila O Dos Santos
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
17
|
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: 29] [Impact Index Per Article: 4.8] [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
|
18
|
Zendehbad Z, Izadi P, Daraei A, Yekaninejad MS, Nafissi N, Younosi N, Khorasani G, Tavakkoly Bazzaz J. Early Parity Epigenetic Footprint of FOXA1 Gene Body in Normal Breast Tissue of Iranian Women. IRANIAN BIOMEDICAL JOURNAL 2019; 23:99-106. [PMID: 30054444 PMCID: PMC6707108 DOI: 10.29252/.23.2.99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/26/2018] [Accepted: 07/04/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Young age at first full-term pregnancy (FFTP) is an important factor in breast cancer risk reduction. It is postulated that this protective effect is the result of stable molecular signatures imprinted by physiological process of pregnancy, but the molecular mechanism of this protective role is unclear. The aim of the current study was to identify the effect of early FFTP on methylation status of FOXA1 gene body. FOXA1 is an essential transcription factor for mammary gland development and estrogen responsiveness of breast tissue. METHODS Fresh frozen normal breast tissues (n = 51) were collected from Iranian women who underwent cosmetic mammoplasty (27 nulliparous women and 24 parous women who have experienced first pregnancy before the age of 25). DNA was extracted and then methylated DNA immunoprecipitation (MeDIP) real-time PCR was used to assess FOXA1 gene body methylation. RESULTS Our results revealed that FOXA1 methylation level is significantly higher in early parous compared with nulliparous group (p = 0.041). CONCLUSION Our study provides new hint about the association between early FFTP and epigenetic modifications within gene body of FOXA1 in normal breast tissue. More investigation is required for clarifying molecular mechanisms underlying this association in order to develop breast cancer prevention strategies.
Collapse
Affiliation(s)
- Zahra Zendehbad
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Pantea Izadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdolreza Daraei
- Genetic Department, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mir Saeed Yekaninejad
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Nahid Nafissi
- Surgical Department, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Nasim Younosi
- Surgical Department, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghasemali Khorasani
- Division of Plastic and Reconstructive Surgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Tavakkoly Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
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: 23] [Impact Index Per Article: 3.8] [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
|
20
|
Mansoori Y, Zendehbad Z, Askari A, Kouhpayeh A, Tavakkoly-Bazzaz J, Nariman-Saleh-Fam Z, Bastami M, Saadatian Z, Mansoori B, Yousefvand A, Mansoori H, Daraei A. Breast cancer-linked lncRNA u-Eleanor is upregulated in breast of healthy women with lack or short duration of breastfeeding. J Cell Biochem 2018; 120:9869-9876. [PMID: 30548300 DOI: 10.1002/jcb.28269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022]
Abstract
Recently, it has been revealed that estrogen-related reproductive factors are linked with some early gene expression lesions associated with malignancy in clinically healthy breasts. Accordingly, the aim of the current study was to evaluate the association of expression levels of estrogen-related long noncoding RNAs (lncRNAs) upstream Eleanor (u-Eleanor) and HOX antisense intergenic RNA (HOTAIR) with the different patterns of reproductive factors in breast tissue of healthy women. The subjects of this study were 98 cancer-free women who had undergone cosmetic mammoplasty. The expression levels of u-Eleanor and HOTAIR were measured using quantitative real-time polymerase chain reaction. The results of the current study showed that the women without a history of breastfeeding had a high-level expression of u-Eleanor compared with the women with a breastfeeding duration greater than 6 to 24 months (P = 0.03) as well as the women with a breastfeeding duration of more than 24 months (P = 0.005). Furthermore, a higher expression of u-Eleanor was found in the women with a short breastfeeding duration for 1 to 6 months than that in the women with a breastfeeding duration of greater than 24 months (P = 0.02). In the same way, the results of correlation test (r = -0.258; P = 0.036) and multivariate regression model (β = -0.321; P = 0.023) are indicative of a significant relationship of elevated expression of u-Eleanor with decreasing breastfeeding duration in the women. These findings could be important to identify the molecular mechanisms behind the relationship between a lack or short duration of the breastfeeding and the risk of breast cancer, which has previously been reported by epidemiological studies.
Collapse
Affiliation(s)
- Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Zahra Zendehbad
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Askari
- Department of Orthopedy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Kouhpayeh
- Department of Pharmacology, Fasa University of Medical Sciences, Fasa, Iran
| | - Javad Tavakkoly-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ziba Nariman-Saleh-Fam
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Bastami
- Immunology Research Center, Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Saadatian
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnam Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Amin Yousefvand
- Department of Food Hygiene, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Hosein Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Abdolreza Daraei
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
21
|
Husby A, Wohlfahrt J, Øyen N, Melbye M. Pregnancy duration and breast cancer risk. Nat Commun 2018; 9:4255. [PMID: 30353005 PMCID: PMC6199327 DOI: 10.1038/s41467-018-06748-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/20/2018] [Indexed: 12/29/2022] Open
Abstract
Full-term pregnancies reduce a woman’s long-term breast cancer risk, while abortions have been shown to have no effect. The precise minimal duration of pregnancy necessary to lower a woman’s breast cancer risk is, however, unknown. Here we provide evidence which point to the protective effect of pregnancy on breast cancer risk arising precisely at the 34th pregnancy week. Using a cohort of 2.3 million Danish women, we found the reduction in breast cancer risk was not observed for pregnancies lasting 33 weeks or less, but restricted to those pregnancies lasting 34 weeks or longer. We further found that parity, socioeconomic status, and vital status of the child at birth did not explain the association, and also replicated our finding in data from 1.6 million women in Norway. We suggest that a distinct biological effect introduced around week 34 of pregnancy holds the key to understand pregnancy-associated breast cancer protection. It is known that full-term pregnancies can reduce a woman’s breast cancer risk. Here, the authors interrogate data from 2.3 million Danish women, showing that this protective effect arises at precisely the 34th week of the pregnancy, and replicated this finding in 1.6 million women from Norway.
Collapse
Affiliation(s)
- Anders Husby
- Department of Epidemiology Research, Statens Serum Institut, DK-2300, Copenhagen, Denmark.,Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jan Wohlfahrt
- Department of Epidemiology Research, Statens Serum Institut, DK-2300, Copenhagen, Denmark
| | - Nina Øyen
- Department of Epidemiology Research, Statens Serum Institut, DK-2300, Copenhagen, Denmark.,Department of Global Public Health and Primary Care, Faculty of Medicine, University of Bergen, N-5020, Bergen, Norway.,Department of Medical Genetics, Haukeland University Hospital, N-5021, Bergen, Norway
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, DK-2300, Copenhagen, Denmark. .,Department of Clinical Medicine, University of Copenhagen, DK-2100, Copenhagen, Denmark. .,Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
22
|
Takabatake M, Daino K, Imaoka T, Blyth BJ, Kokubo T, Nishimura Y, Showler K, Hosoki A, Moriyama H, Nishimura M, Kakinuma S, Fukushi M, Shimada Y. Differential effect of parity on rat mammary carcinogenesis after pre- or post-pubertal exposure to radiation. Sci Rep 2018; 8:14325. [PMID: 30254198 PMCID: PMC6156598 DOI: 10.1038/s41598-018-32406-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 09/03/2018] [Indexed: 01/28/2023] Open
Abstract
Radiation exposure during the peri-pubertal period is a proven risk factor for breast cancer, whereas parity is an established protective factor. The present study investigated whether parity imposes differential protective effects against radiation-induced rat mammary carcinoma depending on the age at exposure. Pre- and post-pubertal female rats, irradiated or left unirradiated, were mated and allowed to nurse until weaning or left unmated. Appearance of mammary tumors was monitored, and serum concentrations of estradiol and progesterone were measured following weaning. Carcinomas were evaluated by immunohistochemistry for estrogen receptor, progesterone receptor, and the cell proliferation marker Ki-67. Parity reduced the risk of carcinoma in unirradiated and pre-pubertally irradiated rats but not post-pubertally irradiated rats. Although radiation exposure increased serum progesterone level, parity after pre-pubertal exposure significantly decreased the elevated progesterone to a normal level, reflecting a protective effect. Moreover, parity significantly decreased the proportion of hormone receptor-positive carcinomas after pre-pubertal exposure. Parity was also related to the observed positive association between progesterone receptor and Ki-67 indices in cancer tissue, implying progesterone receptor-dependent cell proliferation. Thus, parity protects against radiation-induced rat mammary carcinogenesis depending on the age at exposure; the mechanisms may involve changes in hormone levels and cancer tissue.
Collapse
Affiliation(s)
- 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.
- Department of Radiological Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, 116-8551, 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
| | - 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
- Department of Radiological Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, 116-8551, Japan
| | - Benjamin J Blyth
- 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
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Toshiaki Kokubo
- Department of Engineering and Safety, NIRS, QST, Chiba, 263-8555, 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
| | - Kaye Showler
- 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
- Department of Radiological Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, 116-8551, Japan
- Department of Radiology, The Jikei University Hospital, Tokyo, 105-8471, Japan
| | - Ayaka Hosoki
- 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
- Healios K.K. Kobe Research Institute, Kobe, 650-0047, 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
- Department of Radiological Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, 116-8551, 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
| | - 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
| | - Masahiro Fukushi
- Department of Radiological Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, 116-8551, Japan
| | - Yoshiya Shimada
- Department of Radiological Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, 116-8551, Japan.
- Executive Director, QST, Chiba, 263-8555, Japan.
| |
Collapse
|
23
|
McGowan EM, Lin Y, Hatoum D. Good Guy or Bad Guy? The Duality of Wild-Type p53 in Hormone-Dependent Breast Cancer Origin, Treatment, and Recurrence. Cancers (Basel) 2018; 10:cancers10060172. [PMID: 29857525 PMCID: PMC6025368 DOI: 10.3390/cancers10060172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 05/26/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022] Open
Abstract
"Lactation is at one point perilously near becoming a cancerous process if it is at all arrested", Beatson, 1896. Most breast cancers arise from the milk-producing cells that are characterized by aberrant cellular, molecular, and epigenetic translation. By understanding the underlying molecular disruptions leading to the origin of cancer, we might be able to design novel strategies for more efficacious treatments or, ambitiously, divert the cancerous process. It is an established reality that full-term pregnancy in a young woman provides a lifetime reduction in breast cancer risk, whereas delay in full-term pregnancy increases short-term breast cancer risk and the probability of latent breast cancer development. Hormonal activation of the p53 protein (encode by the TP53 gene) in the mammary gland at a critical time in pregnancy has been identified as one of the most important determinants of whether the mammary gland develops latent breast cancer. This review discusses what is known about the protective influence of female hormones in young parous women, with a specific focus on the opportune role of wild-type p53 reprogramming in mammary cell differentiation. The importance of p53 as a protector or perpetrator in hormone-dependent breast cancer, resistance to treatment, and recurrence is also explored.
Collapse
Affiliation(s)
- Eileen M McGowan
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China.
- School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia.
| | - Yiguang Lin
- School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia.
| | - Diana Hatoum
- School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia.
| |
Collapse
|
24
|
Mansoori Y, Tabei MB, Askari A, Izadi P, Daraei A, Naghizadeh MM, Zendehbad Z, Bastami M, Nariman-Saleh-Fam Z, Mansoori H, Tavakkoly-Bazzaz J. A link between expression level of long-non-coding RNA ZFAS1 in breast tissue of healthy women and obesity. Int J Biol Markers 2018; 33:500-506. [PMID: 29690801 DOI: 10.1177/1724600818762258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Epidemiological and experimental literature indicates that the risk of breast cancer incidence is strongly linked to hormone-dependent factors, including reproductive history and obesity. However, the molecular mechanisms underlying the association between these factors and breast cancer risk are poorly understood. The aim of this study, therefore, was to determine whether obesity and reproductive history are associated with expression levels of two breast cancer-related long non-coding RNAs (lncRNAs), namely ZFAS1 and SRA1 in cancer-free breast tissues of women. METHODS In the current research, 145 healthy women were recruited, and the quantitative expression levels of the two lncRNAs were determined through qPCR assay after gathering the mammoplasty breast tissue samples. RESULTS It was found that women with body mass index (BMI)≥30 kg/m2 and BMI 25-29 kg/m2 show a low expression of ZFAS1 compared to the BMI<25 kg/m2 (P=0.031 and P=0.027, respectively). Then, the correlation analysis disclosed a negative correlation of ZFAS1 low expression with increasing BMI (r=-0.194, P=0.019). Interestingly, this analysis demonstrated a negative correlation between low expression of the ZFAS1 and high BMI in women with menarche age below 14 (r=-221; P=0.028). Lastly, it was also revealed that there was a negative association of the low expression level of ZFAS1 with increasing BMI in women through regression models (B=-0.048, P=0.019). CONCLUSIONS These findings suggest interesting clues about the links between high BMI and the expression levels of ZFAS1 in non-diseased breasts that may help us better understand the underlying mechanisms through which obesity contributes to breast carcinogenesis. However, such results need more validations in future research.
Collapse
Affiliation(s)
- Yaser Mansoori
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mohammad Bagher Tabei
- Department of Medical Genetics, School of Medical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Askari
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.,Bone and Joint Reconstraction Research Center, Shafa Orthopedic Haspital, Iran University of Medical Sciences, Tehran, IR Iran
| | - Pantea Izadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdolreza Daraei
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | | | - Zahra Zendehbad
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Bastami
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ziba Nariman-Saleh-Fam
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hosein Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.,Department of Medical Genetics, School of Medical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Javad Tavakkoly-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Richard JLC, Eichhorn PJA. Deciphering the roles of lncRNAs in breast development and disease. Oncotarget 2018; 9:20179-20212. [PMID: 29732012 PMCID: PMC5929455 DOI: 10.18632/oncotarget.24591] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 02/21/2018] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the second leading cause of cancer related deaths in women. It is therefore important to understand the mechanisms underlying breast cancer development as well as raises the need for enhanced, non-invasive strategies for novel prognostic and diagnostic methods. The emergence of long non-coding RNAs (lncRNAs) as potential key players in neoplastic disease has received considerable attention over the past few years. This relatively new class of molecular regulators has been shown from ongoing research to act as critical players for key biological processes. Deregulated expression levels of lncRNAs have been observed in a number of cancers including breast cancer. Furthermore, lncRNAs have been linked to breast cancer initiation, progression, metastases and to limit sensitivity to certain targeted therapeutics. In this review we provide an update on the lncRNAs associated with breast cancer and mammary gland development and illustrate the versatility of such lncRNAs in gene control, differentiation and development both in normal physiological conditions and in diseased states. We also highlight the therapeutic and diagnostic potential of lncRNAs in cancer.
Collapse
Affiliation(s)
- John Lalith Charles Richard
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
- Current Address: Genome Institute of Singapore, Agency for Science Technology and Research, 138672, Singapore
| | - Pieter Johan Adam Eichhorn
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
- School of Pharmacy, Curtin University, Perth, 6845, Australia
| |
Collapse
|
26
|
Bracamontes CG, Lopez-Valdez R, Subramani R, Arumugam A, Nandy S, Rajamanickam V, Ravichandran V, Lakshmanaswamy R. The serum protein profile of early parity which induces protection against breast cancer. Oncotarget 2018; 7:82538-82553. [PMID: 27769065 PMCID: PMC5347712 DOI: 10.18632/oncotarget.12757] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/03/2016] [Indexed: 12/14/2022] Open
Abstract
Early parity reduces the risk of breast cancer in women while nulliparity and late parity increase the risk of breast cancer. In order to translate this protection to women where early pregnancy is not feasible, much work has focused on understanding how parity confers protection against breast cancer, the molecular mechanisms by which this occurs is still not well understood. Healthy parous and nulliparous women were recruited for this study. We assessed serum protein profiles of early parous, late parous, and nulliparous women using the Phospho Explorer antibody array. Significantly altered proteins identified were validated by Western blot analysis. In silico analysis was performed with the data obtained. Our findings indicate increased phosphorylation levels of CDK1, AKT1 and Epo-R increased cell cycle and cell proliferation in late/nulliparous women. Increased levels of LIMK1, paxillin, caveolin-1, and tyrosine hydroxylase in late/nulliparous women demonstrate enhanced cell stress while decreased activity of p-p53 and pRAD51 in late/nulliparous women indicates decreased apoptosis and increased genomic instability. Further, increased levels of pFAK, pCD3zeta, pSTAT5B, MAP3K8 in early parous women favor enhanced innate/adaptive immunity. Overall, we have identified a unique protein signature that is responsible for the decreased risk of breast cancer and these proteins can also serve as biomarkers to predict the risk of breast cancer.
Collapse
Affiliation(s)
- Christina Gutierrez Bracamontes
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, TX 79905, USA
| | - Rebecca Lopez-Valdez
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, TX 79905, USA
| | - Ramadevi Subramani
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, TX 79905, USA
| | - Arunkumar Arumugam
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, TX 79905, USA
| | - Sushmita Nandy
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, TX 79905, USA
| | - Venkatesh Rajamanickam
- Division of Genetic Epidemiology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Vignesh Ravichandran
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | - Rajkumar Lakshmanaswamy
- Center of Emphasis in Cancer Research, Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, TX 79905, USA.,Texas Tech University Health Sciences Center El Paso-Graduate School of Biomedical Sciences, El Paso, TX 79905, USA
| |
Collapse
|
27
|
Subramani R, Lakshmanaswamy R. Pregnancy and Breast Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 151:81-111. [PMID: 29096898 DOI: 10.1016/bs.pmbts.2017.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Breast cancer is the most commonly diagnosed type of cancer among women worldwide. The majority of breast cancers are sporadic and the etiology is not well understood. Several factors have been attributed to altering the risk of breast cancer. A full-term pregnancy is a crucial factor in altering the risk. Early full-term pregnancy has been shown to reduce the lifetime risk of breast cancer, while a later first full-term pregnancy increases breast cancer risk. Epidemiological and experimental data demonstrate that spontaneous or induced abortions do not significantly alter the risk of breast cancer. In this study, we briefly discuss the different types and stages of breast cancer, various risk factors, and potential mechanisms involved in early full-term pregnancy-induced protection against breast cancer. Understanding how early full-term pregnancy induces protection against breast cancer will help design innovative preventive and therapeutic strategies. This understanding can also help in the development of molecular biomarkers that can be of tremendous help in predicting the risk of breast cancer in the general population.
Collapse
Affiliation(s)
- Ramadevi Subramani
- Center of Emphasis in Cancer Research, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Rajkumar Lakshmanaswamy
- Center of Emphasis in Cancer Research, Paul L. Foster School of Medicine, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States.
| |
Collapse
|
28
|
Rao CV. Protective Effects of Human Chorionic Gonadotropin Against Breast Cancer: How Can We Use This Information to Prevent/Treat the Disease? Reprod Sci 2016; 24:1102-1110. [DOI: 10.1177/1933719116676396] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- C. V. Rao
- Department of Cellular Biology and Pharmacology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Department of Molecular and Human Genetics, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Department of Obstetrics and Gynecology, Reproduction and Development Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
29
|
Katz TA. Potential Mechanisms underlying the Protective Effect of Pregnancy against Breast Cancer: A Focus on the IGF Pathway. Front Oncol 2016; 6:228. [PMID: 27833901 PMCID: PMC5080290 DOI: 10.3389/fonc.2016.00228] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 10/11/2016] [Indexed: 01/21/2023] Open
Abstract
A first full-term birth at an early age protects women against breast cancer by reducing lifetime risk by up to 50%. The underlying mechanism resulting in this protective effect remains unclear, but many avenues have been investigated, including lobular differentiation, cell fate, and stromal composition. A single pregnancy at an early age protects women for 30-40 years, and this long-term protection is likely regulated by a relatively stable yet still modifiable method, such as epigenetic reprograming. Long-lasting epigenetic modifications have been shown to be induced by pregnancy and to target the IGF pathway. Understanding how an early first full-term pregnancy protects against breast cancer and the role of epigenetic reprograming of the IGF system may aid in developing new preventative strategies for young healthy women in the future.
Collapse
Affiliation(s)
- Tiffany A Katz
- Center for Precision Environmental Health, Baylor College of Medicine , Houston, TX , USA
| |
Collapse
|
30
|
Russo J. Reproductive history and breast cancer prevention. Horm Mol Biol Clin Investig 2016; 27:3-10. [PMID: 27518906 DOI: 10.1515/hmbci-2016-0033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 07/19/2016] [Indexed: 12/11/2022]
Abstract
The hormonal milieu of an early full-term pregnancy induces lobular development, completing the cycle of differentiation of the breast. This process induces a specific genomic signature in the mammary gland that is represented by the stem cell containing a heterochomatin condensed nucleus (HTN). Even though differentiation significantly reduces cell proliferation in the mammary gland, the mammary epithelium remains capable of responding with proliferation to given stimuli, such as a new pregnancy. The stem cell HTN is able to metabolize the carcinogen and repair the induced DNA damage more efficiently than the stem cell containing an euchromatinic structure (EUN), as it has been demonstrated in the rodent experimental system. The basic biological concept is that pregnancy shifts the stem cell EUN to the stem cell HTN that is refractory to carcinogenesis. Data generated by the use of cDNA micro array techniques have allowed to demonstrate that while lobular development regressed after pregnancy and lactation, programmed cell death genes, DNA repair genes, chromatin remodeling, transcription factors and immune-surveillance gene transcripts all of these genes are upregulated and are part of the genomic signature of pregnancy that is associated with the preventive effect of this physiological process.
Collapse
|
31
|
Alvarenga EM, Rodrigues VL, Moraes AS, Naves LS, Mondin M, Felisbino MB, Mello MLS. Histone epigenetic marks in heterochromatin and euchromatin of the Chagas' disease vector, Triatoma infestans. Acta Histochem 2016; 118:401-12. [PMID: 27079857 DOI: 10.1016/j.acthis.2016.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 03/31/2016] [Accepted: 04/05/2016] [Indexed: 01/23/2023]
Abstract
Triatoma infestans, a vector of Chagas' disease, shows several particular cell biology characteristics, including the presence of conspicuous heterochromatic bodies (chromocenters) where DNA methylation has not been previously detected. Whether histone modifications contribute to the condensed state of these bodies has not yet been studied. Here, we investigated epigenetic modifications of histones H3 and H4 and presence of the non-histone heterochromatin protein (HP1-α) in the chromocenters and euchromatin of T. infestans cell nuclei, using immunocytochemistry. The effect of different concentrations of the histone deacetylase inhibitors valproic acid (VPA) and sodium butyrate (NaBt) on chromocenter condensation was visually examined; in VPA-treated specimens, this effect was also analyzed by image analysis. Trimethylated H3K9 signals, which were revealed in chromocenter and non-chromocenter areas, were strongest in chromocenters, whereas selected acetylated histone marks and mono- and dimethylated H3K9 and H4K20 signals were detected only in euchromatin. Weak trimethylated H4K20 signals and variable distribution of HP1-α were detected in chromocenters of part of the cellular population analyzed. Although specific VPA and NaBt treatment conditions affected the heterochromatin condensation pattern, they did not induce a decrease in survival and molting rates of the T. infestans nymphs. The VPA-induced chromatin remodeling was not accompanied by induction of H3K9 acetylation in chromocenters. Present findings regarding histone modifications and effects following VPA or NaBt treatments did not yet solve the question of which factors are responsible for maintenance of the condensed state of chromocenters in T. infestans. A possibility requiring further investigation remains on histone methylation marks and/or non-histone proteins.
Collapse
|
32
|
Felisbino MB, Alves da Costa T, Gatti MSV, Mello MLS. Differential Response of Human Hepatocyte Chromatin to HDAC Inhibitors as a Function of Microenvironmental Glucose Level. J Cell Physiol 2016; 231:2257-65. [DOI: 10.1002/jcp.25343] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 02/12/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Marina Barreto Felisbino
- Department of Structural and Functional Biology, Institute of Biology; University of Campinas (Unicamp); Campinas São Paulo Brazil
| | - Thiago Alves da Costa
- Department of Structural and Functional Biology, Institute of Biology; University of Campinas (Unicamp); Campinas São Paulo Brazil
| | - Maria Silvia Viccari Gatti
- Department of Genetics, Evolution and Bioagents, Institute of Biology; University of Campinas (Unicamp); Campinas São Paulo Brazil
| | - Maria Luiza Silveira Mello
- Department of Structural and Functional Biology, Institute of Biology; University of Campinas (Unicamp); Campinas São Paulo Brazil
| |
Collapse
|
33
|
Katz TA, Liao SG, Palmieri VJ, Dearth RK, Pathiraja TN, Huo Z, Shaw P, Small S, Davidson NE, Peters DG, Tseng GC, Oesterreich S, Lee AV. Targeted DNA Methylation Screen in the Mouse Mammary Genome Reveals a Parity-Induced Hypermethylation of Igf1r That Persists Long after Parturition. Cancer Prev Res (Phila) 2015; 8:1000-9. [PMID: 26290394 PMCID: PMC4677681 DOI: 10.1158/1940-6207.capr-15-0178] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 08/05/2015] [Indexed: 02/06/2023]
Abstract
The most effective natural prevention against breast cancer is an early first full-term pregnancy. Understanding how the protective effect is elicited will inform the development of new prevention strategies. To better understand the role of epigenetics in long-term protection, we investigated parity-induced DNA methylation in the mammary gland. FVB mice were bred or remained nulliparous and mammary glands harvested immediately after involution (early) or 6.5 months following involution (late), allowing identification of both transient and persistent changes. Targeted DNA methylation (109 Mb of Ensemble regulatory features) analysis was performed using the SureSelectXT Mouse Methyl-seq assay and massively parallel sequencing. Two hundred sixty-nine genes were hypermethylated and 128 hypomethylated persistently at both the early and late time points. Pathway analysis of the persistently differentially methylated genes revealed Igf1r to be central to one of the top identified signaling networks, and Igf1r itself was one of the most significantly hypermethylated genes. Hypermethylation of Igf1r in the parous mammary gland was associated with a reduction of Igf1r mRNA expression. These data suggest that the IGF pathway is regulated at multiple levels during pregnancy and that its modification might be critical in the protective role of pregnancy. This supports the approach of lowering IGF action for prevention of breast cancer, a concept that is currently being tested clinically.
Collapse
Affiliation(s)
- Tiffany A Katz
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Serena G Liao
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vincent J Palmieri
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Robert K Dearth
- Department of Biology, University of Texas-Rio Grande Valley, Edinburg, Texas
| | - Thushangi N Pathiraja
- Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore
| | - Zhiguang Huo
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Patricia Shaw
- Department of Obstetrics. Gynecology, and Reproductive Sciences, University of Pittsburgh, Pennsylvania
| | - Sarah Small
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Nancy E Davidson
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - David G Peters
- Department of Obstetrics. Gynecology, and Reproductive Sciences, University of Pittsburgh, Pennsylvania
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Steffi Oesterreich
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.
| | - Adrian V Lee
- Department of Pharmacology and Chemical Biology, Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania.
| |
Collapse
|
34
|
Aflatoxins ingestion and canine mammary tumors: There is an association? Food Chem Toxicol 2015; 84:74-8. [DOI: 10.1016/j.fct.2015.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 07/30/2015] [Accepted: 08/03/2015] [Indexed: 01/29/2023]
|
35
|
Wang H, Sun Q, Zhao W, Qi L, Gu Y, Li P, Zhang M, Li Y, Liu SL, Guo Z. Individual-level analysis of differential expression of genes and pathways for personalized medicine. ACTA ACUST UNITED AC 2014; 31:62-8. [PMID: 25165092 DOI: 10.1093/bioinformatics/btu522] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
MOTIVATION The differential expression analysis focusing on inter-group comparison can capture only differentially expressed genes (DE genes) at the population level, which may mask the heterogeneity of differential expression in individuals. Thus, to provide patient-specific information for personalized medicine, it is necessary to conduct differential expression analysis at the individual level. RESULTS We proposed a method to detect DE genes in individual disease samples by using the disrupted ordering in individual disease samples. In both simulated data and real paired cancer-normal sample data, this method showed excellent performance. It was found to be insensitive to experimental batch effects and data normalization. The landscape of stable gene pairs in a particular type of normal tissue could be predetermined using previously accumulated data, based on which dysregulated genes and pathways for any disease sample can be readily detected. The usefulness of the RankComp method in clinical settings was exemplified by the identification and application of prognostic markers for lung cancer. AVAILABILITY AND IMPLEMENTATION RankComp is implemented in R script that is freely available from Supplementary Materials.
Collapse
Affiliation(s)
- Hongwei Wang
- College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China
| | - Qiang Sun
- College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China
| | - Wenyuan Zhao
- College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China
| | - Lishuang Qi
- College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China
| | - Yunyan Gu
- College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China
| | - Pengfei Li
- College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China
| | - Mengmeng Zhang
- College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China
| | - Yang Li
- College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China
| | - Shu-Lin Liu
- College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China
| | - Zheng Guo
- College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China College of Bioinformatics Science and Technology, Genomics Research Center, Harbin Medical University, Harbin 150086, China, Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4N1, Canada and Bioinformatics Department, Basic Medical College, Fujian Medical University, Fuzhou 350004, China
| |
Collapse
|
36
|
Rotunno M, Sun X, Figueroa J, Sherman ME, Garcia-Closas M, Meltzer P, Williams T, Schneider SS, Jerry DJ, Yang XR, Troester MA. Parity-related molecular signatures and breast cancer subtypes by estrogen receptor status. Breast Cancer Res 2014; 16:R74. [PMID: 25005139 PMCID: PMC4227137 DOI: 10.1186/bcr3689] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 06/25/2014] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Relationships of parity with breast cancer risk are complex. Parity is associated with decreased risk of postmenopausal hormone receptor-positive breast tumors, but may increase risk for basal-like breast cancers and early-onset tumors. Characterizing parity-related gene expression patterns in normal breast and breast tumor tissues may improve understanding of the biological mechanisms underlying this complex pattern of risk. METHODS We developed a parity signature by analyzing microRNA microarray data from 130 reduction mammoplasty (RM) patients (54 nulliparous and 76 parous). This parity signature, together with published parity signatures, was evaluated in gene expression data from 150 paired tumors and adjacent benign breast tissues from the Polish Breast Cancer Study, both overall and by tumor estrogen receptor (ER) status. RESULTS We identified 251 genes significantly upregulated by parity status in RM patients (parous versus nulliparous; false discovery rate = 0.008), including genes in immune, inflammation and wound response pathways. This parity signature was significantly enriched in normal and tumor tissues of parous breast cancer patients, specifically in ER-positive tumors. CONCLUSIONS Our data corroborate epidemiologic data, suggesting that the etiology and pathogenesis of breast cancers vary by ER status, which may have implications for developing prevention strategies for these tumors.
Collapse
|
37
|
Russo J, Santucci-Pereira J, Russo IH. The genomic signature of breast cancer prevention. Genes (Basel) 2014; 5:65-83. [PMID: 24705287 PMCID: PMC3978512 DOI: 10.3390/genes5010065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/31/2014] [Accepted: 02/08/2014] [Indexed: 11/16/2022] Open
Abstract
The breast of parous postmenopausal women exhibits a specific signature that has been induced by a full term pregnancy. This signature is centered in chromatin remodeling and the epigenetic changes induced by methylation of specific genes which are important regulatory pathways induced by pregnancy. Through the analysis of the genes found to be differentially methylated between women of varying parity, multiple positions at which beta-catenin production and use is inhibited were recognized. The biological importance of the pathways identified in this specific population cannot be sufficiently emphasized because they could represent a safeguard mechanism mediating the protection of the breast conferred by full term pregnancy.
Collapse
Affiliation(s)
- Jose Russo
- The Irma H. Russo MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, 333 Cottman Avenue, Philadelphia, PA 19111, USA.
| | - Julia Santucci-Pereira
- The Irma H. Russo MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, 333 Cottman Avenue, 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 Avenue, Philadelphia, PA 19111, USA
| |
Collapse
|
38
|
Barton M, Santucci-Pereira J, Russo J. Molecular pathways involved in pregnancy-induced prevention against breast cancer. Front Endocrinol (Lausanne) 2014; 5:213. [PMID: 25540638 PMCID: PMC4261797 DOI: 10.3389/fendo.2014.00213] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 11/25/2014] [Indexed: 12/18/2022] Open
Abstract
Pregnancy produces a protective effect against breast cancer in women who had their first full term pregnancy (FTP) in their middle twenties. The later in life the first delivery occurs, the higher the risk of breast cancer development. Also, transiently during the postpartum period, the risk of developing breast cancer increases. This transient increased risk is taken over by a long-lasting protective period. The genomic profile of parous women has shown pregnancy induces a long-lasting "genomic signature" that explains the preventive effect on breast cancer. This signature reveals that chromatin remodeling is the driver of the differentiation process conferred by FTP. The chromatin remodeling process may be the ultimate step mediating the protection of the breast against developing breast cancer in post-menopausal years.
Collapse
Affiliation(s)
- Maria Barton
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA
| | - Julia Santucci-Pereira
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA
| | - Jose Russo
- The Irma H. Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA
- *Correspondence: Jose Russo, The Irma H Russo, MD Breast Cancer Research Laboratory, Fox Chase Cancer Center, Temple University Health System, 333 Cottman Avenue, Room P2037, Philadelphia, PA 19111, USA e-mail:
| |
Collapse
|
39
|
Abstract
Chromatin packaging plays a significant role in regulating gene transcription. Study of the higher-order packing states of chromatin by image analysis at the light microscope level, especially when validated by methods of molecular biology, immunochemistry, and/or immunocytochemistry, enabled the detection of changes involved in the processes associated with or preceding alterations in transcriptional activities. Here, we recommend and describe the use of relatively simple methods for staining and detecting chromatin remodelling by image analysis.
Collapse
Affiliation(s)
- Benedicto de Campos Vidal
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | | | | |
Collapse
|
40
|
Molecular profiling of human mammary gland links breast cancer risk to a p27(+) cell population with progenitor characteristics. Cell Stem Cell 2013; 13:117-30. [PMID: 23770079 DOI: 10.1016/j.stem.2013.05.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 02/11/2013] [Accepted: 05/09/2013] [Indexed: 12/20/2022]
Abstract
Early full-term pregnancy is one of the most effective natural protections against breast cancer. To investigate this effect, we have characterized the global gene expression and epigenetic profiles of multiple cell types from normal breast tissue of nulliparous and parous women and carriers of BRCA1 or BRCA2 mutations. We found significant differences in CD44(+) progenitor cells, where the levels of many stem cell-related genes and pathways, including the cell-cycle regulator p27, are lower in parous women without BRCA1/BRCA2 mutations. We also noted a significant reduction in the frequency of CD44(+)p27(+) cells in parous women and showed, using explant cultures, that parity-related signaling pathways play a role in regulating the number of p27(+) cells and their proliferation. Our results suggest that pathways controlling p27(+) mammary epithelial cells and the numbers of these cells relate to breast cancer risk and can be explored for cancer risk assessment and prevention.
Collapse
|