1
|
Rodrigues I, Fernandes R, Ferreira A, Pereira D, Fernandes R, Soares R, Luís C. Is Progesterone Receptor a Neglected Feature in Breast Cancer? A Retrospective Study Analysing the Clinicopathological Characteristics of Breast Cancer Based on Progesterone Receptor Status. Clin Breast Cancer 2025; 25:e331-e340. [PMID: 39706710 DOI: 10.1016/j.clbc.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/23/2024]
Abstract
PURPOSE The aim of this study is to analyse PR independently and its relationship with demographic and clinicopathological information. INTRODUCTION Steroid hormones, particularly estrogen and progesterone, play a crucial role in breast cancer (BC) etiology. Research attention has focused mainly on estrogen while the progesterone impact on breast cancer has yet to be fully uncover. Hormone receptors, including those for estrogen and progesterone, are crucial in BC molecular classification, shaping prognosis and treatment strategies. Beyond its metabolic effects, progesterone and its receptor (PR) have significant clinical implications, impacting clinical outcomes. MATERIALS AND METHODS The study comprised 2223 women who were diagnosed with BC at the Comprehensive Cancer Centre in Portugal (IPO-Porto) between 2012 and 2016. Variables, including age at diagnosis, body mass index (BMI), laterality, topographic localization, histological type, differentiation grade, tumor stage, estrogen receptor (ER) and Human Epidermal growth factor Receptor 2 (HER2) expression, were stratified according to the expression of Progesterone Receptor. Statistical analysis included Pearson's Chi-squared test, binary and multinomial regression, and Cox proportional hazard model. Statistical significance was set for P < .05. RESULTS The results reveal a statistical association between PR and BMI, histological type, differentiation grade, tumour stage, ER and HER2. Progesterone receptor negativity is associated with adverse clinical outcomes, including advanced tumor stages, and diminished overall survival. CONCLUSION Further research is needed to elucidate the precise contributions of progesterone to breast cancer progression and to optimize therapeutic approaches for improved patient outcomes.
Collapse
Affiliation(s)
- Ilda Rodrigues
- Biochemistry Unit, Department of Biomedicine, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal; i3S ‑ Instituto de Inovação e Investigação em Saúde, University of Porto, Porto, Portugal
| | - Rute Fernandes
- Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Ana Ferreira
- Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal; ICBAS - Abel Salazar Institute of Biomedical Sciences, Porto, Portugal
| | - Deolinda Pereira
- Portuguese Oncology Institute of Porto (IPO-Porto), Porto, Portugal
| | - Rúben Fernandes
- Faculty of Health Sciences, University Fernando Pessoa, Fernando Pessoa Hospital-School (FCS/HEFP/UFP), Porto, Portugal
| | - Raquel Soares
- Biochemistry Unit, Department of Biomedicine, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal; i3S ‑ Instituto de Inovação e Investigação em Saúde, University of Porto, Porto, Portugal
| | - Carla Luís
- Biochemistry Unit, Department of Biomedicine, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal; i3S ‑ Instituto de Inovação e Investigação em Saúde, University of Porto, Porto, Portugal; Faculty of Health Sciences, University Fernando Pessoa, Fernando Pessoa Hospital-School (FCS/HEFP/UFP), Porto, Portugal.
| |
Collapse
|
2
|
Mogus JP, Vandenberg LN. Exposure to ethinyl estradiol during lactation reduces maternal mammary gland complexity and restricts pup growth in mice. Reprod Toxicol 2025; 135:108931. [PMID: 40324638 DOI: 10.1016/j.reprotox.2025.108931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/06/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
The mammary gland undergoes dramatic changes during pregnancy and lactation, remodeling a relatively simple ductal structure into an extensive network of milk secreting alveoli. Estradiol, through activation of estrogen receptor (ER), plays an essential role in the early expansion of the mammary epithelium during pregnancy, but has little role during lactation when circulating levels are low. Yet, ER is present in the epithelium during lactation and exposure to estrogenic endocrine disrupting chemicals (EDCs) could disrupt the lactational capability of the mammary gland. To understand the effects of ER agonists during lactation, we exposed CD-1 mice to ethinyl estradiol [20 μg/kg] from lactation day 1 (LD1) until weaning at LD21. To assess changes in lactational output, pup weight gain was assessed throughout the lactation period. Both maternal weight gain and maternal behavior were observed as well. Maternal mammary glands were collected and assessed for changes in histomorphology and molecular indicators of differentiation and cell death. We observed that EE2 exposure restricted weight growth in dams, increased time nursing on the nest and reduced other maternal activities. EE2 exposed pups had restricted growth and development. At weaning, EE2 exposed dams had less epithelial complexity than VEH controls despite no changes in alveolar structure. Molecular analysis of the mammary gland at weaning indicated that EE2 reduced epithelial pSTAT3 expression, an early indicator of cell degradation, without altering differentiation markers or apoptosis. These findings indicate that EE2 exposure reduces epithelial expansion in lactation prior to weaning, resulting in restricted growth and development in pups.
Collapse
Affiliation(s)
- Joshua P Mogus
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Laura N Vandenberg
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
3
|
Hamilton AM, Srivastava VK, Hiney JK, Dees WL, Dearth RK. Manganese-induced Precocious Puberty Alters Mammary Epithelial Cell Proliferation in Female Rats. Endocrinology 2025; 166:bqaf052. [PMID: 40105700 PMCID: PMC12006721 DOI: 10.1210/endocr/bqaf052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/02/2025] [Accepted: 03/17/2025] [Indexed: 03/20/2025]
Abstract
Precocious puberty (PP) is an established breast cancer risk factor. In the normal mammary gland, hormone receptor-positive (HR+) cells rarely proliferate. In breast cancer, proliferating epithelial cells are often HR+. It is not known if PP can modify this population of proliferating HR+ cells. Previously, we established a manganese-induced precocious puberty (MnPP) model to study the effects of PP on mammary gland development in female rats. Here, we characterized the distribution of HR+ proliferating mammary epithelial cells in prepubertal and adult rodents, in association with precocious puberty. Female rats were exposed daily to 10 mg/kg manganese chloride or saline (control) from postnatal day (PND) 12 to PND 30. Mammary glands were collected on PNDs 30 and 120, processed for western blot analysis and double immunofluorescence staining for proliferating cell nuclear antigen and progesterone receptor or estrogen receptor. MnPP increased the percentage of HR+ mammary epithelial cells coexpressing proliferating cell nuclear antigen relative to normally developed controls at PND 30. This correlated with increased expression of estrogen receptor-regulated proteins in MnPP mammary glands relative to controls at PND 30, including FOXA1, AREG, and c-Myc. Conversely, at PND 120 relative to PND 30, proliferating HR+ cells remained chronically elevated in MnPP mammary glands at PND 120, which coincided with decreased expression of cell-cycle regulator, p27, and increased expression of progesterone receptor-regulated markers, EREG and sp1. Collectively, these results suggest early puberty alters steroidal regulation of classic proliferative mechanisms in the prepubertal gland with increased prevalence of high-risk proliferating HR+ cells.
Collapse
MESH Headings
- Animals
- Female
- Cell Proliferation/drug effects
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/pathology
- Rats
- Puberty, Precocious/chemically induced
- Puberty, Precocious/metabolism
- Puberty, Precocious/pathology
- Rats, Sprague-Dawley
- Receptors, Progesterone/metabolism
- Proliferating Cell Nuclear Antigen/metabolism
- Receptors, Estrogen/metabolism
- Sexual Maturation/drug effects
- Manganese
- Chlorides
- Manganese Compounds
Collapse
Affiliation(s)
- Alina M Hamilton
- School of Integrative Biological and Chemical Sciences, College of Sciences, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27606, USA (Current)
| | - Vinod K Srivastava
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4458, USA
| | - Jill K Hiney
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4458, USA
| | - William L Dees
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4458, USA
| | - Robert K Dearth
- School of Integrative Biological and Chemical Sciences, College of Sciences, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
4
|
Guo Z, Zhu Z, Lin X, Zhang KM, Wu Q, Wang S, Luo M, Lin X, Wang L, Zhou J. Conception and Concern: A Review of Breast Cancer Risk in Assisted Reproductive Technology. Int J Biol Sci 2025; 21:2647-2671. [PMID: 40303287 PMCID: PMC12035894 DOI: 10.7150/ijbs.105357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/25/2025] [Indexed: 05/02/2025] Open
Abstract
This review examines the complex relationship between assisted reproductive technology (ART) and the potential risk of breast cancer. Through a thorough analysis, we explore various aspects of this association, considering both the theoretical and mechanistic overlap between ART and breast cancer, as well as the growing body of empirical research aimed at elucidating this relationship. Theoretical analysis suggests that ART exposure inevitably increases levels of reproductive hormones over a relatively short period, potentially elevating susceptibility to breast cancer. However, current clinical evidence does not strongly support this hypothesis, and no direct correlation between ART and breast cancer development has been established. Our study lays the groundwork for informed discussion and offers recommendations for further research in this area of women's health, based on a comprehensive review of both theoretical and clinical research. The findings provide valuable information to guide both specialists and patients in decision-making regarding ART treatment. As we navigate the complexities surrounding conception, this manuscript serves as an essential resource for understanding and addressing the potential risks of breast cancer in the context of ART.
Collapse
Affiliation(s)
- Zijie Guo
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Ziyu Zhu
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Xixi Lin
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | | | - Qingliang Wu
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
- The Ninth People's Hospital of Hangzhou, Hangzhou, Zhejiang, 310014, China
| | - Shenkangle Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Mingpeng Luo
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310014, China
| | - Xiaona Lin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
| | - Linbo Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Jichun Zhou
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| |
Collapse
|
5
|
Van Keymeulen A. Mechanisms of Regulation of Cell Fate in Breast Development and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:167-184. [PMID: 39821026 DOI: 10.1007/978-3-031-70875-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
This chapter focuses on the mechanisms of regulation of cell fate in breast development, occurring mainly after birth, as well as in breast cancer. First, we will review how the microenvironment of the breast, as well as external cues, plays a crucial role in mammary gland cell specification and will describe how it has been shown to reprogram non-mammary cells into mammary epithelial cells. Then we will focus on the transcription factors and master regulators which have been established to be determinant for basal (BC) and luminal cell (LC) identity, and will describe the experiments of ectopic expression or loss of function of these transcription factors which demonstrated that they were crucial for cell fate. We will also discuss how master regulators are involved in the fate choice of LCs between estrogen receptor (ER)-positive cells and ER- cells, which will give rise to alveolar cells upon pregnancy and lactation. We will describe how oncogene expression induces reprogramming and change of fate of mammary epithelial cells before tumor appearance, which could be an essential step in tumorigenesis. Finally, we will describe the involvement of master regulators of mammary epithelial cells in breast cancer.
Collapse
Affiliation(s)
- Alexandra Van Keymeulen
- Laboratory of Stem Cells and Cancer (LSCC), Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
6
|
Ingthorsson S, Traustadottir GA, Gudjonsson T. Breast Morphogenesis: From Normal Development to Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:29-44. [PMID: 39821019 DOI: 10.1007/978-3-031-70875-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The human breast gland is composed of branching epithelial ducts that culminate in milk-producing units known as terminal duct lobular units (TDLUs). The epithelial compartment comprises an inner layer of luminal epithelial cells (LEP) and an outer layer of contractile myoepithelial cells (MEP). Both LEP and MEP arise from a common stem cell population. The epithelial compartment undergoes dynamic branching morphogenesis and remodelling, which expands the surface area for milk production. The epithelial remodelling that starts at the onset of menarche is largely under hormonal control, first and foremost by estrogen and progesterone from ovaries, the production of which is stimulated by pituitary-derived hormones. Menopause leads to a significant decline in estrogen and progesterone levels, resulting in involution and senescence of the breast epithelium. The branching morphogenesis involves developmental events such as epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET). EMT and MET confer plasticity to the epithelial compartment enabling the migration of epithelial cells through the stroma and restoration of the epithelial phenotype. In the normal breast, the stroma, including the basement membrane (BM), collagen-rich extracellular matrix, and various stromal cells, supports the correct histoarchitecture of the glandular tree. However, in cancer, the stroma can acquire tumour-promoting properties and is referred to as the tumour microenvironment. This chapter will explore the developmental processes including branching morphogenesis in the normal breast gland and discuss the lineage relationship between LEPS and MEPs and their interactions with the surrounding stroma in the normal and neoplastic breast gland. Finally, we will review various in vitro and in vivo models employed in mammary gland research.
Collapse
Affiliation(s)
- Saevar Ingthorsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Faculty of Nursing and Midwifery, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Gunnhildur Asta Traustadottir
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Pathology, Landspitali University Hospital, Reykjavik, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
- Department of Laboratory Hematology, Landspitali University Hospital, Reykjavik, Iceland.
| |
Collapse
|
7
|
Iggo R, MacGrogan G. Classification of Breast Cancer Through the Perspective of Cell Identity Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:185-207. [PMID: 39821027 DOI: 10.1007/978-3-031-70875-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
The mammary epithelium has an inner luminal layer that contains estrogen receptor (ER)-positive hormone-sensing cells and ER-negative alveolar/secretory cells, and an outer basal layer that contains myoepithelial/stem cells. Most human tumours resemble either hormone-sensing cells or alveolar/secretory cells. The most widely used molecular classification, the Intrinsic classification, assigns hormone-sensing tumours to Luminal A/B and human epidermal growth factor 2-enriched (HER2E)/molecular apocrine (MA)/luminal androgen receptor (LAR)-positive classes, and alveolar/secretory tumours to the Basal-like class. Molecular classification is most useful when tumours have classic invasive carcinoma of no special type (NST) histology. It is less useful for special histological types of breast cancer, such as metaplastic breast cancer and adenoid cystic cancer, which are better described with standard pathology terms. Compared to mice, humans show a strong bias towards making tumours that resemble mammary hormone-sensing cells. This could be caused by the formation in adolescence of der(1;16), a translocation through the centromeres of chromosomes 1 and 16, which only occurs in humans and could trap the cells in the hormone-sensing state.
Collapse
Affiliation(s)
- Richard Iggo
- INSERM, Bergonie Cancer Institute, University of Bordeaux, Bordeaux, France.
| | - Gaetan MacGrogan
- INSERM, Bergonie Cancer Institute, University of Bordeaux, Bordeaux, France
| |
Collapse
|
8
|
Clarke RB, Sørlie T. A Guide to Breast Cancer Research: An Introduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:1-5. [PMID: 39821017 DOI: 10.1007/978-3-031-70875-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
"A Guide to Breast Cancer Research: From Cells and Molecular Mechanisms to Therapy" is designed as a comprehensive reference for early career investigators and postgraduate students. This book aims to provide a broad overview of contemporary breast cancer research. It covers key areas including development and cancer, metastasis and immunology, subtypes, signalling, therapy, and resistance. This book is organised into seven sections addressing mammary gland development, model systems, cellular origins and heterogeneity, cellular and molecular bases, signalling pathways, metastasis and immunity, and treatment and resistance mechanisms. A few topics such as specific signalling pathways, some emerging therapies, imaging technologies, and AI applications are only briefly mentioned or omitted, reflecting the ever-evolving nature of breast cancer research. This book emphasises the importance of collaboration in advancing cancer research. Initiatives like the Cancer Moonshot and Cancer Grand Challenges advocate for "radical collaboration" of researchers with shared visions and resources. We also note the significance of global efforts in breast cancer research, the need for addressing disparities in care across different regions and for equity in healthcare. Overall, this book showcases milestones and advances in breast cancer research over the past three decades, reflecting significant progress in understanding and treating the disease, which has led to improved patient outcomes.
Collapse
Affiliation(s)
- Robert B Clarke
- Faculty of Biology, Medicine and Health, Division of Cancer Sciences, Manchester Breast Centre, University of Manchester, Manchester, UK
| | - Therese Sørlie
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
9
|
Dawson CA, Milevskiy MJG, Capaldo BD, Yip RKH, Song X, Vaillant F, Prokopuk L, Jackling FC, Smyth GK, Chen Y, Lindeman GJ, Visvader JE. Hormone-responsive progenitors have a unique identity and exhibit high motility during mammary morphogenesis. Cell Rep 2024; 43:115073. [PMID: 39700014 DOI: 10.1016/j.celrep.2024.115073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Hormone-receptor-positive (HR+) luminal cells largely mediate the response to estrogen and progesterone during mammary gland morphogenesis. However, there remains a lack of consensus on the precise nature of the precursor cells that maintain this essential HR+ lineage. Here we refine the identification of HR+ progenitors and demonstrate their unique regenerative capacity compared to mature HR+ cells. HR+ progenitors proliferate but do not expand, suggesting rapid differentiation. Subcellular resolution, 3D intravital microscopy was performed on terminal end buds (TEBs) during puberty to dissect the contribution of each luminal lineage. Surprisingly, HR+ TEB progenitors were highly elongated and motile compared to columnar HR- progenitors and static, conoid HR+ cells within ducts. This dynamic behavior was also observed in response to hormones. Development of an AI model for motility dynamics analysis highlighted stark behavioral changes in HR+ progenitors as they transitioned to mature cells. This work provides valuable insights into how progenitor behavior contributes to mammary morphogenesis.
Collapse
Affiliation(s)
- Caleb A Dawson
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Michael J G Milevskiy
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Bianca D Capaldo
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Raymond K H Yip
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Xiaoyu Song
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - François Vaillant
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Lexie Prokopuk
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Felicity C Jackling
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yunshun Chen
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Geoffrey J Lindeman
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia; Parkville Familial Cancer Centre and Department of Medical Oncology, The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC 3052, Australia
| | - Jane E Visvader
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia.
| |
Collapse
|
10
|
Rojo MD, Bandyopadhyay I, Burke CM, Sturtz AD, Phillips ES, Matherne MG, Embrey SJ, LaRue R, Qiu Y, Schwertfeger KL, Machado HL. C/EBPβ deletion in macrophages impairs mammary gland alveolar budding during the estrous cycle. Life Sci Alliance 2024; 7:e202302516. [PMID: 39025525 PMCID: PMC11258408 DOI: 10.26508/lsa.202302516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024] Open
Abstract
Macrophages have important roles in mammary gland development and tissue homeostasis, but the specific mechanisms that regulate macrophage function need further elucidation. We have identified C/EBPβ as an important transcription factor expressed by multiple macrophage populations in the normal mammary gland. Mammary glands from mice with C/EBPβ-deficient macrophages (Cebpb ΔM) show a significant decrease in alveolar budding during the diestrus stage of the reproductive cycle, whereas branching morphogenesis remains unchanged. Defects in alveolar budding were found to be the result of both systemic hormones and local macrophage-directed signals. RNA sequencing shows significant changes in PR-responsive genes and alterations in the Wnt landscape of mammary epithelial cells of Cebpb ΔM mice, which regulate stem cell expansion during diestrus. Cebpb ΔM macrophages demonstrate a shift from a pro-inflammatory to a tissue-reparative phenotype, and exhibit increased phagocytic capacity as compared to WT. Finally, Cebpb ΔM macrophages down-regulate Notch2 and Notch3, which normally promote stem cell expansion during alveolar budding. These results suggest that C/EBPβ is an important macrophage factor that facilitates macrophage-epithelial crosstalk during a key stage of mammary gland tissue homeostasis.
Collapse
Affiliation(s)
- Michelle D Rojo
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ishitri Bandyopadhyay
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Caitlin M Burke
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Alexa D Sturtz
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Emily S Phillips
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Megan G Matherne
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Samuel J Embrey
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Rebecca LaRue
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, and Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - Yinjie Qiu
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, and Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - Kathryn L Schwertfeger
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, and Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Heather L Machado
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Cancer Center, Louisiana Cancer Research Consortium, New Orleans, LA, USA
| |
Collapse
|
11
|
Davies BP, Crew RC, Cochrane ALK, Davies K, Figueiredo Baptista A, Jeckel S, McCrone IS, Niu Y, Strugnell BW, Waine K, Fowden AL, Bryant CE, Wills JW, Giussani DA, Hughes K. An ovine model for investigation of the microenvironment of the male mammary gland. J Anat 2024; 245:405-419. [PMID: 38735860 PMCID: PMC11306760 DOI: 10.1111/joa.14055] [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: 12/17/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024] Open
Abstract
The specific biology of the male breast remains relatively unexplored in spite of the increasing global prevalence of male breast cancer. Delineation of the microenvironment of the male breast is restricted by the low availability of human samples and a lack of characterisation of appropriate animal models. Unlike the mouse, the male ovine gland persists postnatally. We suggest that the male ovine mammary gland constitutes a promising adjunctive model for the male breast. In this study, we evaluate the male ovine mammary gland microenvironment, comparing intact and neutered males. Assessment of the glandular histo-anatomy highlights the resemblance of the male gland to that of neonatal female sheep and confirms the presence of rudimentary terminal duct lobular units. Irrespective of neutered status, cell proliferation in epithelial and stromal compartments is similarly low in males, and cell proliferation in epithelial cells and in the intralobular stroma is significantly lower than in pubertal female sheep. Between 42% and 72% of the luminal mammary epithelial cells in the male gland express the androgen receptor and expression is significantly reduced by neutering. Luminal epithelial cells within the intact and neutered male gland also express oestrogen receptor alpha, but minimal progesterone receptor expression is observed. The distribution of leukocytes within the ducts and stroma is similar to the mammary gland of female sheep and females of other species. Both macrophages and T lymphocytes are intercalated in the epithelial bilayer and are more abundant in the intralobular stroma than the interlobular stroma, suggesting that they may have a protective immunological function within the vestigial glandular tissue of the male sheep. Mast cells are also observed within the stroma. These cells cluster near the glandular tissue and are frequently located adjacent to blood vessels. The abundance of mast cells is significantly higher in intact males compared to neutered males, suggesting that hormone signalling may impact mast cell recruitment. In this study, we demonstrate the utility of the male ovine mammary gland as a model for furthering our knowledge of postnatal male mammary biology.
Collapse
Affiliation(s)
| | - Rachael C. Crew
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Department of Obstetrics and GynaecologyUniversity of CambridgeCambridgeUK
- School of Human SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Anna L. K. Cochrane
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Katie Davies
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | | | - Sonja Jeckel
- Farm Animal Pathology and DiagnosticsThe Royal Veterinary CollegeHatfieldUK
| | - Ian S. McCrone
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | - Youguo Niu
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | | | - Katie Waine
- Farm Post Mortems LtdDurhamUK
- Present address:
Faculty of Veterinary MedicineUniversity of CalgaryCalgaryAlbertaCanada.
| | - Abigail L. Fowden
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Clare E. Bryant
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | - John W. Wills
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | - Dino A. Giussani
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Katherine Hughes
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
12
|
Ciwinska M, Messal HA, Hristova HR, Lutz C, Bornes L, Chalkiadakis T, Harkes R, Langedijk NSM, Hutten SJ, Menezes RX, Jonkers J, Prekovic S, Simons BD, Scheele CLGJ, van Rheenen J. Mechanisms that clear mutations drive field cancerization in mammary tissue. Nature 2024; 633:198-206. [PMID: 39232148 PMCID: PMC11374684 DOI: 10.1038/s41586-024-07882-3] [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: 12/13/2022] [Accepted: 07/26/2024] [Indexed: 09/06/2024]
Abstract
Oncogenic mutations are abundant in the tissues of healthy individuals, but rarely form tumours1-3. Yet, the underlying protection mechanisms are largely unknown. To resolve these mechanisms in mouse mammary tissue, we use lineage tracing to map the fate of wild-type and Brca1-/-;Trp53-/- cells, and find that both follow a similar pattern of loss and spread within ducts. Clonal analysis reveals that ducts consist of small repetitive units of self-renewing cells that give rise to short-lived descendants. This offers a first layer of protection as any descendants, including oncogenic mutant cells, are constantly lost, thereby limiting the spread of mutations to a single stem cell-descendant unit. Local tissue remodelling during consecutive oestrous cycles leads to the cooperative and stochastic loss and replacement of self-renewing cells. This process provides a second layer of protection, leading to the elimination of most mutant clones while enabling the minority that by chance survive to expand beyond the stem cell-descendant unit. This leads to fields of mutant cells spanning large parts of the epithelial network, predisposing it for transformation. Eventually, clone expansion becomes restrained by the geometry of the ducts, providing a third layer of protection. Together, these mechanisms act to eliminate most cells that acquire somatic mutations at the expense of driving the accelerated expansion of a minority of cells, which can colonize large areas, leading to field cancerization.
Collapse
Affiliation(s)
- Marta Ciwinska
- VIB-KULeuven Centre for Cancer Biology, Department of Oncology, Leuven, Belgium
| | - Hendrik A Messal
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hristina R Hristova
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Catrin Lutz
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Laura Bornes
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Rolf Harkes
- Bioimaging Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nathalia S M Langedijk
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Stefan J Hutten
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Renée X Menezes
- Biostatistics Centre and Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Stefan Prekovic
- Centre for Molecular Medicine, UMC Utrecht, Utrecht, the Netherlands
| | - Benjamin D Simons
- Gurdon Institute, University of Cambridge, Cambridge, UK.
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK.
| | | | - Jacco van Rheenen
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
13
|
Heijmans N, Wiese KE, Jonkers J, van Amerongen R. Transcriptomic Analysis of Pubertal and Adult Virgin Mouse Mammary Epithelial and Stromal Cell Populations. J Mammary Gland Biol Neoplasia 2024; 29:13. [PMID: 38916673 PMCID: PMC11199289 DOI: 10.1007/s10911-024-09565-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/30/2024] [Indexed: 06/26/2024] Open
Abstract
Conflicting data exist as to how mammary epithelial cell proliferation changes during the reproductive cycle. To study the effect of endogenous hormone fluctuations on gene expression in the mouse mammary gland, we performed bulk RNAseq analyses of epithelial and stromal cell populations that were isolated either during puberty or at different stages of the adult virgin estrous cycle. Our data confirm prior findings that proliferative changes do not occur in every mouse in every cycle. We also show that during the estrous cycle the main gene expression changes occur in adipocytes and fibroblasts. Finally, we present a comprehensive overview of the Wnt gene expression landscape in different mammary gland cell types in pubertal and adult mice. This work contributes to understanding the effects of physiological hormone fluctuations and locally produced signaling molecules on gene expression changes in the mammary gland during the reproductive cycle and should be a useful resource for future studies investigating gene expression patterns in different cell types across different developmental timepoints.
Collapse
Affiliation(s)
- Nika Heijmans
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| | - Katrin E Wiese
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Renée van Amerongen
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Fu S, Ke H, Yuan H, Xu H, Chen W, Zhao L. Dual role of pregnancy in breast cancer risk. Gen Comp Endocrinol 2024; 352:114501. [PMID: 38527592 DOI: 10.1016/j.ygcen.2024.114501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Reproductive history is one of the strongest risk factors for breast cancer in women. Pregnancy can promote short-term breast cancer risk, but also reduce a woman's lifetime risk of breast cancer. Changes in hormone levels before and after pregnancy are one of the key factors in breast cancer risk. This article summarizes the changes in hormone levels before and after pregnancy, and the roles of hormones in mammary gland development and breast cancer progression. Other factors, such as changes in breast morphology and mammary gland differentiation, changes in the proportion of mammary stem cells (MaSCs), changes in the immune and inflammatory environment, and changes in lactation before and after pregnancy, also play key roles in the occurrence and development of breast cancer. This review discusses the dual effects and the potential mechanisms of pregnancy on breast cancer risk from the above aspects, which is helpful to understand the complexity of female breast cancer occurrence.
Collapse
Affiliation(s)
- Shiting Fu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Hao Ke
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | | | - Huaimeng Xu
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China
| | - Wenyan Chen
- Department of Medical Oncology, The Third Hospital of Nanchang, Nanchang 330009, China
| | - Limin Zhao
- Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang 330031, China.
| |
Collapse
|
15
|
Koskinen LM, Nieminen L, Arjonen A, Guzmán C, Peurla M, Peuhu E. Spatial Engineering of Mammary Epithelial Cell Cultures with 3D Bioprinting Reveals Growth Control by Branch Point Proximity. J Mammary Gland Biol Neoplasia 2024; 29:5. [PMID: 38416267 PMCID: PMC10902034 DOI: 10.1007/s10911-024-09557-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024] Open
Abstract
The three-dimensional (3D) structure of the ductal epithelium and the surrounding extracellular matrix (ECM) are integral aspects of the breast tissue, and they have important roles during mammary gland development, function and malignancy. However, the architecture of the branched mammary epithelial network is poorly recapitulated in the current in vitro models. 3D bioprinting is an emerging approach to improve tissue-mimicry in cell culture. Here, we developed and optimized a protocol for 3D bioprinting of normal and cancerous mammary epithelial cells into a branched Y-shape to study the role of cell positioning in the regulation of cell proliferation and invasion. Non-cancerous cells formed continuous 3D cell networks with several organotypic features, whereas the ductal carcinoma in situ (DCIS) -like cancer cells exhibited aberrant basal polarization and defective formation of the basement membrane (BM). Quantitative analysis over time demonstrated that both normal and cancerous cells proliferate more at the branch tips compared to the trunk region of the 3D-bioprinted cultures, and particularly at the tip further away from the branch point. The location-specific rate of proliferation was independent of TGFβ signaling but invasion of the DCIS-like breast cancer cells was reduced upon the inhibition of TGFβ. Thus, our data demonstrate that the 3D-bioprinted cells can sense their position in the branched network of cells and proliferate at the tips, thus recapitulating this feature of mammary epithelial branching morphogenesis. In all, our results demonstrate the capacity of the developed 3D bioprinting method for quantitative analysis of the relationships between tissue structure and cell behavior in breast morphogenesis and cancer.
Collapse
Affiliation(s)
- Leena M Koskinen
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | | | | | - Markus Peurla
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Emilia Peuhu
- Institute of Biomedicine, Cancer Laboratory FICAN West, University of Turku, Turku, Finland.
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
| |
Collapse
|
16
|
Kwon HC, Jung HS, Kim DH, Han JH, Han SG. The Role of Progesterone in Elf5 Activation and Milk Component Synthesis for Cell-Cultured Milk Production in MAC-T Cells. Animals (Basel) 2024; 14:642. [PMID: 38396610 PMCID: PMC10886090 DOI: 10.3390/ani14040642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/12/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Prolactin is essential for mammary gland development and lactation. Progesterone also induces ductal branching and alveolar formation via initial secretory differentiation within the mammary gland. Herein, we aimed to evaluate the role of progesterone as a prolactin substitute for the production of cell-cultured milk components in MAC-T cells. Cells were treated with various hormones such as prolactin (PRL), progesterone (P4), 17β-estradiol (E2), cortisol (COR), and insulin (INS) for 5 d. MAC-T cells cultured in a P4 differentiation media (2500 ng/mL of P4, 25 ng/mL of E2, 25 ng/mL of COR, and 25 ng/mL of INS) showed similar levels of E74-like factor 5 (Elf5) and milk component synthesis (α-casein, β-casein, α-lactalbumin, β-lactoglobulin, and triglycerides) compared to those cultured in a PRL differentiation media (5000 ng/mL of PRL, 500 ng/mL of CORT, and 50 ng/mL of INS). The levels of α-casein and triglycerides in the optimal P4 differentiation media were present at comparable levels to those in the PRL differentiation media. Our results demonstrated that P4 induces the activation of Elf5 and the synthesis of milk components in MAC-T cells, similar to PRL. Therefore, P4 may be used as an effective substitute of PRL for cell-cultured milk production in in vitro frameworks.
Collapse
Affiliation(s)
| | | | | | | | - Sung Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea; (H.C.K.); (H.S.J.); (D.H.K.); (J.H.H.)
| |
Collapse
|
17
|
Geldsetzer-Mendoza C, Riveros JL. Morphophysiological Responses of the Goat Mammary Gland to Water Scarcity in Arid and Semi-Arid Environments: Are They Enough to Generate Adaptation to New Climatic Challenges? Animals (Basel) 2023; 13:3825. [PMID: 38136862 PMCID: PMC10740433 DOI: 10.3390/ani13243825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/11/2023] [Accepted: 11/14/2023] [Indexed: 12/24/2023] Open
Abstract
Due to climate change, diverse territories of the planet will suffer from water restrictions. Goats are perceived as the most resilient ruminants in this scenario. So, various studies have focused on describing how a lower water intake influences milk production, especially in breeds adapted to desert environments. In water-stress situations, goats lose up to 32% of their body weight (BW), the rate of passage is reduced, and the digestibility of the feed increases. When goats consume water again, the rumen prevents hemolysis and osmotic shock from occurring. Regarding milk production, the response varies depending on the breed and the level of water restriction, maintaining the milk volume or reducing it by up to 41%. Systemically, it decreases the urinary volume and glomerular filtration rate, increasing blood osmolality and the vasopressin (ADH) concentration. Studies are scarce regarding changes in blood flow to the mammary gland, but there would be a reduction in blood flow velocity of up to 40% without changing blood pressure. New studies must be undertaken to determine which breeds or crosses are the best adapted to changing environmental conditions and to improve our understanding of the changes that occur at the morphophysiological level of the caprine mammary gland.
Collapse
Affiliation(s)
| | - José Luis Riveros
- Department of Animal Sciences, Faculty of Agronomy and Forestry, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| |
Collapse
|
18
|
Aarts MT, Wagner M, van der Wal T, van Boxtel AL, van Amerongen R. A molecular toolbox to study progesterone receptor signaling. J Mammary Gland Biol Neoplasia 2023; 28:24. [PMID: 38019315 PMCID: PMC10687192 DOI: 10.1007/s10911-023-09550-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/29/2023] [Indexed: 11/30/2023] Open
Abstract
Progesterone receptor (PR) signaling is required for mammary gland development and homeostasis. A major bottleneck in studying PR signaling is the lack of sensitive assays to measure and visualize PR pathway activity both quantitatively and spatially. Here, we develop new tools to study PR signaling in human breast epithelial cells. First, we generate optimized Progesterone Responsive Element (PRE)-luciferase constructs and demonstrate that these new reporters are a powerful tool to quantify PR signaling activity across a wide range of progesterone concentrations in two luminal breast cancer cell lines, MCF7 and T47D. We also describe a fluorescent lentiviral PRE-GFP reporter as a novel tool to visualize PR signaling at the single-cell level. Our reporter constructs are sensitive to physiological levels of progesterone. Second, we show that low background signaling, and high levels of PR expression are a prerequisite for robustly measuring PR signaling. Increasing PR expression by transient transfection, stable overexpression in MCF7 or clonal selection in T47D, drastically improves both the dynamic range of luciferase reporter assays, and the induction of endogenous PR target genes as measured by qRT-PCR. We find that the PR signaling response differs per cell line, target gene and hormone concentration used. Taken together, our tools allow a more rationally designed approach for measuring PR signaling in breast epithelial cells.
Collapse
Affiliation(s)
- Marleen T Aarts
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| | - Muriel Wagner
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| | - Tanne van der Wal
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| | - Antonius L van Boxtel
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| | - Renée van Amerongen
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands.
| |
Collapse
|
19
|
Zeng J, Singh S, Zhou X, Jiang Y, Casarez E, Atkins KA, Janes KA, Zong H. A genetic mosaic mouse model illuminates the pre-malignant progression of basal-like breast cancer. Dis Model Mech 2023; 16:dmm050219. [PMID: 37815460 PMCID: PMC10668031 DOI: 10.1242/dmm.050219] [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: 04/26/2023] [Accepted: 09/11/2023] [Indexed: 10/11/2023] Open
Abstract
Basal-like breast cancer (BLBC) is highly aggressive, and often characterized by BRCA1 and p53 deficiency. Although conventional mouse models enabled the investigation of BLBC at malignant stages, its initiation and pre-malignant progression remain understudied. Here, we leveraged a mouse genetic system known as mosaic analysis with double markers (MADM) to study BLBC initiation by generating rare GFP+Brca1, p53-deficient mammary cells alongside RFP+ wild-type sibling cells. After confirming the close resemblance of mammary tumors arising in this model to human BLBC at both transcriptomic and genomic levels, we focused our studies on the pre-malignant progression of BLBC. Initiated GFP+ mutant cells showed a stepwise pre-malignant progression trajectory from focal expansion to hyper-alveolarization and then to micro-invasion. Furthermore, despite morphological similarities to alveoli, hyper-alveolarized structures actually originate from ductal cells based on twin-spot analysis of GFP-RFP sibling cells. Finally, luminal-to-basal transition occurred exclusively in cells that have progressed to micro-invasive lesions. Our MADM model provides excellent spatiotemporal resolution to illuminate the pre-malignant progression of BLBC, and should enable future studies on early detection and prevention for this cancer.
Collapse
Affiliation(s)
- Jianhao Zeng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Shambhavi Singh
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Xian Zhou
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Ying Jiang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Eli Casarez
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Kristen A. Atkins
- Department of Pathology, University of Virginia Health System, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Kevin A. Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Hui Zong
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
- University of Virginia Comprehensive Cancer Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| |
Collapse
|
20
|
López-Mejía JA, Mantilla-Ollarves JC, Rocha-Zavaleta L. Modulation of JAK-STAT Signaling by LNK: A Forgotten Oncogenic Pathway in Hormone Receptor-Positive Breast Cancer. Int J Mol Sci 2023; 24:14777. [PMID: 37834225 PMCID: PMC10573125 DOI: 10.3390/ijms241914777] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer remains the most frequently diagnosed cancer in women worldwide. Tumors that express hormone receptors account for 75% of all cases. Understanding alternative signaling cascades is important for finding new therapeutic targets for hormone receptor-positive breast cancer patients. JAK-STAT signaling is commonly activated in hormone receptor-positive breast tumors, inducing inflammation, proliferation, migration, and treatment resistance in cancer cells. In hormone receptor-positive breast cancer, the JAK-STAT cascade is stimulated by hormones and cytokines, such as prolactin and IL-6. In normal cells, JAK-STAT is inhibited by the action of the adaptor protein, LNK. However, the role of LNK in breast tumors is not fully understood. This review compiles published reports on the expression and activation of the JAK-STAT pathway by IL-6 and prolactin and potential inhibition of the cascade by LNK in hormone receptor-positive breast cancer. Additionally, it includes analyses of available datasets to determine the level of expression of LNK and various members of the JAK-STAT family for the purpose of establishing associations between expression and clinical outcomes. Together, experimental evidence and in silico studies provide a better understanding of the potential implications of the JAK-STAT-LNK loop in hormone receptor-positive breast cancer progression.
Collapse
Affiliation(s)
- José A. López-Mejía
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 03100, Mexico; (J.A.L.-M.); (J.C.M.-O.)
| | - Jessica C. Mantilla-Ollarves
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 03100, Mexico; (J.A.L.-M.); (J.C.M.-O.)
| | - Leticia Rocha-Zavaleta
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 03100, Mexico; (J.A.L.-M.); (J.C.M.-O.)
- Programa Institucional de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 03100, Mexico
| |
Collapse
|
21
|
Myllymäki SM, Kaczyńska B, Lan Q, Mikkola ML. Spatially coordinated cell cycle activity and motility govern bifurcation of mammary branches. J Cell Biol 2023; 222:e202209005. [PMID: 37367826 PMCID: PMC10300433 DOI: 10.1083/jcb.202209005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 03/03/2023] [Accepted: 06/01/2023] [Indexed: 06/28/2023] Open
Abstract
Branching morphogenesis is an evolutionary solution to maximize epithelial function in a compact organ. It involves successive rounds of branch elongation and branch point formation to generate a tubular network. In all organs, branch points can form by tip splitting, but it is unclear how tip cells coordinate elongation and branching. Here, we addressed these questions in the embryonic mammary gland. Live imaging revealed that tips advance by directional cell migration and elongation relies upon differential cell motility that feeds a retrograde flow of lagging cells into the trailing duct, supported by tip proliferation. Tip bifurcation involved localized repression of cell cycle and cell motility at the branch point. Cells in the nascent daughter tips remained proliferative but changed their direction to elongate new branches. We also report the fundamental importance of epithelial cell contractility for mammary branching morphogenesis. The co-localization of cell motility, non-muscle myosin II, and ERK activities at the tip front suggests coordination/cooperation between these functions.
Collapse
Affiliation(s)
- Satu-Marja Myllymäki
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Beata Kaczyńska
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Qiang Lan
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Marja L. Mikkola
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Githaka JM, Pirayeshfard L, Goping IS. Cancer invasion and metastasis: Insights from murine pubertal mammary gland morphogenesis. Biochim Biophys Acta Gen Subj 2023; 1867:130375. [PMID: 37150225 DOI: 10.1016/j.bbagen.2023.130375] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/20/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Cancer invasion and metastasis accounts for the majority of cancer related mortality. A better understanding of the players that drive the aberrant invasion and migration of tumors cells will provide critical targets to inhibit metastasis. Postnatal pubertal mammary gland morphogenesis is characterized by highly proliferative, invasive, and migratory normal epithelial cells. Identifying the molecular regulators of pubertal gland development is a promising strategy since tumorigenesis and metastasis is postulated to be a consequence of aberrant reactivation of developmental stages. In this review, we summarize the pubertal morphogenesis regulators that are involved in cancer metastasis and revisit pubertal mammary gland transcriptome profiling to uncover both known and unknown metastasis genes. Our updated list of pubertal morphogenesis regulators shows that most are implicated in invasion and metastasis. This review highlights molecular linkages between development and metastasis and provides a guide for exploring novel metastatic drivers.
Collapse
Affiliation(s)
- John Maringa Githaka
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Leila Pirayeshfard
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ing Swie Goping
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Department of Oncology, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
23
|
Morato A, Accornero P, Hovey RC. ERBB Receptors and Their Ligands in the Developing Mammary Glands of Different Species: Fifteen Characters in Search of an Author. J Mammary Gland Biol Neoplasia 2023; 28:10. [PMID: 37219601 DOI: 10.1007/s10911-023-09538-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/26/2023] [Indexed: 05/24/2023] Open
Abstract
The ERBB tyrosine kinase receptors and their ligands belong to a complex family that has diverse biological effects and expression profiles in the developing mammary glands, where its members play an essential role in translating hormone signals into local effects. While our understanding of these processes stems mostly from mouse models, there is the potential for differences in how this family functions in the mammary glands of other species, particularly in light of their unique histomorphological features. Herein we review the postnatal distribution and function of ERBB receptors and their ligands in the mammary glands of rodents and humans, as well as for livestock and companion animals. Our analysis highlights the diverse biology for this family and its members across species, the regulation of their expression, and how their roles and functions might be modulated by varying stromal composition and hormone interactions. Given that ERBB receptors and their ligands have the potential to influence processes ranging from normal mammary development to diseased states such as cancer and/or mastitis, both in human and veterinary medicine, a more complete understanding of their biological functions should help to direct future research and the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Alessia Morato
- Department of Animal Science, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA.
| | - Paolo Accornero
- Department of Veterinary Science, University of Turin, Largo Paolo Braccini 2, Grugliasco, TO, 10095, Italy
| | - Russell C Hovey
- Department of Animal Science, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| |
Collapse
|
24
|
Zeng J, Singh S, Jiang Y, Casarez E, Atkins KA, Janes KA, Zong H. A genetic mosaic mouse model illuminates the pre-malignant progression of basal-like breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.538333. [PMID: 37163037 PMCID: PMC10168298 DOI: 10.1101/2023.04.25.538333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Basal-like breast cancer is an aggressive breast cancer subtype, often characterized by a deficiency in BRCA1 function and concomitant loss of p53 . While conventional mouse models enable the investigation of its malignant stages, one that reveals its initiation and pre-malignant progression is lacking. Here, we leveraged a mouse genetic system known as M osaic A nalysis with D ouble M arkers (MADM) to generate rare GFP-labeled Brca1 , p53 -deficient cells alongside RFP+ wildtype sibling cells in the mammary gland. The mosaicism resembles the sporadic initiation of human cancer and enables spatially resolved analysis of mutant cells in comparison to paired wildtype sibling cells. Mammary tumors arising in the model show transcriptomic and genomic characteristics similar to human basal-like breast cancer. Analysis of GFP+ mutant cells at interval time points before malignancy revealed a stepwise progression of lesions from focal expansion to hyper-alveolarization and then to micro-invasion. These stereotyped morphologies indicate the pre-malignant stage irrespective of the time point at which it is observed. Paired analysis of GFP-RFP siblings during focal expansion suggested that hyper-alveolarized structures originate from ductal rather than alveolar cells, despite their morphological similarities to alveoli. Evidence for luminal-to-basal transition at the pre-malignant stages was restricted to cells that had escaped hyper-alveoli and progressed to micro-invasive lesions. Our MADM-based mouse model presents a useful tool for studying the pre-malignancy of basal-like breast cancer. Summary statement A mouse model recapitulates the process of human basal-like breast tumorigenesis initiated from sporadic Brca1, p53 -deficient cells, empowering spatially-resolved analysis of mutant cells during pre-malignant progression.
Collapse
|
25
|
Campbell CJ, Booth BW. The Influence of the Normal Mammary Microenvironment on Breast Cancer Cells. Cancers (Basel) 2023; 15:cancers15030576. [PMID: 36765535 PMCID: PMC9913214 DOI: 10.3390/cancers15030576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 01/20/2023] Open
Abstract
The tumor microenvironment is recognized as performing a critical role in tumor initiation, progression, and metastasis of many cancers, including breast cancer. The breast cancer microenvironment is a complex mixture of cells consisting of tumor cells, immune cells, fibroblasts, and vascular cells, as well as noncellular components, such as extracellular matrix and soluble products. The interactions between the tumor cells and the tumor microenvironment modulate tumor behavior and affect the responses of cancer patients to therapies. The interactions between tumor cells and the surrounding environment can include direct cell-to-cell contact or through intercellular signals over short and long distances. The intricate functions of the tumor microenvironment in breast cancer have led to increased research into the tumor microenvironment as a possible therapeutic target of breast cancer. Though expanded research has shown the clear importance of the tumor microenvironment, there is little focus on how normal mammary epithelial cells can affect breast cancer cells. Previous studies have shown the normal breast microenvironment can manipulate non-mammary stem cells and tumor-derived cancer stem cells to participate in normal mammary gland development. The tumorigenic cells lose their tumor-forming capacity and are "redirected" to divide into "normal", non-tumorigenic cells. This cellular behavior is "cancer cell redirection". This review will summarize the current literature on cancer cell redirection and the normal mammary microenvironment's influence on breast cancer cells.
Collapse
|
26
|
Khorshid Shamshiri A, Alidoust M, Hemmati Nokandei M, Pasdar A, Afzaljavan F. Genetic architecture of mammographic density as a risk factor for breast cancer: a systematic review. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:1729-1747. [PMID: 36639603 DOI: 10.1007/s12094-022-03071-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Mammography Density (MD) is a potential risk marker that is influenced by genetic polymorphisms and can subsequently modulate the risk of breast cancer. This qualitative systematic review summarizes the genes and biological pathways involved in breast density and discusses the potential clinical implications in view of the genetic risk profile for breast density. METHODS The terms related to "Common genetic variations" and "Breast density" were searched in Scopus, PubMed, and Web of Science databases. Gene pathways analysis and assessment of protein interactions were also performed. RESULTS Eighty-six studies including 111 genes, reported a significant association between mammographic density in different populations. ESR1, IGF1, IGFBP3, and ZNF365 were the most prevalent genes. Moreover, estrogen metabolism, signal transduction, and prolactin signaling pathways were significantly related to the associated genes. Mammography density was an associated phenotype, and eight out of 111 genes, including COMT, CYP19A1, CYP1B1, ESR1, IGF1, IGFBP1, IGFBP3, and LSP1, were modifiers of this trait. CONCLUSION Genes involved in developmental processes and the evolution of secondary sexual traits play an important role in determining mammographic density. Due to the effect of breast tissue density on the risk of breast cancer, these genes may also be associated with breast cancer risk.
Collapse
Affiliation(s)
- Asma Khorshid Shamshiri
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Alidoust
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahboubeh Hemmati Nokandei
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Pasdar
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Division of Applied Medicine, Medical School, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Fahimeh Afzaljavan
- Clinical Research Development Unit, Faculty of Medicine, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, 917794-8564, Iran.
| |
Collapse
|
27
|
Renteria M, Belkin O, Aickareth J, Jang D, Hawwar M, Zhang J. Zinc's Association with the CmPn/CmP Signaling Network in Breast Cancer Tumorigenesis. Biomolecules 2022; 12:1672. [PMID: 36421686 PMCID: PMC9687477 DOI: 10.3390/biom12111672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 08/24/2023] Open
Abstract
It is well-known that serum and cellular concentrations of zinc are altered in breast cancer patients. Specifically, there are notable zinc hyper-aggregates in breast tumor cells when compared to normal mammary epithelial cells. However, the mechanisms responsible for zinc accumulation and the consequences of zinc dysregulation are poorly understood. In this review, we detailed cellular zinc regulation/dysregulation under the influence of varying levels of sex steroids and breast cancer tumorigenesis to try to better understand the intricate relationship between these factors based on our current understanding of the CmPn/CmP signaling network. We also made some efforts to propose a relationship between zinc signaling and the CmPn/CmP signaling network.
Collapse
Affiliation(s)
| | | | | | | | | | - Jun Zhang
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
28
|
Schaduangrat N, Anuwongcharoen N, Moni MA, Lio' P, Charoenkwan P, Shoombuatong W. StackPR is a new computational approach for large-scale identification of progesterone receptor antagonists using the stacking strategy. Sci Rep 2022; 12:16435. [PMID: 36180453 PMCID: PMC9525257 DOI: 10.1038/s41598-022-20143-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022] Open
Abstract
Progesterone receptors (PRs) are implicated in various cancers since their presence/absence can determine clinical outcomes. The overstimulation of progesterone can facilitate oncogenesis and thus, its modulation through PR inhibition is urgently needed. To address this issue, a novel stacked ensemble learning approach (termed StackPR) is presented for fast, accurate, and large-scale identification of PR antagonists using only SMILES notation without the need for 3D structural information. We employed six popular machine learning (ML) algorithms (i.e., logistic regression, partial least squares, k-nearest neighbor, support vector machine, extremely randomized trees, and random forest) coupled with twelve conventional molecular descriptors to create 72 baseline models. Then, a genetic algorithm in conjunction with the self-assessment-report approach was utilized to determine m out of the 72 baseline models as means of developing the final meta-predictor using the stacking strategy and tenfold cross-validation test. Experimental results on the independent test dataset show that StackPR achieved impressive predictive performance with an accuracy of 0.966 and Matthew's coefficient correlation of 0.925. In addition, analysis based on the SHapley Additive exPlanation algorithm and molecular docking indicates that aliphatic hydrocarbons and nitrogen-containing substructures were the most important features for having PR antagonist activity. Finally, we implemented an online webserver using StackPR, which is freely accessible at http://pmlabstack.pythonanywhere.com/StackPR . StackPR is anticipated to be a powerful computational tool for the large-scale identification of unknown PR antagonist candidates for follow-up experimental validation.
Collapse
Affiliation(s)
- Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Nuttapat Anuwongcharoen
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Mohammad Ali Moni
- Artificial Intelligence & Digital Health Data Science, School of Health and Rehabilitation Sciences, Faculty of Health and Behavioural Sciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Pietro Lio'
- Department of Computer Science and Technology, University of Cambridge, Cambridge, CB3 0FD, UK
| | - Phasit Charoenkwan
- Modern Management and Information Technology, College of Arts, Media and Technology, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
29
|
Diagnostic and Prognostic Impact of Progesterone Receptor Immunohistochemistry: A Study Evaluating More Than 16,000 Tumors. Anal Cell Pathol (Amst) 2022; 2022:6412148. [PMID: 35992051 PMCID: PMC9381849 DOI: 10.1155/2022/6412148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
Progesterone receptor (PR) is a member of the nuclear/steroid hormone receptor family of ligand-dependent transcription factors. It plays an important role in reproduction and mammary gland development and has various tissue-specific effects in nonreproductive organs. In diagnostic pathology, positive PR immunostaining is used to support a diagnosis of breast or gynecologic origin in a tumor. In this study, the expression of PR was analyzed by immunohistochemistry in 18,176 (interpretable: 16,445) samples from 147 different tumor types and subtypes in a tissue microarray format. PR immunostaining was detected in 57.4% of breast tumors, 28.6% of other gynecological tumors, and 1.8% of nongynecological and nonmammary tumors. Among the group of nongynecological and nonmammary tumors, particularly high rates of PR positivity were seen in neuroendocrine tumors (54.3%) and neuroendocrine carcinomas (35.7%) of the pancreas. A comparison with clinico-pathological parameters showed that reduced PR immunostaining was significantly associated with adverse histopathological and clinical features in breast carcinoma, endometrioid endometrial carcinoma, and pancreatic neuroendocrine tumors. In summary, our analysis of 147 different tumor types for PR immunostaining provides a ranking list of tumor entities according to their prevalence of PR positivity, helps to better understand the diagnostic utility of PR, and highlights the distinct PR positivity among neuroendocrine neoplasms of pancreatic origin.
Collapse
|
30
|
Lee SH, Yap YHY, Lim CL, Woo ARE, Lin VCL. Activation function 1 of progesterone receptor is required for mammary development and regulation of RANKL during pregnancy. Sci Rep 2022; 12:12286. [PMID: 35854046 PMCID: PMC9296660 DOI: 10.1038/s41598-022-16289-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022] Open
Abstract
Progesterone receptor (PGR) is a member of the nuclear receptor superfamily of transcription factors. It is critical for mammary stem cells expansion, mammary ductal branching and alveologenesis. The transcriptional activity of PGR is mainly mediated by activation functions AF1 and AF2. Although the discovery of AF1 and AF2 propelled the understanding of the mechanism of gene regulation by nuclear receptors, their physiological roles are still poorly understood. This is largely due to the lack of suitable genetic models. The present study reports gain or loss of AF1 function mutant mouse models in the study of mammary development. The gain of function mutant AF1_QQQ exhibits hyperactivity while the loss of function mutant AF1_FFF shows hypoactivity on mammary development. However, the involvement of AF1 is context dependent. Whereas the AF1_FFF mutation causes significant impairment in mammary development during pregnancy or in response to estrogen and progesterone, it has no effect on mammary development in nulliparous mice. Furthermore, Rankl, but not Wnt4 and Areg is a major target gene of AF1. In conclusion, PGR AF1 is a pivotal ligand-dependent activation domain critical for mammary development during pregnancy and it exerts gene specific effect on PGR regulated genes.
Collapse
Affiliation(s)
- Shi Hao Lee
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Yeannie H Y Yap
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.,Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Bandar Saujana Putra, 42610, Jenjarom, Selangor, Malaysia
| | - Chew Leng Lim
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Amanda Rui En Woo
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Valerie C L Lin
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.
| |
Collapse
|
31
|
Regulation of Tight Junctions by Sex Hormones in Goat Mammary Epithelial Cells. Animals (Basel) 2022; 12:ani12111404. [PMID: 35681868 PMCID: PMC9179430 DOI: 10.3390/ani12111404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary How ovarian hormones affect goat lactation by regulating cell–cell junctions is still unclear. Through the in vivo and in vitro assays, we found that ovarian hormones could elevate cell–cell junction protein expression, which may affect the intercellular space and molecule transportation between the goat mammary epithelial cells. Our assessment suggests that ovarian hormones may affect goat milk production by regulating the cell–cell junction protein expression between mammary epithelial cells. Abstract The sex hormones of estrogen and progesterone (P4) play a vital role in mammary gland development and milk lactation in ruminants. The tight junction (TJ) between adjacent secretory epithelial cells is instrumental in establishing the mammary blood–milk barrier. However, whether estrogen and P4 exert their effect on mammary function via regulating TJ remain unclear. Here, to clarify the role of 17-β estradiol (E2) and P4 in the regulation of TJ in goat mammary gland, we first explored the relationships between the concentrations of E2, P4, and the protein expression of claudin-1, claudin-3, occludin, and ZO-1 during the mammary gland development in goat. Then, we further explored the mRNA and protein expression of claudin-1, claudin-3, occludin, and ZO-1 in the goat mammary epithelial cells (GMECs) in vitro under different concentrations of E2 and P4. The results demonstrated that the protein expression of claudin-1 decreased, but occludin and ZO-1 increased with the decline in E2 and P4 during the transition from pregnancy to lactation. In the in vitro studies, E2 exerted a positive effect on the mRNA expression of claudin-1, and accelerated the proteins’ expression of claudin-1 and ZO-1 in GMECs; P4 upregulated the mRNA expression of claudin-1, claudin-3, occludin, and ZO-1, and also improved the protein expression of claudin-1, claudin-3, and ZO-1 in the GMECs. The results demonstrated that E2 and P4 play an important role in regulating the expression of the mammary TJ components, which may ultimately affect the mammary gland development and milk lactation.
Collapse
|
32
|
Alveolar cells in the mammary gland: lineage commitment and cell death. Biochem J 2022; 479:995-1006. [PMID: 35551601 PMCID: PMC9162463 DOI: 10.1042/bcj20210734] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/17/2022]
Abstract
The mammary gland provides a spectacular example of physiological cell death whereby the cells that produce milk during lactation are removed swiftly, efficiently, and without inducing inflammation upon the cessation of lactation. The milk-producing cells arise primarily during pregnancy and comprise the alveolar lineage that is specified by signalling pathways and factors that are activated in response to pregnancy hormones. There are at least two alveolar sub-lineages, one of which is marked by the presence of binucleate cells that are especially susceptible to programmed cell death during involution. This process of post-lactational regression, or involution, is carefully orchestrated and occurs in two phases, the first results in a rapid switch in cell fate with the secretory epithelial cells becoming phagocytes whereupon they destroy dead and dying cells from milk. This reversible phase is followed by the second phase that is marked by an influx of immune cells and a remodelling of the gland to replace the alveolar cells with re-differentiated adipocytes, resulting in a return to the pre-pregnant state in preparation for any subsequent pregnancies. The mouse mammary gland provides an excellent experimental tool with which to investigate lineage commitment and the mechanisms of programmed cell death that occur in a normal physiological process. Importantly, involution has highlighted a role for lysoptosis, a mechanism of cell death that is mediated by lysosomal cathepsins and their endogenous inhibitors, serpins. In this review, I discuss alveolar lineage commitment during pregnancy and the programmed cell death pathways that destroy these cells during involution.
Collapse
|
33
|
Coppola L, Tait S, Fabbrizi E, Perugini M, La Rocca C. Comparison of the Toxicological Effects of Pesticides in Non-Tumorigenic MCF-12A and Tumorigenic MCF-7 Human Breast Cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:4453. [PMID: 35457321 PMCID: PMC9030493 DOI: 10.3390/ijerph19084453] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 11/16/2022]
Abstract
Humans are exposed to residues of organophosphate and neonicotinoid pesticides, commonly used in agriculture. Children are particularly vulnerable and, among possible adverse outcomes, the increased incidence of premature mammary gland development (thelarche) has raised concern. We evaluated the toxicological effects of chlorpyrifos (CPF), imidacloprid (IMI) and glyphosate (GLY) at exposure concentrations occurring in children on the tumorigenic MCF-7 and non-tumorigenic MCF-12A breast cell lines, as representative of the target organ model, assessing cytotoxicity, apoptosis, necrosis, intracellular reactive oxygen species (ROS) and ATP levels, 17β-estradiol secretion and gene expression of nuclear receptors involved in mammary gland development. The pesticides decreased cell vitality in MCF-7 and cell proliferation in MCF-12A cells. ATP levels were decreased in MCF-7 cells by pesticides and apoptosis was increased in MCF-12A cells only by GLY (2.3 nM). ROS production was decreased by pesticides in both cell lines, except IMI (1.6 nM) in MCF-7 cells. Endocrine disrupting activity was highlighted by induction of 17β-estradiol secretion and modulation of the gene expression of estrogen alpha and beta, progesterone, androgen, and aryl hydrocarbon receptors in both cell lines. The use of MCF-7 and MCF-12A cells highlighted dissimilar modes of action of each pesticide at low human relevant concentrations.
Collapse
Affiliation(s)
- Lucia Coppola
- Center for Gender-Specific Medicine, Italian National Institute of Health, 00161 Rome, Italy; (L.C.); (S.T.)
- Department of Physiology and Pharmacology V. Erspamer, Sapienza University of Rome, 00185 Rome, Italy
| | - Sabrina Tait
- Center for Gender-Specific Medicine, Italian National Institute of Health, 00161 Rome, Italy; (L.C.); (S.T.)
| | - Enrica Fabbrizi
- Pediatric Departmental Simple Operative Unit, Civitanova Marche Hospital, ASUR Marche Area Vasta n. 3, 62100 Macerata, Italy;
| | - Monia Perugini
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy;
| | - Cinzia La Rocca
- Center for Gender-Specific Medicine, Italian National Institute of Health, 00161 Rome, Italy; (L.C.); (S.T.)
| |
Collapse
|
34
|
The Mammary Gland: Basic Structure and Molecular Signaling during Development. Int J Mol Sci 2022; 23:ijms23073883. [PMID: 35409243 PMCID: PMC8998991 DOI: 10.3390/ijms23073883] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 01/27/2023] Open
Abstract
The mammary gland is a compound, branched tubuloalveolar structure and a major characteristic of mammals. The mammary gland has evolved from epidermal apocrine glands, the skin glands as an accessory reproductive organ to support postnatal survival of offspring by producing milk as a source of nutrition. The mammary gland development begins during embryogenesis as a rudimentary structure that grows into an elementary branched ductal tree and is embedded in one end of a larger mammary fat pad at birth. At the onset of ovarian function at puberty, the rudimentary ductal system undergoes dramatic morphogenetic change with ductal elongation and branching. During pregnancy, the alveolar differentiation and tertiary branching are completed, and during lactation, the mature milk-producing glands eventually develop. The early stages of mammary development are hormonal independent, whereas during puberty and pregnancy, mammary gland development is hormonal dependent. We highlight the current understanding of molecular regulators involved during different stages of mammary gland development.
Collapse
|
35
|
Li Z, Wei H, Li S, Wu P, Mao X. The Role of Progesterone Receptors in Breast Cancer. Drug Des Devel Ther 2022; 16:305-314. [PMID: 35115765 PMCID: PMC8801368 DOI: 10.2147/dddt.s336643] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 01/12/2022] [Indexed: 12/30/2022] Open
Abstract
The progesterone receptor (PR) modulates estrogen receptors α (ERα) action in breast cancer; it is an upregulated target gene of ER, and its expression is dependent on estrogen. PR is also a valuable prognostic biomarker in breast cancer, especially in hormone-positive breast cancer. High expression of PR is more frequently observed in tumors with a better baseline prognosis (ie, luminal A) than tumors with a poor baseline prognosis (ie, luminal B). In the following review, we present the role of PR in breast cancer, including the genomic characteristics and pathways in breast cancer, PR and endocrine therapy.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Hongrui Wei
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Siyan Li
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Pei Wu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Xiaoyun Mao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| |
Collapse
|
36
|
Pecci A, Ogara MF, Sanz RT, Vicent GP. Choosing the right partner in hormone-dependent gene regulation: Glucocorticoid and progesterone receptors crosstalk in breast cancer cells. Front Endocrinol (Lausanne) 2022; 13:1037177. [PMID: 36407312 PMCID: PMC9672667 DOI: 10.3389/fendo.2022.1037177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Abstract
Steroid hormone receptors (SHRs) belong to a large family of ligand-activated nuclear receptors that share certain characteristics and possess others that make them unique. It was thought for many years that the specificity of hormone response lay in the ligand. Although this may be true for pure agonists, the natural ligands as progesterone, corticosterone and cortisol present a broader effect by simultaneous activation of several SHRs. Moreover, SHRs share structural and functional characteristics that range from similarities between ligand-binding pockets to recognition of specific DNA sequences. These properties are clearly evident in progesterone (PR) and glucocorticoid receptors (GR); however, the biological responses triggered by each receptor in the presence of its ligand are different, and in some cases, even opposite. Thus, what confers the specificity of response to a given receptor is a long-standing topic of discussion that has not yet been unveiled. The levels of expression of each receptor, the differential interaction with coregulators, the chromatin accessibility as well as the DNA sequence of the target regions in the genome, are reliable sources of variability in hormone action that could explain the results obtained so far. Yet, to add further complexity to this scenario, it has been described that receptors can form heterocomplexes which can either compromise or potentiate the respective hormone-activated pathways with its possible impact on the pathological condition. In the present review, we summarized the state of the art of the functional cross-talk between PR and GR in breast cancer cells and we also discussed new paradigms of specificity in hormone action.
Collapse
Affiliation(s)
- Adali Pecci
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
- *Correspondence: Adali Pecci, ; Guillermo Pablo Vicent,
| | - María Florencia Ogara
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| | - Rosario T. Sanz
- Molecular Biology Institute of Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), Barcelona, Spain
| | - Guillermo Pablo Vicent
- Molecular Biology Institute of Barcelona, Consejo Superior de Investigaciones Científicas (IBMB-CSIC), Barcelona, Spain
- *Correspondence: Adali Pecci, ; Guillermo Pablo Vicent,
| |
Collapse
|
37
|
Akinjiyan FA, Han Y, Luo J, Toriola AT. Does circulating progesterone mediate the associations of single nucleotide polymorphisms in progesterone receptor (PGR)-related genes with mammographic breast density in premenopausal women? Discov Oncol 2021; 12:47. [PMID: 34790961 PMCID: PMC8566393 DOI: 10.1007/s12672-021-00438-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/28/2021] [Indexed: 10/31/2022] Open
Abstract
Progesterone is a proliferative hormone in the breast but the associations of genetic variations in progesterone-regulated pathways with mammographic breast density (MD) in premenopausal women and whether these associations are mediated through circulating progesterone are not clearly defined. We, therefore, investigated these associations in 364 premenopausal women with a median age of 44 years. We sequenced 179 progesterone receptor (PGR)-related single nucleotide polymorphisms (SNPs). We measured volumetric percent density (VPD) and non-dense volume (NDV) using Volpara. Linear regression models were fit on circulating progesterone or VPD/NDV separately. We performed mediation analysis to evaluate whether the effect of a SNP on VPD/NDV is mediated through circulating progesterone. All analyses were adjusted for confounders, phase of menstrual cycle and the Benjamini-Hochberg false discovery (FDR) adjusted p-value was applied to correct for multiple testing. In multivariable analyses, only PGR rs657516 had a direct effect on VPD (averaged direct effect estimate = - 0.20, 95%CI = - 0.38 ~ - 0.04, p-value = 0.02) but this was not statistically significant after FDR correction and the effect was not mediated by circulating progesterone (mediation effect averaged across the two genotypes = 0.01, 95%CI = - 0.02 ~ 0.03, p-value = 0.70). Five SNPs (PGR rs11571241, rs11571239, rs1824128, rs11571150, PGRMC1 rs41294894) were associated with circulating progesterone but these were not statistically significant after FDR correction. SNPs in PGR-related genes were not associated with VPD, NDV and circulating progesterone did not mediate the associations, suggesting that the effects, if any, of these SNPs on MD are independent of circulating progesterone. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s12672-021-00438-1.
Collapse
Affiliation(s)
- Favour A. Akinjiyan
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Yunan Han
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, Campus Box 8100, 660 South Euclid Ave, St. Louis, MO 63110 USA
- Department of Breast Surgery, First Hospital of China Medical University, Shenyang, 110001 Liaoning Province China
| | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, Campus Box 8100, 660 South Euclid Ave, St. Louis, MO 63110 USA
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Adetunji T. Toriola
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, Campus Box 8100, 660 South Euclid Ave, St. Louis, MO 63110 USA
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
38
|
Kuang X, McAndrew MJ, Mustachio LM, Chen YJC, Atanassov BS, Lin K, Lu Y, Shen J, Salinger A, Macatee T, Dent SYR, Koutelou E. Usp22 Overexpression Leads to Aberrant Signal Transduction of Cancer-Related Pathways but Is Not Sufficient to Drive Tumor Formation in Mice. Cancers (Basel) 2021; 13:4276. [PMID: 34503086 PMCID: PMC8428332 DOI: 10.3390/cancers13174276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022] Open
Abstract
Usp22 overexpression is observed in several human cancers and is correlated with poor patient outcomes. The molecular basis underlying this correlation is not clear. Usp22 is the catalytic subunit of the deubiquitylation module in the SAGA histone-modifying complex, which regulates gene transcription. Our previous work demonstrated that the loss of Usp22 in mice leads to decreased expression of several components of receptor tyrosine kinase and TGFβ signaling pathways. To determine whether these pathways are upregulated when Usp22 is overexpressed, we created a mouse model that expresses high levels of Usp22 in all tissues. Phenotypic characterization of these mice revealed over-branching of the mammary glands in females. Transcriptomic analyses indicate the upregulation of key pathways involved in mammary gland branching in mammary epithelial cells derived from the Usp22-overexpressing mice, including estrogen receptor, ERK/MAPK, and TGFβ signaling. However, Usp22 overexpression did not lead to increased tumorigenesis in any tissue. Our findings indicate that elevated levels of Usp22 are not sufficient to induce tumors, but it may enhance signaling abnormalities associated with oncogenesis.
Collapse
Affiliation(s)
- Xianghong Kuang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael J. McAndrew
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Luminex Corporation, 12212 Technology Blvd. Suite 130, Austin, TX 78721, USA
| | - Lisa Maria Mustachio
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ying-Jiun C. Chen
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Boyko S. Atanassov
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Shen
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew Salinger
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Timothy Macatee
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sharon Y. R. Dent
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Evangelia Koutelou
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA; (X.K.); (M.J.M.); (L.M.M.); (Y.-J.C.C.); (K.L.); (Y.L.); (J.S.); (A.S.); (T.M.)
- Center for Cancer Epigenetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
39
|
Ruiz TFR, Taboga SR, Leonel ECR. Molecular mechanisms of mammary gland remodeling: A review of the homeostatic versus bisphenol a disrupted microenvironment. Reprod Toxicol 2021; 105:1-16. [PMID: 34343637 DOI: 10.1016/j.reprotox.2021.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 12/30/2022]
Abstract
Mammary gland (MG) undergoes critical points of structural changes throughout a woman's life. During the perinatal and pubertal stages, MG develops through growth and differentiation to establish a pre-mature feature. If pregnancy and lactation occur, the epithelial compartment branches and differentiates to create a specialized structure for milk secretion and nurturing of the newborn. However, the ultimate MG modification consists of a regression process aiming to reestablish the smaller and less energy demanding structure until another production cycle happens. The unraveling of these fascinating physiologic cycles has helped the scientific community elucidate aspects of molecular regulation of proliferative and apoptotic events and remodeling of the stromal compartment. However, greater understanding of the hormonal pathways involved in MG developmental stages led to concern that endocrine disruptors such as bisphenol A (BPA), may influence these specific development/involution stages, called "windows of susceptibility". Since it is used in the manufacture of polycarbonate plastics and epoxy resins, BPA is a ubiquitous chemical present in human everyday life, exerting an estrogenic effect. Thus, descriptions of its deleterious effects on the MG, especially in terms of serum hormone concentrations, hormonal receptor expression, molecular pathways, and epigenetic alterations, have been widely published. Therefore, allied to a didactic description of the main physiological mechanisms involved in different critical points of MG development, the current review provides a summary of key mechanisms by which the endocrine disruptor BPA impacts MG homeostasis at different windows of susceptibility, causing short- and long-term effects.
Collapse
Affiliation(s)
- Thalles Fernando Rocha Ruiz
- São Paulo State University (Unesp), Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São José Do Rio Preto, Brazil.
| | - Sebastião Roberto Taboga
- São Paulo State University (Unesp), Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São José Do Rio Preto, Brazil.
| | - Ellen Cristina Rivas Leonel
- São Paulo State University (Unesp), Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São José Do Rio Preto, Brazil; Federal University of Goiás (UFG), Department of Histology, Embryology and Cell Biology, Institute of Biological Sciences, Goiânia, Brazil.
| |
Collapse
|
40
|
Chen W, Wei W, Yu L, Zhang X, Huang F, Zheng Q, Wang L, Cai C. Baicalin Promotes Mammary Gland Development via Steroid-Like Activities. Front Cell Dev Biol 2021; 9:682469. [PMID: 34295892 PMCID: PMC8290356 DOI: 10.3389/fcell.2021.682469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/07/2021] [Indexed: 12/03/2022] Open
Abstract
Baicalin, the main flavonoid component extracted from Scutellaria roots, has a variety of biological activities and is therefore used in the treatment of many kinds of diseases. However, whether baicalin affects the normal development of tissues and organs is still unclear. Here, using a mouse mammary gland model, we investigated the effects of baicalin on the expansion of mammary stem cells (MaSCs) and mammary development, as well as breast cancer progression. Interestingly, we found that baicalin administration significantly accelerates duct elongation at puberty, and promotes alveolar development and facilitates milk secretion during pregnancy. Furthermore, self-renewal of MaSCs was significantly promoted in the presence of baicalin. Moreover, in a tumor xenograft model, baicalin promoted tumor growth of the MDA-MB-231 cell line, but suppressed tumor growth of the ZR-751 cell line. Mechanistically, baicalin can induce expression of the protein C receptor, while inhibiting the expression of the estrogen receptor. Transcriptome analysis revealed that baicalin is involved in signaling pathways related to mammary gland development, immune response, and cell cycle control. Taken together, our results from comprehensive investigation of the biological activity of baicalin provide a theoretical basis for its rational clinical application.
Collapse
Affiliation(s)
- Weizhen Chen
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Medical Research Institute, Wuhan University, Wuhan, China
| | - Wei Wei
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Medical Research Institute, Wuhan University, Wuhan, China
| | - Liya Yu
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xin Zhang
- Guangzhou University of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou, China
| | - Fujing Huang
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Medical Research Institute, Wuhan University, Wuhan, China
| | - Qiping Zheng
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Hematological Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China.,Shenzhen Academy of Peptide Targeting Technology at Pingshan, Shenzhen Tyercan Bio-pharm Co., Ltd., Shenzhen, China
| | - Lingli Wang
- Guangzhou University of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou, China
| | - Cheguo Cai
- Department of Orthopaedics, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Medical Research Institute, Wuhan University, Wuhan, China.,Dongguan and Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan City, China.,Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| |
Collapse
|
41
|
Rauner G, Kuperwasser C. Microenvironmental control of cell fate decisions in mammary gland development and cancer. Dev Cell 2021; 56:1875-1883. [PMID: 34256927 DOI: 10.1016/j.devcel.2021.06.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 12/28/2022]
Abstract
Cell fate decisions are critical for adequate tissue development, maintenance and regeneration. In the mammary gland, epithelial cell fates are tightly controlled by the microenvironment. Here, we review how cell fate decisions are regulated by components of the microenvironment during mammary gland development and how pathological changes in the microenvironment can alter cell fates, leading to malignancy. Specifically, we describe the current understanding of how mammary cell fate is controlled and directed by three elements: the extracellular matrix, the immune microenvironment, and hormones-and how these elements can converge to create microenvironments that promote a fourth element: DNA damage.
Collapse
Affiliation(s)
- Gat Rauner
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Charlotte Kuperwasser
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA; Laboratory for the Convergence of Biomedical, Physical, and Engineering Sciences, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
42
|
Englund JI, Ritchie A, Blaas L, Cojoc H, Pentinmikko N, Döhla J, Iqbal S, Patarroyo M, Katajisto P. Laminin alpha 5 regulates mammary gland remodeling through luminal cell differentiation and Wnt4-mediated epithelial crosstalk. Development 2021; 148:269157. [PMID: 34128985 PMCID: PMC8254867 DOI: 10.1242/dev.199281] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/10/2021] [Indexed: 11/20/2022]
Abstract
Epithelial attachment to the basement membrane (BM) is essential for mammary gland development, yet the exact roles of specific BM components remain unclear. Here, we show that Laminin α5 (Lama5) expression specifically in the luminal epithelial cells is necessary for normal mammary gland growth during puberty, and for alveologenesis during pregnancy. Lama5 loss in the keratin 8-expressing cells results in reduced frequency and differentiation of hormone receptor expressing (HR+) luminal cells. Consequently, Wnt4-mediated crosstalk between HR+ luminal cells and basal epithelial cells is compromised during gland remodeling, and results in defective epithelial growth. The effects of Lama5 deletion on gland growth and branching can be rescued by Wnt4 supplementation in the in vitro model of branching morphogenesis. Our results reveal a surprising role for BM-protein expression in the luminal mammary epithelial cells, and highlight the function of Lama5 in mammary gland remodeling and luminal differentiation. Summary: Luminal mammary epithelial cells produce basement membrane laminin α5 necessary for mammary epithelial growth and differentiation. Laminin α5 loss compromises hormone receptor-positive luminal cell function and Wnt4-mediated crosstalk between epithelial cells.
Collapse
Affiliation(s)
- Johanna I Englund
- Institute of Biotechnology, Helsinki Institute of Life Sciences (HiLIFE), 00014 University of Helsinki, Helsinki, Finland
| | - Alexandra Ritchie
- Institute of Biotechnology, Helsinki Institute of Life Sciences (HiLIFE), 00014 University of Helsinki, Helsinki, Finland
| | - Leander Blaas
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Hanne Cojoc
- Institute of Biotechnology, Helsinki Institute of Life Sciences (HiLIFE), 00014 University of Helsinki, Helsinki, Finland
| | - Nalle Pentinmikko
- Institute of Biotechnology, Helsinki Institute of Life Sciences (HiLIFE), 00014 University of Helsinki, Helsinki, Finland
| | - Julia Döhla
- Institute of Biotechnology, Helsinki Institute of Life Sciences (HiLIFE), 00014 University of Helsinki, Helsinki, Finland
| | - Sharif Iqbal
- Institute of Biotechnology, Helsinki Institute of Life Sciences (HiLIFE), 00014 University of Helsinki, Helsinki, Finland
| | - Manuel Patarroyo
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 11 Solna, Sweden
| | - Pekka Katajisto
- Institute of Biotechnology, Helsinki Institute of Life Sciences (HiLIFE), 00014 University of Helsinki, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden.,Faculty of Biological and Environmental Sciences, 00014 University of Helsinki, Helsinki, Finland.,Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Solna, Sweden
| |
Collapse
|
43
|
Alsheikh HAM, Metge BJ, Pruitt HC, Kammerud SC, Chen D, Wei S, Shevde LA, Samant RS. Disruption of STAT5A and NMI signaling axis leads to ISG20-driven metastatic mammary tumors. Oncogenesis 2021; 10:45. [PMID: 34078871 PMCID: PMC8172570 DOI: 10.1038/s41389-021-00333-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/23/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Molecular dynamics of developmental processes are repurposed by cancer cells to support cancer initiation and progression. Disruption of the delicate balance between cellular differentiation and plasticity during mammary development leads to breast cancer initiation and metastatic progression. STAT5A is essential for differentiation of secretory mammary alveolar epithelium. Active STAT5A characterizes breast cancer patients for favorable prognosis. N-Myc and STAT Interactor protein (NMI) was initially discovered as a protein that interacts with various STATs; however, the relevance of these interactions to normal mammary development and cancer was not known. We observe that NMI protein is expressed in the mammary ductal epithelium at the onset of puberty and is induced in pregnancy. NMI protein is decreased in 70% of patient specimens with metastatic breast cancer compared to primary tumors. Here we present our finding that NMI and STAT5A cooperatively mediate normal mammary development. Loss of NMI in vivo caused a decrease in STAT5A activity in normal mammary epithelial as well as breast cancer cells. Analysis of STAT5A mammary specific controlled genetic program in the context of NMI knockout revealed ISG20 (interferon stimulated exonuclease gene 20, a protein involved in rRNA biogenesis) as an unfailing negatively regulated target. Role of ISG20 has never been described in metastatic process of mammary tumors. We observed that overexpression of ISG20 is increased in metastases compared to matched primary breast tumor tissues. Our observations reveal that NMI-STAT5A mediated signaling keeps a check on ISG20 expression via miR-17–92 cluster. We show that uncontrolled ISG20 expression drives tumor progression and metastasis.
Collapse
Affiliation(s)
| | - Brandon J Metge
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hawley C Pruitt
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sarah C Kammerud
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dongquan Chen
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shi Wei
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rajeev S Samant
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA. .,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA. .,Birmingham VA Medical Center, Birmingham, AL, USA.
| |
Collapse
|
44
|
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
|
45
|
The risk of developing a meningioma during and after pregnancy. Sci Rep 2021; 11:9153. [PMID: 33911184 PMCID: PMC8080659 DOI: 10.1038/s41598-021-88742-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/09/2021] [Indexed: 01/01/2023] Open
Abstract
Pregnancy has been associated with diagnosis or growth of meningiomas in several case reports, which has led to the hypothesis that pregnancy may be a risk factor for meningiomas. The aim of this study was to test this hypothesis in a large population-based cohort study. Women born in Sweden 1958–2000 (N = 2,204,126) were identified and matched with the Medical Birth Register and the Cancer Register. The expected number of meningioma cases and risk ratios were calculated for parous and nulliparous women and compared to the observed number of cases. Compared to parous women, meningiomas were more common among nulliparous (SIR = 1.73; 95% CI 1.52–1.95). The number of meningioma cases detected during pregnancy was lower than the expected (SIR = 0.40; 95% CI 0.20–0.72). Moreover, no increased risk was found in the first-year post-partum (SIR = 1.04; 95% CI 0.74–1.41). Contrary to our hypothesis, there was no increased risk for diagnosing a meningioma during pregnancy or 1-year post-partum. A lower detection rate during pregnancy, may reflect under-utilization of diagnostic procedures, but the actual number of meningiomas was homogenously lower among parous than nulliparous women throughout the study period, indicating that pregnancy is not a risk factor for meningioma.
Collapse
|
46
|
Ambhore NS, Kalidhindi RSR, Sathish V. Sex-Steroid Signaling in Lung Diseases and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:243-273. [PMID: 33788197 DOI: 10.1007/978-3-030-63046-1_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sex/gender difference exists in the physiology of multiple organs. Recent epidemiological reports suggest the influence of sex-steroids in modulating a wide variety of disease conditions. Sex-based discrepancies have been reported in pulmonary physiology and various chronic inflammatory responses associated with lung diseases like asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, and rare lung diseases. Notably, emerging clinical evidence suggests that several respiratory diseases affect women to a greater degree, with increased severity and prevalence than men. Although sex-specific differences in various lung diseases are evident, such differences are inherent to sex-steroids, which are major biological variables in men and women who play a central role to control these differences. The focus of this chapter is to comprehend the sex-steroid biology in inflammatory lung diseases and to understand the mechanistic role of sex-steroids signaling in regulating these diseases. Exploring the roles of sex-steroid signaling in the regulation of lung diseases and inflammation is crucial for the development of novel and effective therapy. Overall, we will illustrate the importance of differential sex-steroid signaling in lung diseases and their possible clinical implications for the development of complementary and alternative medicine to treat lung diseases.
Collapse
Affiliation(s)
- Nilesh Sudhakar Ambhore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | | | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
47
|
Dawson CA, Visvader JE. The Cellular Organization of the Mammary Gland: Insights From Microscopy. J Mammary Gland Biol Neoplasia 2021; 26:71-85. [PMID: 33835387 DOI: 10.1007/s10911-021-09483-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/25/2021] [Indexed: 12/19/2022] Open
Abstract
Despite rapid advances in our knowledge of the cellular heterogeneity and molecular regulation of the mammary gland, how these relate to 3D cellular organization remains unclear. In addition to hormonal regulation, mammary gland development and function is directed by para- and juxtacrine signaling among diverse cell-types, particularly the immune and mesenchymal populations. Precise mapping of the cellular landscape of the breast will help to decipher this complex coordination. Imaging of thin tissue sections has provided foundational information about cell positioning in the mammary gland and now technological advances in tissue clearing and subcellular-resolution 3D imaging are painting a more complete picture. In particular, confocal, light-sheet and multiphoton microscopy applied to intact tissue can fully capture cell morphology, position and interactions, and have the power to identify spatially rare events. This review will summarize our current understanding of mammary gland cellular organization as revealed by microscopy. We focus on the mouse mammary gland and cover a broad range of immune and stromal cell types at major developmental stages and give insights into important tissue niches and cellular interactions.
Collapse
Affiliation(s)
- Caleb A Dawson
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, 3052, Parkville, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, 3010, Parkville, VIC, Australia.
| | - Jane E Visvader
- Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, 3052, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, 3010, Parkville, VIC, Australia
| |
Collapse
|
48
|
Kim O, Park EY, Kwon SY, Shin S, Emerson RE, Shin YH, DeMayo FJ, Lydon JP, Coffey DM, Hawkins SM, Quilliam LA, Cheon DJ, Fernández FM, Nephew KP, Karpf AR, Widschwendter M, Sood AK, Bast RC, Godwin AK, Miller KD, Cho CH, Kim J. Targeting progesterone signaling prevents metastatic ovarian cancer. Proc Natl Acad Sci U S A 2020; 117:31993-32004. [PMID: 33262282 PMCID: PMC7749341 DOI: 10.1073/pnas.2013595117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Effective cancer prevention requires the discovery and intervention of a factor critical to cancer development. Here we show that ovarian progesterone is a crucial endogenous factor inducing the development of primary tumors progressing to metastatic ovarian cancer in a mouse model of high-grade serous carcinoma (HGSC), the most common and deadliest ovarian cancer type. Blocking progesterone signaling by the pharmacologic inhibitor mifepristone or by genetic deletion of the progesterone receptor (PR) effectively suppressed HGSC development and its peritoneal metastases. Strikingly, mifepristone treatment profoundly improved mouse survival (∼18 human years). Hence, targeting progesterone/PR signaling could offer an effective chemopreventive strategy, particularly in high-risk populations of women carrying a deleterious mutation in the BRCA gene.
Collapse
MESH Headings
- Adult
- Animals
- BRCA1 Protein/genetics
- Breast/pathology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/prevention & control
- Cystadenocarcinoma, Serous/chemistry
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/prevention & control
- Disease Models, Animal
- Estradiol/administration & dosage
- Female
- Humans
- Mice
- Middle Aged
- Mifepristone/pharmacology
- Mifepristone/therapeutic use
- Mutation
- Neoplasms, Experimental/chemically induced
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/prevention & control
- Ovarian Neoplasms/chemically induced
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/prevention & control
- Ovary/pathology
- Ovary/surgery
- Progesterone/administration & dosage
- Progesterone/antagonists & inhibitors
- Progesterone/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Salpingo-oophorectomy
- Signal Transduction/drug effects
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Olga Kim
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Eun Young Park
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Sun Young Kwon
- Department of Pathology, School of Medicine, Keimyung University, 41931 Daegu, Republic of Korea
| | - Sojin Shin
- Department of Obstetrics and Gynecology, School of Medicine, Keimyung University, 41931 Daegu, Republic of Korea
| | - Robert E Emerson
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Yong-Hyun Shin
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709
| | - John P Lydon
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030
| | - Donna M Coffey
- Department of Pathology and Genomic Medicine, Houston Methodist and Weill Cornell Medical College, Houston, TX 77030
| | - Shannon M Hawkins
- Department of Obstetrics and Gynecology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Lawrence A Quilliam
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Dong-Joo Cheon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208
| | - Facundo M Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332
| | - Kenneth P Nephew
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405
| | - Adam R Karpf
- Eppley Institute for Cancer Research, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Martin Widschwendter
- Department of Women's Cancer, Institute for Women's Health, University College London, WC1E 6AU London, United Kingdom
- Research Institute for Biomedical Aging Research, Universität Innsbruck, 6020 Innsbruck, Austria
- European Translational Oncology Prevention and Screening (EUTOPS) Institute, Universität Innsbruck, 6060 Hall in Tirol, Austria
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
- Department of Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Robert C Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Kathy D Miller
- Department of Medicine, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Chi-Heum Cho
- Department of Obstetrics and Gynecology, School of Medicine, Keimyung University, 41931 Daegu, Republic of Korea;
| | - Jaeyeon Kim
- Department of Biochemistry and Molecular Biology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202;
| |
Collapse
|
49
|
Watson CJ, Khaled WT. Mammary development in the embryo and adult: new insights into the journey of morphogenesis and commitment. Development 2020; 147:dev169862. [PMID: 33191272 DOI: 10.1242/dev.169862] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The mammary gland is a unique tissue and the defining feature of the class Mammalia. It is a late-evolving epidermal appendage that has the primary function of providing nutrition for the young, although recent studies have highlighted additional benefits of milk including the provision of passive immunity and a microbiome and, in humans, the psychosocial benefits of breastfeeding. In this Review, we outline the various stages of mammary gland development in the mouse, with a particular focus on lineage specification and the new insights that have been gained by the application of recent technological advances in imaging in both real-time and three-dimensions, and in single cell RNA sequencing. These studies have revealed the complexity of subpopulations of cells that contribute to the mammary stem and progenitor cell hierarchy and we suggest a new terminology to distinguish these cells.
Collapse
Affiliation(s)
- Christine J Watson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Walid T Khaled
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| |
Collapse
|
50
|
Kim G, Lee JG, Cheong SA, Yon JM, Lee MS, Hong EJ, Baek IJ. Progesterone receptor membrane component 1 is required for mammary gland development†. Biol Reprod 2020; 103:1249-1259. [PMID: 32915211 DOI: 10.1093/biolre/ioaa164] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/03/2020] [Accepted: 09/10/2020] [Indexed: 12/18/2022] Open
Abstract
The physiological functions of progesterone (P4) in female reproductive organs including the mammary glands are mediated via the progesterone receptor (PR), but not all P4 functions can be explained by PR-mediated signaling. Progesterone receptor membrane component 1 (PGRMC1), a potential mediator of P4 actions, plays an important role in the ovary and uterus in maintaining female fertility and pregnancy, but its function in mammary glands has not been elucidated. This study investigated the role of PGRMC1 in mouse mammary gland development. Unlike in the uterus, exogenous estrogen (E2) and/or P4 did not alter PGRMC1 expression in the mammary gland, and Pgrmc1-knockout (KO) mice displayed reduced ductal elongation and side branching in response to hormone treatment. During pregnancy, PGRMC1 was expressed within both the luminal and basal epithelium and gradually increased with gestation and decreased rapidly after parturition. Moreover, although lactogenic capacity was normal after parturition, Pgrmc1 KO resulted in defective mammary gland development from puberty until midpregnancy, while the expression of PR and its target genes was not significantly different between wild-type and Pgrmc1-KO mammary gland. These data suggest that PGRMC1 is essential for mammary gland development during puberty and pregnancy in a PR-independent manner.
Collapse
Affiliation(s)
- Globinna Kim
- ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Seoul, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jong Geol Lee
- ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Seung-A Cheong
- ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Jung-Min Yon
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Myeong Sup Lee
- Asan Medical Institute of Convergence Science and Technology (AMIST), Seoul, Republic of Korea.,Department of Biomedical Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - In-Jeoung Baek
- ConveRgence mEDIcine research cenTer (CREDIT), Asan Institute for Life Sciences, Seoul, Republic of Korea.,Asan Medical Institute of Convergence Science and Technology (AMIST), Seoul, Republic of Korea.,Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|