51
|
Classical and Non-Classical Progesterone Signaling in Breast Cancers. Cancers (Basel) 2020; 12:cancers12092440. [PMID: 32867363 PMCID: PMC7563480 DOI: 10.3390/cancers12092440] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/11/2020] [Accepted: 08/24/2020] [Indexed: 12/24/2022] Open
Abstract
Much emphasis is placed on estrogen (E2) and estrogen receptor (ER) signaling as most research is focused on understanding E2 and ER’s ability to enhance proliferative signals in breast cancers. Progesterone (P4) is important for normal mammary gland development, function and menstrual control. However, P4 and its receptors (PRs) in breast cancer etiology continue to be understudied and its role in breast cancer remains controversial. The Women’s Health Initiative (WHI) clinical trial clearly demonstrated the importance of progestogens in breast cancer development. P4 has historically been associated with classical-signaling through nuclear receptors, however non-classical P4 signaling via membrane receptors has been described. Progestogens have the ability to bind to nuclear and membrane receptors and studies have demonstrated that both can promote breast cancer cell proliferation and breast tumor growth. In this review, we attempt to understand the classical and non-classical signaling role of P4 in breast cancers because both nuclear and membrane receptors could become viable therapeutic options for breast cancer patients.
Collapse
|
52
|
Avagliano A, Fiume G, Ruocco MR, Martucci N, Vecchio E, Insabato L, Russo D, Accurso A, Masone S, Montagnani S, Arcucci A. Influence of Fibroblasts on Mammary Gland Development, Breast Cancer Microenvironment Remodeling, and Cancer Cell Dissemination. Cancers (Basel) 2020; 12:E1697. [PMID: 32604738 PMCID: PMC7352995 DOI: 10.3390/cancers12061697] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
The stromal microenvironment regulates mammary gland development and tumorigenesis. In normal mammary glands, the stromal microenvironment encompasses the ducts and contains fibroblasts, the main regulators of branching morphogenesis. Understanding the way fibroblast signaling pathways regulate mammary gland development may offer insights into the mechanisms of breast cancer (BC) biology. In fact, the unregulated mammary fibroblast signaling pathways, associated with alterations in extracellular matrix (ECM) remodeling and branching morphogenesis, drive breast cancer microenvironment (BCM) remodeling and cancer growth. The BCM comprises a very heterogeneous tissue containing non-cancer stromal cells, namely, breast cancer-associated fibroblasts (BCAFs), which represent most of the tumor mass. Moreover, the different components of the BCM highly interact with cancer cells, thereby generating a tightly intertwined network. In particular, BC cells activate recruited normal fibroblasts in BCAFs, which, in turn, promote BCM remodeling and metastasis. Thus, comparing the roles of normal fibroblasts and BCAFs in the physiological and metastatic processes, could provide a deeper understanding of the signaling pathways regulating BC dissemination. Here, we review the latest literature describing the structure of the mammary gland and the BCM and summarize the influence of epithelial-mesenchymal transition (EpMT) and autophagy in BC dissemination. Finally, we discuss the roles of fibroblasts and BCAFs in mammary gland development and BCM remodeling, respectively.
Collapse
Affiliation(s)
- Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.F.); (E.V.)
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy;
| | - Nunzia Martucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Eleonora Vecchio
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.F.); (E.V.)
| | - Luigi Insabato
- Anatomic Pathology Unit, Department of Advanced Biomedical Sciences, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (D.R.)
| | - Daniela Russo
- Anatomic Pathology Unit, Department of Advanced Biomedical Sciences, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (D.R.)
| | - Antonello Accurso
- Department of General, Oncological, Bariatric and Endocrine-Metabolic Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Stefania Montagnani
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| |
Collapse
|
53
|
Asavasupreechar T, Saito R, Miki Y, Edwards DP, Boonyaratanakornkit V, Sasano H. Systemic distribution of progesterone receptor subtypes in human tissues. J Steroid Biochem Mol Biol 2020; 199:105599. [PMID: 31991170 PMCID: PMC9968951 DOI: 10.1016/j.jsbmb.2020.105599] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 11/25/2022]
Abstract
Progesterone receptor (PR) is expressed in a wide variety of human tissues, including both reproductive and non-reproductive tissues. Upon binding to the PR, progesterone can display several non-reproductive functions, including neurosteroid activity in the central nervous system, inhibition of smooth muscle contractile activity in the gastrointestinal tract, and regulating the development and maturation of the lung. PR exists as two major isoforms, PRA and PRB. Differential expression of these PR isoforms reportedly contributes to different biological activities of the hormone. However, the distribution of the PR isoforms in human tissues has remained virtually unexplored. In this study, we immunolocalized PR expression in various human tissues using PR (1294) specific antibody, which is capable of detecting both PRA and PRB, and PRB (250H11) specific antibody. Tissues from the uterus, ovary, breast, placenta, prostate, testis, cerebrum, cerebellum, pituitary, spinal cord, esophagus, stomach, small intestine, colon, pancreas, liver, kidney, urinary bladder, lung, heart, aorta, thymus, adrenal gland, thyroid, spleen, skin, and bone were examined in four different age groups (fetal, pediatric, young, and old) in male and female subjects. PR and PRB were detected in the nuclei of cells in the female reproductive system, in both the nuclei and cytoplasm of pituitary gland and pancreatic acinar cells, and only in the cytoplasm of cells in the testis, stomach, small intestine, colon, liver, kidney, urinary bladder, lung, adrenal gland, and skin. Of particular interest, total PRB expression overlapped with that of total PR expression in most tissues but was negative in the female fetal reproductive system. The findings indicate that progesterone could affect diverse human organs differently than from reproductive organs. These findings provide new insights into the novel biological roles of progesterone in non-reproductive organs.
Collapse
Affiliation(s)
| | - Ryoko Saito
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Miki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Dean P Edwards
- Departments of Molecular & Cellular Biology and Pathology & Immunology, Baylor College of Medicine, Houston, USA
| | - Viroj Boonyaratanakornkit
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand; Age-related Inflammation and Degeneration Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
54
|
Lyons KE, Ryan CA, Dempsey EM, Ross RP, Stanton C. Breast Milk, a Source of Beneficial Microbes and Associated Benefits for Infant Health. Nutrients 2020; 12:E1039. [PMID: 32283875 PMCID: PMC7231147 DOI: 10.3390/nu12041039] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 12/27/2022] Open
Abstract
Human breast milk is considered the optimum feeding regime for newborn infants due to its ability to provide complete nutrition and many bioactive health factors. Breast feeding is associated with improved infant health and immune development, less incidences of gastrointestinal disease and lower mortality rates than formula fed infants. As well as providing fundamental nutrients to the growing infant, breast milk is a source of commensal bacteria which further enhance infant health by preventing pathogen adhesion and promoting gut colonisation of beneficial microbes. While breast milk was initially considered a sterile fluid and microbes isolated were considered contaminants, it is now widely accepted that breast milk is home to its own unique microbiome. The origins of bacteria in breast milk have been subject to much debate, however, the possibility of an entero-mammary pathway allowing for transfer of microbes from maternal gut to the mammary gland is one potential pathway. Human milk derived strains can be regarded as potential probiotics; therefore, many studies have focused on isolating strains from milk for subsequent use in infant health and nutrition markets. This review aims to discuss mammary gland development in preparation for lactation as well as explore the microbial composition and origins of the human milk microbiota with a focus on probiotic development.
Collapse
Affiliation(s)
- Katríona E. Lyons
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - C. Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork T12 YE02, Ireland
| | - Eugene M. Dempsey
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork T12 YE02, Ireland
- INFANT Research Centre, University College Cork, Cork T12 DFK4, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
55
|
Trabert B, Sherman ME, Kannan N, Stanczyk FZ. Progesterone and Breast Cancer. Endocr Rev 2020; 41:5568276. [PMID: 31512725 PMCID: PMC7156851 DOI: 10.1210/endrev/bnz001] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 09/06/2019] [Indexed: 12/31/2022]
Abstract
Synthetic progestogens (progestins) have been linked to increased breast cancer risk; however, the role of endogenous progesterone in breast physiology and carcinogenesis is less clearly defined. Mechanistic studies using cell culture, tissue culture, and preclinical models implicate progesterone in breast carcinogenesis. In contrast, limited epidemiologic data generally do not show an association of circulating progesterone levels with risk, and it is unclear whether this reflects methodologic limitations or a truly null relationship. Challenges related to defining the role of progesterone in breast physiology and neoplasia include: complex interactions with estrogens and other hormones (eg, androgens, prolactin, etc.), accounting for timing of blood collections for hormone measurements among cycling women, and limitations of assays to measure progesterone metabolites in blood and progesterone receptor isotypes (PRs) in tissues. Separating the individual effects of estrogens and progesterone is further complicated by the partial dependence of PR transcription on estrogen receptor (ER)α-mediated transcriptional events; indeed, interpreting the integrated interaction of the hormones may be more essential than isolating independent effects. Further, many of the actions of both estrogens and progesterone, particularly in "normal" breast tissues, are driven by paracrine mechanisms in which ligand binding to receptor-positive cells evokes secretion of factors that influence cell division of neighboring receptor-negative cells. Accordingly, blood and tissue levels may differ, and the latter are challenging to measure. Given conflicting data related to the potential role of progesterone in breast cancer etiology and interest in blocking progesterone action to prevent or treat breast cancer, we provide a review of the evidence that links progesterone to breast cancer risk and suggest future directions for filling current gaps in our knowledge.
Collapse
Affiliation(s)
- Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Mark E Sherman
- Health Sciences Research, Mayo Clinic, Jacksonville, Florida
| | - Nagarajan Kannan
- Laboratory of Stem Cell and Cancer Biology, Division of Experimental Pathology and Laboratory Medicine, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Frank Z Stanczyk
- Departments of Obstetrics and Gynecology, and Preventive Medicine, University of Southern California Keck School of Medicine, Los Angeles, California
| |
Collapse
|
56
|
Ataca D, Aouad P, Constantin C, Laszlo C, Beleut M, Shamseddin M, Rajaram RD, Jeitziner R, Mead TJ, Caikovski M, Bucher P, Ambrosini G, Apte SS, Brisken C. The secreted protease Adamts18 links hormone action to activation of the mammary stem cell niche. Nat Commun 2020; 11:1571. [PMID: 32218432 PMCID: PMC7099066 DOI: 10.1038/s41467-020-15357-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 02/28/2020] [Indexed: 11/21/2022] Open
Abstract
Estrogens and progesterone control breast development and carcinogenesis via their cognate receptors expressed in a subset of luminal cells in the mammary epithelium. How they control the extracellular matrix, important to breast physiology and tumorigenesis, remains unclear. Here we report that both hormones induce the secreted protease Adamts18 in myoepithelial cells by controlling Wnt4 expression with consequent paracrine canonical Wnt signaling activation. Adamts18 is required for stem cell activation, has multiple binding partners in the basement membrane and interacts genetically with the basal membrane-specific proteoglycan, Col18a1, pointing to the basement membrane as part of the stem cell niche. In vitro, ADAMTS18 cleaves fibronectin; in vivo, Adamts18 deletion causes increased collagen deposition during puberty, which results in impaired Hippo signaling and reduced Fgfr2 expression both of which control stem cell function. Thus, Adamts18 links luminal hormone receptor signaling to basement membrane remodeling and stem cell activation. How hormonal signaling in the mammary epithelium controls the surrounding extracellular matrix is unclear. Here, the authors show that a secreted protease, Adamts18, induced by upstream estrogen-progesterone activated Wnt4 in myoepithelial cells, remodels the basement membrane and contributes to mammary epithelial stemness.
Collapse
Affiliation(s)
- Dalya Ataca
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland
| | - Patrick Aouad
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland
| | - Céline Constantin
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland
| | - Csaba Laszlo
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland
| | - Manfred Beleut
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland.,Medoderm GmbH, Robert Koch-Straße 50 D, 55129, Mainz, Germany
| | - Marie Shamseddin
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland.,Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Renuga Devi Rajaram
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland
| | - Rachel Jeitziner
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Agora Swiss Cancer Center Leman, Rue du Bugnon 25a, 1015, Lausanne, Switzerland
| | - Timothy J Mead
- Department of Biomedical Engineering-ND20, Cleveland Clinic Lerner Research Institute, 9500 Euclid Ave., Cleveland, OH, 44195, USA
| | - Marian Caikovski
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Agora Swiss Cancer Center Leman, Rue du Bugnon 25a, 1015, Lausanne, Switzerland
| | - Philipp Bucher
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland
| | - Giovanna Ambrosini
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland
| | - Suneel S Apte
- Department of Biomedical Engineering-ND20, Cleveland Clinic Lerner Research Institute, 9500 Euclid Ave., Cleveland, OH, 44195, USA
| | - Cathrin Brisken
- Ecole Polytechnique Fédérale de Lausanne, Station 19, CH-1015, Lausanne, Switzerland.
| |
Collapse
|
57
|
Gagniac L, Rusidzé M, Boudou F, Cagnet S, Adlanmerini M, Jeannot P, Gaide N, Giton F, Besson A, Weyl A, Gourdy P, Raymond-Letron I, Arnal JF, Brisken C, Lenfant F. Membrane expression of the estrogen receptor ERα is required for intercellular communications in the mammary epithelium. Development 2020; 147:dev.182303. [PMID: 32098763 PMCID: PMC7075076 DOI: 10.1242/dev.182303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 02/06/2020] [Indexed: 01/23/2023]
Abstract
17β-Estradiol induces the postnatal development of mammary gland and influences breast carcinogenesis by binding to the estrogen receptor ERα. ERα acts as a transcription factor but also elicits rapid signaling through a fraction of ERα expressed at the membrane. Here, we have used the C451A-ERα mouse model mutated for the palmitoylation site to understand how ERα membrane signaling affects mammary gland development. Although the overall structure of physiological mammary gland development is slightly affected, both epithelial fragments and basal cells isolated from C451A-ERα mammary glands failed to grow when engrafted into cleared wild-type fat pads, even in pregnant hosts. Similarly, basal cells purified from hormone-stimulated ovariectomized C451A-ERα mice did not produce normal outgrowths. Ex vivo, C451A-ERα basal cells displayed reduced matrix degradation capacities, suggesting altered migration properties. More importantly, C451A-ERα basal cells recovered in vivo repopulating ability when co-transplanted with wild-type luminal cells and specifically with ERα-positive luminal cells. Transcriptional profiling identified crucial paracrine luminal-to-basal signals. Altogether, our findings uncover an important role for membrane ERα expression in promoting intercellular communications that are essential for mammary gland development.
Collapse
Affiliation(s)
- Laurine Gagniac
- INSERM U1048, I2MC, Université de Toulouse, Toulouse 31432, France
| | - Mariam Rusidzé
- INSERM U1048, I2MC, Université de Toulouse, Toulouse 31432, France
| | - Frederic Boudou
- INSERM U1048, I2MC, Université de Toulouse, Toulouse 31432, France
| | - Stephanie Cagnet
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | | - Pauline Jeannot
- LBCMCP, Centre de Biologie Intégrative, Université de Toulouse, CNRS, Toulouse 31062, France
| | - Nicolas Gaide
- LabHPEC Laboratoire d'HistoPathologie Expérimentale et Comparée STROMALab, Université de Toulouse, CNRS ERL5311, EFS, ENVT, Inserm U1031, UPS, Toulouse 31300, France
| | - Frank Giton
- APHP H.Mondor- IMRB - INSERM U955, Créteil 94010, France
| | - Arnaud Besson
- LBCMCP, Centre de Biologie Intégrative, Université de Toulouse, CNRS, Toulouse 31062, France
| | - Ariane Weyl
- INSERM U1048, I2MC, Université de Toulouse, Toulouse 31432, France
| | - Pierre Gourdy
- INSERM U1048, I2MC, Université de Toulouse, Toulouse 31432, France
| | - Isabelle Raymond-Letron
- LabHPEC Laboratoire d'HistoPathologie Expérimentale et Comparée STROMALab, Université de Toulouse, CNRS ERL5311, EFS, ENVT, Inserm U1031, UPS, Toulouse 31300, France
| | | | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | |
Collapse
|
58
|
Zhu H, Jia X, Ren M, Yang L, Chen J, Han L, Ding Y, Ding M. Mifepristone Treatment in Pregnant Murine Model Induced Mammary Gland Dysplasia and Postpartum Hypogalactia. Front Cell Dev Biol 2020; 8:102. [PMID: 32154252 PMCID: PMC7047202 DOI: 10.3389/fcell.2020.00102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 02/07/2020] [Indexed: 12/31/2022] Open
Abstract
Mammary gland dysplasia and postpartum hypogalactia often occur in humans and in the livestock breeding industry. However, their underlying mechanisms are not clear yet. Mifepristone, which has a high affinity for progesterone (P4) and glucocorticoid receptors, was exploited here to induce the disorders of mammary gland development and lactation. Four strategies were devised for treating pregnant mice with mifepristone. In the first strategy, mice were administered 1.20 mg mifepristone/kg body weight (BW) on pregnancy day 4 (Pd4). In the second strategy, mifepristone was administered to mice twice, with 1.20 mg/kg BW on Pd4 and 0.40 mg/kg BW on Pd8. In the third strategy, mice were treated with a single dose of 0.40 mg mifepristone/kg BW on Pd8. In the fourth strategy, mice were administered 0.40 mg mifepristone/kg BW on Pd8 and 0.20 mg mifepristone/kg BW on Pd12. The results suggested that mifepristone administration at the dose of 1.20 mg/kg BW on Pd4 caused significant reduction in milk production on lactation day 1 (Ld1), Ld2, and Ld3, as assessed using a weigh-suckle-weigh assay. Mammary β-casein expression, milk yields, litter growth rates, gland structure, and serum concentrations of 17-β estrogen (E2), P4, prolactin (PRL), growth hormone (GH), corticosterone (CORT) and oxytocin (OT) as well as the receptors of these hormones were determined during pregnancy or lactation after performing the first (Pd4) strategy. The results demonstrated that mifepristone administration during early pregnancy decreased β-casein expression, milk yields and litter growth rates, induced fewer alveoli, enlarged alveolar lumina, and altered the levels of E2, P4, PRL, GH, CORT, and OT as well as the mRNA expression of these hormonal receptors during pregnancy or early lactation. The present study on pregnant mice treated with mifepristone offers an innovative murine model to study the mechanism underlying mammary gland dysplasia and postpartum hypogalactia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mingxing Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
59
|
Cai C, Geng A, Wang M, Yang L, Yu QC, Zeng YA. Amphiregulin mediates the hormonal regulation on Rspondin-1 expression in the mammary gland. Dev Biol 2020; 458:43-51. [DOI: 10.1016/j.ydbio.2019.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 09/23/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023]
|
60
|
Sundaram S, Johnson LK, Yan L. High-Fat Diet Alters Circadian Rhythms in Mammary Glands of Pubertal Mice. Front Endocrinol (Lausanne) 2020; 11:349. [PMID: 32625167 PMCID: PMC7314922 DOI: 10.3389/fendo.2020.00349] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/04/2020] [Indexed: 12/17/2022] Open
Abstract
Childhood obesity in girls is associated with early puberty and menarche. Breast tissue exhibits circadian rhythms. These rhythms may be altered by environmental factors. We hypothesized that a high-fat diet (HFD) disrupts circadian rhythms in pubertal mammary glands. Weanling female C57BL/6 mice were fed the standard AIN93G diet or a HFD (providing 16% or 45% of energy from soybean oil) for 3 weeks. Mammary glands were harvested from 6-week-old mice every 4 h on Zeitgeber time over a 48-h period; rhythmic expressions of circadian genes and genes encoding estrogen receptor and progesterone receptor were analyzed by using the Cosinor model. HFD, compared to AIN93G diet, altered diurnal oscillations of circadian genes in pubertal mammary glands. These included changes in amplitude of Per2, Cry1 (reduced), Clock, Rev-erbα, and Per1 (elevated), a delay in acrophase (the hour at which the rhythm peaks) of Bmal1 by 2.2 h, and changes in mesor (the mean of the rhythm from peak to trough) of Bmal1, Per2, Cry1 (reduced), Rev-rebα, and Per1 (elevated). Furthermore, HFD altered diurnal expression of estrogen receptor and progesterone receptor at both mRNA and protein levels. These findings indicate that HFD alters circadian regulation in pubertal mammary glands, which may contribute to the disturbance of hormonal homeostasis and lead to early development and growth of mammary glands in pubertal mice.
Collapse
|
61
|
Abramson L, Massaro L, Alberty-Oller JJ, Melsaether A. Breast Imaging During Pregnancy and Lactation. JOURNAL OF BREAST IMAGING 2019; 1:342-351. [PMID: 38424810 DOI: 10.1093/jbi/wbz065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/18/2019] [Indexed: 03/02/2024]
Abstract
Breast imaging during pregnancy and lactation is important in order to avoid delays in the diagnosis and treatment of pregnancy-associated breast cancers. Radiologists have an opportunity to improve breast cancer detection by becoming familiar with appropriate breast imaging and providing recommendations to women and their referring physicians. Importantly, during pregnancy and lactation, both screening and diagnostic breast imaging can be safely performed. Here we describe when and how to screen, how to work up palpable masses, and evaluate bloody nipple discharge. The imaging features of common findings in the breasts of pregnant and lactating women are also reviewed. Finally, we address breast cancer staging and provide a brief primer on treatment options for pregnancy-associated breast cancers.
Collapse
Affiliation(s)
- Lisa Abramson
- Icahn School of Medicine at Mount Sinai, Department of Radiology, New York, NY
| | - Lindsey Massaro
- Icahn School of Medicine at Mount Sinai, Department of Radiology, New York, NY
| | | | - Amy Melsaether
- Icahn School of Medicine at Mount Sinai, Department of Radiology, New York, NY
| |
Collapse
|
62
|
Cenciarini ME, Proietti CJ. Molecular mechanisms underlying progesterone receptor action in breast cancer: Insights into cell proliferation and stem cell regulation. Steroids 2019; 152:108503. [PMID: 31562879 DOI: 10.1016/j.steroids.2019.108503] [Citation(s) in RCA: 18] [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: 06/26/2019] [Revised: 09/13/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
The ovarian steroid hormone progesterone and its nuclear receptor, the Progesterone Receptor (PR), play an essential role in the regulation of cell proliferation and differentiation in the mammary gland. In addition, experimental and clinical evidence demonstrate their critical role in controlling mammary gland tumorigenesis and breast cancer development. When bound to its ligand, the main action of PR is as a transcription factor, which regulates the expression of target genes networks. PR also activates signal transduction pathways through a rapid or non-genomic mechanism in breast cancer cells, an event that is fully integrated with its genomic effects. This review summarizes the molecular mechanisms of the ligand-activated PR actions that drive epithelial cell proliferation and the regulation of the stem cell population in the normal breast and in breast cancer.
Collapse
Affiliation(s)
- Mauro E Cenciarini
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina
| | - Cecilia J Proietti
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina.
| |
Collapse
|
63
|
Frank-Kamenetskii A, Booth BW. Redirecting Normal and Cancer Stem Cells to a Mammary Epithelial Cell Fate. J Mammary Gland Biol Neoplasia 2019; 24:285-292. [PMID: 31732837 DOI: 10.1007/s10911-019-09439-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 10/31/2019] [Indexed: 12/21/2022] Open
Abstract
Tissue microenvironments, also known as stem cell niches, influence not only resident cells but also cells in surrounding tissues. Physical and biochemical intercellular signals originating from resident stem cells or non-stem cells participate in the homeostasis of the tissue regulating cell proliferation, differentiation, wound healing, tissue remodeling, and tumorigenesis. In recent publications it has been demonstrated that the normal mouse mammary microenvironment can provide development and differentiation guidance to not only resident mammary cells but also cells of non-mammary origin including tumor-derived cells. When placed in reforming mammary stem cell niches the non-mammary cells proliferate and differentiate along mammary epithelial cell lineages and contribute progeny to reforming mammary gland outgrowths. The tumor-derived cells that are redirected to assume mammary epithelial phenotypes lose their cancer-forming capacity and shift their gene expression profiles from a cancer profile towards a normal mammary epithelial expression profile. This review summarizes the recent discoveries regarding the ability of the normal mouse mammary microenvironment to dictate the cell fates of non-mammary cells introduced into mammary stem cell niches.
Collapse
Affiliation(s)
- Anastasia Frank-Kamenetskii
- Department of Bioengineering, Clemson University, 401-1 Rhodes Engineering Research Center, Clemson, SC, 29634, USA
| | - Brian W Booth
- Department of Bioengineering, Clemson University, 401-1 Rhodes Engineering Research Center, Clemson, SC, 29634, USA.
| |
Collapse
|
64
|
Ito S. Opioids in Breast Milk: Pharmacokinetic Principles and Clinical Implications. J Clin Pharmacol 2019; 58 Suppl 10:S151-S163. [PMID: 30248201 DOI: 10.1002/jcph.1113] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/01/2018] [Indexed: 12/14/2022]
Abstract
Safety of maternal drug therapy during breastfeeding may be assessed from estimated levels of drug exposure of the infant through milk. Pharmacokinetic (PK) principles predict that the lower the clearance is, the higher the infant dose via milk will be. Drugs with low clearance (<1 mL/[kg·min]) are likely to cause an infant exposure level greater than 10% of the weight-adjusted maternal dose even if the milk-to-plasma concentration ratio is 1. Most drugs cause relatively low-level exposure below 10% of the weight-adjusted maternal dose, but opioids require caution because of their potential for severe adverse effects. Furthermore, substantial individual variations of drug clearance exist in both mother and infant, potentially causing drug accumulation over time in some infants even if an estimated dose of the drug through milk is small. Such PK differences among individuals are known not only for codeine and tramadol through pharmacogenetic variants of CYP2D6 but also for non-CYP2D6 substrate opioids including oxycodone, indicating difficulties of eliminating PK uncertainty by simply replacing an opioid with another. Overall, opioid use for pain management during labor and delivery and subsequent short-term use for 2-3 days are compatible with breastfeeding. In contrast, newly initiated and prolonged maternal opioid therapy must follow a close monitoring program of the opioid-naive infants. Until more safety data become available, treatment duration of newly initiated opioids in the postpartum period should be limited to 2-3 days in unsupervised outpatient settings. Opioid addiction treatment with methadone and buprenorphine during pregnancy may continue into breastfeeding, but infant conditions must be monitored.
Collapse
Affiliation(s)
- Shinya Ito
- Division of Clinical Pharmacology and Toxicology, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
65
|
Transporters in the Mammary Gland-Contribution to Presence of Nutrients and Drugs into Milk. Nutrients 2019; 11:nu11102372. [PMID: 31590349 PMCID: PMC6836069 DOI: 10.3390/nu11102372] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/19/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
A large number of nutrients and bioactive ingredients found in milk play an important role in the nourishment of breast-fed infants and dairy consumers. Some of these ingredients include physiologically relevant compounds such as vitamins, peptides, neuroactive compounds and hormones. Conversely, milk may contain substances-drugs, pesticides, carcinogens, environmental pollutants-which have undesirable effects on health. The transfer of these compounds into milk is unavoidably linked to the function of transport proteins. Expression of transporters belonging to the ATP-binding cassette (ABC-) and Solute Carrier (SLC-) superfamilies varies with the lactation stages of the mammary gland. In particular, Organic Anion Transporting Polypeptides 1A2 (OATP1A2) and 2B1 (OATP2B1), Organic Cation Transporter 1 (OCT1), Novel Organic Cation Transporter 1 (OCTN1), Concentrative Nucleoside Transporters 1, 2 and 3 (CNT1, CNT2 and CNT3), Peptide Transporter 2 (PEPT2), Sodium-dependent Vitamin C Transporter 2 (SVCT2), Multidrug Resistance-associated Protein 5 (ABCC5) and Breast Cancer Resistance Protein (ABCG2) are highly induced during lactation. This review will focus on these transporters overexpressed during lactation and their role in the transfer of products into the milk, including both beneficial and harmful compounds. Furthermore, additional factors, such as regulation, polymorphisms or drug-drug interactions will be described.
Collapse
|
66
|
Fu NY, Nolan E, Lindeman GJ, Visvader JE. Stem Cells and the Differentiation Hierarchy in Mammary Gland Development. Physiol Rev 2019; 100:489-523. [PMID: 31539305 DOI: 10.1152/physrev.00040.2018] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mammary gland is a highly dynamic organ that undergoes profound changes within its epithelium during puberty and the reproductive cycle. These changes are fueled by dedicated stem and progenitor cells. Both short- and long-lived lineage-restricted progenitors have been identified in adult tissue as well as a small pool of multipotent mammary stem cells (MaSCs), reflecting intrinsic complexity within the epithelial hierarchy. While unipotent progenitor cells predominantly execute day-to-day homeostasis and postnatal morphogenesis during puberty and pregnancy, multipotent MaSCs have been implicated in coordinating alveologenesis and long-term ductal maintenance. Nonetheless, the multipotency of stem cells in the adult remains controversial. The advent of large-scale single-cell molecular profiling has revealed striking changes in the gene expression landscape through ontogeny and the presence of transient intermediate populations. An increasing number of lineage cell-fate determination factors and potential niche regulators have now been mapped along the hierarchy, with many implicated in breast carcinogenesis. The emerging diversity among stem and progenitor populations of the mammary epithelium is likely to underpin the heterogeneity that characterizes breast cancer.
Collapse
Affiliation(s)
- Nai Yang Fu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Emma Nolan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Geoffrey J Lindeman
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jane E Visvader
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
67
|
Wang QA, Scherer PE. Remodeling of Murine Mammary Adipose Tissue during Pregnancy, Lactation, and Involution. J Mammary Gland Biol Neoplasia 2019; 24:207-212. [PMID: 31512027 PMCID: PMC6790178 DOI: 10.1007/s10911-019-09434-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/14/2019] [Indexed: 01/06/2023] Open
Abstract
White adipocytes in the mammary gland stroma comprise the majority of the mammary gland mass. White adipocytes regulate numerous hormonal and metabolic processes and exhibit compositional and phenotypic plasticity. This plasticity is exemplified by the ability of mammary adipocytes to regress during lactation, when mammary epithelial cells expand to establish sufficient milk-producing alveoli. Upon weaning, the process reverses through mammary involution, during which adipocytes extensively regenerate, and alveolar epithelial cells disappear through cell death, returning the mammary gland to the non-lactating state. Despite intensive studies on the development and involution of the mammary alveolar epithelium, the fate of mammary adipocytes during pregnancy and lactation, and the origins of mammary adipocytes regenerated during mammary involution, is poorly understood. Here, we discuss the recent discoveries of the fate of mammary adipocytes during pregnancy and lactation in a number of different mouse models, and the lineage origin of mammary adipocytes regenerated during involution.
Collapse
Affiliation(s)
- Qiong A Wang
- Department of Molecular & Cellular Endocrinology, Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA.
- Comprehensive Cancer Center, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA.
| | - Philipp E Scherer
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
68
|
Tharmapalan P, Mahendralingam M, Berman HK, Khokha R. Mammary stem cells and progenitors: targeting the roots of breast cancer for prevention. EMBO J 2019; 38:e100852. [PMID: 31267556 PMCID: PMC6627238 DOI: 10.15252/embj.2018100852] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/11/2019] [Accepted: 04/11/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer prevention is daunting, yet not an unsurmountable goal. Mammary stem and progenitors have been proposed as the cells-of-origin in breast cancer. Here, we present the concept of limiting these breast cancer precursors as a risk reduction approach in high-risk women. A wealth of information now exists for phenotypic and functional characterization of mammary stem and progenitor cells in mouse and human. Recent work has also revealed the hormonal regulation of stem/progenitor dynamics as well as intrinsic lineage distinctions between mammary epithelial populations. Leveraging these insights, molecular marker-guided chemoprevention is an achievable reality.
Collapse
Affiliation(s)
| | - Mathepan Mahendralingam
- Princess Margaret Cancer CentreUniversity Health NetworkUniversity of TorontoTorontoONCanada
| | - Hal K Berman
- Princess Margaret Cancer CentreUniversity Health NetworkUniversity of TorontoTorontoONCanada
| | - Rama Khokha
- Princess Margaret Cancer CentreUniversity Health NetworkUniversity of TorontoTorontoONCanada
| |
Collapse
|
69
|
Yoshida K, Line J, Griffith K, Wooldredge A, Cruz YP. Progesterone signaling during pregnancy in the lab opossum, Monodelphis domestica. Theriogenology 2019; 136:101-110. [PMID: 31254723 DOI: 10.1016/j.theriogenology.2019.06.026] [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: 07/08/2018] [Revised: 05/18/2019] [Accepted: 06/12/2019] [Indexed: 10/26/2022]
Abstract
To investigate subtle pregnancy-associated changes in the lab opossum, Monodelphis domestica, an induced ovulator, we compared pregnant with non-pregnant and pseudopregnant animals with regard to serum P4 levels and progesterone receptor (PR) expression. Using video-verified, time-mated lab opossums as sources of biological material, we compared ovaries, uteri and sera obtained on odd-numbered days of the 14.5-day pregnancy in this animal. Females that mated successfully but did not produce embryos were classified as pseudopregnant. P4 levels differed significantly between pregnant (N = 21) and either non-pregnant (N = 3) or pseudopregnant (N = 3) opossums, but not between the non-pregnant and pseudopregnant groups. A significant decline in serum P4 occurred between pregnancy days 3 and 5, coinciding with an elevated probability of pregnancy failure between days 5 and 9. PR was detected in the nuclei of uterine-gland epithelial cells on pregnancy days 5 and 7 as well as variably in the corpora lutea (CL) of animals on pregnancy days 3-11. PR expression in the CL suggests that P4 may be autostimulatory in lab opossums and that certain levels of this steroid are required during normal pregnancy. The significant day-3 drop in P4 may explain why pregnancy failure in this polyovular metatherian is likeliest to occur between days 5 and 9, an interval during which the extended period of blastocyst morphogenesis and expansion occurs. Taken together, these results suggest that P4 may have unrecognized signaling roles not only in pregnancy but perhaps embryonic development as well in the lab opossum.
Collapse
Affiliation(s)
- K Yoshida
- Department of Biology, Oberlin College, Oberlin, OH, 44071, USA
| | - J Line
- Department of Biology, Oberlin College, Oberlin, OH, 44071, USA
| | - K Griffith
- Department of Biology, Oberlin College, Oberlin, OH, 44071, USA
| | - A Wooldredge
- Department of Biology, Oberlin College, Oberlin, OH, 44071, USA
| | - Y P Cruz
- Department of Biology, Oberlin College, Oberlin, OH, 44071, USA.
| |
Collapse
|
70
|
Ma G, Gao A, Yang Y, He Y, Zhang X, Zhang B, Zhang Z, Li M, Fu X, Zhao D, Wu R, Qi L, Hu Q, Li J, Fu L, Zhu Z, Dong JT. Zfhx3 is essential for progesterone/progesterone receptor signaling to drive ductal side-branching and alveologenesis in mouse mammary glands. J Genet Genomics 2019; 46:119-131. [PMID: 30954439 DOI: 10.1016/j.jgg.2019.03.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/05/2019] [Indexed: 01/26/2023]
Abstract
Progesterone (Pg)/progesterone receptor (PR) signaling drives mammary gland side-branching and alveologenesis, but the mechanisms through which Pg/PR signaling functions remain to be clarified. Using in vitro and in vivo models and histological and molecular analyses, we determined the role of Zfhx3 transcription factor in mammary gland development driven by Pg/PR signaling. Postnatal deletion of Zfhx3 in mouse mammary epithelial cells attenuated side-branching morphogenesis and alveologenesis. These effects were undetectable in the absence of Pg/PR signaling. During the estrus cycle, Zfhx3 expression corresponded to that of Pg, being at the highest level at the diestrus stage; Zfhx3 deletion inhibited mammary gland branching more potently at diestrus than estrus stage. Loss of Zfhx3 not only attenuated the expansion of stem/progenitor cells driven by Pg/PR signaling, but also impaired the function of Pg/PR signaling in the transcriptional activation of multiple genes. In addition, Pg/PR signaling significantly expanded PR- and Zfhx3-positive epithelial cells, and induced the physical association of ZFHX3 with PR. These findings establish Zfhx3 as an integral transcription factor of Pg/PR signaling in driving side-branching and alveologenesis during mammary gland development.
Collapse
Affiliation(s)
- Gui Ma
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ang Gao
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yinan Yang
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yuan He
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xi Zhang
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Baotong Zhang
- Department of Hematology and Medical Oncology, School of Medicine, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Zhiqian Zhang
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Mei Li
- Ningbo Institute of Medical Sciences, Ningbo, 315020, China
| | - Xing Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Dan Zhao
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Rui Wu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Leilei Qi
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qingxia Hu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Juan Li
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Liya Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zhengmao Zhu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jin-Tang Dong
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China; Department of Hematology and Medical Oncology, School of Medicine, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
71
|
Enger BD, Tucker HLM, Nickerson SC, Parsons CLM, Akers RM. Effects of Staphylococcus aureus intramammary infection on the expression of estrogen receptor α and progesterone receptor in mammary glands of nonlactating cows administered estradiol and progesterone to stimulate mammary growth. J Dairy Sci 2019; 102:2607-2617. [PMID: 30639023 DOI: 10.3168/jds.2018-15499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/20/2018] [Indexed: 11/19/2022]
Abstract
Intramammary infections (IMI) are prevalent in nonlactating dairy cattle and are known to alter mammary structure and negatively affect the amount of mammary epithelium in the gland. Mechanisms responsible for the observed changes in mammary growth during an IMI are poorly understood, yet the importance of the key mammogenic hormones driving mammary growth is well recognized. This study's objective was to characterize the expression of estrogen receptor α (ESR1) and progesterone receptor (PGR) in mammary glands stimulated to grow and develop in the presence or absence of an IMI as well as preliminarily characterize myoepithelial cell response to IMI. Mammary growth was stimulated in 18 nonpregnant, nonlactating dairy cows using subcutaneous estradiol and progesterone injections, and 2 culture-negative quarters of each cow were subsequently infused with either saline (n = 18) or Staphylococcus aureus (n = 18). Mammary parenchyma tissues were collected 5 d (n = 9) or 10 d (n = 9) postchallenge and examined using immunofluorescence microscopy to quantify positive nuclei and characterize staining features. There tended to be a greater number of ESR1-positive nuclei observed across 8 random mammary parenchyma fields of view in saline quarters than in Staph. aureus quarters (201 vs. 163 ± 44 nuclei). Saline quarters also contained a greater number of PGR-positive nuclei (520 vs. 440 ± 45 nuclei) and myoepithelial cells (971 vs. 863 ± 48 nuclei) than Staph. aureus-challenged quarters. However, when ESR1, PGR, and myoepithelial nuclei counts were adjusted for Staph. aureus quarters containing less epithelium, differences between quarter treatments abated. The examined ESR1 and PGR staining characteristics were similar between saline and Staph. aureus quarters but were differentially affected by day of tissue collection. Additionally, nuclear staining area of myoepithelial cells was greater in Staph. aureus quarters than in saline quarters. These results indicate that IMI had little effect on the number or staining characteristics of ESR1- or PGR-positive nuclei relative to epithelial area, but myoepithelial cells appear to be affected by IMI and the associated inflammation in nonlactating mammary glands that were stimulated to grow rapidly using mammogenic hormones. Accordingly, reductions in mammary epithelium in affected glands are not suspected to be resultant of alterations in the number or staining characteristics of ESR1- or PGR-positive mammary epithelial cells.
Collapse
Affiliation(s)
- B D Enger
- Dairy Science Department, Virginia Polytechnic Institute and State University, Blacksburg 24060
| | - H L M Tucker
- Dairy Science Department, Virginia Polytechnic Institute and State University, Blacksburg 24060
| | - S C Nickerson
- Animal and Dairy Science Department, University of Georgia, Athens 30602
| | - C L M Parsons
- Dairy Science Department, Virginia Polytechnic Institute and State University, Blacksburg 24060
| | - R M Akers
- Dairy Science Department, Virginia Polytechnic Institute and State University, Blacksburg 24060.
| |
Collapse
|
72
|
Oestrogen receptor α AF-1 and AF-2 domains have cell population-specific functions in the mammary epithelium. Nat Commun 2018; 9:4723. [PMID: 30413705 PMCID: PMC6226531 DOI: 10.1038/s41467-018-07175-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/15/2018] [Indexed: 12/20/2022] Open
Abstract
Oestrogen receptor α (ERα) is a transcription factor with ligand-independent and ligand-dependent activation functions (AF)-1 and -2. Oestrogens control postnatal mammary gland development acting on a subset of mammary epithelial cells (MECs), termed sensor cells, which are ERα-positive by immunohistochemistry (IHC) and secrete paracrine factors, which stimulate ERα-negative responder cells. Here we show that deletion of AF-1 or AF-2 blocks pubertal ductal growth and subsequent development because both are required for expression of essential paracrine mediators. Thirty percent of the luminal cells are ERα-negative by IHC but express Esr1 transcripts. This low level ERα expression through AF-2 is essential for cell expansion during puberty and growth-inhibitory during pregnancy. Cell-intrinsic ERα is not required for cell proliferation nor for secretory differentiation but controls transcript levels of cell motility and cell adhesion genes and a stem cell and epithelial mesenchymal transition (EMT) signature identifying ERα as a key regulator of mammary epithelial cell plasticity. Oestrogen receptors α (ERα) are expressed in a subset of mammary epithelial cells. Here, the authors identify cells with low-ERα protein levels and show that distinct cell populations have distinct requirements for the AF1 and AF2 domains of the ERα, and ERα acts in a biphasic manner dependent on developmental stage.
Collapse
|
73
|
Li D, Ji Y, Zhao C, Yao Y, Yang A, Jin H, Chen Y, San M, Zhang J, Zhang M, Zhang L, Feng X, Zheng Y. OXTR overexpression leads to abnormal mammary gland development in mice. J Endocrinol 2018; 239:121-136. [PMID: 30089682 DOI: 10.1530/joe-18-0356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 07/21/2018] [Accepted: 08/07/2018] [Indexed: 01/06/2023]
Abstract
Oxytocin receptor (OXTR) is a G-protein-coupled receptor and known for regulation of maternal and social behaviors. Null mutation (Oxtr-/-) leads to defects in lactation due to impaired milk ejection and maternal nurturing. Overexpression of OXTR has never been studied. To define the functions of OXTR overexpression, a transgenic mouse model that overexpresses mouse Oxtr under β-actin promoter was developed ( ++ Oxtr). ++ Oxtr mice displayed advanced development and maturation of mammary gland, including ductal distention, enhanced secretory differentiation and early milk production at non-pregnancy and early pregnancy. However, ++ Oxtr dams failed to produce adequate amount of milk and led to lethality of newborns due to early involution of mammary gland in lactation. Mammary gland transplantation results indicated the abnormal mammary gland development was mainly from hormonal changes in ++Oxtr mice but not from OXTR overexpression in mammary gland. Elevated OXTR expression increased prolactin-induced phosphorylation and nuclear localization of STAT5 (p-STAT5), and decreased progesterone level, leading to early milk production in non-pregnant and early pregnant females, whereas low prolactin and STAT5 activation in lactation led to insufficient milk production. Progesterone treatment reversed the OXTR-induced accelerated mammary gland development by inhibition of prolactin/p-STAT5 pathway. Prolactin administration rescued lactation deficiency through STAT5 activation. Progesterone plays a negative role in OXTR-regulated prolactin/p-STAT5 pathways. The study provides evidence that OXTR overexpression induces abnormal mammary gland development through progesterone and prolactin-regulated p-STAT5 pathway.
Collapse
Affiliation(s)
- Dan Li
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
| | - Yan Ji
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chunlan Zhao
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
| | - Yapeng Yao
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
| | - Anlan Yang
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
| | - Honghong Jin
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
| | - Yang Chen
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
| | - Mingjun San
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
| | - Jing Zhang
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
| | - Mingjiao Zhang
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
| | - Luqing Zhang
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, China
| | - Xuechao Feng
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, China
| | - Yaowu Zheng
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, Jilin, China
| |
Collapse
|
74
|
van de Ven M, Liu X, van der Burg E, Klarenbeek S, Alexi X, Zwart W, Dijcks F, Bouwman P, Jonkers J. BRCA1-associated mammary tumorigenesis is dependent on estrogen rather than progesterone signaling. J Pathol 2018; 246:41-53. [PMID: 29877575 DOI: 10.1002/path.5105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/01/2018] [Accepted: 05/25/2018] [Indexed: 12/16/2022]
Abstract
Hereditary breast cancers in BRCA1 mutation carriers are mostly estrogen receptor α (ERα)-negative and progesterone receptor (PR)-negative; however, hormone depletion via bilateral oophorectomy does result in a marked reduction in breast cancer risk, suggesting that BRCA1-associated breast tumorigenesis is dependent on hormone signaling. We used geneticaly engineered mouse models to determine the individual influences of ERα and PR signaling on the development of BRCA1-deficient breast cancer. In line with the human data, BRCA1-deficient mouse mammary tumors are ERα-negative, and bilateral ovariectomy leads to abrogation of mammary tumor development. Hormonal replacement experiments in ovariectomized mice showed that BRCA1-deficient mammary tumor formation is promoted by estrogen but not by progesterone. In line with these data, mammary tumorigenesis was significantly delayed by the selective ERα downregulator fulvestrant, but not by the selective PR antagonist Org33628. Together, our results illustrate that BRCA1-associated tumorigenesis is dependent on estrogen signaling rather than on progesterone signaling, and call into question the utility of PR antagonists as a tumor prevention strategy for BRCA1 mutation carriers. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Marieke van de Ven
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Xiaoling Liu
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Eline van der Burg
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sjoerd Klarenbeek
- Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Xanthippi Alexi
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Fred Dijcks
- Synthon Biopharmaceuticals B.V, Nijmegen, The Netherlands
| | - Peter Bouwman
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
75
|
Johnson MB, Hoffmann JN, You HM, Lastra RR, Fernandez S, Strober JW, Allaw AB, Brady MJ, Conzen SD, McClintock MK. Psychosocial Stress Exposure Disrupts Mammary Gland Development. J Mammary Gland Biol Neoplasia 2018; 23:59-73. [PMID: 29687293 PMCID: PMC6207373 DOI: 10.1007/s10911-018-9392-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 03/22/2018] [Indexed: 01/18/2023] Open
Abstract
Exposure to psychosocial stressors and ensuing stress physiology have been associated with spontaneous invasive mammary tumors in the Sprague-Dawley rat model of human breast cancer. Mammary gland (MG) development is a time when physiologic and environmental exposures influence breast cancer risk. However, the effect of psychosocial stress exposure on MG development remains unknown. Here, in the first comprehensive longitudinal study of MG development in nulliparous female rats (from puberty through young adulthood; 8-25 wks of age), we quantify the spatial gradient of differentiation within the MG of socially stressed (isolated) and control (grouped) rats. We then demonstrate that social isolation increased stress reactivity to everyday stressors, resulting in downregulation of glucocorticoid receptor (GR) expression in the MG epithelium. Surprisingly, given that chemical carcinogens increase MG cancer risk by preventing normal terminal end bud (TEB) differentiation, chronic isolation stress did not alter TEBs. Instead, isolation blunted MG growth and alveolobular differentiation and reduced epithelial cell proliferation in these structures. Social isolation also enhanced corpora luteal progesterone at all ages but reduced estrogenization only in early adulthood, a pattern that precludes modulated ovarian function as a sufficient mechanism for the effects of isolation on MG development. This longitudinal study of natural variation provides an integrated view of MG development and the importance of increased GR activation in nulliparous ductal growth and alveolobular differentiation. Thus, social isolation and its physiological sequelae disrupt MG growth and differentiation and suggest a contribution of stress exposure during puberty and young adulthood to the previously observed increase in invasive MG cancer observed in chronically socially-isolated adult Sprague-Dawley rats.
Collapse
Affiliation(s)
- Marianna B Johnson
- Committee on Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | | | - Hannah M You
- Institute for Mind and Biology, The University of Chicago, Chicago, IL, USA
| | - Ricardo R Lastra
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Sully Fernandez
- Committee on Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jordan W Strober
- Committee on Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Ahmad B Allaw
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Matthew J Brady
- Committee on Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Suzanne D Conzen
- Committee on Molecular Metabolism and Nutrition, The University of Chicago, Chicago, IL, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Ben May Department of Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Martha K McClintock
- Institute for Mind and Biology, The University of Chicago, Chicago, IL, USA.
- Departments of Psychology and Comparative Human Development, The University of Chicago, 940 East 57th Street, Chicago, IL, 60637, USA.
| |
Collapse
|
76
|
LaPlante CD, Bansal R, Dunphy KA, Jerry DJ, Vandenberg LN. Oxybenzone Alters Mammary Gland Morphology in Mice Exposed During Pregnancy and Lactation. J Endocr Soc 2018; 2:903-921. [PMID: 30057971 PMCID: PMC6057512 DOI: 10.1210/js.2018-00024] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/08/2018] [Indexed: 02/08/2023] Open
Abstract
Hormones and endocrine-disrupting chemicals are generally thought to have permanent “organizational” effects when exposures occur during development but not adulthood. Yet, an increasing number of studies have shown that pregnant females are disrupted by endocrine-disrupting chemical exposures, with some effects that are permanent. Here, we examined the long-term effects of exposure to oxybenzone, an estrogenic chemical found in sunscreen and personal care products, on the morphology of the mammary gland in mice exposed during pregnancy and lactation. Female mice were exposed to vehicle or 30, 212, or 3000 µg oxybenzone/kg/d, from pregnancy day 0 until weaning. A nulliparous group, receiving vehicle treatment, was also evaluated. Mammary glands were collected 5 weeks after involution for whole-mount, histological, immunohistochemical, and molecular analyses. Exposure to 3000 µg oxybenzone/kg/d induced permanent changes to ductal density that was significantly different from both the nulliparous and vehicle groups. The two highest doses of oxybenzone similarly induced an intermediate phenotype for expression of progesterone receptor. A monotonic, dose-dependent increase in cell proliferation was also observed in the oxybenzone-treated females, becoming statistically significant at the highest dose. Finally, oxybenzone exposure induced an intermediate phenotype for Esr1 expression in all oxybenzone-treated groups. These data suggest that oxybenzone, at doses relevant to human exposures, produces long-lasting alterations to mammary gland morphology and function. Further studies are needed to determine if exposure to this chemical during pregnancy and lactation will interfere with the known protection that pregnancy provides against breast cancer.
Collapse
Affiliation(s)
- Charlotte D LaPlante
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Ruby Bansal
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Karen A Dunphy
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - D Joseph Jerry
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| | - Laura N Vandenberg
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, Massachusetts
| |
Collapse
|
77
|
Hilton HN, Clarke CL, Graham JD. Estrogen and progesterone signalling in the normal breast and its implications for cancer development. Mol Cell Endocrinol 2018; 466:2-14. [PMID: 28851667 DOI: 10.1016/j.mce.2017.08.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/11/2017] [Accepted: 08/18/2017] [Indexed: 12/31/2022]
Abstract
The ovarian hormones estrogen and progesterone are master regulators of the development and function of a broad spectrum of human tissues, including the breast, reproductive and cardiovascular systems, brain and bone. Acting through the nuclear estrogen (ER) and progesterone receptors (PR), both play complex and essential coordinated roles in the extensive development of the lobular alveolar epithelial structures of the normal breast during puberty, the normal menstrual cycle and pregnancy. The past decade has seen major advances in understanding the mechanisms of action of estrogen and progesterone in the normal breast and in the delineation of the complex hierarchy of cell types regulated by ovarian hormones in this tissue. There is evidence for a role for both ER and PR in driving breast cancer, and both are favourable prognostic markers with respect to outcome. In this review, we summarize current knowledge of the mechanisms of action of ER and PR in the normal breast, and implications for the development and management of breast cancer.
Collapse
Affiliation(s)
- Heidi N Hilton
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW 2145, Australia
| | - Christine L Clarke
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW 2145, Australia
| | - J Dinny Graham
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney Medical School - Westmead, The University of Sydney, Westmead, NSW 2145, Australia.
| |
Collapse
|
78
|
CHOUDHARY RATANKUMAR, PATHAK DEVENDRA, CHOUDHARY SHANTI, VERMA RAMNEEK. Immunolocalization of estrogen alpha and progesterone beta receptors in goat mammary gland. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2018. [DOI: 10.56093/ijans.v88i4.78803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Steroid receptors particularly estrogen receptor alpha and progesterone receptor beta are essential for mammary gland development. Objective of this study was to explore transcript and protein expression profile of steroid receptors in goat mammary glands. A varied expression of ER-alpha and PR-B was observed during lactation, nonlactating/ dry, mastitic and mammary pre-cancer/cancer in goats. During lactation, immunopositivity of ER-alpha was observed only in the nuclei of mammary epithelial cells (MEC) and stromal cells. However, in non-lactating stage, ER-alpha immunopositivity was observed both in nucleus and cytoplasm of MEC. In mammary pre-cancer (based on aberrant expression of CD10, FNDC3B and MUC1) immunoreactivity of ER-alpha (38±12.5%) varied from non-lactating (14.8±3.1%) and lactating (7.9±2.6%) glands. During naturally infected mastitis, a reduction in the expression of ER-alpha and PR-B was observed.We observed similar expression patterns of ER-alpha and PR-B as that of their protein expression. Transcripts of these receptors were highest in mammary precancer. In comparison to lactating glands, expressions of ER-alpha and PR-B was upregulated in mammary precancers by 17- folds and 9.2-folds, respectively. These results showed a reduction in expression of steroid receptors in mastitic glands and upregulation in mammary precancer indicating role of these receptors in cell proliferation.
Collapse
|
79
|
McQueen CM, Schmitt EE, Sarkar TR, Elswood J, Metz RP, Earnest D, Rijnkels M, Porter WW. PER2 regulation of mammary gland development. Development 2018; 145:dev.157966. [PMID: 29490985 PMCID: PMC5897596 DOI: 10.1242/dev.157966] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 02/19/2018] [Indexed: 12/24/2022]
Abstract
The molecular clock plays key roles in daily physiological functions, development and cancer. Period 2 (PER2) is a repressive element, which inhibits transcription activated by positive clock elements, resulting in diurnal cycling of genes. However, there are gaps in our understanding of the role of the clock in normal development outside of its time-keeping function. Here, we show that PER2 has a noncircadian function that is crucial to mammalian mammary gland development. Virgin Per2-deficient mice, Per2-/- , have underdeveloped glands, containing fewer bifurcations and terminal ducts than glands of wild-type mice. Using a transplantation model, we show that these changes are intrinsic to the gland and further identify changes in cell fate commitment. Per2-/- mouse mammary glands have a dual luminal/basal phenotypic character in cells of the ductal epithelium. We identified colocalization of E-cadherin and keratin 14 in luminal cells. Similar results were demonstrated using MCF10A and shPER2 MCF10A human cell lines. Collectively this study reveals a crucial noncircadian function of PER2 in mammalian mammary gland development, validates the Per2-/- model, and describes a potential role for PER2 in breast cancer.
Collapse
Affiliation(s)
- Cole M McQueen
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Emily E Schmitt
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Tapasree R Sarkar
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Jessica Elswood
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Richard P Metz
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - David Earnest
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807, USA
| | - Monique Rijnkels
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Weston W Porter
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Center for Biological Clocks Research, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
80
|
ElShamy WM. The protective effect of longer duration of breastfeeding against pregnancy-associated triple negative breast cancer. Oncotarget 2018; 7:53941-53950. [PMID: 27248476 PMCID: PMC5288234 DOI: 10.18632/oncotarget.9690] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 05/23/2016] [Indexed: 12/24/2022] Open
Abstract
Parity associated breast cancer (PABC) often diagnosed within the 2-5 years after a full term pregnancy. PABC is usually present with more advanced, poorly differentiated, high-grade cancers that show shorter time to progression and often of the triple negative breast cancer (TNBC) subtype. Data from around the world show that pregnancy-associated TNBC is independently associated with poor survival, underscoring the impact of the pregnant breast microenvironment on the biology and consequently the prognosis of these tumors. Although it is not yet clear, a link between pregnancy-associated TNBCs and lack or shorter duration of breastfeeding (not pregnancy per se) has been proposed. Here, we present epidemiological and experimental evidence for the protective effect of longer duration of lactation against pregnancy-associated TNBCs, and propose a putative molecular mechanism for this protective effect and its effect in eliminating any potential TNBC precursors from the breast by the end of the natural breast involution.
Collapse
Affiliation(s)
- Wael M ElShamy
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
81
|
Shull JD, Dennison KL, Chack AC, Trentham-Dietz A. Rat models of 17β-estradiol-induced mammary cancer reveal novel insights into breast cancer etiology and prevention. Physiol Genomics 2018; 50:215-234. [PMID: 29373076 DOI: 10.1152/physiolgenomics.00105.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Numerous laboratory and epidemiologic studies strongly implicate endogenous and exogenous estrogens in the etiology of breast cancer. Data summarized herein suggest that the ACI rat model of 17β-estradiol (E2)-induced mammary cancer is unique among rodent models in the extent to which it faithfully reflects the etiology and biology of luminal types of breast cancer, which together constitute ~70% of all breast cancers. E2 drives cancer development in this model through mechanisms that are largely dependent upon estrogen receptors and require progesterone and its receptors. Moreover, mammary cancer development appears to be associated with generation of oxidative stress and can be modified by multiple dietary factors, several of which may attenuate the actions of reactive oxygen species. Studies of susceptible ACI rats and resistant COP or BN rats provide novel insights into the genetic bases of susceptibility and the biological processes regulated by genetic determinants of susceptibility. This review summarizes research progress resulting from use of these physiologically relevant rat models to advance understanding of breast cancer etiology and prevention.
Collapse
Affiliation(s)
- James D Shull
- McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison , Madison, Wisconsin.,University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison , Madison, Wisconsin
| | - Kirsten L Dennison
- McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison , Madison, Wisconsin
| | - Aaron C Chack
- McArdle Laboratory for Cancer Research, Department of Oncology, School of Medicine and Public Health, University of Wisconsin-Madison , Madison, Wisconsin
| | - Amy Trentham-Dietz
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison , Madison, Wisconsin.,University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison , Madison, Wisconsin
| |
Collapse
|
82
|
Park YH. The nuclear factor-kappa B pathway and response to treatment in breast cancer. Pharmacogenomics 2017; 18:1697-1709. [PMID: 29182047 DOI: 10.2217/pgs-2017-0044] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The nuclear factor-kappa B (NF-κB) pathway is known to contribute to critical signaling in cancer biology, including breast cancer, through promotion of proliferation, angiogenesis, metastasis, tumor progression, inflammation and cell survival. In this review, in vivo and in vitro studies of the NF-κB pathway in breast cancer are discussed, focusing on DNA damage and the epithelial-mesenchymal transition associated with breast cancer stem cell properties. The relationships between NF-κB signaling and conventional cancer treatments in terms of response to chemo- and radiotherapy will also be discussed. Then contribution and involvement of immune system in the NF-κB pathway will be covered. Furthermore, the future perspective of NF-κB targeting as an innovative strategy to overcome refractory breast cancer, including recent updates on out-receptor activator of NF-κB (RANKing), will be covered.
Collapse
Affiliation(s)
- Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| |
Collapse
|
83
|
Rädler PD, Wehde BL, Wagner KU. Crosstalk between STAT5 activation and PI3K/AKT functions in normal and transformed mammary epithelial cells. Mol Cell Endocrinol 2017; 451:31-39. [PMID: 28495456 PMCID: PMC5515553 DOI: 10.1016/j.mce.2017.04.025] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 04/29/2017] [Indexed: 01/01/2023]
Abstract
Janus kinases (JAKs) and signal transducers and activators of transcription (STATs) have been shown to function downstream of several peptide hormones and cytokines that are required for postnatal development and secretory function of the mammary gland. As part of an extended network, these signal transducers can engage in crosstalk with other pathways to facilitate synergistic, and sometimes antagonistic, actions of different growth factors. Specifically, signaling through the JAK2/STAT5 cascade has been demonstrated to be indispensable for the specification, proliferation, differentiation, and survival of secretory mammary epithelial cells. Following a concise description of major cellular programs in mammary gland development and the role of growth factors that rely on JAK/STAT signaling to orchestrate these programs, this review highlights the significance of active STAT5 and its crosstalk with the PI3 kinase and AKT1 for mediating the proliferation of alveolar progenitors and survival of their functionally differentiated descendants in the mammary gland. Based on its ability to provide self-sufficiency in growth signals that are also capable of overriding intrinsic cell death programs, persistently active STAT5 can serve as a potent oncoprotein that contributes to the genesis of breast cancer. Recent experimental evidence demonstrated that, similar to normal developmental programs, oncogenic functions of STAT5 rely on molecular crosstalk with PI3K/AKT signaling for the initiation, and in some instances the progression, of breast cancer. The multitude by which STATs can interact with individual mediators of the PI3K/AKT signaling cascade may provide novel avenues for targeting signaling nodes within molecular networks that are crucial for the survival of cancer cells.
Collapse
Affiliation(s)
- Patrick D Rädler
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, USA
| | - Barbara L Wehde
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, USA
| | - Kay-Uwe Wagner
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, USA; Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE 68198-6805, USA.
| |
Collapse
|
84
|
Knutson TP, Truong TH, Ma S, Brady NJ, Sullivan ME, Raj G, Schwertfeger KL, Lange CA. Posttranslationally modified progesterone receptors direct ligand-specific expression of breast cancer stem cell-associated gene programs. J Hematol Oncol 2017; 10:89. [PMID: 28412963 PMCID: PMC5392969 DOI: 10.1186/s13045-017-0462-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/03/2017] [Indexed: 12/18/2022] Open
Abstract
Background Estrogen and progesterone are potent breast mitogens. In addition to steroid hormones, multiple signaling pathways input to estrogen receptor (ER) and progesterone receptor (PR) actions via posttranslational events. Protein kinases commonly activated in breast cancers phosphorylate steroid hormone receptors (SRs) and profoundly impact their activities. Methods To better understand the role of modified PRs in breast cancer, we measured total and phospho-Ser294 PRs in 209 human breast tumors represented on 2754 individual tissue spots within a tissue microarray and assayed the regulation of this site in human tumor explants cultured ex vivo. To complement this analysis, we assayed PR target gene regulation in T47D luminal breast cancer models following treatment with progestin (promegestone; R5020) and antiprogestins (mifepristone, onapristone, or aglepristone) in conditions under which the receptor is regulated by Lys388 SUMOylation (K388 intact) or is SUMO-deficient (via K388R mutation to mimic persistent Ser294 phosphorylation). Selected phospho-PR-driven target genes were validated by qRT-PCR and following RUNX2 shRNA knockdown in breast cancer cell lines. Primary and secondary mammosphere assays were performed to implicate phospho-Ser294 PRs, epidermal growth factor signaling, and RUNX2 in breast cancer stem cell biology. Results Phospho-Ser294 PR species were abundant in a majority (54%) of luminal breast tumors, and PR promoter selectivity was exquisitely sensitive to posttranslational modifications. Phospho-PR expression and target gene programs were significantly associated with invasive lobular carcinoma (ILC). Consistent with our finding that activated phospho-PRs undergo rapid ligand-dependent turnover, unique phospho-PR gene signatures were most prevalent in breast tumors clinically designated as PR-low to PR-null (luminal B) and included gene sets associated with cancer stem cell biology (HER2, PAX2, AHR, AR, RUNX). Validation studies demonstrated a requirement for RUNX2 in the regulation of selected phospho-PR target genes (SLC37A2). In vitro mammosphere formation assays support a role for phospho-Ser294-PRs via growth factor (EGF) signaling as well as RUNX2 as potent drivers of breast cancer stem cell fate. Conclusions We conclude that PR Ser294 phosphorylation is a common event in breast cancer progression that is required to maintain breast cancer stem cell fate, in part via cooperation with growth factor-initiated signaling pathways and key phospho-PR target genes including SLC37A2 and RUNX2. Clinical measurement of phosphorylated PRs should be considered a useful marker of breast tumor stem cell potential. Alternatively, unique phospho-PR target gene sets may provide useful tools with which to identify patients likely to respond to selective PR modulators that block PR Ser294 phosphorylation as part of rational combination (i.e., with antiestrogens) endocrine therapies designed to durably block breast cancer recurrence. Electronic supplementary material The online version of this article (doi:10.1186/s13045-017-0462-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Todd P Knutson
- Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, Masonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer and Cardiovascular Research Building, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Thu H Truong
- Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, Masonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer and Cardiovascular Research Building, 2231 6th St SE, Minneapolis, MN, 55455, USA
| | - Shihong Ma
- Department of Urology, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, J8.130C, Dallas, TX, 75390-9110, USA
| | - Nicholas J Brady
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Megan E Sullivan
- Department of Pathology, Evanston Hospital, University of Chicago, NorthShore University HealthSystem, Evanston, IL, 60201, USA
| | - Ganesh Raj
- Department of Urology, UT Southwestern Medical Center at Dallas, 5323 Harry Hines Blvd, J8.130C, Dallas, TX, 75390-9110, USA
| | - Kathryn L Schwertfeger
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Carol A Lange
- Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, Masonic Cancer Center, University of Minnesota, Delivery Code 2812, Cancer and Cardiovascular Research Building, 2231 6th St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
85
|
Leonel EC, Falleiros LR, Campos SG, Taboga SR. Histological and immunohistochemical characterization of the Mongolian gerbil's mammary gland during gestation, lactation and involution. Acta Histochem 2017; 119:273-283. [PMID: 28238410 DOI: 10.1016/j.acthis.2017.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 02/10/2017] [Accepted: 02/10/2017] [Indexed: 01/22/2023]
Abstract
The morphological description of normal tissues is fundamental for making comparisons and in order to identify injuries and lesions. The aim of this work was to describe the morphological characteristics of the female Mongolian gerbil's (Meriones unguiculatus) normal mammary gland, the average expression of hormone receptors, and the average proliferation rates in the epithelial cells during the periods of lactation, pregnancy and involution. Dams were euthanized on the 14th and 21st gestational days, 7 and 14days after parturition, and 3 and 5days after weaning. The dams' mammary tissues were processed and were submitted to haematoxylin and eosin staining, Periodic Acid Schiff (PAS) staining, and Gomori's Reticulin staining. Additionally, immunohistochemistry was performed for the characterization of myoepithelial cells with α-actin, the proliferation rates with proliferating cell nuclear antigen (PCNA), the estrogen hormonal receptors (ESR1 and ESR2), and progesterone receptor (PR) quantifications. It was observed that the abundant adipose tissues were replaced by glandular epithelia and there was an increase in the epithelial cell's height (from 5.97 to 32.4μm in 14th and 21st gestational days and from 20.64 to 25.4μm in 7th and 14th lactational days, respectively) and the acini diameters (from 24.88 to 69.92μm in 14th and 21st gestational days and from 139.69 to 118.59μm in 7th and 14th lactational days, respectively) with the progression of gestation and lactation. The PAS staining intensity varied throughout the glands and between the stages that were evaluated. The extracellular matrix showed different phenotypes too, with more of a presence of the Type I collagen during the early gestation and involution and with more reticular fibers (Type III collagen) during the late gestation period and lactation. The myoepithelial layers showed alterations in their distribution with thick patterns as verified by the α-actin labeling. The PCNA showed higher rates of the marked cells in 14th and 21st gestational days (40.25 and 60.28%) and in 7th and 14th lactational days (64.08 and 65.08%). The hormone receptor quantifications showed a high variation in the rates: the average PR staining decreased from 14th to 21st gestational days (from 42.3 to 8.54%), from 7th to 14th lactational days (from 59.83 to 23.18%) and from 3rd to 5th days after weaning (from 39.98 to 12.72). There were higher averages of ESR1 staining in gestational days 14 and 21(from 58.06 to 30.02%). ESR2 staining decreased during gestation (25.7 and 12.94% in 14th and 21st gestational days)and involution (from 50.97 to 30.18% in 3rd and 5th days after weaning). The Mongolian gerbils showed similar morphological characteristics when they were compared to mice and rats. However, the higher proliferation rates with a smaller involution period compared to other murine characterized this species as being adequate for mammary pathologies studies.
Collapse
|
86
|
Able AA, Burrell JA, Stephens JM. STAT5-Interacting Proteins: A Synopsis of Proteins that Regulate STAT5 Activity. BIOLOGY 2017; 6:biology6010020. [PMID: 28287479 PMCID: PMC5372013 DOI: 10.3390/biology6010020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/27/2017] [Accepted: 03/06/2017] [Indexed: 01/17/2023]
Abstract
Signal Transducers and Activators of Transcription (STATs) are key components of the JAK/STAT pathway. Of the seven STATs, STAT5A and STAT5B are of particular interest for their critical roles in cellular differentiation, adipogenesis, oncogenesis, and immune function. The interactions of STAT5A and STAT5B with cytokine/hormone receptors, nuclear receptors, transcriptional regulators, proto-oncogenes, kinases, and phosphatases all contribute to modulating STAT5 activity. Among these STAT5 interacting proteins, some serve as coactivators or corepressors to regulate STAT5 transcriptional activity and some proteins can interact with STAT5 to enhance or repress STAT5 signaling. In addition, a few STAT5 interacting proteins have been identified as positive regulators of STAT5 that alter serine and tyrosine phosphorylation of STAT5 while other proteins have been identified as negative regulators of STAT5 via dephosphorylation. This review article will discuss how STAT5 activity is modulated by proteins that physically interact with STAT5.
Collapse
Affiliation(s)
- Ashley A Able
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Jasmine A Burrell
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Jacqueline M Stephens
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
87
|
Gargiulo L, May M, Rivero EM, Copsel S, Lamb C, Lydon J, Davio C, Lanari C, Lüthy IA, Bruzzone A. A Novel Effect of β-Adrenergic Receptor on Mammary Branching Morphogenesis and its Possible Implications in Breast Cancer. J Mammary Gland Biol Neoplasia 2017; 22:43-57. [PMID: 28074314 DOI: 10.1007/s10911-017-9371-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 01/02/2017] [Indexed: 12/20/2022] Open
Abstract
Understanding the mechanisms that govern normal mammary gland development is crucial to the comprehension of breast cancer etiology. β-adrenergic receptors (β-AR) are targets of endogenous catecholamines such as epinephrine that have gained importance in the context of cancer biology. Differences in β2-AR expression levels may be responsible for the effects of epinephrine on tumor vs non-tumorigenic breast cell lines, the latter expressing higher levels of β2-AR. To study regulation of the breast cell phenotype by β2-AR, we over-expressed β2-AR in MCF-7 breast cancer cells and knocked-down the receptor in non-tumorigenic MCF-10A breast cells. In MCF-10A cells having knocked-down β2-AR, epinephrine increased cell proliferation and migration, similar to the response by tumor cells. In contrast, in MCF-7 cells overexpressing the β2-AR, epinephrine decreased cell proliferation and migration and increased adhesion, mimicking the response of the non-tumorigenic MCF-10A cells, thus underscoring that β2-AR expression level is a key player in cell behavior. β-adrenergic stimulation with isoproterenol induced differentiation of breast cells growing in 3-dimension cell culture, and also the branching of murine mammary epithelium in vivo. Branching induced by isoproterenol was abolished in fulvestrant or tamoxifen-treated mice, demonstrating that the effect of β-adrenergic stimulation on branching is dependent on the estrogen receptor (ER). An ER-independent effect of isoproterenol on lumen architecture was nonetheless found. Isoproterenol significantly increased the expression of ERα, Ephrine-B1 and fibroblast growth factors in the mammary glands of mice, and in MCF-10A cells. In a poorly differentiated murine ductal carcinoma, isoproterenol also decreased tumor growth and induced tumor differentiation. This study highlights that catecholamines, through β-AR activation, seem to be involved in mammary gland development, inducing mature duct formation. Additionally, this differentiating effect could be resourceful in a breast tumor context.
Collapse
Affiliation(s)
- Lucía Gargiulo
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - María May
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - Ezequiel M Rivero
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - Sabrina Copsel
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
- Laboratorio de Farmacología de Receptores, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, 1113, Buenos Aires, CABA, Argentina
| | - Caroline Lamb
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - John Lydon
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Carlos Davio
- Laboratorio de Farmacología de Receptores, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, 1113, Buenos Aires, CABA, Argentina
| | - Claudia Lanari
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - Isabel A Lüthy
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - Ariana Bruzzone
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), CONICET-Universidad Nacional del Sur, Camino La Carrindanga km 7, 8000, Bahía Blanca, Argentina.
| |
Collapse
|
88
|
Perinatal Exposure to Bisphenol A or Diethylstilbestrol Increases the Susceptibility to Develop Mammary Gland Lesions After Estrogen Replacement Therapy in Middle-Aged Rats. Discov Oncol 2017; 8:78-89. [DOI: 10.1007/s12672-016-0282-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/23/2016] [Indexed: 11/26/2022] Open
|
89
|
Abstract
The mouse mammary gland is widely used as a model for human breast cancer and has greatly added to our understanding of the molecular mechanisms involved in breast cancer development and progression. To fully appreciate the validity and limitations of the mouse model, it is essential to be aware of the similarities and also the differences that exist between the mouse mammary gland and the human breast. This introduction therefore describes the parallels and contrasts in mouse mammary gland and human breast morphogenesis from an early embryonic phase through to puberty, adulthood, pregnancy, parturition, and lactation, and finally the regressive stage of involution.
Collapse
Affiliation(s)
- Sara McNally
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, 4, Ireland.
| | - Torsten Stein
- Institute of Cancer Sciences, College of MVLS, University of Glasgow, Glasgow, UK
| |
Collapse
|
90
|
Carroll JS, Hickey TE, Tarulli GA, Williams M, Tilley WD. Deciphering the divergent roles of progestogens in breast cancer. Nat Rev Cancer 2017; 17:54-64. [PMID: 27885264 DOI: 10.1038/nrc.2016.116] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Most breast cancers are driven by oestrogen receptor-α. Anti-oestrogenic drugs are the standard treatment for these breast cancers; however, treatment resistance is common, necessitating new therapeutic strategies. Recent preclinical and historical clinical studies support the use of progestogens to activate the progesterone receptor (PR) in breast cancers. However, widespread controversy exists regarding the role of progestogens in this disease, hindering the clinical implementation of PR-targeted therapies. Herein, we present and discuss data at the root of this controversy and clarify the confusion and misinterpretations that have consequently arisen. We then present our view on how progestogens may be safely and effectively used in treating breast cancer.
Collapse
Affiliation(s)
- Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - Theresa E Hickey
- Dame Roma Mitchell Cancer Research Laboratories, Hanson Institute and School of Medicine, University of Adelaide, Adelaide SA 5005, Australia
| | - Gerard A Tarulli
- Dame Roma Mitchell Cancer Research Laboratories, Hanson Institute and School of Medicine, University of Adelaide, Adelaide SA 5005, Australia
| | - Michael Williams
- Division of Epidemiology, Department of Public Health and Preventive Medicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239-3098, USA
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Hanson Institute and School of Medicine, University of Adelaide, Adelaide SA 5005, Australia
| |
Collapse
|
91
|
McBryan J, Howlin J. Pubertal Mammary Gland Development: Elucidation of In Vivo Morphogenesis Using Murine Models. Methods Mol Biol 2017; 1501:77-114. [PMID: 27796948 DOI: 10.1007/978-1-4939-6475-8_3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
During the past 25 years, the combination of increasingly sophisticated gene targeting technology with transplantation techniques has allowed researchers to address a wide array of questions about postnatal mammary gland development. These in turn have significantly contributed to our knowledge of other branched epithelial structures. This review chapter highlights a selection of the mouse models exhibiting a pubertal mammary gland phenotype with a focus on how they have contributed to our overall understanding of in vivo mammary morphogenesis. We discuss mouse models that have enabled us to assign functions to particular genes and proteins and, more importantly, have determined when and where these factors are required for completion of ductal outgrowth and branch patterning. The reason for the success of the mouse mammary gland model is undoubtedly the suitability of the postnatal mammary gland to experimental manipulation. The gland itself is very amenable to investigation and the combination of genetic modification with accessibility to the tissue has allowed an impressive number of studies to inform biology. Excision of the rudimentary epithelial structure postnatally allows genetically modified tissue to be readily transplanted into wild type stroma or vice versa, and has thus defined the contribution of each compartment to particular phenotypes. Similarly, whole gland transplantation has been used to definitively discern local effects from indirect systemic effects of various growth factors and hormones. While appreciative of the power of these tools and techniques, we are also cognizant of some of their limitations, and we discuss some shortcomings and future strategies that can overcome them.
Collapse
Affiliation(s)
- Jean McBryan
- Department of Molecular Medicine Royal College of Surgeons in Ireland Education and Research Centre, Beaumont Hospital, Dublin, 9, Ireland
| | - Jillian Howlin
- Division of Oncology-Pathology, Lund University Cancer Center/Medicon Village, Building 404:B2, Scheelevägen 2, 223 81, Lund, Sweden.
| |
Collapse
|
92
|
Ingthorsson S, Briem E, Bergthorsson JT, Gudjonsson T. Epithelial Plasticity During Human Breast Morphogenesis and Cancer Progression. J Mammary Gland Biol Neoplasia 2016; 21:139-148. [PMID: 27815674 PMCID: PMC5159441 DOI: 10.1007/s10911-016-9366-3] [Citation(s) in RCA: 15] [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: 09/27/2016] [Accepted: 10/23/2016] [Indexed: 01/05/2023] Open
Abstract
Understanding the complex events leading to formation of an epithelial-based organ such as the breast requires a detailed insight into the crosstalk between epithelial and stromal compartments. These interactions occur both through heterotypic cellular interactions and between cells and matrix components. While in vivo models may partially capture these complex interactions, there is a need for in- vitro models to study these events. In this review we discuss cell-cell interactions in breast development focusing on the stem cell niche and branching morphogenesis. Given the recent understanding that the basic developmental events underlying branching morphogenesis are closely related to pathways important to cancer progression, i.e. epithelial plasticity and epithelial to mesenchymal transition (EMT), we will also discuss aspects relevant to cancer progression. In cancer, the adoption of mesenchymal phenotype by the malignant cells allows stromal invasion and subsequent intravasation to blood- or lymphatic vessels, a route that is a prerequisite for metastasis. A number of publications have demonstrated that tumor initiating cells, sometimes referred to as cancer stem cells adapt an EMT phenotype that renders them more resistant to apoptosis and drug therapy. The mechanism behind this phenomenon is currently unknown but this may partially explain relapse in breast cancer patients. Increased understanding of branching morphogenesis in the breast gland and the regulation of EMT and its reverse process mesenchymal to epithelial transition (MET) may hold the keys for future development of methods/drugs that neutralize the invading properties of cancer cells.
Collapse
Affiliation(s)
- Saevar Ingthorsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavík, Iceland
| | - Eirikur Briem
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavík, Iceland
| | - Jon Thor Bergthorsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavík, Iceland
- Department of Laboratory Hematology, Landspitali, University Hospital, Reykjavík, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavík, Iceland.
- Department of Laboratory Hematology, Landspitali, University Hospital, Reykjavík, Iceland.
| |
Collapse
|
93
|
Abstract
The mammary gland is the only organ to undergo most of its development after birth and therefore particularly attractive for studying developmental processes. In the mouse, powerful tissue recombination techniques are available that can be elegantly combined with the use of different genetically engineered mouse models to study development and differentiation in vivo.In this chapter, we describe how epithelial intrinsic gene function can by discerned by grafting mammary epithelial cells of different genotypes to wild-type recipients. Either pieces of mammary epithelial tissue or dissociated mammary epithelial cells are isolated from donor mice and subsequently transplanted into recipients whose mammary fat pads were divested of their endogenous epithelium. This is followed by phenotypic characterization of the epithelial outgrowth either by fluorescence stereomicroscopy for the fluorescently marked grafts or carmine alum whole mount for the unmarked epithelia.
Collapse
|
94
|
Berryhill GE, Brust-Mascher I, Huynh JH, Famula TR, Reardon C, Hovey RC. A Convenient Method for Evaluating Epithelial Cell Proliferation in the Whole Mammary Glands of Female Mice. Endocrinology 2016; 157:3742-3748. [PMID: 27571136 PMCID: PMC5045499 DOI: 10.1210/en.2016-1480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The mammary glands (MG) undergo rapid expansion of the ductal network during puberty in response to endocrine cues including the potent mitogenic effects of estrogen. The proliferation of mammary epithelial cells occurs in a spatially distinctive manner, where terminal end buds located at the ductal termini are the primary site of cell division. Here, we present a relatively high throughput approach to spatially assess epithelial cell proliferation in whole mouse MG using histochemical detection of 5-ethynyl-2'-deoxyuridine in conjunction with a standard curve-based data deconvolution technique to semiquantitatively measure proliferation via wide-field epifluorescent microscopy. This approach was validated against the "gold standard" of counting labeled nuclei from confocal images utilizing computer-assisted image analysis. Our method proved sensitive enough to describe the significant and spatially variable proliferative response to low-dose estrogen after 108 hours. This flexible method presents a timely and economical approach to obtaining spatial information regarding epithelial cell proliferation in the mouse MG.
Collapse
Affiliation(s)
- Grace E Berryhill
- Department of Animal Science (G.E.B., J.H.H., T.R.F., R.C.H.) and University of California Davis Health Sciences District Advanced Imaging Facility, School of Veterinary Medicine, (I.B.-M., C.R.), University of California, Davis, California 95616-8521
| | - Ingrid Brust-Mascher
- Department of Animal Science (G.E.B., J.H.H., T.R.F., R.C.H.) and University of California Davis Health Sciences District Advanced Imaging Facility, School of Veterinary Medicine, (I.B.-M., C.R.), University of California, Davis, California 95616-8521
| | - Jill H Huynh
- Department of Animal Science (G.E.B., J.H.H., T.R.F., R.C.H.) and University of California Davis Health Sciences District Advanced Imaging Facility, School of Veterinary Medicine, (I.B.-M., C.R.), University of California, Davis, California 95616-8521
| | - Thomas R Famula
- Department of Animal Science (G.E.B., J.H.H., T.R.F., R.C.H.) and University of California Davis Health Sciences District Advanced Imaging Facility, School of Veterinary Medicine, (I.B.-M., C.R.), University of California, Davis, California 95616-8521
| | - Colin Reardon
- Department of Animal Science (G.E.B., J.H.H., T.R.F., R.C.H.) and University of California Davis Health Sciences District Advanced Imaging Facility, School of Veterinary Medicine, (I.B.-M., C.R.), University of California, Davis, California 95616-8521
| | - Russell C Hovey
- Department of Animal Science (G.E.B., J.H.H., T.R.F., R.C.H.) and University of California Davis Health Sciences District Advanced Imaging Facility, School of Veterinary Medicine, (I.B.-M., C.R.), University of California, Davis, California 95616-8521
| |
Collapse
|
95
|
|
96
|
Grimm SL, Hartig SM, Edwards DP. Progesterone Receptor Signaling Mechanisms. J Mol Biol 2016; 428:3831-49. [PMID: 27380738 DOI: 10.1016/j.jmb.2016.06.020] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/25/2016] [Accepted: 06/27/2016] [Indexed: 12/27/2022]
Abstract
Progesterone receptor (PR) is a master regulator in female reproductive tissues that controls developmental processes and proliferation and differentiation during the reproductive cycle and pregnancy. PR also plays a role in progression of endocrine-dependent breast cancer. As a member of the nuclear receptor family of ligand-dependent transcription factors, the main action of PR is to regulate networks of target gene expression in response to binding its cognate steroid hormone, progesterone. This paper summarizes recent advances in understanding the structure-function properties of the receptor protein and the tissue/cell-type-specific PR signaling pathways that contribute to the biological actions of progesterone in the normal breast and in breast cancer.
Collapse
Affiliation(s)
- Sandra L Grimm
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA
| | - Sean M Hartig
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA
| | - Dean P Edwards
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA.
| |
Collapse
|
97
|
Sau A, Lau R, Cabrita MA, Nolan E, Crooks PA, Visvader JE, Pratt MAC. Persistent Activation of NF-κB in BRCA1-Deficient Mammary Progenitors Drives Aberrant Proliferation and Accumulation of DNA Damage. Cell Stem Cell 2016; 19:52-65. [PMID: 27292187 DOI: 10.1016/j.stem.2016.05.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 02/20/2016] [Accepted: 05/06/2016] [Indexed: 10/21/2022]
Abstract
Human BRCA1 mutation carriers and BRCA1-deficient mouse mammary glands contain an abnormal population of mammary luminal progenitors that can form 3D colonies in a hormone-independent manner. The intrinsic cellular regulatory defect in these presumptive breast cancer precursors is not known. We have discovered that nuclear factor kappaB (NF-κB) (p52/RelB) is persistently activated in a subset of BRCA1-deficient mammary luminal progenitors. Hormone-independent luminal progenitor colony formation required NF-κB, ataxia telangiectasia-mutated (ATM), and the inhibitor of kappaB kinase, IKKα. Progesterone (P4)-stimulated proliferation resulted in a marked enhancement of DNA damage foci in Brca1(-/-) mouse mammary. In vivo, NF-κB inhibition prevented recovery of Brca1(-/-) hormone-independent colony-forming cells. The majority of human BRCA1(mut/+) mammary glands showed marked lobular expression of nuclear NF-κB. We conclude that the aberrant proliferative capacity of Brca1(-/-) luminal progenitor cells is linked to the replication-associated DNA damage response, where proliferation of mammary progenitors is perpetuated by damage-induced, autologous NF-κB signaling.
Collapse
Affiliation(s)
- Andrea Sau
- Breast Cancer Research Laboratory, Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Rosanna Lau
- Breast Cancer Research Laboratory, Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Miguel A Cabrita
- Breast Cancer Research Laboratory, Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Emma Nolan
- Stem Cells and Cancer 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
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jane E Visvader
- Stem Cells and Cancer 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
| | - M A Christine Pratt
- Breast Cancer Research Laboratory, Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
98
|
Cordero A, Pellegrini P, Sanz-Moreno A, Trinidad EM, Serra-Musach J, Deshpande C, Dougall WC, Pujana MA, González-Suárez E. Rankl Impairs Lactogenic Differentiation Through Inhibition of the Prolactin/Stat5 Pathway at Midgestation. Stem Cells 2016; 34:1027-39. [PMID: 26695351 DOI: 10.1002/stem.2271] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/06/2015] [Accepted: 11/04/2015] [Indexed: 11/12/2022]
Abstract
Prolactin and progesterone both orchestrate the proliferation and differentiation of the mammary gland during gestation. Differentiation of milk secreting alveoli depends on the presence of prolactin receptor, the downstream Jak2-Stat5 pathway and the transcription factor Elf5. A strict regulation of Rank signaling is essential for the differentiation of the mammary gland and in particular for alveolar commitment. Impaired alveologenesis and lactation failure are observed in both, knockout and Rank overexpressing mice; however, the underlying molecular mechanism responsible for these phenotypes remains largely unknown. Using genome-wide expression analyses and functional studies, we show here that Rankl (RL) exposure leads to impaired secretory differentiation of alveolar cells not only in MMTV-RANK but also in wild-type (WT) mammary acini. Conversely, pharmacological blockage of Rank signaling at midgestation in WT mice leads to precocious and exacerbated lactogenesis. Mechanistically, RL negatively regulates Stat5 phosphorylation and Elf5 expression at the onset of lactogenesis. Continuous RL exposure leads to the expansion of basal and bipotent cells in WT and MMTV-RANK acini. Overall, we demonstrate that enhanced Rank signaling impairs secretory differentiation during pregnancy by inhibition of the prolactin/p-Stat5 pathway.
Collapse
Affiliation(s)
- Alex Cordero
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Pasquale Pellegrini
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Adrián Sanz-Moreno
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Eva M Trinidad
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | | | - Chetan Deshpande
- Department of Molecular Sciences and Computational Biology, Amgen Inc., Thousand Oaks, California, USA
| | | | | | - Eva González-Suárez
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| |
Collapse
|
99
|
Jonas W, Woodside B. Physiological mechanisms, behavioral and psychological factors influencing the transfer of milk from mothers to their young. Horm Behav 2016; 77:167-81. [PMID: 26232032 DOI: 10.1016/j.yhbeh.2015.07.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 07/12/2015] [Accepted: 07/23/2015] [Indexed: 12/13/2022]
Abstract
This article is part of a Special Issue "Parental Care".Producing milk to support the growth of their young is a central element of maternal care in mammals. In spite of the facts that ecological constraints influence nursing frequency, length of time until weaning and the composition of milk, there is considerable similarity in the anatomy and physiology of milk production and delivery across mammalian species. Here we provide an overview of cross species variation in nursing patterns and milk composition as well as the mechanisms underlying mammary gland development, milk production and letdown. Not all women breastfeed their infants, thus in later sections we review studies of factors that facilitate or impede the initiation and duration of breastfeeding. The results of these investigations suggest that the decisions to initiate and maintain breastfeeding are influenced by an array of personal, social and biological factors. Finally, studies comparing the development of breastfed and formula fed infants as well as those investigating associations between breastfeeding, maternal health and mother/infant interaction are reviewed. Leading health agencies including the World Health Organization and CDC advocate breastfeeding for at least the first 6months postpartum. To achieve these rates will require not only institutional support but also a focus on individual mother/infant dyads and their experience.
Collapse
Affiliation(s)
- Wibke Jonas
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Fraser Mustard Institute of Human Development, University of Toronto, Toronto, Canada
| | - Barbara Woodside
- Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC, Canada.
| |
Collapse
|
100
|
Zong Y, Goldstein AS, Witte ON. Tissue Recombination Models for the Study of Epithelial Cancer. Cold Spring Harb Protoc 2015; 2015:pdb.top069880. [PMID: 26631129 DOI: 10.1101/pdb.top069880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Animal models of cancer provide fundamental insight into the cellular and molecular mechanisms of human cancer development. As an alternative to genetically engineered mouse models, increasing evidence shows that tissue recombination and transplantation models represent an efficient approach to faithfully recapitulate solid epithelial cancer in mice. Cancer can be rapidly initiated through lentiviral delivery of defined genetic alterations into target cells that are grown in a physiological milieu with an appropriate epithelial-stromal interaction. Through genetic manipulation of distinct subpopulations of epithelial cells and mesenchymal cells, this powerful system can readily test both cell-autonomous roles of genetic events in the epithelial compartment and the paracrine effects of the microenvironment. Here we review the recent advances in mouse models of several epithelial cancers achieved using orthotopic transplantation and tissue recombination strategies, with an emphasis on the dissociated cell in vivo prostate regeneration model to investigate prostate cancer.
Collapse
Affiliation(s)
- Yang Zong
- Howard Hughes Medical Institute, University of California, Los Angeles, California 90095
| | - Andrew S Goldstein
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California 90095; Department of Urology, University of California, Los Angeles, California 90095; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California 90095; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California 90095
| | - Owen N Witte
- Howard Hughes Medical Institute, University of California, Los Angeles, California 90095; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California 90095; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90095; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, California 90095; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, California 90095
| |
Collapse
|