1
|
Schierano-Marotti G, Altamirano GA, Oddi S, Gomez AL, Meyer N, Muñoz-de-Toro M, Zenclussen AC, Rodríguez HA, Kass L. Branching morphogenesis of the mouse mammary gland after exposure to benzophenone-3. Toxicol Appl Pharmacol 2024; 484:116868. [PMID: 38382712 DOI: 10.1016/j.taap.2024.116868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/14/2024] [Accepted: 02/17/2024] [Indexed: 02/23/2024]
Abstract
Pubertal mammary branching morphogenesis is a hormone-regulated process susceptible to exposure to chemicals with endocrine disruptive capacity, such as the UV-filter benzophenone-3 (BP3). Our aim was to assess whether intrauterine or in vitro exposure to BP3 modified the branching morphogenesis of the female mouse mammary gland. For this, pregnant mice were dermally exposed to BP3 (0.15 or 50 mg/kg/day) from gestation day (GD) 8.5 to GD18.5. Sesame oil treatment served as control. Changes of the mammary glands of the offspring were studied on postnatal day 45. Further, mammary organoids from untreated mice were cultured under branching induction conditions and exposed for 9 days to BP3 (1 × 10-6 M, 1 × 10-9 M, or 1 × 10-12 M with 0.01% ethanol as control) to evaluate the branching progression. Mice that were exposed to BP3 in utero showed decreased mRNA levels of progesterone receptor (PR) and WNT4. However, estradiol and progesterone serum levels, mammary histomorphology, proliferation, and protein expression of estrogen receptor alpha (ESR1) and PR were not significantly altered. Interestingly, direct exposure to BP3 in vitro also decreased the mRNA levels of PR, RANKL, and amphiregulin without affecting the branching progression. Most effects were found after exposure to 50 mg/kg/day or 1 × 10-6 M of BP3, both related to sunscreen application in humans. In conclusion, exposure to BP3 does not impair mammary branching morphogenesis in our models. However, BP3 affects PR transcriptional expression and its downstream mediators, suggesting that exposure to BP3 might affect other developmental stages of the mammary gland.
Collapse
Affiliation(s)
- Gonzalo Schierano-Marotti
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Gabriela A Altamirano
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Sofia Oddi
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Ayelen L Gomez
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Nicole Meyer
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research and Perinatal Immunology, Saxonian Incubator for Clinical Translation, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Ana C Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research and Perinatal Immunology, Saxonian Incubator for Clinical Translation, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Horacio A Rodríguez
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Laura Kass
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
2
|
Bhattacharya A, Alam K, Roy NS, Kaur K, Kaity S, Ravichandiran V, Roy S. Exploring the interaction between extracellular matrix components in a 3D organoid disease model to replicate the pathophysiology of breast cancer. J Exp Clin Cancer Res 2023; 42:343. [PMID: 38102637 PMCID: PMC10724947 DOI: 10.1186/s13046-023-02926-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
In vitro models are necessary to study the pathophysiology of the disease and the development of effective, tailored treatment methods owing to the complexity and heterogeneity of breast cancer and the large population affected by it. The cellular connections and tumor microenvironments observed in vivo are often not recapitulated in conventional two-dimensional (2D) cell cultures. Therefore, developing 3D in vitro models that mimic the complex architecture and physiological circumstances of breast tumors is crucial for advancing our understanding of the illness. A 3D scaffold-free in vitro disease model mimics breast cancer pathophysiology by allowing cells to self-assemble/pattern into 3D structures, in contrast with other 3D models that rely on artificial scaffolds. It is possible that this model, whether applied to breast tumors using patient-derived primary cells (fibroblasts, endothelial cells, and cancer cells), can accurately replicate the observed heterogeneity. The complicated interactions between different cell types are modelled by integrating critical components of the tumor microenvironment, such as the extracellular matrix, vascular endothelial cells, and tumor growth factors. Tissue interactions, immune cell infiltration, and the effects of the milieu on drug resistance can be studied using this scaffold-free 3D model. The scaffold-free 3D in vitro disease model for mimicking tumor pathophysiology in breast cancer is a useful tool for studying the molecular basis of the disease, identifying new therapeutic targets, and evaluating treatment modalities. It provides a more physiologically appropriate high-throughput platform for screening large compound library in a 96-384 well format. We critically discussed the rapid development of personalized treatment strategies and accelerated drug screening platforms to close the gap between traditional 2D cell culture and in vivo investigations.
Collapse
Affiliation(s)
- Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
3
|
Li T, Ren X, Li T, Yu L, Teng M, Zheng Y, Lei A. Low-Dose Sodium Salicylate Promotes Ovulation by Regulating Steroids via CYP17A1. Int J Mol Sci 2023; 24:ijms24032579. [PMID: 36768902 PMCID: PMC9916436 DOI: 10.3390/ijms24032579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
To meet the current demand of assisted reproduction and animal breeding via superovulation and reduce the impact of hormone drugs, it is necessary to develop new superovulation drugs. This study examined the role of inflammation and steroids in ovulation. Sodium salicylate can regulate inflammation and steroids. However, the effect of sodium salicylate on ovulation has not been studied. In this study, mice were intraperitoneally injected with different concentrations of sodium salicylate for four consecutive days. The effects of sodium salicylate on oocyte quality and on the number of ovulations were examined, and these effects were compared with those of pregnant horse serum gonadotropin (PMSG)/follicle-stimulating hormone (FSH) treatment. We found that low-dose sodium salicylate increased the levels of ovulation hormones and inflammation by promoting the expression of CYP17A1. Sodium salicylate had the same effect as the commonly used superovulation drug PMSG/FSH and reduced the histone methylation level. Sodium salicylate can promote ovulation in mice and Awang sheep. It can greatly decrease the use of hormone drugs, reduce breeding costs and physical impacts, and can thus be used for livestock breeding.
Collapse
Affiliation(s)
- Tao Li
- Shaanxi Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Xuehua Ren
- Shaanxi Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Tianjiao Li
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Lian Yu
- Shaanxi Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Mingming Teng
- Shaanxi Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Yi Zheng
- Key Laboratory for Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, China
| | - Anmin Lei
- Shaanxi Stem Cell Engineering and Technology Center, College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang 712100, China
- Correspondence:
| |
Collapse
|
4
|
Trott JF, Schennink A, Horigan KC, Lemay DG, Cohen JR, Famula TR, Dragon JA, Hovey RC. Unique Transcriptomic Changes Underlie Hormonal Interactions During Mammary Histomorphogenesis in Female Pigs. Endocrinology 2022; 163:bqab256. [PMID: 34918063 PMCID: PMC10409904 DOI: 10.1210/endocr/bqab256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Indexed: 11/19/2022]
Abstract
Successful lactation and the risk for developing breast cancer depend on growth and differentiation of the mammary gland (MG) epithelium that is regulated by ovarian steroids (17β-estradiol [E] and progesterone [P]) and pituitary-derived prolactin (PRL). Given that the MG of pigs share histomorphogenic features present in the normal human breast, we sought to define the transcriptional responses within the MG of pigs following exposure to all combinations of these hormones. Hormone-ablated female pigs were administered combinations of E, medroxyprogesterone 17-acetate (source of P), and either haloperidol (to induce PRL) or 2-bromo-α-ergocryptine. We subsequently monitored phenotypic changes in the MG including mitosis, receptors for E and P (ESR1 and PGR), level of phosphorylated STAT5 (pSTAT5), and the frequency of terminal ductal lobular unit (TDLU) subtypes; these changes were then associated with all transcriptomic changes. Estrogen altered the expression of approximately 20% of all genes that were mostly associated with mitosis, whereas PRL stimulated elements of fatty acid metabolism and an inflammatory response. Several outcomes, including increased pSTAT5, highlighted the ability of E to enhance PRL action. Regression of transcriptomic changes against several MG phenotypes revealed 1669 genes correlated with proliferation, among which 29 were E inducible. Additional gene expression signatures were associated with TDLU formation and the frequency of ESR1 or PGR. These data provide a link between the hormone-regulated genome and phenome of the MG in a species having a complex histoarchitecture like that in the human breast, and highlight an underexplored synergy between the actions of E and PRL during MG development.
Collapse
Affiliation(s)
- Josephine F Trott
- Department of Animal Science, University of California, Davis, Davis, California 95616, USA
| | - Anke Schennink
- Department of Animal Science, University of California, Davis, Davis, California 95616, USA
| | - Katherine C Horigan
- Department of Animal Science, University of Vermont, Burlington, Vermont 05405, USA
| | - Danielle G Lemay
- US Department of Agriculture ARS Western Human Nutrition Research Center, Davis, California 95616, USA
| | - Julia R Cohen
- Department of Animal Science, University of California, Davis, Davis, California 95616, USA
| | - Thomas R Famula
- Department of Animal Science, University of California, Davis, Davis, California 95616, USA
| | - Julie A Dragon
- Vermont Integrative Genomics Resource, University of Vermont, Burlington, Vermont 05405, USA
| | - Russell C Hovey
- Department of Animal Science, University of California, Davis, Davis, California 95616, USA
| |
Collapse
|
5
|
Effect of acetate, β-hydroxybutyrate and their interaction on lipogenic gene expression, triglyceride contents and lipid droplet formation in dairy cow mammary epithelial cells. In Vitro Cell Dev Biol Anim 2021; 57:66-75. [PMID: 33403623 DOI: 10.1007/s11626-020-00538-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/09/2020] [Indexed: 10/22/2022]
Abstract
The purpose of this study was to assess the effects of acetate and β-hydroxybutyrate alone or in combination on lipogenic genes and their associated regulatory proteins in dairy cow mammary epithelial cells (DCMEC) using quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blotting, lipid droplet staining and a triglyceride content detection kit, to determine whether SCFA are related to milk fat synthesis regulation in DCMEC. Our experiment shows that addition of different concentrations of acetate, β-hydroxybutyrate and their combinations to DCMEC increase in relative mRNA abundance of lipogenic genes and key transcription factors suggest an increase in lipogenic capacity, which is supported by an increased in cytosolic triglyceride content. Similarly, the protein expression level of acetyl-coenzyme A carboxylase (ACACA), fatty acid synthase (FASN) and sterol-coenzyme desaturase-1 (SCD1) genes and the transcription factor sterol regulatory element-binding protein-1 (SREBP1) were found to be increased by addition of acetate, β-hydroxybutyrate and their combinations. The expression pattern of fat-related genes and proteins showed similar trends in almost all treatments, suggesting that common transcription factor are regulating these genes. These results show that acetate and β-hydroxybutyrate regulate fat synthesis, further confirming that SCFAs work by targeting genes to activate the SREBP1 and insulin-induced gene 1 protein (INSIG1) signalling pathways in DCMEC.
Collapse
|
6
|
Li W, Li C, Lu J, Zhao Y. MiR-145 is involved in the proliferation of bovine mammary epithelial cells and regulates bovine insulin receptor substrate 1. ITALIAN JOURNAL OF ANIMAL SCIENCE 2020. [DOI: 10.1080/1828051x.2020.1732234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Wenqing Li
- College of Life Science, Henan Agricultural University, Zhengzhou, China
| | - Chenwan Li
- College of Life Science, Henan Agricultural University, Zhengzhou, China
| | - Jinghui Lu
- College of Life Science, Henan Agricultural University, Zhengzhou, China
| | - Yangning Zhao
- College of Life Science, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
7
|
Huang T, Shafrir AL, Eliassen AH, Rexrode KM, Tworoger SS. Estimated Number of Lifetime Ovulatory Years and Its Determinants in Relation to Levels of Circulating Inflammatory Biomarkers. Am J Epidemiol 2020; 189:660-670. [PMID: 31845729 DOI: 10.1093/aje/kwz264] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 12/16/2022] Open
Abstract
Reproductive events, such as ovulation, trigger an inflammatory cascade. Few studies have examined their long-term influence on inflammatory profiles. We included 3,393 premenopausal and 3,915 postmenopausal women with intact ovaries/uterus from the Nurses' Health studies (Nurses' Health Study (1989-1990) and Nurses' Health Study II (1996-1999)) in an analysis of the association between lifetime ovulatory years (LOY) and levels of inflammatory biomarkers. We estimated LOY as age at menopause (age at blood collection for premenopausal women) minus age at menarche, subtracting years of oral contraceptive (OC) use and 1 year per pregnancy. After adjustment for other inflammation-related factors (e.g., body mass index, exercise, diet), every 5-year increase in LOY was associated with lower C-reactive protein (CRP) levels in both premenopausal (difference = -11.5%, 95% confidence interval: -15.0, -8.0; P < 0.0001) and postmenopausal (difference = -7.2%, 95% confidence interval: -10.0, -4.3; P < 0.0001) women. Older age at menopause (P = 0.007), earlier menarche (P = 0.007), and shorter duration of OC use (P = 0.002) were associated with lower CRP levels in postmenopausal women, whereas duration of OC use was positively associated with CRP levels in premenopausal women (P < 0.0001). LOY was modestly inversely associated with interleukin 6 in postmenopausal women (P = 0.03). Notably, the associations of CRP with LOY were similar in magnitude to associations with exercise and a healthy diet, though weaker than the association with body mass index. Although many reproductive events induce acute inflammation, increased LOY was associated with lower chronic systemic inflammation even after menopause.
Collapse
Affiliation(s)
- Tianyi Huang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Amy L Shafrir
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, Massachusetts
- Division of Adolescent and Young Adult Medicine, Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - A Heather Eliassen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kathryn M Rexrode
- Division of Women’s Health, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shelley S Tworoger
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
8
|
Goldstuck ND. Modern menstruation: Is it abnormal and unhealthy? Med Hypotheses 2020; 144:109955. [PMID: 32526510 DOI: 10.1016/j.mehy.2020.109955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/04/2020] [Indexed: 12/17/2022]
Abstract
The menstrual cycle appears to have evolved in humans and some other species to improve reproductive efficiency by enabling progesterone production without the presence of the fetus. This phenomenon is termed spontaneous decidualization. Repeated menstruation is produced in modern women because of better general health and having fewer pregnancies later in life and limiting breast feeding. The repeated breakdown of a progesterone primed endometrium releases an inflammatory cascade which appears to have short and long term adverse consequences. Repetitive modern menstruation is no longer a sign of good general and reproductive health but a harbinger of possible future health problems. Cyclical menstruation has no intrinsic biological value or necessity in modern life. Spontaneous decidualization can be prevented by using estrogen-progestin combinations or progestins on their own. Newer and safer hormonal products are now available and should become even safer in the future. Hormonal suppression of spontaneous decidualization should be encouraged in adolescent and young women as a health promotional option.
Collapse
Affiliation(s)
- Norman D Goldstuck
- Department of Obstetrics and Gynaecology, Faculty of Medicine and Health Sciences, University of Stellenbosch, Francie van Zyl Dr, Tygerberg Hospital, Bellville, Cape Town 7505, South Africa.
| |
Collapse
|
9
|
Di Lollo V, Canciello A, Orsini M, Bernabò N, Ancora M, Di Federico M, Curini V, Mattioli M, Russo V, Mauro A, Cammà C, Barboni B. Transcriptomic and computational analysis identified LPA metabolism, KLHL14 and KCNE3 as novel regulators of Epithelial-Mesenchymal Transition. Sci Rep 2020; 10:4180. [PMID: 32144311 PMCID: PMC7060278 DOI: 10.1038/s41598-020-61017-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 02/17/2020] [Indexed: 12/15/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a complex biological program between physiology and pathology. Here, amniotic epithelial cells (AEC) were used as in vitro model of transiently inducible EMT in order to evaluate the transcriptional insights underlying this process. Therefore, RNA-seq was used to identify the differentially expressed genes and enrichment analyses were carried out to assess the intracellular pathways involved. As a result, molecules exclusively expressed in AEC that experienced EMT (GSTA1-1 and GSTM3) or when this process is inhibited (KLHL14 and KCNE3) were identified. Lastly, the network theory was used to obtain a computational model able to recognize putative controller genes involved in the induction and in the prevention of EMT. The results suggested an opposite role of lysophosphatidic acid (LPA) synthesis and degradation enzymes in the regulation of EMT process. In conclusion, these molecules may represent novel EMT regulators and also targets for developing new therapeutic strategies.
Collapse
Affiliation(s)
- V Di Lollo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy. .,Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy.
| | - A Canciello
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy.
| | - M Orsini
- Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - N Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - M Ancora
- Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - M Di Federico
- Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - V Curini
- Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - M Mattioli
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - V Russo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - A Mauro
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - C Cammà
- Molecular biology and genomic Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise "G. Caporale", Teramo, Italy
| | - B Barboni
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
10
|
Duffy DM, Ko C, Jo M, Brannstrom M, Curry TE. Ovulation: Parallels With Inflammatory Processes. Endocr Rev 2019; 40:369-416. [PMID: 30496379 PMCID: PMC6405411 DOI: 10.1210/er.2018-00075] [Citation(s) in RCA: 277] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 11/18/2018] [Indexed: 12/14/2022]
Abstract
The midcycle surge of LH sets in motion interconnected networks of signaling cascades to bring about rupture of the follicle and release of the oocyte during ovulation. Many mediators of these LH-induced signaling cascades are associated with inflammation, leading to the postulate that ovulation is similar to an inflammatory response. First responders to the LH surge are granulosa and theca cells, which produce steroids, prostaglandins, chemokines, and cytokines, which are also mediators of inflammatory processes. These mediators, in turn, activate both nonimmune ovarian cells as well as resident immune cells within the ovary; additional immune cells are also attracted to the ovary. Collectively, these cells regulate proteolytic pathways to reorganize the follicular stroma, disrupt the granulosa cell basal lamina, and facilitate invasion of vascular endothelial cells. LH-induced mediators initiate cumulus expansion and cumulus oocyte complex detachment, whereas the follicular apex undergoes extensive extracellular matrix remodeling and a loss of the surface epithelium. The remainder of the follicle undergoes rapid angiogenesis and functional differentiation of granulosa and theca cells. Ultimately, these functional and structural changes culminate in follicular rupture and oocyte release. Throughout the ovulatory process, the importance of inflammatory responses is highlighted by the commonalities and similarities between many of these events associated with ovulation and inflammation. However, ovulation includes processes that are distinct from inflammation, such as regulation of steroid action, oocyte maturation, and the eventual release of the oocyte. This review focuses on the commonalities between inflammatory responses and the process of ovulation.
Collapse
Affiliation(s)
- Diane M Duffy
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - CheMyong Ko
- Department of Comparative Biosciences, University of Illinois Urbana Champaign, Urbana, Illinois
| | - Misung Jo
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, Kentucky
| | - Mats Brannstrom
- Department of Obstetrics and Gynecology, University of Gothenburg, Gothenburg, Sweden.,Stockholm IVF, Stockholm, Sweden
| | - Thomas E Curry
- Department of Obstetrics and Gynecology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
11
|
Wang W, Do HN, Aupperlee MD, Durairaj S, Flynn EE, Miksicek RJ, Haslam SZ, Schwartz RC. C/EBPβ LIP and c-Jun synergize to regulate expression of the murine progesterone receptor. Mol Cell Endocrinol 2018; 477:57-69. [PMID: 29870755 PMCID: PMC6153074 DOI: 10.1016/j.mce.2018.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 01/11/2023]
Abstract
CCAAT/enhancer binding protein β (C/EBPβ) is required for murine mammary ductal morphogenesis and alveologenesis. Progesterone is critical for proliferation and alveologenesis in adult mammary glands, and there is a similar requirement for progesterone receptor isoform B (PRB) in alveologenesis. We examined C/EBPβ regulation of PR expression. All three C/EBPβ isoforms, including typically inhibitory LIP, transactivated the PR promoter. LIP, particularly, strongly synergized with c-Jun to drive PR transcription. Endogenous C/EBPβ and c-Jun stimulated a PR promoter-reporter and these two factors showed promoter occupancy on the endogenous PR gene. Additionally, LIP overexpression elevated endogenous PR protein expression. In pregnancy, both PRB and the relative abundance of LIP among C/EBPβ isoforms increase. Consistent with a role in PRB expression, in vivo C/EBPβ and PR isoform A expression showed mutually exclusive localization in mammary epithelium, while C/EBPβ and PRB largely co-localized. We suggest a critical role for C/EBPβ, particularly LIP, in PRB expression.
Collapse
Affiliation(s)
- Weizhong Wang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | - Han Ngoc Do
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Mark D Aupperlee
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA; Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Srinivasan Durairaj
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Emily E Flynn
- Genetics Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Richard J Miksicek
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Sandra Z Haslam
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA; Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Richard C Schwartz
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA; Breast Cancer and the Environment Research Program, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
12
|
Bian Y, Lei Y, Wang C, Wang J, Wang L, Liu L, Liu L, Gao X, Li Q. Epigenetic Regulation of miR-29s Affects the Lactation Activity of Dairy Cow Mammary Epithelial Cells. J Cell Physiol 2015; 230:2152-63. [DOI: 10.1002/jcp.24944] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 01/23/2015] [Indexed: 01/17/2023]
Affiliation(s)
- Yanjie Bian
- Research Department of Lactation Biology and Regulation of Mammary Gland Function; Northeast Agricultural University; Harbin 150030 China
| | - Yu Lei
- Research Department of Lactation Biology and Regulation of Mammary Gland Function; Northeast Agricultural University; Harbin 150030 China
| | - Chunmei Wang
- Key Laboratory of Dairy Science of Ministry of Education; Northeast Agricultural University; Harbin 150030 China
| | - Jie Wang
- Key Laboratory of Dairy Science of Ministry of Education; Northeast Agricultural University; Harbin 150030 China
| | - Lina Wang
- Research Department of Lactation Biology and Regulation of Mammary Gland Function; Northeast Agricultural University; Harbin 150030 China
| | - Lili Liu
- Key Laboratory of Dairy Science of Ministry of Education; Northeast Agricultural University; Harbin 150030 China
| | - Lixin Liu
- Key Laboratory of Dairy Science of Ministry of Education; Northeast Agricultural University; Harbin 150030 China
| | - Xuejun Gao
- Key Laboratory of Dairy Science of Ministry of Education; Northeast Agricultural University; Harbin 150030 China
| | - Qingzhang Li
- Research Department of Lactation Biology and Regulation of Mammary Gland Function; Northeast Agricultural University; Harbin 150030 China
- Key Laboratory of Dairy Science of Ministry of Education; Northeast Agricultural University; Harbin 150030 China
| |
Collapse
|
13
|
The role of ovarian sex steroids in metabolic homeostasis, obesity, and postmenopausal breast cancer: molecular mechanisms and therapeutic implications. BIOMED RESEARCH INTERNATIONAL 2015; 2015:140196. [PMID: 25866757 PMCID: PMC4383469 DOI: 10.1155/2015/140196] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/06/2014] [Indexed: 12/20/2022]
Abstract
Obese postmenopausal women have an increased risk of breast cancer and are likely to have a worse prognosis than nonobese postmenopausal women. The cessation of ovarian function after menopause results in withdrawal of ovarian sex steroid hormones, estrogen, and progesterone. Accumulating evidence suggests that the withdrawal of estrogen and progesterone causes homeostasis imbalances, including decreases in insulin sensitivity and leptin secretion and changes in glucose and lipid metabolism, resulting in a total reduction in energy expenditure. Together with a decrease in physical activity and consumption of a high fat diet, these factors significantly contribute to obesity in postmenopausal women. Obesity may contribute to breast cancer development through several mechanisms. Obesity causes localized inflammation, an increase in local estrogen production, and changes in cellular metabolism. In addition, obese women have a higher risk of insulin insensitivity, and an increase in insulin and other growth factor secretion. In this review, we describe our current understanding of the molecular actions of estrogen and progesterone and their contributions to cellular metabolism, obesity, inflammation, and postmenopausal breast cancer. We also discuss how modifications of estrogen and progesterone actions might be used as a therapeutic approach for obesity and postmenopausal breast cancer.
Collapse
|
14
|
Lu J, Reese J, Zhou Y, Hirsch E. Progesterone-induced activation of membrane-bound progesterone receptors in murine macrophage cells. J Endocrinol 2015; 224:183-94. [PMID: 25472814 PMCID: PMC4297269 DOI: 10.1530/joe-14-0470] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Parturition is an inflammatory process mediated to a significant extent by macrophages. Progesterone (P4) maintains uterine quiescence in pregnancy, and a proposed functional withdrawal of P4 classically regulated by nuclear progesterone receptors (nPRs) leads to labor. P4 can affect the functions of macrophages despite the reported lack of expression of nPRs in these immune cells. Therefore, in this study we investigated the effects of the activation of the putative membrane-associated PR on the function of macrophages (a key cell for parturition) and discuss the implications of these findings for pregnancy and parturition. In murine macrophage cells (RAW 264.7), activation of mPRs by P4 modified to be active only extracellularly by conjugation to BSA (P4BSA, 1.0×10(-7) mol/l) caused a pro-inflammatory shift in the mRNA expression profile, with significant upregulation of the expression of cyclooxygenase 2 (COX2 (Ptgs2)), Il1B, and Tnf and downregulation of membrane progesterone receptor alpha (Paqr7) and oxytocin receptor (Oxtr). Pretreatment with PD98059, a MEK1/2 inhibitor, significantly reduced P4BSA-induced expression of mRNA of Il1B, Tnf, and Ptgs2. Inhibition of protein kinase A (PKA) by H89 blocked P4BSA-induced expression of Il1B and Tnf mRNA. P4BSA induced rapid phosphorylation of MEK1/2 and CREB (a downstream target of PKA). This phosphorylation was inhibited by pretreatment with PD98059 and H89, respectively, revealing that MEK1/2 and PKA are two of the components involved in mPR signaling. Taken together, these results indicate that changes in membrane progesterone receptor alpha expression and signaling in macrophages are associated with the inflammatory responses; and that these changes might contribute to the functional withdrawal of P4 related to labor.
Collapse
Affiliation(s)
- Jing Lu
- Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA
| | - Joshua Reese
- Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA
| | - Ying Zhou
- Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA
| | - Emmet Hirsch
- Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA Department of OB/GYNNorthShore University HealthSystem, 2650 Ridge Avenue, Evanston, Illinois 60201, USADepartment of OB/GYNPritzker School of Medicine, University of Chicago, 924 East 57th Street Suite 104, Chicago, Illinois 60637, USA
| |
Collapse
|
15
|
Wang Z, Hou X, Qu B, Wang J, Gao X, Li Q. Pten regulates development and lactation in the mammary glands of dairy cows. PLoS One 2014; 9:e102118. [PMID: 25009983 PMCID: PMC4092105 DOI: 10.1371/journal.pone.0102118] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 06/15/2014] [Indexed: 12/31/2022] Open
Abstract
Pten is a tumor suppressor gene regulating many cellular processes, including growth, adhesion, and apoptosis. In the aim of investigating the role of Pten during mammary gland development and lactation of dairy cows, we analyzed Pten expression levels in the mammary glands of dairy cows by using western blotting, immunohistochemistry, and quantitative polymerase chain reaction (qPCR) assays. Dairy cow mammary epithelial cells (DCMECs) were used to study the function of Pten in vitro. We determined concentrations of β-casein, triglyceride, and lactose in the culture medium following Pten overexpression and siRNA inhibition. To determine whether Pten affected DCMEC viability and proliferation, cells were analyzed by CASY-TT and flow cytometry. Genes involved in lactation-related signaling pathways were detected. Pten expression was also assessed by adding prolactin and glucose to cell cultures. When Pten was overexpressed, proliferation of DCMECs and concentrations for β-casein, triglyceride, and lactose were significantly decreased. Overexpression of Pten down-regulated expression of MAPK, CYCLIN D1, AKT, MTOR, S6K1, STAT5, SREBP1, PPARγ, PRLR, and GLUT1, but up-regulated 4EBP1 in DCMECs. The Pten siRNA inhibition experiments revealed results that opposed those from the gene overexpression experiments. Introduction of prolactin (PRL) increased secretion of β-casein, triglyceride, and lactose, but decreased Pten expression levels. Introduction of glucose also increased β-casein and triglyceride concentrations, but did not significantly alter Pten expression levels. The Pten mRNA and protein expression levels were decreased 0.3- and 0.4-fold in mammary glands of lactating cows producing high quality milk (milk protein >3.0%, milk fat >3.5%), compared with those cows producing low quality milk (milk protein <3.0%, milk fat <3.5%). In conclusion, Pten functions as an inhibitor during mammary gland development and lactation in dairy cows. It can down-regulate DCMECs secretion of β-casein, triglyceride, and lactose, and plays a critical role in lactation related signaling pathways.
Collapse
Affiliation(s)
- Zhuoran Wang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xiaoming Hou
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Bo Qu
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Jie Wang
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xuejun Gao
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Qingzhang Li
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, Heilongjiang, China
- * E-mail:
| |
Collapse
|
16
|
Aupperlee MD, Zhao Y, Tan YS, Leipprandt JR, Bennett J, Haslam SZ, Schwartz RC. Epidermal growth factor receptor (EGFR) signaling is a key mediator of hormone-induced leukocyte infiltration in the pubertal female mammary gland. Endocrinology 2014; 155:2301-13. [PMID: 24693965 PMCID: PMC4020926 DOI: 10.1210/en.2013-1933] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is well documented that macrophages and eosinophils play important roles in normal murine pubertal mammary gland development. Although it is accepted that estrogen (E) and progesterone (P) are key players in mammary gland development, the roles these hormones might play in regulating the actions of leukocytes in that process is an understudied area. We show here that P and E, respectively, induce unique, but overlapping, sets of proinflammatory and angiogenic cytokines and chemokines, in the pubertal female BALB/c mammary gland, as well as induce infiltration of macrophages and eosinophils to the mammary periepithelium. This extends earlier studies showing P induction of proinflammatory products in pubertal and adult mammary epithelial organoids and P-induced in vivo infiltration of leukocytes to the adult mammary periepithelium. Importantly, epidermal growth factor receptor-signaling, which is likely mediated by amphiregulin (Areg), a downstream mediator of E and P, is both necessary and sufficient for both E- and P-induced recruitment of macrophages and eosinophils to the pubertal mammary periepithelium. We further show that receptor activator of nuclear factor κB ligand (RANKL), although not sufficient of itself to cause macrophage and eosinophil recruitment, contributes to an optimal response to P. The potency of Areg is highlighted by the fact that it is sufficient to induce macrophage and eosinophil recruitment at levels equivalent to that induced by either E or P. Our finding of a dominant role for Areg in hormonally induced leukocyte recruitment to the pubertal mammary gland parallels its dominance in regulating ductal outgrowth and its role in P-induced proliferation in the pubertal gland.
Collapse
Affiliation(s)
- Mark D Aupperlee
- Breast Cancer and the Environment Research Program, Departments of Physiology (M.D.A., Y.Z., Y.S.T., J.R.L., J.B., S.Z.H.) and Microbiology and Molecular Genetics (R.C.S.), Michigan State University, East Lansing, Michigan 48824
| | | | | | | | | | | | | |
Collapse
|
17
|
Wang L, Lin Y, Bian Y, Liu L, Shao L, Lin L, Qu B, Zhao F, Gao X, Li Q. Leucyl-tRNA synthetase regulates lactation and cell proliferation via mTOR signaling in dairy cow mammary epithelial cells. Int J Mol Sci 2014; 15:5952-69. [PMID: 24722568 PMCID: PMC4013607 DOI: 10.3390/ijms15045952] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 03/28/2014] [Accepted: 03/28/2014] [Indexed: 01/06/2023] Open
Abstract
The role of LeuRS, an aminoacyl-tRNA synthetase, as an intracellular l-leucine sensor for the mTORC1 pathway has been the subject of much research recently. Despite this, the association between LeuRS and lactation in dairy cow mammary epithelial cells (DCMECs) remains unknown. In this study, we found that LeuRS expression in mammary gland tissue was significantly higher during lactation than pregnancy. Moreover, our data demonstrates that LeuRS is localized in the cytoplasm. Treatment with leucine increased DCMECs viability and proliferation, as well as mammalian target of rapamycin (mTOR), p-mTOR, ribosomal protein S6 kinase 1 (S6K1), p-S6K1, β-Casein, sterol regulatory element binding protein 1c (SREBP-1c), glucose transporter 1 (GLUT1), and Cyclin D1 mRNA and protein expression. Secretion of lactose and triglyceride were also increased. siRNA-mediated knockdown of LeuRS led to reduction in all of these processes. Based on these data, LeuRS up-regulates the mTOR pathway to promote proliferation and lactation of DCMECs in response to changes in the intracellular leucine concentration.
Collapse
Affiliation(s)
- Lina Wang
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | - Ye Lin
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | - Yanjie Bian
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | - Lili Liu
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | - Li Shao
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | - Lin Lin
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | - Bo Qu
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | - Feng Zhao
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | - Xuejun Gao
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| | - Qingzhang Li
- Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
18
|
Obr AE, Grimm SL, Bishop KA, Pike JW, Lydon JP, Edwards DP. Progesterone receptor and Stat5 signaling cross talk through RANKL in mammary epithelial cells. Mol Endocrinol 2013; 27:1808-24. [PMID: 24014651 PMCID: PMC3805851 DOI: 10.1210/me.2013-1077] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 08/21/2013] [Indexed: 02/08/2023] Open
Abstract
Progesterone (P4) stimulates proliferation of the mammary epithelium by a mechanism that involves paracrine signaling mediated from progesterone receptor (PR)-positive to neighboring PR-negative cells. Here we used a primary mouse mammary epithelial cell (MEC) culture system to define the molecular mechanism by which P4 regulates the expression of target gene effectors of proliferation including the paracrine factor receptor and activator of nuclear factor κB ligand (RANKL). MECs from adult virgin mice grown and embedded in three-dimensional basement-membrane medium resemble mammary ducts in vivo structurally and with respect to other properties including a heterogeneous pattern of PR expression, P4 induction of RANKL and other target genes in a PR-dependent manner, and a proliferative response to progestin. RANKL was demonstrated to have multiple functional P4-responsive enhancers that bind PR in a hormone-dependent manner as detected by chromatin immunoprecipitation assay. P4 also stimulated recruitment of signal transducer and activator of transcription (Stat)5a to RANKL enhancers through an apparent tethering with PR. Analysis of primary MECs from Stat5a knockout mice revealed that P4 induction of RANKL and a broad range of other PR target genes required Stat5a, as did P4-stimulated cell proliferation. In the absence of Stat5a, PR binding was lost at selective RANKL enhancers but was retained with others, suggesting that Stat5a acts to facilitate PR DNA binding at selective sites and to function as a coactivator with DNA-bound PR at others. These results show that RANKL is a direct PR target gene and that Stat5a has a novel role as a cofactor in PR-mediated transcriptional signaling in the mammary gland.
Collapse
Affiliation(s)
- Alison E Obr
- PhD, Department of Molecular & Cellular Biology, Baylor College of Medicine, BCM Box 130, One Baylor Plaza, Houston, Texas 77030.
| | | | | | | | | | | |
Collapse
|
19
|
Jeurink P, van Bergenhenegouwen J, Jiménez E, Knippels L, Fernández L, Garssen J, Knol J, Rodríguez J, Martín R. Human milk: a source of more life than we imagine. Benef Microbes 2013; 4:17-30. [DOI: 10.3920/bm2012.0040] [Citation(s) in RCA: 225] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The presence of bacteria in human milk has been acknowledged since the seventies. For a long time, microbiological analysis of human milk was only performed in case of infections and therefore the presence of non-pathogenic bacteria was yet unknown. During the last decades, the use of more sophisticated culture-dependent and -independent techniques, and the steady development of the -omic approaches are opening up the new concept of the ‘milk microbiome’, a complex ecosystem with a greater diversity than previously anticipated. In this review, possible mechanisms by which bacteria can reach the mammary gland (contamination versus active migration) are discussed. In addition, the potential roles of human milk for both infant and maternal health are summarised. A better understanding of the link between the milk microbiome and health benefit, the potential factors influencing this relationship and whether or not it can be influenced by nutrition is required to open new avenues in the field of pregnancy and lactation.
Collapse
Affiliation(s)
- P.V. Jeurink
- Danone Research, Centre for Specialised Nutrition, P.O. Box 7005, 6700 CA Wageningen, the Netherlands
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, P.O. Box 80082, 3508 TB Utrecht, the Netherlands
| | - J. van Bergenhenegouwen
- Danone Research, Centre for Specialised Nutrition, P.O. Box 7005, 6700 CA Wageningen, the Netherlands
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, P.O. Box 80082, 3508 TB Utrecht, the Netherlands
| | - E. Jiménez
- Dpto Nutrición, Bromatología y Tecnología de los Alimentos, UCM, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - L.M.J. Knippels
- Danone Research, Centre for Specialised Nutrition, P.O. Box 7005, 6700 CA Wageningen, the Netherlands
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, P.O. Box 80082, 3508 TB Utrecht, the Netherlands
| | - L. Fernández
- Dpto Nutrición, Bromatología y Tecnología de los Alimentos, UCM, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - J. Garssen
- Danone Research, Centre for Specialised Nutrition, P.O. Box 7005, 6700 CA Wageningen, the Netherlands
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, P.O. Box 80082, 3508 TB Utrecht, the Netherlands
| | - J. Knol
- Danone Research, Centre for Specialised Nutrition, P.O. Box 7005, 6700 CA Wageningen, the Netherlands
- Laboratory of Microbiology, Wageningen University, P.O. Box 8033, 6700 EJ Wageningen, the Netherlands
| | - J.M. Rodríguez
- Dpto Nutrición, Bromatología y Tecnología de los Alimentos, UCM, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - R. Martín
- Danone Research, Centre for Specialised Nutrition, P.O. Box 7005, 6700 CA Wageningen, the Netherlands
| |
Collapse
|
20
|
Proteomic and functional analyses reveal MAPK1 regulates milk protein synthesis. Molecules 2012; 18:263-75. [PMID: 23271465 PMCID: PMC6270553 DOI: 10.3390/molecules18010263] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 12/11/2012] [Accepted: 12/18/2012] [Indexed: 01/15/2023] Open
Abstract
L-Lysine (L-Lys) is an essential amino acid that plays fundamental roles in protein synthesis. Many nuclear phosphorylated proteins such as Stat5 and mTOR regulate milk protein synthesis. However, the details of milk protein synthesis control at the transcript and translational levels are not well known. In this current study, a two-dimensional gel electrophoresis (2-DE)/MS-based proteomic technology was used to identify phosphoproteins responsible for milk protein synthesis in dairy cow mammary epithelial cells (DCMECs). The effect of L-Lys on DCMECs was analyzed by CASY technology and reversed phase high performance liquid chromatography (RP-HPLC). The results showed that cell proliferation ability and β-casein expression were enhanced in DCMECs treated with L-Lys. By phosphoproteomics analysis, six proteins, including MAPK1, were identified up-expressed in DCMECs treated with 1.2 mM L-Lys for 24 h, and were verified by quantitative real-time PCR (qRT-PCR) and western blot. Overexpression and siRNA inhibition of MAPK1 experiments showed that MAPK1 upregulated milk protein synthesis through Stat5 and mTOR pathway. These findings that MAPK1 involves in regulation of milk synthesis shed new insights for understanding the mechanisms of milk protein synthesis.
Collapse
|
21
|
Lu L, Gao X, Li Q, Huang J, Liu R, Li H. Comparative phosphoproteomics analysis of the effects of L-methionine on dairy cow mammary epithelial cells. CANADIAN JOURNAL OF ANIMAL SCIENCE 2012. [DOI: 10.4141/cjas2012-063] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lu, L., Gao, X., Li, Q., Huang, J., Liu, R. and Li, H. 2012. Comparative phosphoproteomics analysis of the effects of L-methionine on dairy cow mammary epithelial cells. Can. J. Anim. Sci. 92: 433–442. L-methionine is an essential amino acid that plays fundamental roles in protein synthesis. Many nuclear phosphorylated proteins such as Stat5 (signal transducer and activator of transcription 5) and mTOR (mammalian target of rapamycin) regulate milk protein synthesis. But a comprehensive understanding of transcriptional and posttranscriptional regulation of milk protein synthesis is lacking. In the current study, two-dimensional gel electrophoresis (2-DE)/MS-based proteomics analysis was used to identify phosphoproteins responsible for milk protein synthesis in dairy cow mammary epithelial cells (DCMECs). The effects of L-methionine on DCMECs were analyzed by CASY (Counter Analyser System) technique, reversed phase high performance liquid chromatography. The results showed that rate of cell proliferation and expression of β-casein were increased in DCMECs treated with 0.6 mM L-methionine for 24 h. Five proteins for which expression was significantly increased in DCMECs were selected, and their expression changes were verified by quantitative real-time PCR and Western blot analysis. The five up-regulated expressed phosphoproteins included Staphylococcal nuclease domain-containing protein 1(SND1), Septin-6, Glycyl-tRNA synthetase (GARS), Twinfilin-1 and eukaryotic elongation factor1-beta (eEF1B). This study revealed that availability of L-methionine influences the levels of nuclear phosphorylated proteins of DCMECs which opens a new avenue for the study of the molecular mechanism linking to milk protein synthesis.
Collapse
Affiliation(s)
- Limin Lu
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Xuejun Gao
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Qingzhang Li
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Jianguo Huang
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Rong Liu
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| | - Huiming Li
- The Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
22
|
Meyer G, Leipprandt J, Xie J, Aupperlee MD, Haslam SZ. A potential role of progestin-induced laminin-5/α6-integrin signaling in the formation of side branches in the mammary gland. Endocrinology 2012; 153:4990-5001. [PMID: 22910029 PMCID: PMC3512027 DOI: 10.1210/en.2012-1518] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/31/2012] [Indexed: 11/19/2022]
Abstract
Mammary organoids from adult mice produce tubules, analogous to mammary ducts in vivo, in response to hepatocyte growth factor (HGF) when cultured in collagen gels. The combination of HGF plus progestin (R5020) causes reduced tubule number and length. We hypothesized that the inhibitory effect on tubulogenesis was due to progestin-mediated alteration of HGF/c-Met signaling. Using molecular inhibitors and short hairpin RNA, it was determined that HGF activation of Ras-related C3 botulinum toxin substrate (Rac1) was required for the formation of cytoplasmic extensions, the first step of tubulogenesis, and that Rac1 activity was Src kinase (Src) and focal adhesion kinase (FAK) dependent. The highly novel finding was that R5020 reduced tubulogenesis by up-regulating and increasing extracellular laminin and α6-integrin ligation to reduce activation of the Src, focal adhesion kinase, and Rac1 pathway. Receptor activator of nuclear factor-κB ligand, another progesterone-induced paracrine factor, did not replicate this effect of R5020. The inhibitory effect of R5020 on tubulogenesis was likely mediated through progesterone receptor (PR) isoform A (PRA), because PRA is the predominant PR isoform expressed in the organoids, and the progestin-induced effect was prevented by the PR antagonist RU486. These results provide a plausible mechanism that explains progestin/PRA-mediated blunting of HGF-induced tubulogenesis in vitro and is proposed to be relevant to progesterone/PRA-induced side-branching in vivo during pregnancy.
Collapse
Affiliation(s)
- Gabriele Meyer
- Department of Physiology and Breast Cancer and the Environment Research Center, Michigan State University, East Lansing, Michigan 48824, USA
| | | | | | | | | |
Collapse
|
23
|
Jacobsen BM, Horwitz KB. Progesterone receptors, their isoforms and progesterone regulated transcription. Mol Cell Endocrinol 2012; 357:18-29. [PMID: 21952082 PMCID: PMC3272316 DOI: 10.1016/j.mce.2011.09.016] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 08/11/2011] [Accepted: 09/11/2011] [Indexed: 01/16/2023]
Abstract
This review discusses mechanisms by which progesterone receptors (PR) regulate transcription. We examine available data in different species and tissues regarding: (1) regulation of PR levels; and (2) expression profiling of progestin-regulated genes by total PRs, or their PRA and PRB isoforms. (3) We address current views about the composition of progesterone response elements, and postulate that PR monomers acting through "half-site" elements are common, entailing cooperativity with neighboring DNA-bound transcription factors. (4) We summarize transcription data for multiple progestin-regulated promoters as directed by total PR, or PRA vs. PRB. We conclude that current models and methods used to study PR function are problematical, and recommend that future work employ cells and receptors appropriate to the species, focusing on analyses of the effects of endogenous receptors targeting endogenous genes in native chromatin.
Collapse
Affiliation(s)
- Britta M Jacobsen
- Department of Medicine/Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States.
| | | |
Collapse
|
24
|
Axlund SD, Sartorius CA. Progesterone regulation of stem and progenitor cells in normal and malignant breast. Mol Cell Endocrinol 2012; 357:71-9. [PMID: 21945473 PMCID: PMC3288619 DOI: 10.1016/j.mce.2011.09.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2011] [Revised: 08/26/2011] [Accepted: 09/11/2011] [Indexed: 12/17/2022]
Abstract
Progesterone plays an important, if not controversial, role in mammary epithelial cell proliferation and differentiation. Evidence supports that progesterone promotes rodent mammary carcinogenesis under some conditions, progesterone receptors (PR) are necessary for murine mammary gland tumorigenesis, and exogenous progestin use in post-menopausal women increases breast cancer risk. Thus, the progesterone/PR signaling axis can promote mammary tumorigenesis, albeit in a context-dependent manner. A mechanistic basis for the tumor promoting actions of progesterone has thus far remained unknown. Recent studies, however, have identified a novel role for progesterone in controlling the number and function of stem and progenitor cell populations in the normal human and mouse mammary glands, and in human breast cancers. These discoveries promise to reshape our perception of progesterone function in the mammary gland, and have spawned new hypotheses for how progestins may increase the risk of breast cancer. Here we review studies on progesterone regulation of mammary stem cells in normal and malignant tissue, and their implications for breast cancer risk, tumorigenesis, and tumor behavior.
Collapse
Affiliation(s)
| | - Carol A. Sartorius
- Corresponding author at: University of Colorado Anschutz Medical Center, 12801 E 17th Ave. MS8104, Aurora, CO 80045, United States. Tel: +1 303-724-3937; Fax: +1 303-724-3712. (C.A. Sartorius)
| |
Collapse
|
25
|
Haslam SZ, Schwartz RC. Is there a link between a high-fat diet during puberty and breast cancer risk? ACTA ACUST UNITED AC 2011; 7:1-3. [PMID: 21175383 DOI: 10.2217/whe.10.83] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
26
|
Cui W, Li Q, Feng L, Ding W. MiR-126-3p regulates progesterone receptors and involves development and lactation of mouse mammary gland. Mol Cell Biochem 2011; 355:17-25. [PMID: 21526342 DOI: 10.1007/s11010-011-0834-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Accepted: 04/15/2011] [Indexed: 01/19/2023]
Abstract
MicroRNAs (miRNAs) are small (18-22 nucleotide) non-coding, endogenous regulatory RNA molecules, and they regulate gene expression at the post-transcriptional level through binding to their target mRNAs by base-pairing and subsequently inducing either translational repression or mRNA destabilization by plants, animals, and some viruses. In this study, combining microarray techniques with qRT-PCR, we found that miR-126-3p expression showed significant difference in the mouse mammary cycle during pregnancy, particularly on transition from pregnancy to lactation. Bioinformatics were used to predict target gene of miR-126-3p, and luciferase activity assay to test it, it showed that the progesterone receptor (PGR) 3'UTR is directly targeted by miR-126-3p. In this study, mouse mammary epithelial cells as cell model in vitro were used to study the function of miR-126-3p. Using gene silencing and over-expression for miR-126-3p, the expression of PGR protein and the secretion of casein were detected by western blotting and HPLC, respectively. To determine whether miR-126-3p can affect mouse mammary epithelial cells viability, cells were analyzed by CASY-YY. In conclusion, PGR gene confirmed miR-126-3p target genes through luciferase activity and western blotting. And miR-126-3p could also inhibit proliferation of mouse mammary epithelial cells (P < 0.01) and expression of β-casein (P < 0.01), and down-regulate PGR protein (P < 0.05). Our results suggested that miR-126-3p inhibited expression of PGR protein level as well as the proliferation of mammary epithelial cells, therefore miR-126-3p could play an important role in the process of mammary gland development.
Collapse
Affiliation(s)
- Wei Cui
- Key Laboratory of Dairy Science of Education Ministry, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| | | | | | | |
Collapse
|
27
|
Coussens LM, Pollard JW. Leukocytes in mammary development and cancer. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a003285. [PMID: 21123394 DOI: 10.1101/cshperspect.a003285] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Leukocytes, of both the innate and adaptive lineages, are normal cellular components of all tissues. These important cells not only are critical for regulating normal tissue homeostasis, but also are significant paracrine regulators of all physiologic and pathologic tissue repair processes. This article summarizes recent insights regarding the trophic roles of leukocytes at each stage of mammary gland development and during cancer development, with a focus on Murids and humans.
Collapse
Affiliation(s)
- Lisa M Coussens
- Department of Pathology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 94143, USA
| | | |
Collapse
|
28
|
Poffet F, Abraham S, Taramarcaz P, Fontao L, Borradori L. Autoimmune Progesterone Dermatitis: Potential Role of Cutaneous Angiogenin Expression? Dermatology 2011; 223:32-5. [DOI: 10.1159/000329427] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
29
|
MPA-induced gene expression and stromal and parenchymal gene expression profiles in luminal murine mammary carcinomas with different hormonal requirements. Breast Cancer Res Treat 2010; 129:49-67. [DOI: 10.1007/s10549-010-1185-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 09/17/2010] [Indexed: 12/21/2022]
|
30
|
Hilton HN, Kalyuga M, Cowley MJ, Alles MC, Lee HJ, Caldon CE, Blazek K, Kaplan W, Musgrove EA, Daly RJ, Naylor MJ, Graham JD, Clarke CL, Ormandy CJ. The antiproliferative effects of progestins in T47D breast cancer cells are tempered by progestin induction of the ETS transcription factor Elf5. Mol Endocrinol 2010; 24:1380-92. [PMID: 20519331 DOI: 10.1210/me.2009-0516] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Prolactin and progesterone act together to regulate mammary alveolar development, and both hormones have been implicated in breast cancer initiation and progression. Here we show that Elf5, a prolactin-induced ETS transcription factor that specifies the mammary secretory cell lineage, is also induced by progestins in breast cancer cells via a direct mechanism. To define the transcriptional response to progestin elicited via Elf5, we made an inducible Elf5 short hairpin-RNA knock-down model in T47D breast cancer cells and used it to prevent the progestin-induction of Elf5. Functional analysis of Affymetrix gene expression data using Gene Ontologies and Gene Set Enrichment Analysis showed enhancement of the progestin effects on cell cycle gene expression. Cell proliferation assays showed a more efficacious progestin-induced growth arrest when Elf5 was kept at baseline levels. These results showed that progestin induction of Elf5 expression tempered the antiproliferative effects of progestins in T47D cells, providing a further mechanistic link between prolactin and progestin in the regulation of mammary cell phenotype.
Collapse
Affiliation(s)
- H N Hilton
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|