1
|
Zheng S, Chen X, Fang J, Li Y, Xiao X, Zhang X, Zhang L, Cheng Y, Hao L. The role of insulin-like growth factor-1 in lactation. Gene 2025; 962:149577. [PMID: 40404070 DOI: 10.1016/j.gene.2025.149577] [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: 01/03/2025] [Revised: 04/21/2025] [Accepted: 05/18/2025] [Indexed: 05/24/2025]
Abstract
Generally, Insulin-like growth factor 1 (IGF-1) is believed to regulate lactation activity by promoting cell proliferation and differentiation. With the advancement of research, IGF-1 has been discovered to play an important role in different stages of lactation. In actual animal production, lactation ability directly affects milk yield and milk quality, which not only affects the survival and future growth of pups, but also is an important economic trait of some animals. In this paper, it is introduced that IGF-1 plays an important role in the whole lactation process, and what factors are involved in the regulation of IGF-1 in this process and how to improve lactation ability through IGF-1 in animal production, providing a theoretical basis for further exploration of IGF-1 in lactation, and also brings a theoretical foundation for the improvement of animal lactation ability.
Collapse
Affiliation(s)
- Shuo Zheng
- College of Animal Science, Jilin University, Changchun 130062, China
| | - Xi Chen
- College of Animal Science, Jilin University, Changchun 130062, China
| | - JiaY Fang
- College of Animal Science, Jilin University, Changchun 130062, China
| | - Yi Li
- College of Animal Science, Jilin University, Changchun 130062, China
| | - XingY Xiao
- College of Animal Science, Jilin University, Changchun 130062, China
| | - XunM Zhang
- College of Animal Science, Jilin University, Changchun 130062, China
| | - LiB Zhang
- College of Animal Science, Jilin University, Changchun 130062, China
| | - YunY Cheng
- College of Public Health, Jilin University, Changchun 130061, China
| | - LinL Hao
- College of Animal Science, Jilin University, Changchun 130062, China.
| |
Collapse
|
2
|
Zhu Q, Lu X, Chen M, Zhang T, Shi M, Yao W, Zhang H, Gao R, Li X, Zhou Y, Liao S. IGFBP5 affects cardiomyocyte survival and functional recovery in mice following myocardial ischemia. Commun Biol 2024; 7:1594. [PMID: 39613849 DOI: 10.1038/s42003-024-07304-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
Insulin-like growth factor-binding protein 5 (IGFBP5) has been shown to be useful for the diagnosis and treatment of multiple tumors and cerebrovascular diseases. However, it is unknown whether IGFBP5 is involved in myocardial repair following myocardial infarction (MI). Here we show high expression of IGFBP5 in multiple models of ischemic and hypoxic injury. IGFBP5 affected the proliferation of neonatal rat cardiomyocytes (NRCMs) and the cardiomyocyte apoptosis induced by oxygen-glucose deprivation (OGD). Subsequently, heart-specific IGFBP5 knockdown inhibited myocardial apoptosis and increased cardiomyocyte proliferation in mice with MI. During the chronic remodeling stage, heart-specific regulation of IGFBP5 ameliorated pathological cardiac remodeling and dysfunction. Mechanistically, IGFBP5 regulated cardiomyocyte survival through the insulin-like growth factor 1 (IGF1) receptor (IGF1R)/protein kinase B (PKB/AKT) pathway. In summary, our results provide mechanistic insights into the effect of IGFBP5 on cardiomyocyte during cardiac repair. IGFBP5 may represent a therapeutic target for myocardial ischemic injury.
Collapse
Affiliation(s)
- Qingqing Zhu
- Division of Cardiac Surgery Intensive Care Unit, Department of Cardiac Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xinyi Lu
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Mengli Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Ting Zhang
- Department of Cardiovascular Medicine, The Air Force Hospital from Eastern Theater, Nanjing, China
| | - Mengsha Shi
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wenming Yao
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Haifeng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Rongrong Gao
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Xinli Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yanli Zhou
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China.
| | - Shengen Liao
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China.
| |
Collapse
|
3
|
Hoeflich A, Galow AM, Brenmoehl J, Hadlich F. Growth and development of the mammary gland in mice-control of the insulin-like growth factor system by hormones and metalloproteases, and putative interference with micro RNAs. Anim Front 2023; 13:77-85. [PMID: 37324202 PMCID: PMC10266761 DOI: 10.1093/af/vfad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023] Open
Affiliation(s)
| | - Anne-Marie Galow
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl Allee 2, 18196 Dummerstorf, Germany
| | - Julia Brenmoehl
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl Allee 2, 18196 Dummerstorf, Germany
| | - Frieder Hadlich
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl Allee 2, 18196 Dummerstorf, Germany
| |
Collapse
|
4
|
Cohick WS. The role of the IGF system in mammary physiology of ruminants. Domest Anim Endocrinol 2022; 79:106709. [PMID: 35078102 DOI: 10.1016/j.domaniend.2021.106709] [Citation(s) in RCA: 2] [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: 08/29/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022]
Abstract
The IGF system plays a central role in all stages of mammary development, lactation and involution. IGFs exert their effects on the mammary gland through both endocrine and paracrine/autocrine mechanisms and the importance of circulating versus local IGF action remains an open question, especially in ruminants. At the whole organ level, a critical role for IGFs in ductal morphogenesis and lobuloalveolar development has been established, while at the cellular level the ability of IGFs to stimulate cell proliferation and control cell survival contributes to the number of milk-secreting cells in the gland. Much of this work has been conducted in rodents which provide an affordable research model and allow for genetic manipulation of specific components of the IGF system. Research into the role of the IGF system in dairy cows has generally supported information obtained with rodents though large gaps in our knowledge remain and species differences are not well defined. Examples include whether exogenous somatotropin exerts its effects on the mammary gland through local IGF-1 synthesis which is accepted dogma in rodents, what the role of IGF-1 versus IGF-2 is in the mammary gland, and how the IGFBPs regulate IGF bioactivity. This last area is particularly under-investigated in ruminants both at the whole animal and the cellular and molecular levels. Given that the IGF system may underlie many management practices that could contribute to enhancing productive efficiency of lactation, more research into the basic biology of this important system is warranted.
Collapse
Affiliation(s)
- Wendie S Cohick
- Rutgers, The State University of New Jersey, Department of Animal Science, New Brunswick, NJ 08901, USA.
| |
Collapse
|
5
|
Qian Y, Berryman DE, Basu R, List EO, Okada S, Young JA, Jensen EA, Bell SRC, Kulkarni P, Duran-Ortiz S, Mora-Criollo P, Mathes SC, Brittain AL, Buchman M, Davis E, Funk KR, Bogart J, Ibarra D, Mendez-Gibson I, Slyby J, Terry J, Kopchick JJ. Mice with gene alterations in the GH and IGF family. Pituitary 2022; 25:1-51. [PMID: 34797529 PMCID: PMC8603657 DOI: 10.1007/s11102-021-01191-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 01/04/2023]
Abstract
Much of our understanding of GH's action stems from animal models and the generation and characterization of genetically altered or modified mice. Manipulation of genes in the GH/IGF1 family in animals started in 1982 when the first GH transgenic mice were produced. Since then, multiple laboratories have altered mouse DNA to globally disrupt Gh, Ghr, and other genes upstream or downstream of GH or its receptor. The ability to stay current with the various genetically manipulated mouse lines within the realm of GH/IGF1 research has been daunting. As such, this review attempts to consolidate and summarize the literature related to the initial characterization of many of the known gene-manipulated mice relating to the actions of GH, PRL and IGF1. We have organized the mouse lines by modifications made to constituents of the GH/IGF1 family either upstream or downstream of GHR or to the GHR itself. Available data on the effect of altered gene expression on growth, GH/IGF1 levels, body composition, reproduction, diabetes, metabolism, cancer, and aging are summarized. For the ease of finding this information, key words are highlighted in bold throughout the main text for each mouse line and this information is summarized in Tables 1, 2, 3 and 4. Most importantly, the collective data derived from and reported for these mice have enhanced our understanding of GH action.
Collapse
Affiliation(s)
- Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Darlene E Berryman
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Shigeru Okada
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Pediatrics, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Jonathan A Young
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Elizabeth A Jensen
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Translational Biomedical Sciences Doctoral Program, Ohio University, Athens, OH, USA
| | - Stephen R C Bell
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Prateek Kulkarni
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | | | - Patricia Mora-Criollo
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Translational Biomedical Sciences Doctoral Program, Ohio University, Athens, OH, USA
| | - Samuel C Mathes
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Alison L Brittain
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | - Mat Buchman
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Emily Davis
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | - Kevin R Funk
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, USA
| | - Jolie Bogart
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - Diego Ibarra
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Chemistry and Biochemistry, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - Isaac Mendez-Gibson
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- College of Health Sciences and Professions, Ohio University, Athens, OH, USA
| | - Julie Slyby
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - Joseph Terry
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH, USA.
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| |
Collapse
|
6
|
Jenkins EC, Brown SO, Germain D. The Multi-Faced Role of PAPP-A in Post-Partum Breast Cancer: IGF-Signaling is Only the Beginning. J Mammary Gland Biol Neoplasia 2020; 25:181-189. [PMID: 32901383 DOI: 10.1007/s10911-020-09456-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/24/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Insulin-like growth factor (IGF) signaling and control of local bioavailability of free IGF by the IGF binding proteins (IGFBP) are important regulators of both mammary development and breast cancer. A recent genome-wide association study (GWAS) identified small nucleotide polymorphisms that reduce the expression of IGFBP-5 as a risk factor of developing breast cancer. This observation suggests that genetic alterations leading to a decreased level of IGFBP-5 may also contribute to breast cancer. In the current review, we focus on Pregnancy-Associated Plasma Protein A (PAPP-A), a protease involved in the degradation of IGFBP-5. PAPP-A is overexpressed in the majority of breast cancers but its role in cancer has only begun to be explored. More specifically, this review aims at highlighting the role of post-partum involution in the oncogenic function of PAPP-A. Notably, we summarize recent studies indicating that PAPP-A plays a role not only in the degradation of IGFBP-5 but also in the deposition of collagen and activation of the collagen receptor discoidin 2 (DDR2) during post-partum involution. Finally, considering the immunosuppressive microenvironment of post-partum involution, we also discuss the unexpected finding made in Ewing Sarcoma that PAPP-A plays a role in immune evasion. While the immunosuppressive role of PAPP-A in breast cancer remains to be determined, collectively these studies highlight the multifaced role of PAPP-A in cancer that extends well beyond its effect on IGF-signaling.
Collapse
Affiliation(s)
- Edmund Charles Jenkins
- Department of Medicine, Division of Hematology/ Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA
| | - Samantha O Brown
- Department of Medicine, Division of Hematology/ Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA
| | - Doris Germain
- Department of Medicine, Division of Hematology/ Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA.
| |
Collapse
|
7
|
Zhang X, Cheng Z, Wang L, Jiao B, Yang H, Wang X. MiR-21-3p Centric Regulatory Network in Dairy Cow Mammary Epithelial Cell Proliferation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:11137-11147. [PMID: 31532202 DOI: 10.1021/acs.jafc.9b04059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
MicroRNA-mediated gene regulation is important for the development of the mammary gland and the lactating process. A previous study has shown that the expression of microRNA-21 (miR-21) is different in the dry and early lactation period of the dairy cow mammary gland, but the molecular mechanisms underlying the lactation cycle are not fully understood. Here, the function of miR-21-3p on bovine mammary gland epithelial cells (BMECs) was detected by MTT assay and flow cytometry analysis, which showed that miR-21-3p significantly promoted the cell viability and proliferation. Then, the regulating mechanism of miR-21-3p on cell viability and proliferation was elucidated. Dual luciferase assay, RT-qPCR, and Western blot results revealed that IGFBP5 was a target gene of miR-21-3p. It was known that lncRNA could act as a competing endogenous RNA to sequester miRNAs and reduce the regulatory effect of miRNA-targeted genes. Based on our previous lncRNA-seq data and bioinformatics analysis, lncRNA NONBTAT017009.2 was potentially associated with miR-21-3p, and its expression was specifically inhibited with the transfection of miR-21-3p mimic into BMECs. Inversely, the overexpression of NONBTAT017009.2 significantly decreased the expression level of miR-21-3p in BMECs, while the expression of IGFBP5, the target gene of miR-21-3p, was significantly upregulated. In addition, the promoter region of miR-21 contained two STAT3 binding sites, and the dual luciferase reporter assays revealed that the overexpression of STAT3 significantly reduced the promoter activity of miR-21, implying that the transcription factor STAT3 may act as an upstream regulator affecting the regulation process of miR-21-3p. The overexpression of STAT3 significantly inhibited the expression of miR-21-3p, while the mRNA expression of IGFBP5 was significantly increased compared with the control group. Besides, there are no STAT3 binding sites in the promoter region of IGFBP5 as we predicted by gene-regulation and JASPAR software. Therefore, it could infer that STAT3 might regulate the expression of IGFBP5 by miR-21-3p. Taken together, these results established a regulatory network of miR-21-3p to illustrate the regulating mechanism on promoting cow mammary epithelial cell proliferation.
Collapse
Affiliation(s)
- Xiaolan Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi 712100 , China
| | - Zixi Cheng
- The Middle School Attached to Northwestern Polytechnical University , Xi'an , Shaanxi 710072 , China
| | - Lixian Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi 712100 , China
| | - Beilei Jiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi 712100 , China
| | - Hua Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi 712100 , China
| | - Xin Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi 712100 , China
| |
Collapse
|
8
|
Zhao X, Ponchon B, Lanctôt S, Lacasse P. Invited review: Accelerating mammary gland involution after drying-off in dairy cattle. J Dairy Sci 2019; 102:6701-6717. [PMID: 31202662 DOI: 10.3168/jds.2019-16377] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/22/2019] [Indexed: 01/20/2023]
Abstract
Bovine mammary gland involution, as a part of the reproductive cycle in dairy cows, is a very important remodeling transformation of the mammary gland for the subsequent lactation. There is considerable incentive to accelerate mammary gland involution to improve udder health, shorten the dry period, and simplify the management process by reducing dietary changes. The complex process of mammary involution is characterized by morphological changes in the epithelial cells and mammary tissue, changes in the composition of mammary secretions, and changes in the integrity of tight junctions. Involution is facilitated by elements of the immune system and several types of proteases and is coordinated by various types of hormones. This review first describes the involution process and then argues for the need to accelerate it. Last, this review focuses on various intervention methods for accelerating involution. Our aim is to provide a comprehensive overview of bovine mammary gland involution as well as potential techniques and new opinions for dry cow management.
Collapse
Affiliation(s)
- X Zhao
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada H9X 3V9.
| | - B Ponchon
- Department of Animal Science, McGill University, Sainte-Anne-de-Bellevue, QC, Canada H9X 3V9
| | - S Lanctôt
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada J1M 0C8
| | - P Lacasse
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC, Canada J1M 0C8
| |
Collapse
|
9
|
Abstract
Insulin-like growth factor-binding proteins (IGFBPs) 1-6 bind IGFs but not insulin with high affinity. They were initially identified as serum carriers and passive inhibitors of IGF actions. However, subsequent studies showed that, although IGFBPs inhibit IGF actions in many circumstances, they may also potentiate these actions. IGFBPs are widely expressed in most tissues, and they are flexible endocrine and autocrine/paracrine regulators of IGF activity, which is essential for this important physiological system. More recently, individual IGFBPs have been shown to have IGF-independent actions. Mechanisms underlying these actions include (i) interaction with non-IGF proteins in compartments including the extracellular space and matrix, the cell surface and intracellular space, (ii) interaction with and modulation of other growth factor pathways including EGF, TGF-β and VEGF, and (iii) direct or indirect transcriptional effects following nuclear entry of IGFBPs. Through these IGF-dependent and IGF-independent actions, IGFBPs modulate essential cellular processes including proliferation, survival, migration, senescence, autophagy and angiogenesis. They have been implicated in a range of disorders including malignant, metabolic, neurological and immune diseases. A more complete understanding of their cellular roles may lead to the development of novel IGFBP-based therapeutic opportunities.
Collapse
Affiliation(s)
- L A Bach
- Department of Medicine (Alfred)Monash University, Melbourne, Australia
- Department of Endocrinology and DiabetesAlfred Hospital, Melbourne, Australia
| |
Collapse
|
10
|
Meyer Z, Höflich C, Wirthgen E, Olm S, Hammon HM, Hoeflich A. Analysis of the IGF-system in milk from farm animals - Occurrence, regulation, and biomarker potential. Growth Horm IGF Res 2017; 35:1-7. [PMID: 28544872 DOI: 10.1016/j.ghir.2017.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/04/2017] [Accepted: 05/09/2017] [Indexed: 12/18/2022]
Abstract
IGFs and IGF-binding proteins (IGFBPs) are abundantly present in milk and in dairy products. Compared to the IGFs, the IGFBP have received less attention in milk, although truncated IGFBPs and IGFBP-glycosylation have been described in milk. Thereby, complex control of local IGF-effects can be assumed on the levels of IGFBPs, proteases, and protease inhibitors. The present review collects the current knowledge both on presence and regulation of IGFs and IGFBPs in milk particularly from dairy animal species. As a rule higher levels of IGF-I, IGF-II, and IGFBPs are measured around parturition if compared to later time-points of lactation. In all farm animal species included in this review, it is found that the relative abundancies of IGFBPs in milk and serum are similar, with IGFBP-3 and -2 characterized by higher concentrations if compared to IGFBP-4 or -5. The concentrations of IGFs and IGFBPs in milk or dairy products can be altered by hormones, dairy processing, or fermentation. Because milk can be used for non-invasive biomarker research, quality management, and health monitoring, we discuss novel directions of IGF-analysis and potential on-site biomarker research in milk.
Collapse
Affiliation(s)
- Zianka Meyer
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | | | - Elisa Wirthgen
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany; Ligandis GbR, Gülzow-Prüzen, Germany
| | - Sven Olm
- MQD M-V mbH, Institut für Analytik und Hygiene, Güstrow, Germany
| | - Harald M Hammon
- Institute of Nutritional Physiology "Oskar Kellner", Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), Dummerstorf, Germany.
| |
Collapse
|
11
|
Law AMK, Lim E, Ormandy CJ, Gallego-Ortega D. The innate and adaptive infiltrating immune systems as targets for breast cancer immunotherapy. Endocr Relat Cancer 2017; 24:R123-R144. [PMID: 28193698 PMCID: PMC5425956 DOI: 10.1530/erc-16-0404] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 02/13/2017] [Indexed: 12/15/2022]
Abstract
A cancer cell-centric view has long dominated the field of cancer biology. Research efforts have focussed on aberrant cancer cell signalling pathways and on changes to cancer cell DNA. Mounting evidence demonstrates that many cancer-associated cell types within the tumour stroma co-evolve and support tumour growth and development, greatly modifying cancer cell behaviour, facilitating invasion and metastasis and controlling dormancy and sensitivity to drug therapy. Thus, these stromal cells represent potential targets for cancer therapy. Among these cell types, immune cells have emerged as a promising target for therapy. The adaptive and the innate immune system play an important role in normal mammary development and breast cancer. The number of infiltrating adaptive immune system cells with tumour-rejecting capacity, primarily, T lymphocytes, is lower in breast cancer compared with other cancer types, but infiltration occurs in a large proportion of cases. There is strong evidence demonstrating the importance of the immunosuppressive role of the innate immune system during breast cancer progression. A consideration of components of both the innate and the adaptive immune system is essential for the design and development of immunotherapies in breast cancer. In this review, we focus on the importance of immunosuppressive myeloid-derived suppressor cells (MDSCs) as potential targets for breast cancer therapy.
Collapse
Affiliation(s)
- Andrew M K Law
- Tumour Development GroupThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Cancer Biology LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Elgene Lim
- Connie Johnson Breast Cancer Research LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Christopher J Ormandy
- Cancer Biology LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - David Gallego-Ortega
- Tumour Development GroupThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| |
Collapse
|
12
|
LPA receptor activity is basal specific and coincident with early pregnancy and involution during mammary gland postnatal development. Sci Rep 2016; 6:35810. [PMID: 27808166 PMCID: PMC5093903 DOI: 10.1038/srep35810] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 10/06/2016] [Indexed: 01/08/2023] Open
Abstract
During pregnancy, luminal and basal epithelial cells of the adult mammary gland proliferate and differentiate resulting in remodeling of the adult gland. While pathways that control this process have been characterized in the gland as a whole, the contribution of specific cell subtypes, in particular the basal compartment, remains largely unknown. Basal cells provide structural and contractile support, however they also orchestrate the communication between the stroma and the luminal compartment at all developmental stages. Using RNA-seq, we show that basal cells are extraordinarily transcriptionally dynamic throughout pregnancy when compared to luminal cells. We identified gene expression changes that define specific basal functions acquired during development that led to the identification of novel markers. Enrichment analysis of gene sets from 24 mouse models for breast cancer pinpoint to a potential new function for insulin-like growth factor 1 (Igf1r) in the basal epithelium during lactogenesis. We establish that β-catenin signaling is activated in basal cells during early pregnancy, and demonstrate that this activity is mediated by lysophosphatidic acid receptor 3 (Lpar3). These findings identify novel pathways active during functional maturation of the adult mammary gland.
Collapse
|
13
|
Wyszynski A, Hong CC, Lam K, Michailidou K, Lytle C, Yao S, Zhang Y, Bolla MK, Wang Q, Dennis J, Hopper JL, Southey MC, Schmidt MK, Broeks A, Muir K, Lophatananon A, Fasching PA, Beckmann MW, Peto J, Dos-Santos-Silva I, Sawyer EJ, Tomlinson I, Burwinkel B, Marme F, Guénel P, Truong T, Bojesen SE, Nordestgaard BG, González-Neira A, Benitez J, Neuhausen SL, Brenner H, Dieffenbach AK, Meindl A, Schmutzler RK, Brauch H, Nevanlinna H, Khan S, Matsuo K, Ito H, Dörk T, Bogdanova NV, Lindblom A, Margolin S, Mannermaa A, Kosma VM, Wu AH, Van Den Berg D, Lambrechts D, Wildiers H, Chang-Claude J, Rudolph A, Radice P, Peterlongo P, Couch FJ, Olson JE, Giles GG, Milne RL, Haiman CA, Henderson BE, Dumont M, Teo SH, Wong TY, Kristensen V, Zheng W, Long J, Winqvist R, Pylkäs K, Andrulis IL, Knight JA, Devilee P, Seynaeve C, García-Closas M, Figueroa J, Klevebring D, Czene K, Hooning MJ, van den Ouweland AMW, Darabi H, Shu XO, Gao YT, Cox A, Blot W, Signorello LB, Shah M, Kang D, Choi JY, Hartman M, Miao H, Hamann U, Jakubowska A, Lubinski J, Sangrajrang S, McKay J, Toland AE, Yannoukakos D, Shen CY, Wu PE, Swerdlow A, Orr N, et alWyszynski A, Hong CC, Lam K, Michailidou K, Lytle C, Yao S, Zhang Y, Bolla MK, Wang Q, Dennis J, Hopper JL, Southey MC, Schmidt MK, Broeks A, Muir K, Lophatananon A, Fasching PA, Beckmann MW, Peto J, Dos-Santos-Silva I, Sawyer EJ, Tomlinson I, Burwinkel B, Marme F, Guénel P, Truong T, Bojesen SE, Nordestgaard BG, González-Neira A, Benitez J, Neuhausen SL, Brenner H, Dieffenbach AK, Meindl A, Schmutzler RK, Brauch H, Nevanlinna H, Khan S, Matsuo K, Ito H, Dörk T, Bogdanova NV, Lindblom A, Margolin S, Mannermaa A, Kosma VM, Wu AH, Van Den Berg D, Lambrechts D, Wildiers H, Chang-Claude J, Rudolph A, Radice P, Peterlongo P, Couch FJ, Olson JE, Giles GG, Milne RL, Haiman CA, Henderson BE, Dumont M, Teo SH, Wong TY, Kristensen V, Zheng W, Long J, Winqvist R, Pylkäs K, Andrulis IL, Knight JA, Devilee P, Seynaeve C, García-Closas M, Figueroa J, Klevebring D, Czene K, Hooning MJ, van den Ouweland AMW, Darabi H, Shu XO, Gao YT, Cox A, Blot W, Signorello LB, Shah M, Kang D, Choi JY, Hartman M, Miao H, Hamann U, Jakubowska A, Lubinski J, Sangrajrang S, McKay J, Toland AE, Yannoukakos D, Shen CY, Wu PE, Swerdlow A, Orr N, Simard J, Pharoah PDP, Dunning AM, Chenevix-Trench G, Hall P, Bandera E, Amos C, Ambrosone C, Easton DF, Cole MD. An intergenic risk locus containing an enhancer deletion in 2q35 modulates breast cancer risk by deregulating IGFBP5 expression. Hum Mol Genet 2016; 25:3863-3876. [PMID: 27402876 PMCID: PMC5216618 DOI: 10.1093/hmg/ddw223] [Show More Authors] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/11/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is the most diagnosed malignancy and the second leading cause of cancer mortality in females. Previous association studies have identified variants on 2q35 associated with the risk of breast cancer. To identify functional susceptibility loci for breast cancer, we interrogated the 2q35 gene desert for chromatin architecture and functional variation correlated with gene expression. We report a novel intergenic breast cancer risk locus containing an enhancer copy number variation (enCNV; deletion) located approximately 400Kb upstream to IGFBP5, which overlaps an intergenic ERα-bound enhancer that loops to the IGFBP5 promoter. The enCNV is correlated with modified ERα binding and monoallelic-repression of IGFBP5 following oestrogen treatment. We investigated the association of enCNV genotype with breast cancer in 1,182 cases and 1,362 controls, and replicate our findings in an independent set of 62,533 cases and 60,966 controls from 41 case control studies and 11 GWAS. We report a dose-dependent inverse association of 2q35 enCNV genotype (percopy OR = 0.68 95%CI 0.55-0.83, P = 0.0002; replication OR = 0.77 95% CI 0.73-0.82, P = 2.1 × 10-19) and identify 13 additional linked variants (r2 > 0.8) in the 20Kb linkage block containing the enCNV (P = 3.2 × 10-15 - 5.6 × 10-17). These associations were independent of previously reported 2q35 variants, rs13387042/rs4442975 and rs16857609, and were stronger for ER-positive than ER-negative disease. Together, these results suggest that 2q35 breast cancer risk loci may be mediating their effect through IGFBP5.
Collapse
Affiliation(s)
- Asaf Wyszynski
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Chi-Chen Hong
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | - Kyriaki Michailidou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Christian Lytle
- Molecular Biology Core Facility, Dartmouth College, Hanover, NH 03755 USA
| | - Song Yao
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Yali Zhang
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Manjeet K Bolla
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Qin Wang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Melissa C Southey
- Department of Pathology, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Marjanka K Schmidt
- Netherlands Cancer Institute, Antoni van Leeuwenhoek hospital, 1066 CX Amsterdam, The Netherlands
| | - Annegien Broeks
- Wellcome Trust Centre for Human Genetics and Oxford Biomedical Research Centre, University of Oxford, OX3 7BN, UK
| | - Kenneth Muir
- Division of Health Sciences, Warwick Medical school, Warwick University, Coventry, CV4 7AL, UK
- Institute of Population Health, University of Manchester, Manchester, M13 9PL, UK
| | - Artitaya Lophatananon
- Department of Obstetrics and Gynecology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Peter A Fasching
- University Breast Center Franconia, Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, 91054 Erlangen, Germany
- David Geffen School of Medicine, Department of Medicine Division of Hematology and Oncology, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Matthias W Beckmann
- University Breast Center Franconia, Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, 91054 Erlangen, Germany
| | - Julian Peto
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Isabel Dos-Santos-Silva
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Elinor J Sawyer
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, SE1 9RT, UK
| | - Ian Tomlinson
- Wellcome Trust Centre for Human Genetics and Oxford Biomedical Research Centre, University of Oxford, OX3 7BN, UK
| | - Barbara Burwinkel
- Department of Obstetrics and Gynecology, University of Heidelberg, 69120 Heidelberg, Germany
- Molecular Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Frederik Marme
- Department of Obstetrics and Gynecology, University of Heidelberg, 69120 Heidelberg, Germany
- National Center for Tumor Diseases, University of Heidelberg, 69120 Heidelberg, Germany
| | - Pascal Guénel
- Inserm (National Institute of Health and Medical Research), CESP (Center for Research in Epidemiology and Population Health), U1018, Environmental Epidemiology of Cancer, 94807 Villejuif, France
- University Paris-Sud, UMRS 1018, 94807 Villejuif, France
| | - Thérèse Truong
- Inserm (National Institute of Health and Medical Research), CESP (Center for Research in Epidemiology and Population Health), U1018, Environmental Epidemiology of Cancer, 94807 Villejuif, France
- University Paris-Sud, UMRS 1018, 94807 Villejuif, France
| | - Stig E Bojesen
- Copenhagen General Population Study, Herlev Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Børge G Nordestgaard
- Copenhagen General Population Study, Herlev Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Anna González-Neira
- Human Genotyping-CEGEN Unit, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Javier Benitez
- Human Genetics Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
- Centro de Investigación en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | | | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Aida Karina Dieffenbach
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Alfons Meindl
- Division of Gynaecology and Obstetrics, Technische Universität München, 81675 Munich, Germany
| | - Rita K Schmutzler
- Division of Molecular Gyneco-Oncology, Department of Gynaecology and Obstetrics, University Hospital of Cologne, 50931 Cologne, Germany
- Center of Familial Breast and Ovarian Cancer, University Hospital of Cologne, 50931 Cologne, Germany
- Center for Integrated Oncology (CIO), University Hospital of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Hiltrud Brauch
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tübingen, 72074 Tübingen, Germany
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, FI-00029 HUS, Finland
| | - Sofia Khan
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Central Hospital, Helsinki, FI-00029 HUS, Finland
| | - Keitaro Matsuo
- Department of Preventive Medicine, Kyushu University Faculty of Medical Sciences, Fukuoka, Japan
| | - Hidemi Ito
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Aichi, 464-8681, Japan
| | - Thilo Dörk
- Department of Obstetrics and Gynaecology, Hannover Medical School, 30625 Hannover, Germany
| | - Natalia V Bogdanova
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Sara Margolin
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Arto Mannermaa
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
- Cancer Center of Eastern Finland, University of Eastern Finland, FI-70211 Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Veli-Matti Kosma
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
- Cancer Center of Eastern Finland, University of Eastern Finland, FI-70211 Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Anna H Wu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - David Van Den Berg
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Diether Lambrechts
- Vesalius Research Center (VRC), VIB, 3000 Leuven, Belgium
- Laboratory for Translational Genetics, Department of Oncology, University of Leuven, 3000 Leuven, Belgium
| | - Hans Wildiers
- Multidisciplinary Breast Center, Department of General Medical Oncology, University Hospitals Leuven, B-3000 Leuven, Belgium
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anja Rudolph
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Paolo Radice
- Unit of Molecular Bases of Genetic Risk and Genetic Testing, Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori (INT), 20133 Milan, Italy
| | - Paolo Peterlongo
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, 20139 Milan, Italy
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Janet E Olson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Graham G Giles
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Victoria 3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Roger L Milne
- Cancer Epidemiology Centre, Cancer Council Victoria, Melbourne, Victoria 3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Christopher A Haiman
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Brian E Henderson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Martine Dumont
- Centre Hospitalier Universitaire de Québec Research Center and Laval University, QC, G1V 4G2, Canada
| | - Soo Hwang Teo
- Cancer Research Initiatives Foundation, Sime Darby Medical Centre, 47500 Subang Jaya, Selangor, Malaysia
- Breast Cancer Research Unit, University Malaya Cancer Research Institute, University Malaya Medical Centre (UMMC), 59100 Kuala Lumpur, Malaysia
| | - Tien Y Wong
- Singapore Eye Research Institute, National University of Singapore, Singapore 168751
| | - Vessela Kristensen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, N-0310 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo (UiO), 0450 Oslo, Norway
- Department of Clinical Molecular Biology (EpiGen), University of Oslo (UiO), 0450 Oslo, Norway
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Robert Winqvist
- Laboratory of Cancer Genetics and Tumor Biology, Department of Clinical Chemistry and Biocenter Oulu, University of Oulu, NordLab Oulu/Oulu University Hospital, FI-90220 Oulu, Finland
| | - Katri Pylkäs
- Laboratory of Cancer Genetics and Tumor Biology, Department of Clinical Chemistry and Biocenter Oulu, University of Oulu, NordLab Oulu/Oulu University Hospital, FI-90220 Oulu, Finland
| | - Irene L Andrulis
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Julia A Knight
- Prosserman Centre for Health Research, Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Peter Devilee
- Department of Human Genetics & Department of Pathology, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Caroline Seynaeve
- Family Cancer Clinic, Department of Medical Oncology, Erasmus MC-Daniel den Hoed Cancer Center, 3075 EA Rotterdam, The Netherlands
| | - Montserrat García-Closas
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, SM2 5NG, UK
- Breakthrough Breast Cancer Research Centre, Division of Breast Cancer Research, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Jonine Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD 20850, USA
| | - Daniel Klevebring
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Maartje J Hooning
- Department of Medical Oncology, Erasmus University Medical Center, 3075 EA Rotterdam, The Netherlands
| | - Ans M W van den Ouweland
- Department of Clinical Genetics, Erasmus University Medical Center, 3075 EA Rotterdam, The Netherlands
| | - Hatef Darabi
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute, Xuhui, Shanghai, China
| | - Angela Cox
- CRUK/YCR Sheffield Cancer Research Centre, Department of Oncology, University of Sheffield, Sheffield, S10 2RX, UK
| | - William Blot
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
- International Epidemiology Institute, Rockville, MD 20850, USA
| | - Lisa B Signorello
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mitul Shah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, CB1 8RN, UK
| | - Daehee Kang
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 110-799, Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 151-742, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Ji-Yeob Choi
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 151-742, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Mikael Hartman
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117597
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore 117597
| | - Hui Miao
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117597
| | - Ute Hamann
- Molecular Genetics of Breast Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, 70-115 Szczecin, Poland
| | - Jan Lubinski
- Department of Genetics and Pathology, Pomeranian Medical University, 70-115 Szczecin, Poland
| | | | - James McKay
- International Agency for Research on Cancer, 69372 Lyon, CEDEX 08, France
| | - Amanda E Toland
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Drakoulis Yannoukakos
- Molecular Diagnostics Laboratory, IRRP, National Centre for Scientific Research "Demokritos", Aghia Paraskevi Attikis, 153 10 Athens, Greece
| | - Chen-Yang Shen
- Taiwan Biobank, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- School of Public Health, China Medical University, Taichung 404, Taiwan
| | - Pei-Ei Wu
- Taiwan Biobank, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Anthony Swerdlow
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, SM2 5NG, UK
- Division of Breast Cancer Research, Institute of Cancer Research, Sutton, SM2 5NG, UK
| | - Nick Orr
- Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Jacques Simard
- Centre Hospitalier Universitaire de Québec Research Center and Laval University, QC, G1V 4G2, Canada
| | - Paul D P Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, CB1 8RN, UK
| | - Alison M Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, CB1 8RN, UK
| | - Georgia Chenevix-Trench
- Department of Genetics, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm SE-17177, Sweden
| | - Elisa Bandera
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901 USA
| | - Chris Amos
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Christine Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Michael D Cole
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
14
|
Murney R, Stelwagen K, Wheeler TT, Margerison JK, Singh K. The effects of milking frequency on insulin-like growth factor I signaling within the mammary gland of dairy cows. J Dairy Sci 2016; 98:5422-8. [PMID: 26074231 DOI: 10.3168/jds.2015-9425] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 04/22/2015] [Indexed: 11/19/2022]
Abstract
In dairy cows, short-term changes in milking frequency (MF) in early lactation have been shown to produce both an immediate and a long-term effect on milk yield. The effect of MF on milk yield is controlled locally within mammary glands and could be a function of changes in either number or activity of secretory mammary epithelial cells (MEC). Insulin-like growth factor I (IGF-I) signaling is one candidate factor that could mediate these effects, as it can be controlled locally within mammary glands. Both MEC number and activity can be affected by IGF-I signaling by activating the phosphoinositide 3-kinase (PI3K)/Akt and extracellular-signal-regulated kinase (ERK)1/2 pathways. To investigate the relationship between MF and IGF-I signaling, udder halves of 17 dairy cows were milked either 4 times a day (4×) or once a day (1×) for 14 d in early lactation. On d 14, between 3 and 5 h following milking, mammary biopsies were obtained from 10 cows from both udder halves, and changes in the expression of genes associated with IGF-I signaling and the activation of the PI3K/Akt and ERK1/2 pathways were measured. The mRNA abundance of IGF type I receptor, IGF binding protein (IGFBP)-3, and IGFBP-5 were lower following 4× milking relative to 1× milking. However, the mRNA abundance of IGF-I was not affected by MF. Both IGFBP3 and IGFBP5 are thought to inhibit IGF-I; therefore, decreases in their mRNA abundance may serve to stimulate the IGF-I signal in the 4×-milked mammary gland. The activation of PI3K/Akt pathway was lower in response to 4× milking relative to 1×, and the activation of the ERK1/2 was unaffected by MF, suggesting that they do not mediate the effects of MF.
Collapse
Affiliation(s)
- R Murney
- AgResearch Ltd., Ruakura Research Centre, PB 3123, Hamilton 3240, New Zealand; Institute of Agriculture and Environment, College of Sciences, Massey University, PB 11222, Palmerston North 4442, New Zealand.
| | - K Stelwagen
- SciLactis Ltd., Waikato Innovation Park, Hamilton 3240, New Zealand
| | - T T Wheeler
- AgResearch Ltd., Ruakura Research Centre, PB 3123, Hamilton 3240, New Zealand
| | - J K Margerison
- Institute of Agriculture and Environment, College of Sciences, Massey University, PB 11222, Palmerston North 4442, New Zealand
| | - K Singh
- AgResearch Ltd., Ruakura Research Centre, PB 3123, Hamilton 3240, New Zealand
| |
Collapse
|
15
|
Abstract
Insulin-like growth factor binding proteins (IGFBPs) 4-6 have important roles as modulators of IGF actions. IGFBP-4 and IGFBP-6 predominantly inhibit IGF actions, whereas IGFBP-5 may enhance these actions under some circumstances. IGFBP-6 is unique among the IGFBPs for its marked IGF-II binding preference. IGFBPs 4-6 are found in the circulation as binary complexes with IGFs that can enter tissues. Additionally, about half of the circulating IGFBP-5 is found in ternary complexes with IGFs and an acid labile subunit; this high molecular complex cannot leave the circulation and acts as an IGF reservoir. IGFBPs 4-6 also have IGF-independent actions. These IGFBPs are regulated in a cell-specific manner and their dysregulation may play a role in a range of diseases including cancer. However, there is no clear clinical indication for measuring serum levels of these IGFBPs at present.
Collapse
Affiliation(s)
- Leon A Bach
- Department of Medicine (Alfred), Monash University, Prahran, 3181, Australia; Department of Endocrinology and Diabetes, Alfred Hospital, Melbourne, 3004, Australia.
| |
Collapse
|
16
|
Christians JK, King AY, Rogowska MD, Hessels SM. Pappa2 deletion in mice affects male but not female fertility. Reprod Biol Endocrinol 2015; 13:109. [PMID: 26416573 PMCID: PMC4587772 DOI: 10.1186/s12958-015-0108-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/16/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Recent studies have found associations between the gene encoding pregnancy associated plasma protein-A2 (PAPP-A2), a protease of insulin-like growth factor binding protein -5 (IGFBP-5), and measures of female reproductive performance in cattle. The purpose of the present study was to test the effects of Pappa2 deletion on reproduction in mice. FINDINGS We measured the fertility and offspring growth of Pappa2 deletion females, and also performed reciprocal matings (i.e., deletion males mated to control females) to control for the effects of offspring genotype. Ovarian and testicular IGFBP-5 levels were measured by Western blotting. As expected, deletion of Pappa2 increased ovarian IGFBP-5 levels. However, Pappa2 deletion in females had no effect on the interval between pairing and the birth of the first litter, the interval between the births of the first and second litters, or litter size. Offspring weight was lower in the offspring of Pappa2 deletion females, but effects of similar magnitude were observed in the offspring of Pappa2 deletion males, suggesting that the effects were due to heterozygosity for the deletion in the offspring. Pappa2 deletion in males had no effect on litter size or the interval between pairing and the birth of the first litter. However, the interval between the births of the first and second litters was significantly longer in deletion males. CONCLUSIONS Pappa2 deletion had no effect on female reproductive performance. In contrast, Pappa2 deletion had subtle effects on male fertility, although the underlying mechanism remains to be elucidated.
Collapse
Affiliation(s)
- Julian K Christians
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| | - Avery Y King
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| | - Monika D Rogowska
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| | - Sonia M Hessels
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
17
|
Beattie J, Hawsawi Y, Alkharobi H, El-Gendy R. IGFBP-2 and -5: important regulators of normal and neoplastic mammary gland physiology. J Cell Commun Signal 2015; 9:151-8. [PMID: 25645979 DOI: 10.1007/s12079-015-0260-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/12/2015] [Indexed: 01/16/2023] Open
Abstract
The insulin-like growth factor (IGF) axis plays an important role in mammary gland physiology. In addition, dysregulation of this molecular axis may have a causal role in the aetiology and development of breast cancer (BC). This report discusses the IGF axis in normal and neoplastic mammary gland with special reference to IGF binding proteins (IGFBPs) -2 and -5. We describe how these high affinity binders of IGF-1 and IGF-2 may regulate local actions of growth factors in an autocrine and/or paracrine manner and how they also have IGF-independent effects in mammary gland. We discuss clinical studies which investigate both the prognostic value of IGFBP-2 and -5 expression in BC and possible involvement of these genes in the development of resistance to adjuvant endocrine therapies.
Collapse
Affiliation(s)
- James Beattie
- Department of Oral Biology, School of Dentistry, St James University Hospital, Level 7, Wellcome Trust Brenner Building, Leeds, LS9 7TF, UK,
| | | | | | | |
Collapse
|
18
|
De Silva D, Kunasegaran K, Ghosh S, Pietersen AM. Transcriptome analysis of the hormone-sensing cells in mammary epithelial reveals dynamic changes in early pregnancy. BMC DEVELOPMENTAL BIOLOGY 2015; 15:7. [PMID: 25623114 PMCID: PMC4314744 DOI: 10.1186/s12861-015-0058-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 01/15/2015] [Indexed: 12/18/2022]
Abstract
Background Alveoli, the milk-producing units of the mammary gland, are generated during pregnancy by collaboration of different epithelial cell types. We present the first analysis of transcriptional changes within the hormone sensing population during pregnancy. Hormone-receptor positive (HR+) cells play a key role in the initiation of alveologenesis as they sense systemic hormonal changes and translate these into local instructions for neighboring HR- cells. We recently showed that IGF2 is produced specifically by HR+ cells in early pregnancy, but is undetectable in the virgin state. Here, we define the transcriptome of HR+ cells in early pregnancy with the aim to elucidate additional changes that are unique for this dynamic developmental time window. Results We harvested mammary glands from virgin, 3-day and 7-day pregnant mice and isolated a few hundred hormone-sensing cells per animal by FACS for microarray analysis. There was a high concordance between animals with a clear induction of cell cycle progression genes at day 3 of pregnancy and molecules involved in paracrine signalling at day 7. Conclusions These findings underscore the proliferative capacity of HR+ cells upon specific stimuli and elucidate developmentally-restricted changes in cellular communication. Since the majority of breast cancers are HR+, with a variable proportion of HR+ cells per tumor, we anticipate that this data set will aid further studies into the regulation of HR+ cell proliferation and the role of heterotypic signalling within tumors. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0058-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Duvini De Silva
- Laboratory of Mammary Gland Biology, National Cancer Centre Singapore, 11 Hospital Dr, Singapore, 169610, Singapore. .,Program in Cancer & Stem Cell Biology, Duke-NUS Graduate Medical School, 8 College, Rd, 169857, Singapore, Singapore.
| | - Kamini Kunasegaran
- Laboratory of Mammary Gland Biology, National Cancer Centre Singapore, 11 Hospital Dr, Singapore, 169610, Singapore. .,Program in Cancer & Stem Cell Biology, Duke-NUS Graduate Medical School, 8 College, Rd, 169857, Singapore, Singapore.
| | - Sujoy Ghosh
- Program in Cardiovascular & Metabolic Disorders, Duke-NUS Graduate Medical School, 8 College Rd, Singapore, 169857, Singapore.
| | - Alexandra M Pietersen
- Laboratory of Mammary Gland Biology, National Cancer Centre Singapore, 11 Hospital Dr, Singapore, 169610, Singapore. .,Program in Cancer & Stem Cell Biology, Duke-NUS Graduate Medical School, 8 College, Rd, 169857, Singapore, Singapore. .,Department of Physiology, National University of Singapore, 21 Lower Kent Ridge Rd, Singapore, 119077, Singapore.
| |
Collapse
|
19
|
Comparative expression profiling of insulin-like growth factor binding protein-5 in milk of Bos indicus and Bubalus bubalis during lactation. Animal 2015; 9:643-9. [DOI: 10.1017/s1751731114002985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
20
|
Ghoussaini M, Edwards SL, Michailidou K, Nord S, Cowper-Sal·lari R, Desai K, Kar S, Hillman KM, Kaufmann S, Glubb DM, Beesley J, Dennis J, Bolla MK, Wang Q, Dicks E, Guo Q, Schmidt MK, Shah M, Luben R, Brown J, Czene K, Darabi H, Eriksson M, Klevebring D, Bojesen SE, Nordestgaard BG, Nielsen SF, Flyger H, Lambrechts D, Thienpont B, Neven P, Wildiers H, Broeks A, Van’t Veer LJ, Th Rutgers EJ, Couch FJ, Olson JE, Hallberg E, Vachon C, Chang-Claude J, Rudolph A, Seibold P, Flesch-Janys D, Peto J, dos-Santos-Silva I, Gibson L, Nevanlinna H, Muranen TA, Aittomäki K, Blomqvist C, Hall P, Li J, Liu J, Humphreys K, Kang D, Choi JY, Park SK, Noh DY, Matsuo K, Ito H, Iwata H, Yatabe Y, Guénel P, Truong T, Menegaux F, Sanchez M, Burwinkel B, Marme F, Schneeweiss A, Sohn C, Wu AH, Tseng CC, Van Den Berg D, Stram DO, Benitez J, Zamora MP, Perez JIA, Menéndez P, Shu XO, Lu W, Gao YT, Cai Q, Cox A, Cross SS, Reed MWR, Andrulis IL, Knight JA, Glendon G, Tchatchou S, Sawyer EJ, Tomlinson I, Kerin MJ, Miller N, Haiman CA, Henderson BE, Schumacher F, Le Marchand L, Lindblom A, Margolin S, TEO SH, et alGhoussaini M, Edwards SL, Michailidou K, Nord S, Cowper-Sal·lari R, Desai K, Kar S, Hillman KM, Kaufmann S, Glubb DM, Beesley J, Dennis J, Bolla MK, Wang Q, Dicks E, Guo Q, Schmidt MK, Shah M, Luben R, Brown J, Czene K, Darabi H, Eriksson M, Klevebring D, Bojesen SE, Nordestgaard BG, Nielsen SF, Flyger H, Lambrechts D, Thienpont B, Neven P, Wildiers H, Broeks A, Van’t Veer LJ, Th Rutgers EJ, Couch FJ, Olson JE, Hallberg E, Vachon C, Chang-Claude J, Rudolph A, Seibold P, Flesch-Janys D, Peto J, dos-Santos-Silva I, Gibson L, Nevanlinna H, Muranen TA, Aittomäki K, Blomqvist C, Hall P, Li J, Liu J, Humphreys K, Kang D, Choi JY, Park SK, Noh DY, Matsuo K, Ito H, Iwata H, Yatabe Y, Guénel P, Truong T, Menegaux F, Sanchez M, Burwinkel B, Marme F, Schneeweiss A, Sohn C, Wu AH, Tseng CC, Van Den Berg D, Stram DO, Benitez J, Zamora MP, Perez JIA, Menéndez P, Shu XO, Lu W, Gao YT, Cai Q, Cox A, Cross SS, Reed MWR, Andrulis IL, Knight JA, Glendon G, Tchatchou S, Sawyer EJ, Tomlinson I, Kerin MJ, Miller N, Haiman CA, Henderson BE, Schumacher F, Le Marchand L, Lindblom A, Margolin S, TEO SH, YIP CH, Lee DSC, Wong TY, Hooning MJ, Martens JWM, Collée JM, van Deurzen CHM, Hopper JL, Southey MC, Tsimiklis H, Kapuscinski MK, Shen CY, Wu PE, Yu JC, Chen ST, Alnæs GG, Borresen-Dale AL, Giles GG, Milne RL, McLean C, Muir K, Lophatananon A, Stewart-Brown S, Siriwanarangsan P, Hartman M, Miao H, Buhari SABS, Teo YY, Fasching PA, Haeberle L, Ekici AB, Beckmann MW, Brenner H, Dieffenbach AK, Arndt V, Stegmaier C, Swerdlow A, Ashworth A, Orr N, Schoemaker MJ, García-Closas M, Figueroa J, Chanock SJ, Lissowska J, Simard J, Goldberg MS, Labrèche F, Dumont M, Winqvist R, Pylkäs K, Jukkola-Vuorinen A, Brauch H, Brüning T, Koto YD, Radice P, Peterlongo P, Bonanni B, Volorio S, Dörk T, Bogdanova NV, Helbig S, Mannermaa A, Kataja V, Kosma VM, Hartikainen JM, Devilee P, Tollenaar RAEM, Seynaeve C, Van Asperen CJ, Jakubowska A, Lubinski J, Jaworska-Bieniek K, Durda K, Slager S, Toland AE, Ambrosone CB, Yannoukakos D, Sangrajrang S, Gaborieau V, Brennan P, McKay J, Hamann U, Torres D, Zheng W, Long J, Anton-Culver H, Neuhausen SL, Luccarini C, Baynes C, Ahmed S, Maranian M, Healey CS, González-Neira A, Pita G, Alonso MR, Álvarez N, Herrero D, Tessier DC, Vincent D, Bacot F, de Santiago I, Carroll J, Caldas C, Brown MA, Lupien M, Kristensen VN, Pharoah PDP, Chenevix-Trench G, French JD, Easton DF, Dunning AM. Evidence that breast cancer risk at the 2q35 locus is mediated through IGFBP5 regulation. Nat Commun 2014; 4:4999. [PMID: 25248036 PMCID: PMC4321900 DOI: 10.1038/ncomms5999] [Show More Authors] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 08/14/2014] [Indexed: 02/07/2023] Open
Abstract
GWAS have identified a breast cancer susceptibility locus on 2q35. Here we report the fine mapping of this locus using data from 101,943 subjects from 50 case-control studies. We genotype 276 SNPs using the 'iCOGS' genotyping array and impute genotypes for a further 1,284 using 1000 Genomes Project data. All but two, strongly correlated SNPs (rs4442975 G/T and rs6721996 G/A) are excluded as candidate causal variants at odds against >100:1. The best functional candidate, rs4442975, is associated with oestrogen receptor positive (ER+) disease with an odds ratio (OR) in Europeans of 0.85 (95% confidence interval=0.84-0.87; P=1.7 × 10(-43)) per t-allele. This SNP flanks a transcriptional enhancer that physically interacts with the promoter of IGFBP5 (encoding insulin-like growth factor-binding protein 5) and displays allele-specific gene expression, FOXA1 binding and chromatin looping. Evidence suggests that the g-allele confers increased breast cancer susceptibility through relative downregulation of IGFBP5, a gene with known roles in breast cell biology.
Collapse
Affiliation(s)
- Maya Ghoussaini
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
| | - Stacey L. Edwards
- Department of Genetics, QIMR Berghofer Medical Research
Institute, Brisbane, Queensland
4029, Australia
- School of Chemistry and Molecular Biosciences, The University of
Queensland, Brisbane, Queensland
4072, Australia
| | - Kyriaki Michailidou
- Centre for Cancer Genetic Epidemiology, Department of Public
Health and Primary Care, University of Cambridge, Cambridge
CB1 8RN, UK
| | - Silje Nord
- Department of Genetics, Institute for Cancer Research, Oslo
University Hospital, Radiumhospitalet, N-0310
Oslo, Norway
| | - Richard Cowper-Sal·lari
- The Princess Margaret Cancer Centre, University Health
Network, Toronto, Ontario, Canada
M5T 2M9
| | - Kinjal Desai
- The Princess Margaret Cancer Centre, University Health
Network, Toronto, Ontario, Canada
M5T 2M9
- Geisel School of Medicine, Dartmouth College,
Hanover, New Hampshire
03755, USA
| | - Siddhartha Kar
- Centre for Cancer Genetic Epidemiology, Department of Public
Health and Primary Care, University of Cambridge, Cambridge
CB1 8RN, UK
| | - Kristine M. Hillman
- Department of Genetics, QIMR Berghofer Medical Research
Institute, Brisbane, Queensland
4029, Australia
- School of Chemistry and Molecular Biosciences, The University of
Queensland, Brisbane, Queensland
4072, Australia
| | - Susanne Kaufmann
- Department of Genetics, QIMR Berghofer Medical Research
Institute, Brisbane, Queensland
4029, Australia
- School of Chemistry and Molecular Biosciences, The University of
Queensland, Brisbane, Queensland
4072, Australia
| | - Dylan M. Glubb
- Department of Genetics, QIMR Berghofer Medical Research
Institute, Brisbane, Queensland
4029, Australia
| | - Jonathan Beesley
- Department of Genetics, QIMR Berghofer Medical Research
Institute, Brisbane, Queensland
4029, Australia
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public
Health and Primary Care, University of Cambridge, Cambridge
CB1 8RN, UK
| | - Manjeet K. Bolla
- Centre for Cancer Genetic Epidemiology, Department of Public
Health and Primary Care, University of Cambridge, Cambridge
CB1 8RN, UK
| | - Qin Wang
- Centre for Cancer Genetic Epidemiology, Department of Public
Health and Primary Care, University of Cambridge, Cambridge
CB1 8RN, UK
| | - Ed Dicks
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
| | - Qi Guo
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
| | - Marjanka K. Schmidt
- Netherlands Cancer Institute, Antoni van Leeuwenhoek
hospital, 1066 CX
Amsterdam, The Netherlands
| | - Mitul Shah
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
| | - Robert Luben
- Centre for Cancer Genetic Epidemiology, Department of Public
Health and Primary Care, University of Cambridge, Cambridge
CB1 8RN, UK
| | - Judith Brown
- Centre for Cancer Genetic Epidemiology, Department of Public
Health and Primary Care, University of Cambridge, Cambridge
CB1 8RN, UK
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska
Institutet, Stockholm
SE-17177, Sweden
| | - Hatef Darabi
- Department of Medical Epidemiology and Biostatistics, Karolinska
Institutet, Stockholm
SE-17177, Sweden
| | - Mikael Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska
Institutet, Stockholm
SE-17177, Sweden
| | - Daniel Klevebring
- Department of Medical Epidemiology and Biostatistics, Karolinska
Institutet, Stockholm
SE-17177, Sweden
| | - Stig E. Bojesen
- Copenhagen General Population Study, Herlev Hospital,
2730
Herlev, Copenhagen, Denmark
- Department of Clinical Biochemistry, Herlev Hospital,
Copenhagen University Hospital, 2730
Herlev, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of
Copenhagen, 2200
Copenhagen, Denmark
| | - Børge G. Nordestgaard
- Copenhagen General Population Study, Herlev Hospital,
2730
Herlev, Copenhagen, Denmark
- Department of Clinical Biochemistry, Herlev Hospital,
Copenhagen University Hospital, 2730
Herlev, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of
Copenhagen, 2200
Copenhagen, Denmark
| | - Sune F. Nielsen
- Copenhagen General Population Study, Herlev Hospital,
2730
Herlev, Copenhagen, Denmark
- Department of Clinical Biochemistry, Herlev Hospital,
Copenhagen University Hospital, 2730
Herlev, Copenhagen, Denmark
| | - Henrik Flyger
- Department of Breast Surgery, Herlev Hospital, Copenhagen
University Hospital, 2730
Herlev, Copenhagen, Denmark
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Oncology,
University of Leuven, 3000
Leuven, Belgium
- Vesalius Research Center (VRC), VIB, 3000
Leuven, Belgium
| | - Bernard Thienpont
- Vesalius Research Center (VRC), VIB, 3000
Leuven, Belgium
- Vesalius Research Center, University of Leuven,
3000
Leuven, Belgium
| | - Patrick Neven
- Department of Oncology, University of Leuven,
3000
Leuven, Belgium
- Multidisciplinary Breast Center, Department of General Medical
Oncology, University Hospitals Leuven, 3000
Leuven, Belgium
| | - Hans Wildiers
- Department of Oncology, University of Leuven,
3000
Leuven, Belgium
- Multidisciplinary Breast Center, Department of General Medical
Oncology, University Hospitals Leuven, 3000
Leuven, Belgium
| | - Annegien Broeks
- Netherlands Cancer Institute, Antoni van Leeuwenhoek
hospital, 1066 CX
Amsterdam, The Netherlands
| | - Laura J. Van’t Veer
- Netherlands Cancer Institute, Antoni van Leeuwenhoek
hospital, 1066 CX
Amsterdam, The Netherlands
| | - Emiel J. Th Rutgers
- Netherlands Cancer Institute, Antoni van Leeuwenhoek
hospital, 1066 CX
Amsterdam, The Netherlands
| | - Fergus J. Couch
- Department of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester, Minnesota
55905, USA
| | - Janet E. Olson
- Department of Health Sciences Research, Mayo Clinic,
Rochester, Minnesota
55905, USA
| | - Emily Hallberg
- Department of Health Sciences Research, Mayo Clinic,
Rochester, Minnesota
55905, USA
| | - Celine Vachon
- Department of Health Sciences Research, Mayo Clinic,
Rochester, Minnesota
55905, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center
(DKFZ), 69120
Heidelberg, Germany
| | - Anja Rudolph
- Division of Cancer Epidemiology, German Cancer Research Center
(DKFZ), 69120
Heidelberg, Germany
| | - Petra Seibold
- Division of Cancer Epidemiology, German Cancer Research Center
(DKFZ), 69120
Heidelberg, Germany
| | - Dieter Flesch-Janys
- Department of Cancer Epidemiology/Clinical Cancer Registry and
Institute for Medical Biometrics and Epidemiology, University Clinic
Hamburg-Eppendorf, 20246
Hamburg, Germany
| | - Julian Peto
- Department of Non-Communicable Disease Epidemiology, London
School of Hygiene and Tropical Medicine, London
WC1E 7HT, UK
| | - Isabel dos-Santos-Silva
- Department of Non-Communicable Disease Epidemiology, London
School of Hygiene and Tropical Medicine, London
WC1E 7HT, UK
| | - Lorna Gibson
- Department of Non-Communicable Disease Epidemiology, London
School of Hygiene and Tropical Medicine, London
WC1E 7HT, UK
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, Helsinki University
Central Hospital, Helsinki, FI-00029
HUS, Finland
| | - Taru A. Muranen
- Department of Obstetrics and Gynecology, Helsinki University
Central Hospital, Helsinki, FI-00029
HUS, Finland
| | - Kristiina Aittomäki
- Department of Clinical Genetics, University of Helsinki,
Helsinki University Central Hospital, Helsinki,
FI-00029
HUS, Finland
| | - Carl Blomqvist
- Department of Oncology, University of Helsinki, Helsinki
University Central Hospital, Helsinki, FI-00029
HUS, Finland
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska
Institutet, Stockholm
SE-17177, Sweden
| | - Jingmei Li
- Human Genetics Division, Genome Institute of Singapore,
Singapore
138672, Singapore
| | - Jianjun Liu
- Human Genetics Division, Genome Institute of Singapore,
Singapore
138672, Singapore
| | - Keith Humphreys
- Department of Medical Epidemiology and Biostatistics, Karolinska
Institutet, Stockholm
SE-17177, Sweden
| | - Daehee Kang
- Cancer Research Institute, Seoul National University College of
Medicine, Seoul
110-799, Korea
- Department of Biomedical Sciences, Seoul National University
Graduate School, Seoul
151-742, Korea
- Department of Preventive Medicine, Seoul National University
College of Medicine, Seoul
110-799, Korea
| | - Ji-Yeob Choi
- Cancer Research Institute, Seoul National University College of
Medicine, Seoul
110-799, Korea
- Department of Biomedical Sciences, Seoul National University
Graduate School, Seoul
151-742, Korea
| | - Sue K. Park
- Cancer Research Institute, Seoul National University College of
Medicine, Seoul
110-799, Korea
- Department of Biomedical Sciences, Seoul National University
Graduate School, Seoul
151-742, Korea
- Department of Preventive Medicine, Seoul National University
College of Medicine, Seoul
110-799, Korea
| | - Dong-Young Noh
- Department of Surgery, Seoul National University College of
Medicine, Seoul
110-799, Korea
| | - Keitaro Matsuo
- Department of Preventive Medicine, Kyushu University Faculty of
Medical Sciences, Fukuoka
812-8582, Japan
| | - Hidemi Ito
- Division of Epidemiology and Prevention, Aichi Cancer Center
Research Institute, Nagoya, Aichi
464-8681, Japan
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center
Hospital, Nagoya
484-8681, Japan
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer
Center Hospital, Nagoya
484-8681, Japan
| | - Pascal Guénel
- Inserm (National Institute of Health and Medical Research),
CESP (Center for Research in Epidemiology and Population Health), U1018,
Environmental Epidemiology of Cancer, 94807
Villejuif, France
- University Paris-Sud, UMRS 1018, 94807
Villejuif, France
| | - Thérèse Truong
- Inserm (National Institute of Health and Medical Research),
CESP (Center for Research in Epidemiology and Population Health), U1018,
Environmental Epidemiology of Cancer, 94807
Villejuif, France
- University Paris-Sud, UMRS 1018, 94807
Villejuif, France
| | - Florence Menegaux
- Inserm (National Institute of Health and Medical Research),
CESP (Center for Research in Epidemiology and Population Health), U1018,
Environmental Epidemiology of Cancer, 94807
Villejuif, France
- University Paris-Sud, UMRS 1018, 94807
Villejuif, France
| | - Marie Sanchez
- Inserm (National Institute of Health and Medical Research),
CESP (Center for Research in Epidemiology and Population Health), U1018,
Environmental Epidemiology of Cancer, 94807
Villejuif, France
- University Paris-Sud, UMRS 1018, 94807
Villejuif, France
| | - Barbara Burwinkel
- Department of Obstetrics and Gynecology, University of
Heidelberg, 69120
Heidelberg, Germany
- Molecular Epidemiology Group, German Cancer Research Center
(DKFZ), 69120
Heidelberg, Germany
| | - Frederik Marme
- Department of Obstetrics and Gynecology, University of
Heidelberg, 69120
Heidelberg, Germany
- National Center for Tumor Diseases, University of
Heidelberg, 69120
Heidelberg, Germany
| | - Andreas Schneeweiss
- Department of Obstetrics and Gynecology, University of
Heidelberg, 69120
Heidelberg, Germany
- National Center for Tumor Diseases, University of
Heidelberg, 69120
Heidelberg, Germany
| | - Christof Sohn
- Department of Obstetrics and Gynecology, University of
Heidelberg, 69120
Heidelberg, Germany
| | - Anna H. Wu
- Department of Preventive Medicine, Keck School of Medicine,
University of Southern California, Los Angeles,
California
90033, USA
| | - Chiu-chen Tseng
- Department of Preventive Medicine, Keck School of Medicine,
University of Southern California, Los Angeles,
California
90033, USA
| | - David Van Den Berg
- Department of Preventive Medicine, Keck School of Medicine,
University of Southern California, Los Angeles,
California
90033, USA
| | - Daniel O. Stram
- Department of Preventive Medicine, Keck School of Medicine,
University of Southern California, Los Angeles,
California
90033, USA
| | - Javier Benitez
- Centro de Investigación en Red de Enfermedades Raras
(CIBERER), 46010
Valencia, Spain
- Human Genetics Group, Human Cancer Genetics Program, Spanish
National Cancer Research Centre (CNIO), 28029
Madrid, Spain
| | - M. Pilar Zamora
- Servicio de Oncología Médica, Hospital
Universitario La Paz, 28046
Madrid, Spain
| | | | - Primitiva Menéndez
- Servicio de Anatomía Patológica, Hospital
Monte Naranco, 33013
Oviedo, Spain
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt
Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University
School of Medicine, Nashville, Tennessee
37203, USA
| | - Wei Lu
- Shanghai Center for Disease Control and Prevention,
Shanghai
200336, China
| | - Yu-Tang Gao
- Department of Epidemiology, Shanghai Cancer Institute,
Shanghai
200032, China
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt
Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University
School of Medicine, Nashville, Tennessee
37203, USA
| | - Angela Cox
- CRUK/YCR Sheffield Cancer Research Centre, Department of
Oncology, University of Sheffield, Sheffield
S10 2RX, UK
| | - Simon S. Cross
- Academic Unit of Pathology, Department of Neuroscience,
University of Sheffield, Sheffield
S10 2HQ, UK
| | - Malcolm W. R. Reed
- CRUK/YCR Sheffield Cancer Research Centre, Department of
Oncology, University of Sheffield, Sheffield
S10 2RX, UK
| | - Irene L. Andrulis
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai
Hospital, Toronto, Ontario, Canada
M5G 1X5
- Department of Molecular Genetics, University of Toronto,
Toronto, Ontario, Canada
M5S 1A8
| | - Julia A. Knight
- Division of Epidemiology, Dalla Lana School of Public Health,
University of Toronto, Toronto, Ontario,
Canada
M5T 3M7
- Prosserman Centre for Health Research, Lunenfeld-Tanenbaum
Research Institute of Mount Sinai Hospital, Toronto,
Ontario, Canada
M5G 1X5
| | - Gord Glendon
- Ontario Cancer Genetics Network, Lunenfeld-Tanenbaum Research
Institute of Mount Sinai Hospital, Toronto, Ontario,
Canada
M5G 1X5
| | - Sandrine Tchatchou
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai
Hospital, Toronto, Ontario, Canada
M5G 1X5
| | - Elinor J. Sawyer
- Division of Cancer Studies, NIHR Comprehensive Biomedical
Research Centre, Guy’s & St Thomas’ NHS Foundation
Trust in partnership with King's College London, London
SE1 9RT, UK
| | - Ian Tomlinson
- Wellcome Trust Centre for Human Genetics, Oxford Biomedical
Research Centre, University of Oxford, Oxford
OX3 7BN, UK
| | - Michael J. Kerin
- Clinical Science Institute, University Hospital Galway,
Galway, Ireland
| | - Nicola Miller
- Clinical Science Institute, University Hospital Galway,
Galway, Ireland
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine,
University of Southern California Norris Comprehensive Cancer Center,
Los Angeles, California
90033, USA
| | - Brian E. Henderson
- Department of Preventive Medicine, Keck School of Medicine,
University of Southern California Norris Comprehensive Cancer Center,
Los Angeles, California
90033, USA
| | - Fredrick Schumacher
- Department of Preventive Medicine, Keck School of Medicine,
University of Southern California Norris Comprehensive Cancer Center,
Los Angeles, California
90033, USA
| | - Loic Le Marchand
- Epidemiology Program, Cancer Research Center, University of
Hawaii, Honolulu, Hawaii
96813, USA
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska
Institutet, Stockholm
SE-17177, Sweden
| | - Sara Margolin
- Department of Oncology—Pathology, Karolinska
Institutet, Stockholm
SE-17177, Sweden
| | - Soo Hwang TEO
- Breast Cancer Research Unit, University Malaya Cancer Research
Institute, University Malaya Medical Centre, 59100
Kuala Lumpur, Malaysia
- Cancer Research Initiatives Foundation, Sime Darby Medical
Centre, Subang Jaya
47500
Selangor, Malaysia
| | - Cheng Har YIP
- Breast Cancer Research Unit, University Malaya Cancer Research
Institute, University Malaya Medical Centre, 59100
Kuala Lumpur, Malaysia
| | - Daphne S. C. Lee
- Cancer Research Initiatives Foundation, Sime Darby Medical
Centre, Subang Jaya
47500
Selangor, Malaysia
| | - Tien Y. Wong
- Singapore Eye Research Institute, National University of
Singapore, Singapore
168751, Singapore
| | - Maartje J. Hooning
- Department of Medical Oncology, Erasmus MC Cancer
Institute, 3008 AE
Rotterdam, The Netherlands
| | - John W. M. Martens
- Department of Medical Oncology, Erasmus MC Cancer
Institute, 3008 AE
Rotterdam, The Netherlands
| | - J. Margriet Collée
- Department of Clinical Genetics, Erasmus University Medical
Center, 3000 CA
Rotterdam, The Netherlands
| | | | - John L. Hopper
- Centre for Molecular, Environmental, Genetic and Analytical
Epidemiology, Melbourne School of Population Health, University of
Melbourne, Melbourne, Victoria
3010, Australia
| | - Melissa C. Southey
- Department of Pathology, The University of Melbourne,
Melbourne, Victoria
3010, Australia
| | - Helen Tsimiklis
- Department of Pathology, The University of Melbourne,
Melbourne, Victoria
3010, Australia
| | - Miroslav K. Kapuscinski
- Centre for Molecular, Environmental, Genetic and Analytical
Epidemiology, Melbourne School of Population Health, University of
Melbourne, Melbourne, Victoria
3010, Australia
| | - Chen-Yang Shen
- College of Public Health, China Medical University,
Taichung
40402, Taiwan, China
- Institute of Biomedical Sciences, Academia Sinica,
Taipei
115, Taiwan, China
| | - Pei-Ei Wu
- Taiwan Biobank, Institute of Biomedical Sciences, Academia
Sinica, Taipei
115, Taiwan, China
| | - Jyh-Cherng Yu
- Department of Surgery, Tri-Service General Hospital,
Taipei
114, Taiwan, China
| | - Shou-Tung Chen
- Department of Surgery, Changhua Christian Hospital,
Changhua City
500, Taiwan, China
| | - Grethe Grenaker Alnæs
- Department of Genetics, Institute for Cancer Research, Oslo
University Hospital, Radiumhospitalet, N-0310
Oslo, Norway
| | - Anne-Lise Borresen-Dale
- Department of Genetics, Institute for Cancer Research, Oslo
University Hospital, Radiumhospitalet, N-0310
Oslo, Norway
- Institute of Clinical Medicine, University of Oslo (UiO),
0318
Oslo, Norway
| | - Graham G. Giles
- Cancer Epidemiology Centre, Cancer Council Victoria,
Melbourne, Victoria
3004, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of
Population and Global Health, The University of Melbourne,
Melbourne, Victoria
3010, Australia
| | - Roger L. Milne
- Centre for Epidemiology and Biostatistics, Melbourne School of
Population and Global Health, The University of Melbourne,
Melbourne, Victoria
3010, Australia
- Cancer Epidemiology Centre, The Cancer Council Victoria,
Melbourne, Victoria
3053, Australia
| | - Catriona McLean
- Anatomical Pathology, The Alfred Hospital,
Melbourne, Victoria
3004, Australia
| | - Kenneth Muir
- Division of Health Sciences, Warwick Medical School, Warwick
University, Coventry
CV4 7AL, UK
- Institute of Population Health, University of Manchester,
Manchester
M13 9PL, UK
| | - Artitaya Lophatananon
- Division of Health Sciences, Warwick Medical School, Warwick
University, Coventry
CV4 7AL, UK
| | - Sarah Stewart-Brown
- Division of Health Sciences, Warwick Medical School, Warwick
University, Coventry
CV4 7AL, UK
| | | | - Mikael Hartman
- Department of Surgery, Yong Loo Lin School of Medicine,
National University of Singapore and National University Health System,
Singapore
119228, Singapore
- Saw Swee Hock School of Public Health, National University of
Singapore and National University Health System, Singapore
117597, Singapore
| | - Hui Miao
- Saw Swee Hock School of Public Health, National University of
Singapore and National University Health System, Singapore
117597, Singapore
| | | | - Yik Ying Teo
- Saw Swee Hock School of Public Health, National University of
Singapore and National University Health System, Singapore
117597, Singapore
- Department of Statistics and Applied Probability, National
University of Singapore, Singapore
117546, Singapore
| | - Peter A. Fasching
- Division of Hematology and Oncology, Department of Medicine,
David Geffen School of Medicine, University of California at Los Angeles,
Los Angeles, California
90095, USA
- Department of Gynecology and Obstetrics, University Breast
Center Franconia, University Hospital Erlangen, Friedrich-Alexander University
Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN,
91054
Erlangen, Germany
| | - Lothar Haeberle
- Department of Gynecology and Obstetrics, University Breast
Center Franconia, University Hospital Erlangen, Friedrich-Alexander University
Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN,
91054
Erlangen, Germany
| | - Arif B. Ekici
- Institute of Human Genetics, University Hospital Erlangen,
Friedrich Alexander University Erlangen-Nuremberg, 91054
Erlangen, Germany
| | - Matthias W. Beckmann
- Department of Gynecology and Obstetrics, University Breast
Center Franconia, University Hospital Erlangen, Friedrich-Alexander University
Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN,
91054
Erlangen, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German
Cancer Research Center (DKFZ), 69120
Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120
Heidelberg, Germany
| | - Aida Karina Dieffenbach
- Division of Clinical Epidemiology and Aging Research, German
Cancer Research Center (DKFZ), 69120
Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120
Heidelberg, Germany
| | - Volker Arndt
- Division of Clinical Epidemiology and Aging Research, German
Cancer Research Center (DKFZ), 69120
Heidelberg, Germany
| | | | - Anthony Swerdlow
- Division of Breast Cancer Research, Institute of Cancer
Research, London
SM2 5NG, UK
- Division of Genetics and Epidemiology, Institute of Cancer
Research, London
SM2 5NG, UK
| | - Alan Ashworth
- Breakthrough Breast Cancer Research Centre, Division of Breast
Cancer Research, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Nick Orr
- Breakthrough Breast Cancer Research Centre, Division of Breast
Cancer Research, The Institute of Cancer Research, London
SW3 6JB, UK
| | - Minouk J. Schoemaker
- Division of Genetics and Epidemiology, Institute of Cancer
Research, London
SM2 5NG, UK
| | - Montserrat García-Closas
- Breakthrough Breast Cancer Research Centre, Division of Breast
Cancer Research, The Institute of Cancer Research, London
SW3 6JB, UK
- Division of Genetics and Epidemiology, Institute of Cancer
Research, Sutton, Surrey
SM2 5NG, UK
| | - Jonine Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer
Institute, Rockville, Maryland
20850, USA
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer
Institute, Rockville, Maryland
20850, USA
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, M.
Sklodowska-Curie Memorial Cancer Center and Institute of Oncology,
02-781
Warsaw, Poland
| | - Jacques Simard
- Cancer Genomics Laboratory, Centre Hospitalier Universitaire
de Québec Research Center, Laval University, Quebec,
Canada
G1V 4G2
| | - Mark S. Goldberg
- Department of Medicine, McGill University,
Montreal, Quebec, Canada
H3G 2M1
- Division of Clinical Epidemiology, McGill University Health
Centre, Royal Victoria Hospital, Montreal, Quebec,
Canada
H3A 1A8
| | - France Labrèche
- Département de médecine sociale et
préventive, Département de santé environnementale et
santé au travail, Université de Montréal,
Montreal, Quebec, Canada
H3T 1A8
| | - Martine Dumont
- Cancer Genomics Laboratory, Centre Hospitalier Universitaire
de Québec Research Center, Laval University, Quebec,
Canada
G1V 4G2
| | - Robert Winqvist
- Laboratory of Cancer Genetics and Tumor Biology, Department of
Clinical Chemistry and Biocenter Oulu, NordLab Oulu/Oulu University Hospital,
University of Oulu, FI-90220
Oulu, Finland
| | - Katri Pylkäs
- Laboratory of Cancer Genetics and Tumor Biology, Department of
Clinical Chemistry and Biocenter Oulu, NordLab Oulu/Oulu University Hospital,
University of Oulu, FI-90220
Oulu, Finland
| | - Arja Jukkola-Vuorinen
- Department of Oncology, Oulu University Hospital, University
of Oulu, FI-90220
Oulu, Finland
| | - Hiltrud Brauch
- Dr Margarete Fischer-Bosch-Institute of Clinical
Pharmacology, 70376
Stuttgart, Germany
- University of Tübingen, 72074
Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research
Center (DKFZ), 69120
Heidelberg, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the
German Social Accident Insurance, Institute of the Ruhr University Bochum
(IPA), 44789
Bochum, Germany
| | - Yon-Dschun Koto
- Department of Internal Medicine, Evangelische Kliniken Bonn
gGmbH, Johanniter Krankenhaus, 53113
Bonn, Germany
| | - Paolo Radice
- Unit of Molecular Bases of Genetic Risk and Genetic Testing,
Department of Preventive and Predictive Medicine, Fondazione IRCCS Istituto
Nazionale dei Tumori (INT), 20133
Milan, Italy
| | - Paolo Peterlongo
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare,
20139
Milan, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, Istituto Europeo
di Oncologia (IEO), 20141
Milan, Italy
| | - Sara Volorio
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare,
20139
Milan, Italy
- Cogentech Cancer Genetic Test Laboratory,
20133
Milan, Italy
| | - Thilo Dörk
- Department of Obstetrics and Gynaecology, Hannover Medical
School, 30625
Hannover, Germany
| | - Natalia V. Bogdanova
- Department of Radiation Oncology, Hannover Medical
School, 30625
Hannover, Germany
| | - Sonja Helbig
- Department of Obstetrics and Gynaecology, Hannover Medical
School, 30625
Hannover, Germany
| | - Arto Mannermaa
- Cancer Center of Eastern Finland, University of Eastern
Finland, FI-70211
Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio
University Hospital, FI-70210
Kuopio, Finland
- School of Medicine, Institute of Clinical Medicine, Oncology,
University of Eastern Finland, FI-70211
Kuopio, Finland
| | - Vesa Kataja
- Cancer Center of Eastern Finland, University of Eastern
Finland, FI-70211
Kuopio, Finland
- School of Medicine, Institute of Clinical Medicine, Oncology,
University of Eastern Finland, FI-70211
Kuopio, Finland
| | - Veli-Matti Kosma
- Cancer Center of Eastern Finland, University of Eastern
Finland, FI-70211
Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio
University Hospital, FI-70210
Kuopio, Finland
- School of Medicine, Institute of Clinical Medicine, Oncology,
University of Eastern Finland, FI-70211
Kuopio, Finland
| | - Jaana M. Hartikainen
- Cancer Center of Eastern Finland, University of Eastern
Finland, FI-70211
Kuopio, Finland
- Imaging Center, Department of Clinical Pathology, Kuopio
University Hospital, FI-70210
Kuopio, Finland
- School of Medicine, Institute of Clinical Medicine, Oncology,
University of Eastern Finland, FI-70211
Kuopio, Finland
| | - Peter Devilee
- Department of Human Genetics & Department of
Pathology, Leiden University Medical Center, 2300 RC
Leiden, The Netherlands
| | | | - Caroline Seynaeve
- Family Cancer Clinic, Department of Medical Oncology, Erasmus
MC-Daniel den Hoed Cancer Centre, 3075 EA
Rotterdam, The Netherlands
| | - Christi J. Van Asperen
- Department of Clinical Genetics, Erasmus University Medical
Center, 3000 CA
Rotterdam, The Netherlands
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical
University, 70-115
Szczecin, Poland
| | - Jan Lubinski
- Department of Genetics and Pathology, Pomeranian Medical
University, 70-115
Szczecin, Poland
| | | | - Katarzyna Durda
- Department of Genetics and Pathology, Pomeranian Medical
University, 70-115
Szczecin, Poland
| | - Susan Slager
- Department of Health Sciences Research, Mayo Clinic,
Rochester, Minnesota
55905, USA
| | - Amanda E. Toland
- Department of Molecular Virology, Immunology and Medical
Genetics, Comprehensive Cancer Center, The Ohio State University,
Columbus, Ohio
43210, USA
| | | | - Drakoulis Yannoukakos
- Molecular Diagnostics Laboratory, IRRP, National Centre for
Scientific Research ‘Demokritos’, Aghia Paraskevi
Attikis, Athens
15310, Greece
| | | | | | - Paul Brennan
- International Agency for Research on Cancer,
Lyon, Cedex 08, France
| | - James McKay
- International Agency for Research on Cancer,
Lyon, Cedex 08, France
| | - Ute Hamann
- Molecular Genetics of Breast Cancer, German Cancer Research
Center (DKFZ), 69120
Heidelberg, Germany
| | - Diana Torres
- Molecular Genetics of Breast Cancer, German Cancer Research
Center (DKFZ), 69120
Heidelberg, Germany
- Institute of Human Genetics, Pontificia University
Javeriana, Bogota, DC
11001000, Colombia
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt
Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University
School of Medicine, Nashville, Tennessee
37203, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt
Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University
School of Medicine, Nashville, Tennessee
37203, USA
| | - Hoda Anton-Culver
- Department of Epidemiology, University of California
Irvine, Irvine, California
92697, USA
| | - Susan L. Neuhausen
- Department of Population Sciences, Beckman Research Institute
of City of Hope, Duarte, California
92697, USA
| | - Craig Luccarini
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
| | - Caroline Baynes
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
| | - Shahana Ahmed
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
| | - Mel Maranian
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
| | - Catherine S. Healey
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
| | - Anna González-Neira
- Human Genotyping-CEGEN Unit, Human Cancer Genetics Program,
Spanish National Cancer Research Centre (CNIO), 28029
Madrid, Spain
| | - Guillermo Pita
- Human Genotyping-CEGEN Unit, Human Cancer Genetics Program,
Spanish National Cancer Research Centre (CNIO), 28029
Madrid, Spain
| | - M. Rosario Alonso
- Human Genotyping-CEGEN Unit, Human Cancer Genetics Program,
Spanish National Cancer Research Centre (CNIO), 28029
Madrid, Spain
| | - Nuria Álvarez
- Human Genotyping-CEGEN Unit, Human Cancer Genetics Program,
Spanish National Cancer Research Centre (CNIO), 28029
Madrid, Spain
| | - Daniel Herrero
- Human Genotyping-CEGEN Unit, Human Cancer Genetics Program,
Spanish National Cancer Research Centre (CNIO), 28029
Madrid, Spain
| | - Daniel C. Tessier
- Centre d’innovation Génome Québec
et Université McGill, Montréal,
Quebec, Canada
H3A OG1
| | | | | | - Ines de Santiago
- Cancer Research UK, Cambridge Institute, University of
Cambridge, Robinson Way, Cambridge
CB2 0RE, UK
| | - Jason Carroll
- Cancer Research UK, Cambridge Institute, University of
Cambridge, Robinson Way, Cambridge
CB2 0RE, UK
| | - Carlos Caldas
- Cancer Research UK, Cambridge Institute, University of
Cambridge, Robinson Way, Cambridge
CB2 0RE, UK
| | - Melissa A. Brown
- School of Chemistry and Molecular Biosciences, The University of
Queensland, Brisbane, Queensland
4072, Australia
| | - Mathieu Lupien
- The Princess Margaret Cancer Centre, University Health
Network, Toronto, Ontario, Canada
M5T 2M9
- Ontario Cancer Genetics Network, Lunenfeld-Tanenbaum Research
Institute of Mount Sinai Hospital, Toronto, Ontario,
Canada
M5G 1X5
- Department of Medical Biophysics, University of Toronto,
Toronto, Ontario, Canada
M5G 1L7
| | - Vessela N. Kristensen
- Department of Genetics, Institute for Cancer Research, Oslo
University Hospital, Radiumhospitalet, N-0310
Oslo, Norway
- Institute of Clinical Medicine, University of Oslo (UiO),
0318
Oslo, Norway
- Department of Clinical Molecular Biology (EpiGen), University
of Oslo (UiO), 0450
Oslo, Norway
| | - Paul D P Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
- Centre for Cancer Genetic Epidemiology, Department of Public
Health and Primary Care, University of Cambridge, Cambridge
CB1 8RN, UK
| | - Georgia Chenevix-Trench
- Department of Genetics, QIMR Berghofer Medical Research
Institute, Brisbane, Queensland
4029, Australia
| | - Juliet D French
- Department of Genetics, QIMR Berghofer Medical Research
Institute, Brisbane, Queensland
4029, Australia
- School of Chemistry and Molecular Biosciences, The University of
Queensland, Brisbane, Queensland
4072, Australia
| | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
- Centre for Cancer Genetic Epidemiology, Department of Public
Health and Primary Care, University of Cambridge, Cambridge
CB1 8RN, UK
| | - Alison M. Dunning
- Centre for Cancer Genetic Epidemiology, Department of Oncology,
University of Cambridge, Cambridge
CB1 8RN, UK
| |
Collapse
|
21
|
Leung KP, Qu YH, Qiao DF, Xie WB, Li DR, Xu JT, Wang HJ, Yue X. Critical role of insulin‑like growth factor binding protein‑5 in methamphetamine‑induced apoptosis in cardiomyocytes. Mol Med Rep 2014; 10:2306-12. [PMID: 25230843 PMCID: PMC4214346 DOI: 10.3892/mmr.2014.2572] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 05/14/2014] [Indexed: 11/29/2022] Open
Abstract
Methamphetamine (MA) is a highly abused amphetamine-like psychostimulant. At present, the mechanisms underlying MA-induced cardiotoxicity are poorly understood. The cardiotoxic effects have yet not been clearly elucidated with respect to the apoptotic pathway. Insulin-like growth factor binding protein-5 (IGFBP5) is important for cell growth control and the induction of apoptosis. The aim of the present study was to analyze whether IGFBP5 is involved in MA-induced apoptosis as a novel target. MA-induced apoptosis was observed in neonatal rat ventricular myocytes (NRVMs) in a concentration-dependent manner using a terminal deoxyribonucleotide transferase-mediated dUTP nick end-labeling assay. Using reverse transcription polymerase chain reaction and western blotting, MA was demonstrated to induce concentration-dependent increases in the expression of IGFBP5. Silencing IGFBP5 with small interfering RNA significantly reduced apoptosis and suppressed the expression of caspase-3 in NRVMs following treatment with MA. To the best of our knowledge, the present study provided the first evidence suggesting that IGFBP5 is a potential therapeutic target in MA-induced apoptosis in vitro, providing a foundation for future in vivo studies.
Collapse
Affiliation(s)
- Ka-Pui Leung
- Department of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yi-Hong Qu
- Department of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Dong-Fang Qiao
- Department of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wei-Bing Xie
- Department of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Dong-Ri Li
- Department of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jing-Tao Xu
- Department of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hui-Jun Wang
- Department of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xia Yue
- Department of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
22
|
Vijayan A, Guha D, Ameer F, Kaziri I, Mooney C, Bennett L, Sureshbabu A, Tonner E, Beattie J, Allan G, Edwards J, Flint D. IGFBP-5 enhances epithelial cell adhesion and protects epithelial cells from TGFβ1-induced mesenchymal invasion. Int J Biochem Cell Biol 2013; 45:2774-85. [DOI: 10.1016/j.biocel.2013.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 09/12/2013] [Accepted: 10/01/2013] [Indexed: 12/16/2022]
|
23
|
Gajewska M, Zielniok K, Debski B, Motyl T. IGF-I retards proper development of acinar structures formed by bovine mammary epithelial cells via sustained activation of Akt kinase. Domest Anim Endocrinol 2013; 45:111-21. [PMID: 23932581 DOI: 10.1016/j.domaniend.2013.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 06/24/2013] [Accepted: 06/26/2013] [Indexed: 11/29/2022]
Abstract
Insulin-like growth factor-I is involved in mammary gland development, promoting proliferation and inhibiting apoptosis of mammary epithelial cells (MECs). Mitogenic actions of IGF-I are mainly mediated by the phosphatidylinositol-3 kinase (PI3K)/Akt signaling pathway. We have found that in the presence of IGF-I bovine BME-UV1 MECs cultured on reconstituted basement membrane form large spheroids with disrupted polarity and no cavity in the center. These cells showed enhanced phosphorylation of Akt, decreased level of cleaved caspase-3, and sustained proliferative activity throughout the 16-d period of 3-dimensional culture. Inhibition of the PI3K/Akt pathway by a specific inhibitor of PI3K, LY294002, resulted in the restoration of the normal acinar phenotype. However, this effect was noted only when LY294002 was added in the second week of 3-dimensional culture, which corresponded with the time of cell cycle arrest and polarity formation under control conditions. Normal development of acini was also obtained when BME-UV1 cells were treated simultaneously with IGF-I and 17β-estradiol. The addition of 17β-estradiol regulated Akt activation, enabling the subsequent initiation of polarization processes. 17β-Estradiol also increased the level of IGFBP-3 protein in MECs cultured on Matrigel in the presence of IGF-I. The presented results indicate important interactions between signaling pathways activated by estrogen and IGF-I, which regulate alveologenesis in bovine mammary gland.
Collapse
Affiliation(s)
- M Gajewska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska 159, 02-776 Warsaw, Poland.
| | | | | | | |
Collapse
|
24
|
Song SE, Kim YW, Kim JY, Lee DH, Kim JR, Park SY. IGFBP5 mediates high glucose-induced cardiac fibroblast activation. J Mol Endocrinol 2013; 50:291-303. [PMID: 23417767 DOI: 10.1530/jme-12-0194] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This study examined whether IGF-binding protein 5 (IGFBP5) is involved in the high glucose-induced deteriorating effects in cardiac cells. Cardiac fibroblasts and cardiomyocytes were isolated from the hearts of 1- to 3-day-old Sprague Dawley rats. Treatment of fibroblasts with 25 mM glucose increased the number of cells and the mRNA levels of collagen III, matrix metalloproteinase 2 (MMP2), and MMP9. High glucose increased ERK1/2 activity, and the ERK1/2 inhibitor PD98059 suppressed high glucose-mediated fibroblast proliferation and increased collagen III mRNA levels. Whereas high glucose increased both mRNA and protein levels of IGFBP5 in fibroblasts, high glucose did not affect IGFBP5 protein levels in cardiomyocytes. The high glucose-induced increase in IGFBP5 protein levels was inhibited by PD98059 in fibroblasts. While recombinant IGFBP5 increased ERK phosphorylation, cell proliferation, and the mRNA levels of collagen III, MMP2, and MMP9 in fibroblasts, IGFBP5 increased c-Jun N-terminal kinase phosphorylation and induced apoptosis in cardiomyocytes. The knockdown of IGFBP5 inhibited high glucose-induced cell proliferation and collagen III mRNA levels in fibroblasts. Although high glucose increased IGF1 levels, IGF1 did not increase IGFBP5 levels in fibroblasts. The hearts of Otsuka Long-Evans Tokushima Fatty rats and the cardiac fibroblasts of streptozotocin-induced diabetic rats showed increased IGFBP5 expression. These results suggest that IGFBP5 mediates high glucose-induced profibrotic effects in cardiac fibroblasts.
Collapse
Affiliation(s)
- Seung Eun Song
- Department of Physiology, College of Medicine, Yeungnam University, Daegu 705-717, South Korea
| | | | | | | | | | | |
Collapse
|
25
|
Intramammary infusion of Panax ginseng extract in the bovine mammary gland at cessation of milking modifies components of the insulin-like growth factor system during involution. Res Vet Sci 2013; 94:462-70. [DOI: 10.1016/j.rvsc.2013.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 12/10/2012] [Accepted: 01/10/2013] [Indexed: 02/07/2023]
|
26
|
Li Q, Seo JH, Stranger B, McKenna A, Pe'er I, Laframboise T, Brown M, Tyekucheva S, Freedman ML. Integrative eQTL-based analyses reveal the biology of breast cancer risk loci. Cell 2013; 152:633-41. [PMID: 23374354 DOI: 10.1016/j.cell.2012.12.034] [Citation(s) in RCA: 257] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 10/20/2012] [Accepted: 12/20/2012] [Indexed: 01/16/2023]
Abstract
Germline determinants of gene expression in tumors are infrequently studied due to the complexity of transcript regulation caused by somatically acquired alterations. We performed expression quantitative trait locus (eQTL)-based analyses using the multi-level information provided in The Cancer Genome Atlas (TCGA). Of the factors we measured, cis-acting eQTLs accounted for 1.2% of the total variation of tumor gene expression, while somatic copy-number alteration and CpG methylation accounted for 7.3% and 3.3%, respectively. eQTL analyses of 15 previously reported breast cancer risk loci resulted in the discovery of three variants that are significantly associated with transcript levels (false discovery rate [FDR] < 0.1). Our trans-based analysis identified an additional three risk loci to act through ESR1, MYC, and KLF4. These findings provide a more comprehensive picture of gene expression determinants in breast cancer as well as insights into the underlying biology of breast cancer risk loci.
Collapse
Affiliation(s)
- Qiyuan Li
- Department of Medical Oncology, The Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Mammary gland development is delayed in mice deficient for aminopeptidase N. Transgenic Res 2012; 22:425-34. [PMID: 22983824 PMCID: PMC7088532 DOI: 10.1007/s11248-012-9654-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 09/05/2012] [Indexed: 02/02/2023]
Abstract
Development of the mammary gland requires the coordinated action of proteolytic enzymes during two phases of remodelling. Firstly, new ducts and side-branches thereof need to be established during pregnancy to generate an extensive ductal tree allowing the secretion and transport of milk. A second wave of remodelling occurs during mammary involution after weaning. We have analysed the role of the cell surface protease aminopeptidase N (Anpep, APN, CD13) during these processes using Anpep deficient and Anpep over-expressing mice. We find that APN deficiency significantly delays mammary gland morphogenesis during gestation. The defect is characterised by a reduction in alveolar buds and duct branching at mid-pregnancy. Conversely over-expression of Anpep leads to accelerated ductal development. This indicates that Anpep plays a critical role in the proteolytic remodelling of mammary tissue during adult mammary development.
Collapse
|
28
|
Sureshbabu A, Okajima H, Yamanaka D, Tonner E, Shastri S, Maycock J, Szymanowska M, Shand J, Takahashi SI, Beattie J, Allan G, Flint D. IGFBP5 induces cell adhesion, increases cell survival and inhibits cell migration in MCF-7 human breast cancer cells. J Cell Sci 2012; 125:1693-705. [PMID: 22328518 DOI: 10.1242/jcs.092882] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024] Open
Abstract
Maintenance of tissue boundaries is crucial for control of metastasis. We describe a new signalling pathway in which epithelial cell disruption can be minimised and thereby restricts epithelial-mesenchymal transgressions. This involves the release of insulin-like growth factor (IGF)-binding protein 5 (IGFBP5) from apoptotic cells, which increases the adhesion of epithelial cells on mesenchymal but not epithelial extracellular matrix (ECM), and involves the direct interaction of IGFBP5 and α2β1 integrins. IGFBP5 also induced cell adhesion to vitronectin in the absence of αVβ3 integrin, the vitronectin receptor, again through an α2β1-integrin-dependent action, suggesting that IGFBP5 can induce spreading on matrices, even in the absence of the integrins normally used in this process. Using IGFBP5 mutants we demonstrate that the effect is IGF-independent but requires the heparin-binding domain in the C-terminus of IGFBP5. A truncated mutant containing only the C-terminal of IGFBP5 also induced adhesion. Adhesion induced by IGFBP5 was dependent on Cdc42 and resulted in activation of integrin-linked kinase (ILK) and Akt. Consistent with these changes, IGFBP5 facilitated prolonged cell survival in nutrient-poor conditions and decreased phosphorylation of the stress-activated kinase p38 MAPK (MAPK14). Whereas IGFBP5 enhanced adhesion, it inhibited cell migration, although this was not evident using the truncated C-terminal mutant, suggesting that effects of IGFBP5 on adhesion and migration involve different mechanisms. We anticipate that these responses to IGFBP5 would reduce the metastatic potential of cells.
Collapse
Affiliation(s)
- Angara Sureshbabu
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Liu BY, Soloviev I, Huang X, Chang P, Ernst JA, Polakis P, Sakanaka C. Mammary tumor regression elicited by Wnt signaling inhibitor requires IGFBP5. Cancer Res 2012; 72:1568-78. [PMID: 22307840 DOI: 10.1158/0008-5472.can-11-3668] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Wnt ligand-driven tumor growth is inhibited by the soluble Wnt inhibitor Fzd8CRD, but the mechanism through which this effect is mediated is unknown. In the MMTV-Wnt1 mouse model, regression of mammary tumors by Fzd8CRD treatment coincides with an acute and strong induction of insulin-like growth factor (IGF)-binding protein IGFBP5, an antagonist of IGF signaling that mediates involution of mammary gland in females after offspring are weaned. In this study, we show that repression of this IGF inhibitory pathway is crucial for Wnt-driven growth of mammary tumors. We found that IGFBP5 regulation was mediated by the β-catenin-dependent Wnt pathway. Wnt, in addition to IGF ligands, facilitated tumor growth by paracrine communication among tumor cells. In addition, Fzd8CRD caused precocious induction of IGFBP5 in normal mammary glands undergoing involution, implying an acceleration of the involution process by inhibition of Wnt signaling. The molecular and phenotypic parallel between tumor regression and mammary gland involution suggests that Wnt-driven mammary tumors use the same growth mechanism as proliferating normal mammary glands.
Collapse
Affiliation(s)
- Bob Y Liu
- Department of Cancer Targets, Genentech Inc., South San Francisco, California 94080, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Rucker EB, Hale AN, Durtschi DC, Sakamoto K, Wagner KU. Forced involution of the functionally differentiated mammary gland by overexpression of the pro-apoptotic protein bax. Genesis 2011; 49:24-35. [PMID: 21254334 DOI: 10.1002/dvg.20691] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 10/27/2010] [Accepted: 11/02/2010] [Indexed: 01/14/2023]
Abstract
The mammary gland is a developmentally dynamic, hormone-responsive organ that undergoes proliferation and differentiation within the secretory epithelial compartment during pregnancy. The epithelia are maintained by pro-survival signals (e.g., Stat5, Akt1) during lactation, but undergo apoptosis during involution through inactivation of cell survival pathways and upregulation of pro-apoptotic proteins. To assess if the survival signals in the functionally differentiated mammary epithelial cells can override a pro-apoptotic signal, we generated transgenic mice that express Bax under the whey acidic protein (WAP) promoter. WAP-Bax females exhibited a lactation defect and were unable to nourish their offspring. Mammary glands demonstrated: (1) a reduction in epithelial content, (2) hallmark signs of mitochondria-mediated cell death, (3) an increase in apoptotic cells by TUNEL assay, and (4) precocious Stat3 activation. This suggests that upregulation of a single pro-apoptotic factor of the Bcl-2 family is sufficient to initiate apoptosis of functionally differentiated mammary epithelial cells in vivo.
Collapse
Affiliation(s)
- Edmund B Rucker
- Biology Department, University of Kentucky, Lexington, Kentucky, USA.
| | | | | | | | | |
Collapse
|
31
|
Polanco TA, Crismale-Gann C, Reuhl KR, Sarkar DK. Fetal alcohol exposure increases mammary tumor susceptibility and alters tumor phenotype in rats. Alcohol Clin Exp Res 2010; 34:1879-87. [PMID: 20662802 PMCID: PMC4634124 DOI: 10.1111/j.1530-0277.2010.01276.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Altered fetal programming because of a suboptimal in utero environment has been shown to increase susceptibility to many diseases later in life. This study examined the effect of alcohol exposure in utero on N-nitroso-N-methylurea (NMU)-induced mammary cancer risk during adulthood. METHODS Study 1: Pregnant Sprague Dawley rats were fed a liquid diet containing 6.7% ethanol (alcohol-fed), an isocaloric liquid diet (pair-fed), or rat chow ad libitum (ad lib-fed) from day 11 to 21 of gestation. At birth, female pups were cross-fostered to ad lib-fed control dams. Adult offspring were given an I.P. injection of NMU at a dose of 50 mg/kg body weight. Mammary glands were palpated for tumors twice a week, and rats were euthanized at 23 weeks postinjection. Study 2: To investigate the role of estradiol (E2), animals were exposed to the same in utero treatments but were not given NMU. Serum was collected during the preovulatory phase of the estrous cycle. RESULTS At 16 weeks postinjection, overall tumor multiplicity was greater in the offspring from the alcohol-fed group compared to the control groups, indicating a decrease in tumor latency. At study termination, 70% of all animals possessed tumors. Alcohol-exposed animals developed more malignant tumors and more estrogen receptor-α-negative tumors relative to the control groups. In addition, IGF-binding protein-5 (IGFBP-5) mRNA and protein were decreased in tumors of alcohol-exposed animals. Study 2 showed that alcohol-fed animals had significantly increased circulating E2 when compared to either control group. CONCLUSIONS These data indicate that alcohol exposure in utero increases susceptibility to mammary tumorigenesis in adulthood and suggest that alterations in the IGF and E2 systems may play a role in the underlying mechanism.
Collapse
Affiliation(s)
- Tiffany A. Polanco
- Rutgers Endocrine Program, Department of Animal Sciences; Rutgers, The State University of New Jersey, 59 Dudley Rd, New Brunswick, NJ 08901,
| | - Catina Crismale-Gann
- Rutgers Endocrine Program, Department of Animal Sciences; Rutgers, The State University of New Jersey, 59 Dudley Rd, New Brunswick, NJ 08901,
| | - Kenneth R. Reuhl
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy; Rutgers, The State University of New Jersey, 41 Gordon Rd, Piscataway, NJ 08854,
| | - Dipak K. Sarkar
- Rutgers Endocrine Program, Department of Animal Sciences; Rutgers, The State University of New Jersey, 67 Poultry Farm Rd, New Brunswick, NJ 08901,
| |
Collapse
|
32
|
Foote MR, Giesy SL, Bernal-Santos G, Bauman DE, Boisclair YR. t10,c12-CLA decreases adiposity in peripubertal mice without dose-related detrimental effects on mammary development, inflammation status, and metabolism. Am J Physiol Regul Integr Comp Physiol 2010; 299:R1521-8. [PMID: 20844263 DOI: 10.1152/ajpregu.00445.2010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The trans 10, cis 12-conjugated linoleic acid (10,12-CLA) isomer reduces adiposity in several animal models. In the mouse, however, this effect is associated with adipose tissue inflammation, hyperinsulinemia and hepatic lipid accumulation. Moreover, 10,12-CLA was recently shown to promote mammary ductal hyperplasia and ErbB2/Her2-driven mammary cancer in the mouse. Reasons for detrimental effects of 10,12-CLA on the mouse mammary gland could relate to its effect on the mammary fat pad (MFP), which is essential for normal development. Accordingly, we hypothesized that mammary effects of 10,12-CLA were mediated through the MFP in a dose-dependent manner. Female FVB mice were fed 10,12-CLA at doses of 0%, 0.1%, 0.2%, or 0.5% of the diet from day 24 of age, and effects on mammary development and metabolism were measured on day 49. The 0.5% dose reduced ductal elongation and caused premature alveolar budding. These effects were associated with increased expression of inflammatory markers and genes shown to alter epithelial growth (IGF binding protein-5) and alveolar budding (TNF-α and receptor of activated NF-κB ligand). The 0.5% dose also caused hyperinsulinemia and hepatic lipid accumulation. In contrast, the 0.1% 10,12-CLA dose had no adverse effects on mammary development, metabolic events, and inflammatory responses, but remained effective in decreasing adipose weights and lipogenic gene expression. These results show that a low dose of 10,12-CLA reduces adiposity in the mouse without negative effects on mammary development, inflammation, and metabolism, and suggest that previously reported detrimental effects relate to the use of excessive doses.
Collapse
Affiliation(s)
- M R Foote
- Dept. of Animal Science, Cornell Univ., 259 Morrison Hall, Ithaca, NY 14853, USA
| | | | | | | | | |
Collapse
|
33
|
Littlejohn MD, Walker CG, Ward HE, Lehnert KB, Snell RG, Verkerk GA, Spelman RJ, Clark DA, Davis SR. Effects of reduced frequency of milk removal on gene expression in the bovine mammary gland. Physiol Genomics 2009; 41:21-32. [PMID: 19996161 DOI: 10.1152/physiolgenomics.00108.2009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Regulation of milk synthesis and secretion is controlled mostly through local (intramammary) mechanisms. To gain insight into the molecular pathways comprising this response, an analysis of mammary gene expression was conducted in 12 lactating cows shifted from twice daily to once daily milking. Tissues were sampled by biopsy from adjacent mammary quarters of these animals during the two milking frequencies, allowing changes in gene expression to be assessed within each animal. Using bovine-specific, oligonucleotide arrays representing 21,495 unique transcripts, a range of differentially expressed genes were found as a result of less frequent milk removal, constituting transcripts and pathways related to apoptotic signaling (NF-kappaB, JUN, ATF3, IGFBP5, TNFSF12A) mechanical stress and epithelial tight junction synthesis (CYR61, CTGF, THBS1, CLDN4, CLDN8), and downregulated milk synthesis (LALBA, B4GALT1, UGP2, CSN2, GPAM, LPL). Quantitative real-time PCR was used to assess the expression of 13 genes in the study, and all 13 of these were correlated (P < 0.05) with values derived from array analysis. It can be concluded that the physiological changes that occur in the bovine mammary gland as a result of reduced milk removal frequency likely comprise the earliest stages of the involution response and that mechano-signal transduction cascades associated with udder distension may play a role in triggering these events.
Collapse
|
34
|
Sureshbabu A, Okajima H, Yamanaka D, Shastri S, Tonner E, Rae C, Szymanowska M, Shand JH, Takahashi SI, Beattie J, Allan GJ, Flint DJ. IGFBP-5 induces epithelial and fibroblast responses consistent with the fibrotic response. Biochem Soc Trans 2009; 37:882-5. [PMID: 19614612 DOI: 10.1042/bst0370882] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Fibrosis involves activation of fibroblasts, increased production of collagen and fibronectin and transdifferentiation into contractile myofibroblasts. The process resembles aspects of wound-healing but remains unresolved and can be life-threatening when manifest in the kidneys, lungs and liver, in particular. The causes are largely unknown, but recent suggestions that repetitive micro-injury results in the eventual failure of epithelial cell repair due to replicative senescence are gaining favour. This is consistent with the onset of fibrotic diseases in middle age. Because epithelial injury often involves blood loss, inflammatory responses associated with the fibrotic response have been considered as therapeutic targets. However, this has proved largely unsuccessful and focus is now switching to earlier events in the process. These include EMT (epithelial-mesenchymal transition) and fibroblast activation in the absence of inflammation. TGFbeta1 (transforming growth factor-beta1) induces both EMT and fibroblast activation and is considered to be a major pro-fibrotic factor. Recently, IGFBP-5 [IGF (insulin-like growth factor)-binding protein-5] has also been shown to induce similar effects on TGFbeta1, and is strongly implicated in the process of senescence. It also stimulates migration of peripheral blood mononuclear cells, implicating it in the inflammatory response. In this paper, we examine the evidence for a role of IGFBP-5 in fibrosis and highlight its structural relationship with other matrix proteins and growth factors also implicated in tissue remodelling.
Collapse
|
35
|
Stein T, Salomonis N, Nuyten DSA, van de Vijver MJ, Gusterson BA. A mouse mammary gland involution mRNA signature identifies biological pathways potentially associated with breast cancer metastasis. J Mammary Gland Biol Neoplasia 2009; 14:99-116. [PMID: 19408105 DOI: 10.1007/s10911-009-9120-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Accepted: 04/06/2009] [Indexed: 12/15/2022] Open
Abstract
Mouse mammary gland involution resembles a wound healing response with suppressed inflammation. Wound healing and inflammation are also associated with tumour development, and a 'wound-healing' gene expression signature can predict metastasis formation and survival. Recent studies have shown that an involuting mammary gland stroma can promote metastasis. It could therefore be hypothesised that gene expression signatures from an involuting mouse mammary gland may provide new insights into the physiological pathways that promote breast cancer progression. Indeed, using the HOPACH clustering method, the human orthologues of genes that were differentially regulated at day 3 of mammary gland involution and showed prolonged expression throughout the first 4 days of involution distinguished breast cancers in the NKI 295 breast cancer dataset with low and high metastatic activity. Most strikingly, genes associated with copper ion homeostasis and with HIF-1 promoter binding sites were the most over-represented, linking this signature to hypoxia. Further, six out of the ten mRNAs with strongest up-regulation in cancers with poor survival code for secreted factors, identifying potential candidates that may be involved in stromal/matrix-enhanced metastasis formation/breast cancer development. This method therefore identified biological processes that occur during mammary gland involution, which may be critical in promoting breast cancer metastasis that could form a basis for future investigation, and supports a role for copper in breast cancer development.
Collapse
Affiliation(s)
- Torsten Stein
- Division of Cancer Sciences and Molecular Pathology, Section of Gene Regulation and Mechanisms of Disease, Western Infirmary, University of Glasgow, Glasgow, UK.
| | | | | | | | | |
Collapse
|
36
|
Tripathi G, Salih DAM, Drozd AC, Cosgrove RA, Cobb LJ, Pell JM. IGF-independent effects of insulin-like growth factor binding protein-5 (Igfbp5) in vivo. FASEB J 2009; 23:2616-26. [PMID: 19332648 DOI: 10.1096/fj.08-114124] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
IGF activity is regulated tightly by a family of IGF binding proteins (IGFBPs). IGFBP-5 is the most conserved of these and is up-regulated significantly during differentiation of several key lineages and in some cancers. The function of IGFBP-5 in these physiological and pathological situations is unclear, however, several IGFBP-5 sequence motifs and studies in vitro suggest IGF-independent actions. Therefore, we aimed to compare the phenotypes of mice overexpressing wild-type Igfbp5 or an N-terminal mutant Igfbp5 with negligible IGF binding affinity. Both significantly inhibited growth, even at low expression levels. Even though wild-type IGFBP-5 severely disrupted the IGF axis, we found no evidence for interaction of mutant IGFBP-5 with the IGF system. Further, overexpression of wild-type IGFBP-5 rescued the lethal phenotype induced by "excess" IGF-II in type 2 receptor-null mice; mutant IGFBP-5 overexpression could not. Therefore, wild-type IGFBP-5 provides a very effective mechanism for the inhibition of IGF activity and a powerful in vivo mechanism to inhibit IGF activity in pathologies such as cancer. This study is also the first to suggest significant IGF-independent actions for IGFBP-5 during development.
Collapse
Affiliation(s)
- Gyanendra Tripathi
- Laboratory of Molecular Signalling, The Babraham Institute, Cambridge CB22 3AT, UK
| | | | | | | | | | | |
Collapse
|
37
|
Kleinberg DL, Wood TL, Furth PA, Lee AV. Growth hormone and insulin-like growth factor-I in the transition from normal mammary development to preneoplastic mammary lesions. Endocr Rev 2009; 30:51-74. [PMID: 19075184 PMCID: PMC5393153 DOI: 10.1210/er.2008-0022] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Accepted: 11/18/2008] [Indexed: 12/20/2022]
Abstract
Adult female mammary development starts at puberty and is controlled by tightly regulated cross-talk between a group of hormones and growth factors. Although estrogen is the initial driving force and is joined by luteal phase progesterone, both of these hormones require GH-induced IGF-I in the mammary gland in order to act. The same group of hormones, when experimentally perturbed, can lead to development of hyperplastic lesions and increase the chances, or be precursors, of mammary carcinoma. For example, systemic administration of GH or IGF-I causes mammary hyperplasia, and overproduction of IGF-I in transgenic animals can cause the development of usual or atypical hyperplasias and sometimes carcinoma. Although studies have clearly demonstrated the transforming potential of both GH and IGF-I receptor in cell culture and in animals, debate remains as to whether their main role is actually instructive or permissive in progression to cancer in vivo. Genetic imprinting has been shown to occur in precursor lesions as early as atypical hyperplasia in women. Thus, the concept of progression from normal development to cancer through precursor lesions sensitive to hormones and growth factors discussed above is gaining support in humans as well as in animal models. Indeed, elevation of estrogen receptor, GH, IGF-I, and IGF-I receptor during progression suggests a role for these pathways in this process. New agents targeting the GH/IGF-I axis may provide a novel means to block formation and progression of precursor lesions to overt carcinoma. A novel somatostatin analog has recently been shown to prevent mammary development in rats via targeted IGF-I action inhibition at the mammary gland. Similarly, pegvisomant, a GH antagonist, and other IGF-I antagonists such as IGF binding proteins 1 and 5 also block mammary gland development. It is, therefore, possible that inhibition of IGF-I action, or perhaps GH, in the mammary gland may eventually play a role in breast cancer chemoprevention by preventing actions of both estrogen and progesterone, especially in women at extremely high risk for developing breast cancer such as BRCA gene 1 or 2 mutations.
Collapse
Affiliation(s)
- David L Kleinberg
- Neuroendocrine Unit, Department of Medicine, New York University School of Medicine, 550 First Avenue, New York, New York 10016, USA.
| | | | | | | |
Collapse
|
38
|
Manickam R, Pena RN, Whitelaw CBA. Mammary gland differentiation inversely correlates with GDF-8 expression. Mol Reprod Dev 2008; 75:1783-8. [PMID: 18389502 DOI: 10.1002/mrd.20918] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
GDF-8 is recognised as an inhibitor of muscle cell growth and differentiation. Although initially thought to be restricted to muscle cells it is now accepted that GDF-8 expression has a broader tissue distribution. We demonstrate GDF-8 expression in the mouse mammary gland, which is predominantly associated with epithelial cells and displays an inverse correlation to the differentiated state of the gland. Specifically, the highest GDF-8 mRNA levels correlate with periods of maximal ductal growth, diminish as pregnancy progressed and are down-regulated to minimal levels by the onset of lactation as the epithelium differentiates. A similar profile is observed for both GDF-8 protein processing and reflects Smad2/3 phosphorylation profile. However, in contrast to muscle cells, GDF-8 neither reduces proliferation nor induces p21 expression levels in mammary epithelial cells. These data implicate a role for GDF-8 in mammary epithelial cell differentiation and demonstrate that GDF-8 has cell-type specific activities.
Collapse
Affiliation(s)
- Ravikumar Manickam
- Division of Gene Function and Development, Roslin Institute, Roslin, Edinburgh, United Kingdom
| | | | | |
Collapse
|
39
|
Flint DJ, Tonner E, Beattie J, Allan GJ. Role of insulin-like growth factor binding proteins in mammary gland development. J Mammary Gland Biol Neoplasia 2008; 13:443-53. [PMID: 18998203 DOI: 10.1007/s10911-008-9095-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Accepted: 10/28/2008] [Indexed: 01/08/2023] Open
Abstract
Insulin-like growth factors (IGFs) play an important role in mammary gland development and their effects are, in turn, influenced by a family of 6 IGF-binding proteins (IGFBPs). The IGFBPs are expressed in time- and tissue-specific fashion during the periods of rapid growth and involution of the mammary gland. The precise roles of these proteins in vivo have, however, been difficult to determine. This review examines the indirect evidence (evolution, chromosomal location and roles in lower life-forms) the evidence from in vitro studies and the attempts to examine their roles in vivo, using IGFBP-deficient and over-expression models. Evidence exists for a role of the IGFBPs in inhibition of the survival effects of IGFs as well as in IGF-enhancing effects from in vitro studies. The location of the IGFBPs, often associated with the extracellular matrix, suggests roles as a reservoir of IGFs or as a potential barrier, restricting access of IGFs to distinct cellular compartments. We also discuss the relative importance of IGF-dependent versus IGF-independent effects. IGF-independent effects include nuclear localization, activation of proteases and interaction with a variety of extracellular matrix and cell surface proteins. Finally, we examine the increasing evidence for the IGFBPs to be considered as part of a larger family of extracellular matrix proteins involved in morphogenesis and tissue re-modeling.
Collapse
Affiliation(s)
- D J Flint
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0NR, UK.
| | | | | | | |
Collapse
|
40
|
Abstract
Dramatic changes in cell composition and function occur in the mammary gland during a pregnancy-lactation-involution cycle. We investigated the transcriptional changes associated with these biological events by using microarray analysis and identified the critical genes involved by using genetically modified mice. Two surprising findings arose from these studies. First, the microarray data showed that postlactational regression was associated with an acute phase inflammatory response, in addition to cell death. Conditional deletion of signal transducer and activator of transcription (Stat)3 or the nuclear factor-kappaB regulatory kinase inhibitor of kappa B kinase beta resulted in a failure of cell death induction during involution, an indication that these signaling pathways are essential mediators of the involution process. Both Stat3 and nuclear factor-kappaB have been shown to regulate acute phase gene expression in addition to apoptosis regulators. Four distinct transcriptional profiles are present in the first 4 d of involution, whereas there are 3 in lactation. At the peak of lactation (i.e., d 10 in mouse), more than 400 genes reach their maximum expression before declining dramatically in the first 12 h of involution. A reciprocal pattern was observed for more than 500 genes that were specifically upregulated within the first 12 h of forced involution. We are now investigating the role of a subset of these genes in involution. We also uncovered a role for genes normally associated with immune cell signaling in the differentiation of luminal mammary epithelial cells during pregnancy. Genetic deletion of the transcription factor Stat6 resulted in delayed development during pregnancy, and this phenotype was recapitulated in mammary tissue from IL-4 and IL-13 doubly deficient mice. Furthermore, we showed that mammary epithelial cells secrete T-cell regulatory cytokines. T-helper type 1 cytokines, such as interferon-gamma and IL-12a, are secreted by undifferentiated mammary epithelial cells, whereas T-helper type 2 cytokines, including IL-4 and IL-13, are secreted by differentiated cells. This unexpected finding demonstrates a role for immune cell signaling in mammary epithelial cell fate and function.
Collapse
Affiliation(s)
- C J Watson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom.
| |
Collapse
|
41
|
Akkiprik M, Feng Y, Wang H, Chen K, Hu L, Sahin A, Krishnamurthy S, Ozer A, Hao X, Zhang W. Multifunctional roles of insulin-like growth factor binding protein 5 in breast cancer. Breast Cancer Res 2008; 10:212. [PMID: 18710598 PMCID: PMC2575530 DOI: 10.1186/bcr2116] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The insulin-like growth factor axis, which has been shown to protect cells from apoptosis, plays an essential role in normal cell physiology and in cancer development. The family of insulin-like growth factor binding proteins (IGFBPs) has been shown to have a diverse spectrum of functions in cell growth, death, motility, and tissue remodeling. Among the six IGFBP family members, IGFBP-5 has recently been shown to play an important role in the biology of breast cancer, especially in breast cancer metastasis; however, the exact mechanisms of action remain obscure and sometimes paradoxical. An in-depth understanding of IGFBP-5 would shed light on its potential role as a target for breast cancer therapeutics.
Collapse
Affiliation(s)
- Mustafa Akkiprik
- Department of Medical Biology, Marmara University, School of Medicine, 34668 Istanbul, Turkey.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Watkin H, Richert MM, Lewis A, Terrell K, McManaman JP, Anderson SM. Lactation failure in Src knockout mice is due to impaired secretory activation. BMC DEVELOPMENTAL BIOLOGY 2008; 8:6. [PMID: 18215306 PMCID: PMC2266720 DOI: 10.1186/1471-213x-8-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Accepted: 01/23/2008] [Indexed: 11/17/2022]
Abstract
Background Mammary gland development culminates in lactation and is orchestrated by numerous stimuli and signaling pathways. The Src family of nonreceptor tyrosine kinases plays a pivotal role in cell signaling. In order to determine if Src plays a role in mammary gland development we have examined mammary gland development and function during pregnancy and lactation in mice in which expression of Src has been eliminated. Results We have characterized a lactation defect in the Src-/- mice which results in the death of over 80% of the litters nursed by Src-/- dams. Mammary gland development during pregnancy appears normal in these mice; however secretory activation does not seem to occur. Serum prolactin levels are normal in Src-/- mice compared to wildtype controls. Expression of the prolactin receptor at both the RNA and protein level was decreased in Src-/- mice following the transition from pregnancy to lactation, as was phosphorylation of STAT5 and expression of milk protein genes. These results suggest that secretory activation, which occurs following parturition, does not occur completely in Src-/- mice. Failed secretory activation results in precocious involution in the mammary glands of Src-/- even when pups were suckling. Involution was accelerated following pup withdrawal perhaps as a result of incomplete secretory activation. In vitro differentiation of mammary epithelial cells from Src-/- mice resulted in diminished production of milk proteins compared to the amount of milk proteins produced by Src+/+ cells, indicating a direct role for Src in regulating the transcription/translation of milk protein genes in mammary epithelial cells. Conclusion Src is an essential signaling modulator in mammary gland development as Src-/- mice exhibit a block in secretory activation that results in lactation failure and precocious involution. Src appears to be required for increased expression of the prolactin receptor and successful downstream signaling, and alveolar cell organization.
Collapse
Affiliation(s)
- Harriet Watkin
- Department of Pathology, University of Colorado Health Sciences Center, Research Complex I, South Tower, Mail Stop 8104, 12801 East 17th Avenue, Aurora, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Jurgeit A, Berlato C, Obrist P, Ploner C, Massoner P, Schmölzer J, Haffner MC, Klocker H, Huber LA, Geley S, Doppler W. Insulin-like growth factor-binding protein-5 enters vesicular structures but not the nucleus. Traffic 2007; 8:1815-1828. [PMID: 17892529 DOI: 10.1111/j.1600-0854.2007.00655.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In addition to its extracellular function as a secreted protein, IGF-binding protein (IGFBP)-5 has been postulated to act as a signaling molecule in the nucleus. This study aims to assess the significance of this postulated nuclear localization. By confocal immunofluorescence microscopy, we detected IGFBP-5 in the vesicular compartment of mammary epithelial cells in culture, while no nuclear staining was observed. Immunohistochemistry performed on paraffin sections of the involuting mammary gland revealed IGFBP-5 positive staining of epithelial cells only outside the nucleus. To evaluate the contribution of reuptake of extracellular IGFBP-5, T47D cells were incubated with Alexa Fluor 647-labeled IGFBP-5. The protein was taken up into intracellular vesicles and again was neither detectable in the cytoplasm outside of vesicular structures nor in the nucleus. Quantification of the time and concentration dependence of uptake by immunoblotting revealed that the process was saturable at IGFBP-5 concentrations between 1 and 2 mum and partially reversible with 30% remaining in the cell after a 1-h chase. The observation of nuclear uptake of IGFBP-5 was restricted to artificial conditions such as expression of non-secreted forms of IGFBP-5 or selective permeabilization of the plasma membrane by digitonin.
Collapse
Affiliation(s)
- Andreas Jurgeit
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
- Present address: Institute of Zoology, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Chiara Berlato
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Peter Obrist
- Department of Pathology, St. Vinzenz Hospital Zams, 6511 Zams, Austria
| | - Christian Ploner
- Division of Molecular Pathophysiology, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Petra Massoner
- Department of Urology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Judith Schmölzer
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Michael C Haffner
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Helmut Klocker
- Department of Urology, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Lukas A Huber
- Division of Cell Biology, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Stephan Geley
- Division of Molecular Pathophysiology, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| | - Wolfgang Doppler
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Fritz-Pregl-Strasse 3, 6020 Innsbruck, Austria
| |
Collapse
|
44
|
Ning Y, Hoang B, Schuller AGP, Cominski TP, Hsu MS, Wood TL, Pintar JE. Delayed mammary gland involution in mice with mutation of the insulin-like growth factor binding protein 5 gene. Endocrinology 2007; 148:2138-47. [PMID: 17255210 DOI: 10.1210/en.2006-0041] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
IGFs (IGF-I and IGF-II) are essential for development, and their bioactivities are tightly regulated by six related IGF-binding proteins (IGFBPs). IGFBP-5 is the most highly conserved binding protein and is expressed in several key developmental lineages as well as in multiple adult tissues including the mammary gland. To explore IGFBP-5 actions in vivo, we produced IGFBP-5 knockout (KO) mice. Whole-body growth, selected organ weights, and body composition were essentially normal in IGFBP-5 KO mice, presumably because of substantial compensation by remaining IGFBP family members. The IGFBP-5 KO mice also exhibited normal mammary gland development and were capable of nursing their pups. We then directly evaluated the proposed role of IGFBP-5 in apoptosis and remodeling of mammary gland during involution. We found that the process of involution after forced weaning was delayed in IGFBP-5 KO mice, with both the appearance of apoptotic cells and the reappearance of adipocytes retarded in mutant mice, compared with controls. We also determined the effects of IGFBP-5 deletion on mammary gland development in pubertal females after ovariectomy and stimulation with estradiol/progesterone. In this paradigm, IGFBP-5 KO mammary glands exhibited enhanced alveolar bud formation consistent with enhanced IGF-I action. These results demonstrate that IGFBP-5, although not essential for normal growth, is required for normal mammary gland involution and can regulate mammary gland morphogenesis in response to hormone stimulation.
Collapse
Affiliation(s)
- Yun Ning
- Department of Neuroscience, University of Medicine and Dentistry of New Jersey, Piscataway, New Jersey 08854, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Sakamoto K, Yano T, Kobayashi T, Hagino A, Aso H, Obara Y. Growth hormone suppresses the expression of IGFBP-5, and promotes the IGF-I-induced phosphorylation of Akt in bovine mammary epithelial cells. Domest Anim Endocrinol 2007; 32:260-72. [PMID: 16698222 DOI: 10.1016/j.domaniend.2006.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2005] [Revised: 03/24/2006] [Accepted: 03/27/2006] [Indexed: 11/25/2022]
Abstract
Growth hormone (GH) plays a specific role to inhibit apoptosis in the bovine mammary gland through the insulin-like growth factor (IGF)-I system, however, the mechanism of GH action is poorly understood. In this study, we show that GH dramatically inhibits the expression of IGFBP-5, and GH along with IGF-I enhanced the phosphorylation of Akt through the reduction of IGF binding protein (IGFBP)-5. To determine how GH affects Akt through IGF-I in bovine mammary epithelial cells (BMECs), we examined the phosphorylation of Akt in GH treated BMECs and found that IGF-I induced phosphorylation of Akt was significantly enhanced by the treatment with GH. We demonstrated that GH reduces mRNA and protein expression of IGFBP-5 in BMECs, but it does not affect the expression of IGFBP-3. To determine that the enhanced effect of the Akt phosphorylation by the treatment of GH is due to the inhibition of the expression of IGFBP-5, we examined the effect of IGFBP-3 and -5 on the phosphorylation of Akt through IGF-I in the GH-treated BMECs. The phosphorylation of Akt was inhibited in a dose-dependent manner when IGFBP-5 was added at varying concentrations and was also inhibited in the presence of IGFBP-3. The results of this study suggest that GH plays an important role on mammary gland involution in bovine mammary epithelial cells.
Collapse
Affiliation(s)
- Kazuhito Sakamoto
- Laboratory of Animal Physiology, Graduate School of Agriculture, Tohoku University, Sendai 981-8555, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Mammary gland involution is a highly complex multi-step process in which the lactating gland returns to a morphologically near pre-pregnant state. This developmental stage is characterized by a high degree of epithelial cell death, redevelopment of the mammary adipose tissue and tissue remodelling. Many factors involved have been described and these have been reviewed intensively in this journal (Furth, P. A., J. Mammary Gland Biol. Neoplasia, 4:123-127, 1999) and elsewhere. Microarray analysis technology has now not only allowed us to identify genes not previously associated with involution (Stein, T., Morris, J.S., Davis, C.R.,Weber-Hall, S.J., Duffy, M.A., Heath, V.J., et al., Breast Cancer Res., 6: R75-R91, 2004; Clarkson, R.W., Wayland, M.T., Lee, J., Freeman, T., Watson, C.J., Breast Cancer Res., 6: R92-R109, 2004; Clarkson, R.W., Watson, C.J., J. Mammary Gland Biol. Neoplasia, 8: 309-319, 2003), it has also enabled us to define multiple phases of the controlled regulatory response to forced weaning on the basis of their transcriptional profiles. This review provides a synthesis of published data, integrating the time course of transcriptional activity in the mouse mammary gland with a gene ontology approach to identify the pathways involved.
Collapse
Affiliation(s)
- Torsten Stein
- Division of Cancer Sciences and Molecular Pathology, Section of Gene Regulation and Mechanisms of Disease, Western Infirmary, University of Glasgow, Glasgow G11 6NT, UK
| | | | | |
Collapse
|
47
|
Sutherland KD, Lindeman GJ, Visvader JE. The molecular culprits underlying precocious mammary gland involution. J Mammary Gland Biol Neoplasia 2007; 12:15-23. [PMID: 17323120 DOI: 10.1007/s10911-007-9034-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mammary gland involution, characterized by extensive apoptosis and structural remodelling of the gland, is the process by which the gland is returned to the pre-pregnant state. A key advantage of the mammary gland is the ability to synchronize involution through forced weaning, thus allowing the dissection of biochemical pathways involved in the involution process. Over the past few years, significant advances have been made in understanding the signaling pathways and downstream effectors that regulate epithelial cell apoptosis in the first phase of involution, and the importance of matrix metalloproteinases and their inhibitors in both phases of involution. The precise nature of the triggers for apoptosis, however, and the ultimate perpetrators of cell death are not yet clear. This review focuses on genes whose perturbation, either by targeted deletion or overexpression in transgenic mouse models, leads to precocious involution. The accumulating data point to a complex network of signal transduction pathways that synergize to regulate apoptosis in the involuting mammary gland.
Collapse
Affiliation(s)
- Kate D Sutherland
- VBCRC Laboratory, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3050, Australia
| | | | | |
Collapse
|
48
|
Insulin-like growth factors and breast cancer therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 608:101-12. [PMID: 17993235 DOI: 10.1007/978-0-387-74039-3_7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite improvements in breast cancer therapy in recent years, additional therapies need to be developed. New therapies may have activity by themselves or may have utility in combination with other agents. Population, preclinical, and basic data suggest the insulin-like growth factor (IGF) system functions to maintain the malignant phenotype in breast cancer. Since the IGFs act via transmembrane tyrosine kinase receptors, targeting of the key receptors could provide a new pathway in breast cancer. In addition, IGF action enhances cell survival, so combination of anti-IGF therapy with conventional cytotoxic drugs could lead to synergistic effects. In this review, we will discuss the rationale for targeting the IGF system, potential methods to disrupt IGF signaling, and identify potential interactions between IGF inhibitors and other anti-tumor strategies. We will also identify important issues to consider when designing clinical trials.
Collapse
|
49
|
Sun Y, Boyd K, Xu W, Ma J, Jackson CW, Fu A, Shillingford JM, Robinson GW, Hennighausen L, Hitzler JK, Ma Z, Morris SW. Acute myeloid leukemia-associated Mkl1 (Mrtf-a) is a key regulator of mammary gland function. Mol Cell Biol 2006; 26:5809-26. [PMID: 16847333 PMCID: PMC1592762 DOI: 10.1128/mcb.00024-06] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Transcription of immediate-early genes--as well as multiple genes affecting muscle function, cytoskeletal integrity, apoptosis control, and wound healing/angiogenesis--is regulated by serum response factor (Srf). Extracellular signals regulate Srf in part via a pathway involving megakaryoblastic leukemia 1 (Mkl1, also known as myocardin-related transcription factor A [Mrtf-a]), which coactivates Srf-responsive genes downstream of Rho GTPases. Here we investigate Mkl1 function using gene targeting and show the protein to be essential for the physiologic preparation of the mammary gland during pregnancy and the maintenance of lactation. Lack of Mkl1 causes premature involution and impairs expression of Srf-dependent genes in the mammary myoepithelial cells, which control milk ejection following oxytocin-induced contraction. Despite the importance of Srf in multiple transcriptional pathways and widespread Mkl1 expression, the spectrum of abnormalities associated with Mkl1 absence appears surprisingly restricted.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis
- Child
- Failure to Thrive
- Female
- Gene Expression Profiling
- Gene Expression Regulation
- Gene Targeting
- Heart/anatomy & histology
- Heart/embryology
- Humans
- Infant
- Lactation/physiology
- Leukemia, Megakaryoblastic, Acute
- Male
- Mammary Glands, Animal/abnormalities
- Mammary Glands, Animal/anatomy & histology
- Mammary Glands, Animal/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Milk
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/ultrastructure
- Oligonucleotide Array Sequence Analysis
- Oxytocin/metabolism
- Pregnancy
- Prolactin/metabolism
- STAT3 Transcription Factor
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Yi Sun
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lochrie JD, Phillips K, Tonner E, Flint DJ, Allan GJ, Price NC, Beattie J. Insulin-like growth factor binding protein (IGFBP)-5 is upregulated during both differentiation and apoptosis in primary cultures of mouse mammary epithelial cells. J Cell Physiol 2006; 207:471-9. [PMID: 16419030 DOI: 10.1002/jcp.20587] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We have previously demonstrated that insulin-like growth factor binding protein-5 (IGFBP-5) is upregulated following treatment of the mouse mammary epithelial cell line HC11 with lactogenic hormones (dexamethasone, insulin, and prolactin-DIP). In addition, we have also shown that IGFBP-5 is upregulated in mammary epithelial cells in vivo during involution of the rodent mammary gland. We have, therefore, postulated that there may be a dual regulation of IGFBP-5 expression during the temporally separated processes of differentiation and apoptosis of mammary epithelial cells. To test this hypothesis further, we have used a phenotypically differentiated model, which comprises primary cultures of mouse mammary epithelial cells grown on a layer of EHS (Engelbreth-Holm-Swarm) extracellular matrix. We show that lactogenic hormone treatment (hydrocortisone, insulin, and prolactin-HIP) of these cultures induces the upregulation of IGFBP-5 thus replicating the results obtained with the HC11 cell line. In addition, following the induction of apoptosis in primary cultures of mammary epithelial cells by treatment with TGFbeta-3, IGFBP-5 expression is also upregulated. In parallel with this upregulation of IGFBP-5, there is also an increase in the levels of cleaved caspase-3, a well-characterized marker of cellular apoptosis. These findings confirm previous in vivo work demonstrating an increase in IGFBP-5 expression during involution of the mouse mammary gland. When HC11 cells are cultured under serum-free conditions (a well-characterized apoptotic insult in cell culture), there is also an increase in cleaved caspase-3 levels. Unexpectedly, in the presence of TGFbeta-3, caspase-3 levels are attenuated. In the presence of DIP, caspase-3 levels are also decreased in HC11 cells. As described previously, TGFbeta-3 inhibits beta-casein synthesis in HC11 cells. In the HC11 cell line (in contrast to primary cultures of mammary epithelial cells), there is no evidence for TGFbeta-3 induction of IGFBP-5 under either serum-free or DIP-supplemented conditions. We believe our data with primary cultures of mammary epithelial cells support the hypothesis of dual regulation of IGFBP-5 expression during both differentiation and apoptosis in the mammary gland and emphasizes the importance of using appropriate cell culture models to investigate such phenomena in this tissue. We discuss the possible implications of our observations in relation to the physiological processes of pregnancy, lactation, and involution in the mammary gland and the associated changes in mammary epithelial cell function.
Collapse
|