1
|
Hand AR, Abramson CXG, Dressler KA. Tlx1 regulates acinar and duct development in mouse salivary glands. J Anat 2024; 244:343-357. [PMID: 37837237 PMCID: PMC10780161 DOI: 10.1111/joa.13964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
Tlx1 encodes a transcription factor expressed in several craniofacial structures of developing mice. The role of Tlx1 in salivary gland development was examined using morphological and immunohistochemical analyses of Tlx1 null mice. Tlx1 is expressed in submandibular and sublingual glands but not parotid glands of neonatal and adult male and female C57Bl/6J (Tlx1+/+ ) mice. TLX1 protein was localized to the nuclei of terminal tubule cells, developing duct cells and mesenchymal cells in neonatal submandibular and sublingual glands, and to nuclei of duct cells and connective tissue cells in adult glands. Occasionally, TLX1 was observed in nuclei of epithelial cells in or adjacent to the acini. Submandibular glands were smaller and sublingual glands were larger in size in mutant mice (Tlx1-/- ) compared to wild-type mice. Differentiation of terminal tubule and proacinar cells of neonatal Tlx1-/- submandibular glands was abnormal; expression of their characteristic products, submandibular gland protein C and parotid secretory protein, respectively, was reduced. At 3 weeks postnatally, terminal tubule cells at the acinar-intercalated duct junction were poorly developed or absent in Tlx1-/- mice. Granular convoluted ducts in adult mutant mice were decreased, and epidermal growth factor and nerve growth factor expression were reduced. Along with normal acinar cell proteins, adult acinar cells of Tlx1-/- mice continued to express neonatal proteins and expressed parotid proteins not normally present in submandibular glands. Sublingual gland mucous acinar and serous demilune cell differentiation were altered. Tlx1 is necessary for proper differentiation of submandibular and sublingual gland acinar cells, and granular convoluted ducts. The mechanism(s) underlying Tlx1 regulation of salivary gland development and differentiation remains unknown.
Collapse
Affiliation(s)
- Arthur R Hand
- Department of Craniofacial Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut, USA
| | - Cailyn X G Abramson
- Department of Craniofacial Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut, USA
| | - Keith A Dressler
- Department of Craniofacial Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut, USA
| |
Collapse
|
2
|
The Mammary Gland: Basic Structure and Molecular Signaling during Development. Int J Mol Sci 2022; 23:ijms23073883. [PMID: 35409243 PMCID: PMC8998991 DOI: 10.3390/ijms23073883] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 01/27/2023] Open
Abstract
The mammary gland is a compound, branched tubuloalveolar structure and a major characteristic of mammals. The mammary gland has evolved from epidermal apocrine glands, the skin glands as an accessory reproductive organ to support postnatal survival of offspring by producing milk as a source of nutrition. The mammary gland development begins during embryogenesis as a rudimentary structure that grows into an elementary branched ductal tree and is embedded in one end of a larger mammary fat pad at birth. At the onset of ovarian function at puberty, the rudimentary ductal system undergoes dramatic morphogenetic change with ductal elongation and branching. During pregnancy, the alveolar differentiation and tertiary branching are completed, and during lactation, the mature milk-producing glands eventually develop. The early stages of mammary development are hormonal independent, whereas during puberty and pregnancy, mammary gland development is hormonal dependent. We highlight the current understanding of molecular regulators involved during different stages of mammary gland development.
Collapse
|
3
|
Valdebenito-Maturana B, Torres F, Carrasco M, Tapia JC. Differential regulation of transposable elements (TEs) during the murine submandibular gland development. Mob DNA 2021; 12:23. [PMID: 34686213 PMCID: PMC8540199 DOI: 10.1186/s13100-021-00251-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 10/05/2021] [Indexed: 11/29/2022] Open
Abstract
The submandibular gland (SG) is a relatively simple organ formed by three cell types: acinar, myoepithelial, and an intricate network of duct-forming epithelial cells, that together fulfills several physiological functions from assisting food digestion to acting as an immune barrier against pathogens. Successful SG organogenesis is the product of highly controlled and orchestrated genetic and transcriptional programs. Mounting evidence links Transposable Elements (TEs), originally thought to be selfish genetic elements, to different aspects of gene regulation in mammalian development and disease. To our knowledge, the role of TEs during murine SG organogenesis has not been studied. Using novel bioinformatic tools and publicly available RNA-Seq datasets, our results indicate that a significant number of genic and intergenic TEs are differentially expressed during the SG development. Furthermore, changes in expression of specific TEs correlated with that of genes involved in cellular division and differentiation, critical aspects for SG maturation. Altogether, we propose that TEs modulate gene networks that operate during SG development.
Collapse
Affiliation(s)
| | - Francisca Torres
- Stem Cells and Neuroscience Center, School of Medicine, University of Talca, Campus Talca, Talca, Chile
| | - Mónica Carrasco
- Stem Cells and Neuroscience Center, School of Medicine, University of Talca, Campus Talca, Talca, Chile.
| | - Juan Carlos Tapia
- Stem Cells and Neuroscience Center, School of Medicine, University of Talca, Campus Talca, Talca, Chile.
| |
Collapse
|
4
|
Liu S, Xu DS, Li M, Zhang Y, Li Q, Li TT, Ren LQ. Icariin attenuates endothelial-mesenchymal transition via H19/miR-148b-3p/ELF5 in ox-LDL-stimulated HUVECs. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 23:464-475. [PMID: 33510936 PMCID: PMC7809175 DOI: 10.1016/j.omtn.2020.11.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/27/2020] [Indexed: 01/04/2023]
Abstract
Atherosclerosis is the main cause of cardio-cerebrovascular diseases. Endothelial-mesenchymal transition plays an important role in atherosclerosis. Icariin has a protective effect on atherosclerosis; however, the underlying mechanism remains unclear. In this study, we explored the molecular mechanism underlying the protective function of icariin in oxidized low-density lipoprotein-stimulated human umbilical vein endothelial cells. H19, a long non-coding RNA, was identified to be downregulated in the background of the oxidized low-density lipoprotein-induced endothelial-mesenchymal transition in human umbilical vein endothelial cells. Icariin upregulated H19 expression and inhibited the transformation of endothelial cells into interstitial cells. Overexpression of H19 affected endothelial-mesenchymal transition in oxidized low-density lipoprotein-stimulated human umbilical vein endothelial cells, whereas H19 knockdown reversed endothelial protective effects of icariin and reduced human umbilical vein endothelial cell migration. Knockdown of H19 significantly downregulated oxidized low-density lipoprotein-induced E74-like factor 5 and upregulated miR-148b-3p, which was reversed by icariin. Thus, icariin may play a protective role in atherosclerosis, and H19 may be a potential therapeutic target.
Collapse
Affiliation(s)
- Shan Liu
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun 130021, Jilin Province, China
| | - Dong-Sheng Xu
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Min Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun 130021, Jilin Province, China
| | - Yang Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun 130021, Jilin Province, China
| | - Qi Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun 130021, Jilin Province, China.,The Third Hospital Affiliated of Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Teng-Teng Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun 130021, Jilin Province, China
| | - Li-Qun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
5
|
Li H, Jones KL, Hooper JE, Williams T. The molecular anatomy of mammalian upper lip and primary palate fusion at single cell resolution. Development 2019; 146:dev.174888. [PMID: 31118233 DOI: 10.1242/dev.174888] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Abstract
The mammalian lip and primary palate form when coordinated growth and morphogenesis bring the nasal and maxillary processes into contact, and the epithelia co-mingle, remodel and clear from the fusion site to allow mesenchyme continuity. Although several genes required for fusion have been identified, an integrated molecular and cellular description of the overall process is lacking. Here, we employ single cell RNA sequencing of the developing mouse face to identify ectodermal, mesenchymal and endothelial populations associated with patterning and fusion of the facial prominences. This analysis indicates that key cell populations at the fusion site exist within the periderm, basal epithelial cells and adjacent mesenchyme. We describe the expression profiles that make each population unique, and the signals that potentially integrate their behaviour. Overall, these data provide a comprehensive high-resolution description of the various cell populations participating in fusion of the lip and primary palate, as well as formation of the nasolacrimal groove, and they furnish a powerful resource for those investigating the molecular genetics of facial development and facial clefting that can be mined for crucial mechanistic information concerning this prevalent human birth defect.
Collapse
Affiliation(s)
- Hong Li
- Department of Craniofacial Biology, University of Colorado School of Dental Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Kenneth L Jones
- Department of Pediatrics, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Joan E Hooper
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado School of Dental Medicine, 12801 E 17th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
6
|
Singh S, Elenio E, Leu NA, Romano RA, Vaughan AE, DeRiso J, Surendran K, Chakrabarti R. A new Elf5 Cre ERT 2- GFP BAC transgenic mouse model for tracing Elf5 cell lineages in adult tissues. FEBS Lett 2019; 593:1030-1039. [PMID: 31002388 DOI: 10.1002/1873-3468.13390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/03/2019] [Accepted: 04/08/2019] [Indexed: 12/11/2022]
Abstract
Elf5 is a transcription factor known to regulate critical developmental processes and has been shown to act as a tumour suppressor in multiple cancers. Elf5 knockout mice are embryonically lethal, limiting in vivo studies pertaining to its function. Moreover, haploinsufficiency of Elf5 limits the use of current mouse models to investigate adult tissue distribution of Elf5. Here, we successfully generated Elf5Cre ERT 2- GFP bacterial artificial chromosome (BAC) transgenic mice and show that Elf5+ cells are present in several adult tissues, where its expression was previously not known. Our study demonstrates the unique distribution of Elf5+ cells in multiple adult organs, which will facilitate future studies investigating the function of Elf5 in these tissues during homeostasis, repair and cancer.
Collapse
Affiliation(s)
- Snahlata Singh
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emily Elenio
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolae A Leu
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rose-Anne Romano
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, NY, USA
| | - Andrew E Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer DeRiso
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | | | - Rumela Chakrabarti
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Song EAC, Min S, Oyelakin A, Smalley K, Bard JE, Liao L, Xu J, Romano RA. Genetic and scRNA-seq Analysis Reveals Distinct Cell Populations that Contribute to Salivary Gland Development and Maintenance. Sci Rep 2018; 8:14043. [PMID: 30232460 PMCID: PMC6145895 DOI: 10.1038/s41598-018-32343-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/06/2018] [Indexed: 02/06/2023] Open
Abstract
Stem and progenitor cells of the submandibular salivary gland (SMG) give rise to, maintain, and regenerate the multiple lineages of mature epithelial cells including those belonging to the ductal, acinar, basal and myoepithelial subtypes. Here we have exploited single cell RNA-sequencing and in vivo genetic lineage tracing technologies to generate a detailed map of the cell fate trajectories and branch points of the basal and myoepithelial cell populations of the mouse SMG during embryonic development and in adults. Our studies show that the transcription factor p63 and alpha-smooth muscle actin (SMA) serve as faithful markers of the basal and myoepithelial cell lineages, respectively and that both cell types are endowed with progenitor cell properties. However, p63+ basal and SMA+ myoepithelial cells exhibit distinct cell fates by virtue of maintaining different cellular lineages during morphogenesis and in adults. Collectively, our results reveal the dynamic and complex nature of the diverse SMG cell populations and highlight the distinct differentiation potential of the p63 and SMA expressing subtypes in the stem and progenitor cell hierarchy. Long term these findings have profound implications towards a better understanding of the molecular mechanisms that dictate lineage commitment and differentiation programs during development and adult gland maintenance.
Collapse
Affiliation(s)
- Eun-Ah Christine Song
- 0000 0004 1936 9887grid.273335.3Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York 14214 USA
| | - Sangwon Min
- 0000 0004 1936 9887grid.273335.3Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York 14214 USA
| | - Akinsola Oyelakin
- 0000 0004 1936 9887grid.273335.3Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York 14214 USA
| | - Kirsten Smalley
- 0000 0004 1936 9887grid.273335.3Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203 USA
| | - Jonathan E. Bard
- 0000 0004 1936 9887grid.273335.3Genomics and Bioinformatics Core, State University of New York at Buffalo, Buffalo, New York 14222 USA
| | - Lan Liao
- 0000 0001 2160 926Xgrid.39382.33Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030 USA
| | - Jianming Xu
- 0000 0001 2160 926Xgrid.39382.33Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030 USA
| | - Rose-Anne Romano
- 0000 0004 1936 9887grid.273335.3Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York 14214 USA ,0000 0004 1936 9887grid.273335.3Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203 USA
| |
Collapse
|
8
|
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
9
|
Yan H, Qiu L, Xie X, Yang H, Liu Y, Lin X, Huang H. ELF5 in epithelial ovarian carcinoma tissues and biological behavior in ovarian carcinoma cells. Oncol Rep 2017; 37:1412-1418. [PMID: 28184931 PMCID: PMC5364829 DOI: 10.3892/or.2017.5418] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 01/23/2017] [Indexed: 01/09/2023] Open
Abstract
The expression of E74-like factor 5 (ELF5) in epithelial ovarian carcinoma tissues and its effects on biological behavior in ovarian carcinoma cells were assessed in search for a new approach for gene treatment of epithelial ovarian carcinoma. RT-PCR technology was applied to detect the expression of ELF5 mRNA in epithelial ovarian carcinoma (n=49), borderline ovarian epithelial tumor (n=19), benign ovarian epithelial tumor (n=31) and normal ovarian tissues (n=40). Then, we transfected recombinant plasmid pcDNA3.1‑ELF5+EGFP into human ovarian carcinoma SKOV3 cells (recombinant plasmid group) in vitro and screened out stably transfected cells to conduct multiplication culture. Western blot analysis was performed to detect the expression of ELF5 protein in the different groups. Flow cytometry was employed to detect cell apoptosis and cycles. ELF5 mRNA in epithelial ovarian carcinoma and borderline ovarian epithelial tumor tissues were significantly lower (P<0.05) than those in benign ovarian epithelial tumor and normal ovarian tissues. ELF5 protein expression in the cells of recombinant plasmid group was significantly higher compared with empty plasmid and blank control groups. The capacity of cell reproductive recombinant plasmid group at each time point decreased (P<0.05). Flow cytometry detection showed that 67.03% of cells in recombinant plasmid group was blocked in G0/G1 phase (P<0.05), compared with empty plasmid group (37.17%) and blank control group (38.24%). Apoptotic rate of recombinant plasmid group was significantly lower (31.4±1.9%; P<0.05), compared with that of empty plasmid group (9.1±2.2%) and blank control group (8.7±1.5%), and the differences were statistically significant. In conclusion, ELF5 interfered with cell cycle of human ovarian carcinoma SKOV3 cells and promoted apoptosis of human ovarian carcinoma SKOV3 cells inhibiting their growth and invasive capacity; and thus providing a new approach to gene treatment of ovarian carcinoma.
Collapse
Affiliation(s)
- Hongchao Yan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Linglin Qiu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiaolei Xie
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - He Yang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Yongli Liu
- Department of Obstetrics and Gynecology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221000, P.R. China
| | - Xiaoman Lin
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Hongxiang Huang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
10
|
Sano R, Nakajima T, Takahashi Y, Kubo R, Kobayashi M, Takahashi K, Takeshita H, Ogasawara K, Kominato Y. Epithelial Expression of Human ABO Blood Group Genes Is Dependent upon a Downstream Regulatory Element Functioning through an Epithelial Cell-specific Transcription Factor, Elf5. J Biol Chem 2016; 291:22594-22606. [PMID: 27587399 DOI: 10.1074/jbc.m116.730655] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/30/2016] [Indexed: 01/03/2023] Open
Abstract
The human ABO blood group system is of great importance in blood transfusion and organ transplantation. The ABO system is composed of complex carbohydrate structures that are biosynthesized by A- and B-transferases encoded by the ABO gene. However, the mechanisms regulating ABO gene expression in epithelial cells remain obscure. On the basis of DNase I-hypersensitive sites in and around ABO in epithelial cells, we prepared reporter plasmid constructs including these sites. Subsequent luciferase assays and histone modifications indicated a novel positive regulatory element, designated the +22.6-kb site, downstream from ABO, and this was shown to enhance ABO promoter activity in an epithelial cell-specific manner. Expression of ABO and B-antigen was reduced in gastric cancer KATOIII cells by biallelic deletion of the +22.6-kb site using the CRISPR/Cas9 system. Electrophoretic mobility shift assay and chromatin immunoprecipitation assay demonstrated that the site bound to an epithelial cell-specific transcription factor, Elf5. Mutation of the Ets binding motifs to abrogate binding of this factor reduced the regulatory activity of the +22.6-kb site. Furthermore, ELF5 knockdown with shRNA reduced both endogenous transcription from ABO and B-antigen expression in KATOIII cells. Thus, Elf5 appeared to be involved in the enhancer potential of the +22.6-kb site. These results support the contention that ABO expression is dependent upon a downstream positive regulatory element functioning through a tissue-restricted transcription factor, Elf5, in epithelial cells.
Collapse
Affiliation(s)
- Rie Sano
- From the Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, 371-8511 Japan,
| | - Tamiko Nakajima
- From the Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, 371-8511 Japan
| | - Yoichiro Takahashi
- From the Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, 371-8511 Japan
| | - Rieko Kubo
- From the Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, 371-8511 Japan
| | - Momoko Kobayashi
- From the Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, 371-8511 Japan
| | - Keiko Takahashi
- From the Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, 371-8511 Japan
| | - Haruo Takeshita
- the Department of Legal Medicine, Shimane University School of Medicine, Izumo, Japan, and
| | | | - Yoshihiko Kominato
- From the Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, 371-8511 Japan
| |
Collapse
|
11
|
Piggin CL, Roden DL, Gallego-Ortega D, Lee HJ, Oakes SR, Ormandy CJ. ELF5 isoform expression is tissue-specific and significantly altered in cancer. Breast Cancer Res 2016; 18:4. [PMID: 26738740 PMCID: PMC4704400 DOI: 10.1186/s13058-015-0666-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/16/2015] [Indexed: 12/14/2022] Open
Abstract
Background E74-like factor 5 (ELF5) is an epithelial-specific member of the E26 transforming sequence (ETS) transcription factor family and a critical regulator of cell fate in the placenta, pulmonary bronchi, and milk-producing alveoli of the mammary gland. ELF5 also plays key roles in malignancy, particularly in basal-like and endocrine-resistant forms of breast cancer. Almost all genes undergo alternative transcription or splicing, which increases the diversity of protein structure and function. Although ELF5 has multiple isoforms, this has not been considered in previous studies of ELF5 function. Methods RNA-sequencing data for 6757 samples from The Cancer Genome Atlas were analyzed to characterize ELF5 isoform expression in multiple normal tissues and cancers. Extensive in vitro analysis of ELF5 isoforms, including a 116-gene quantitative polymerase chain reaction panel, was performed in breast cancer cell lines. Results ELF5 isoform expression was found to be tissue-specific due to alternative promoter use but altered in multiple cancer types. The normal breast expressed one main isoform, while in breast cancer there were subtype-specific alterations in expression. Expression of other ETS factors was also significantly altered in breast cancer, with the basal-like subtype demonstrating a distinct ETS expression profile. In vitro inducible expression of the full-length isoforms 1 and 2, as well as isoform 3 (lacking the Pointed domain) had similar phenotypic and transcriptional effects. Conclusions Alternative promoter use, conferring differential regulatory responses, is the main mechanism governing ELF5 action rather than differential transcriptional activity of the isoforms. This understanding of expression and function at the isoform level is a vital first step in realizing the potential of transcription factors such as ELF5 as prognostic markers or therapeutic targets in cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13058-015-0666-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Catherine L Piggin
- Cancer Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia.
| | - Daniel L Roden
- Cancer Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia.
| | - David Gallego-Ortega
- Cancer Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia.
| | - Heather J Lee
- Cancer Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia. .,Babraham Institute, Cambridge, CB22 3AT, UK.
| | - Samantha R Oakes
- Cancer Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia.
| | - Christopher J Ormandy
- Cancer Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia.
| |
Collapse
|
12
|
Prokesch A, Smorlesi A, Perugini J, Manieri M, Ciarmela P, Mondini E, Trajanoski Z, Kristiansen K, Giordano A, Bogner-Strauss JG, Cinti S. Molecular aspects of adipoepithelial transdifferentiation in mouse mammary gland. Stem Cells 2015; 32:2756-66. [PMID: 24898182 DOI: 10.1002/stem.1756] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/28/2014] [Accepted: 04/20/2014] [Indexed: 01/09/2023]
Abstract
The circular, reversible conversion of the mammary gland during pregnancy and involution is a paradigm of physiological tissue plasticity. The two most prominent cell types in mammary gland, adipocytes and epithelial cells, interact in an orchestrated way to coordinate this process. Previously, we showed that this conversion is at least partly achieved by reciprocal transdifferentiation between mammary adipocytes and lobulo-alveolar epithelial cells. Here, we aim to shed more light on the regulators of mammary transdifferentiation. Using immunohistochemistry with cell type-specific lipid droplet-coating markers (Perilipin1 and 2), we show that cells with an intermediate adipoepithelial phenotype exist during and after pregnancy. Nuclei of cells with similar transitional structural characteristics are highly positive for Elf5, a master regulator of alveologenesis. In cultured adipocytes, we could show that transient and stable ectopic expression of Elf5 induces expression of the milk component whey acidic protein, although the general adipocyte phenotype is not affected suggesting that additional pioneering factors are necessary. Furthermore, the lack of transdifferentiation of adipocytes during pregnancy after clearing of the epithelial compartment indicates that transdifferentiation signals must emanate from the epithelial part. To explore candidate genes potentially involved in the transdifferentiation process, we devised a high-throughput gene expression study to compare cleared mammary fat pads with developing, contralateral controls at several time points during pregnancy. Incorporation of bioinformatic predictions of secretory proteins provides new insights into possible paracrine signaling pathways and downstream transdifferentiation factors. We discuss a potential role for osteopontin (secreted phosphoprotein 1 [Spp1]) signaling through integrins to induce adipoepithelial transdifferentiation.
Collapse
Affiliation(s)
- A Prokesch
- Institute for Genomics and Bioinformatics, Graz University of Technology, Petersgasse, Graz, Austria; Institute of Biochemistry, Graz University of Technology, Petersgasse, Graz, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Koyama K, Takahara K, Inamoto T, Ibuki N, Minami K, Uehara H, Komura K, Nishida T, Sakamoto T, Hirano H, Nomi H, Kiyama S, Azuma H. E74-like factor inhibition induces reacquisition of hormone sensitiveness decreasing period circadian protein homolog 1 expression in prostate cancer cells. Prostate Int 2015; 3:16-21. [PMID: 26288799 PMCID: PMC4495571 DOI: 10.1016/j.prnil.2015.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 12/27/2014] [Indexed: 01/09/2023] Open
Abstract
Purpose Initiating as an androgen-dependent adenocarcinoma, prostate cancer (PCa) gradually progresses to a castrate-resistant disease following androgen deprivation therapy with a propensity to metastasize. Methods In order to resolve the mechanism of castrate-resistant PCa, we performed a cDNA-microarray assay of two PCa cell lines, LNCaP (androgen dependent) and C4-2 (androgen independent). Among them, we focused on a novel Ets transcription factor, E74-like factor 5 (ELF5), the expression level of which was extremely high in C4-2 in comparison with LNCaP both in the microarray analysis and real-time polymerase chain reaction analysis, and investigated the biological role in acquisition of androgen-refractory PCa growth. Results Western blot analysis and morphological analysis using confocal immunofluorescence microscopy demonstrated that ELF5 was expressed mainly in cytosol both in LNCaP and C4-2. Inhibition of ELF5 expression using ELF5-small interfering RNA in C4-2 induced decreased expression of androgen receptor corepressor, period circadian protein homolog 1, and MTT assay of C4-2 after ELF5 small interfering RNA transfection showed the same cell growth pattern of LNCaP. Conclusions Our in vitro experiments of cell growth and microarray analysis have demonstrated for the first time that decreased expression of period circadian protein homolog 1 due to ELF5 inhibition may induce the possibility of reacquisition of hormone sensitiveness of PCa cells. We suggest that ELF5 could be a novel potential target for the treatment of hormone-refractory PCa patients.
Collapse
Affiliation(s)
- Kohei Koyama
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Kiyoshi Takahara
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Teruo Inamoto
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Naokazu Ibuki
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Koichiro Minami
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Hirofumi Uehara
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Kazumasa Komura
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Takeshi Nishida
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Takeshi Sakamoto
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Hajime Hirano
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Hayahito Nomi
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Satoshi Kiyama
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Haruhito Azuma
- Department of Urology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| |
Collapse
|
14
|
Browne JA, Yang R, Song L, Crawford GE, Leir SH, Harris A. Open chromatin mapping identifies transcriptional networks regulating human epididymis epithelial function. Mol Hum Reprod 2014; 20:1198-207. [PMID: 25180270 DOI: 10.1093/molehr/gau075] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The epithelium lining the epididymis in the male reproductive tract maintains a luminal environment that promotes sperm cell maturation. This process is dependent on the coordinated expression of many genes that encode proteins with a role in epithelial transport. We previously generated genome-wide maps of open chromatin in primary human epididymis epithelial (HEE) cells to identify potential regulatory elements controlling coordinated gene expression in the epididymis epithelium. Subsequent in silico analysis identified transcription factor-binding sites (TFBS) that were over-represented in the HEE open chromatin, including the motif for paired box 2 (PAX2). PAX2 is a critical transcriptional regulator of urogenital tract development, which has been well studied in the kidney but is unexplored in the epididymis. Due to the limited lifespan of primary HEE cells in culture, we investigated the role of PAX2 in an immortalized HEE cell line (REP). First, REP cells were evaluated by DNase I digestion followed by high-throughput sequencing and the PAX2-binding motif was again identified as an over-represented TFBS within intergenic open chromatin, though on fewer chromosomes than in the primary HEE cells. To identify PAX2-target genes in REP cells, RNA-seq analysis was performed after siRNA-mediated depletion of PAX2 and compared with that with a non-targeting siRNA. In response to PAX2-repression, 3135 transcripts were differentially expressed (1333 up-regulated and 1802 down-regulated). Novel PAX2 targets included multiple genes encoding proteins with predicted functions in the epididymis epithelium.
Collapse
Affiliation(s)
- James A Browne
- Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, IL, USA Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rui Yang
- Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, IL, USA Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lingyun Song
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical School, Durham, NC, USA Center for Genomic and Computational Biology, Duke University Medical School, Durham, NC, USA
| | - Gregory E Crawford
- Department of Pediatrics, Division of Medical Genetics, Duke University Medical School, Durham, NC, USA Center for Genomic and Computational Biology, Duke University Medical School, Durham, NC, USA
| | - Shih-Hsing Leir
- Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, IL, USA Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ann Harris
- Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, IL, USA Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
15
|
Takayama M, Miyatake K, Nishida E. Identification and characterization of retinoic acid-responsive genes in mouse kidney development. Genes Cells 2014; 19:637-49. [PMID: 24962468 DOI: 10.1111/gtc.12163] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 05/20/2014] [Indexed: 12/12/2022]
Abstract
Retinoic acid (RA) signaling regulates a variety of developmental processes through controlling the expression of numerous genes. Here, we have identified and characterized RA-responsive genes in mouse kidney development. Analysis of isolated embryonic kidneys cultured in the presence and absence of RA identified 33 candidates of RA-responsive genes. Most of these candidate genes were down-regulated by treatment with the RA receptor antagonist. Many of them have potential binding sites for Elf5, one of the RA-responsive genes, in their promoter region. Whole-mount in situ hybridization showed that specific expression of Elf5 in the ureteric trunk depends on RA. RA-dependent expression in the ureteric trunk was also showed for the sodium channel subunit Scnn1b, which has been shown to be the marker gene of the collecting duct. In contrast, the expression of Ecm1, Tnfsf13b and IL-33 was detected in the stromal mesenchymal cells. Both Tnfsf13b and IL-33 were previously shown to cause nuclear factor κB (NF-κB) activation. We have showed that the inhibition of NF-κB signaling with specific inhibitors suppresses branching morphogenesis of the ureteric bud. Our study thus identifies and characterizes RA-dependent up-regulated genes in kidney development, and suggests an involvement of NF-κB signaling in the branching morphogenesis.
Collapse
Affiliation(s)
- Mami Takayama
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, 606-8502, Japan
| | | | | |
Collapse
|
16
|
Gallego-Ortega D, Oakes SR, Lee HJ, Piggin CL, Ormandy CJ. ELF5, normal mammary development and the heterogeneous phenotypes of breast cancer. BREAST CANCER MANAGEMENT 2013. [DOI: 10.2217/bmt.13.50] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
SUMMARY The ETS transcription factor ELF5 specifies the formation of the secretory cell lineage of the mammary gland during pregnancy, by directing cell fate decisions of the mammary progenitor cells. The decision-making activity continues in breast cancer, where in luminal breast cancer cells forced ELF5 expression suppresses estrogen sensitivity and shifts gene expression toward the basal molecular subtype. The development of anti-estrogen resistance in luminal breast cancer is accompanied by increased expression of ELF5 and acquired dependence on ELF5 for continued proliferation, providing a potential new therapeutic target or prognostic marker to improve the treatment of this stage of the disease. Forced ELF5 expression suppresses the mesenchymal phenotype, making cells more epithelial and producing lower rates of invasion and motility. Conversely, loss of ELF5 promotes metastasis, with a clear corollary in the claudin-low subtype of breast cancer, which does not express ELF5 and is highly metastatic, or during the final stages of tumor progression, where loss of ELF5 expression may be involved in the acquisition of the lethal phenotype. In circumstances where ELF5 expression increases in parallel with metastatic potential, such as anti-estrogen resistant luminal breast cancers and basal breast cancer, there is much more to be understood about ELF5 and metastasis.
Collapse
Affiliation(s)
- David Gallego-Ortega
- Cancer Research Program, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, 384 Victoria Street, Darlinghurst, NSW 2010, Australia
- St Vincent‘s Clinical School, St Vincent‘s Hospital Faculty of Medicine, University of New South Wales, NSW, Australia
| | - Samantha R Oakes
- Cancer Research Program, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, 384 Victoria Street, Darlinghurst, NSW 2010, Australia
- St Vincent‘s Clinical School, St Vincent‘s Hospital Faculty of Medicine, University of New South Wales, NSW, Australia
| | - Heather J Lee
- Cancer Research Program, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, 384 Victoria Street, Darlinghurst, NSW 2010, Australia
- St Vincent‘s Clinical School, St Vincent‘s Hospital Faculty of Medicine, University of New South Wales, NSW, Australia
| | - Catherine L Piggin
- Cancer Research Program, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, 384 Victoria Street, Darlinghurst, NSW 2010, Australia
- St Vincent‘s Clinical School, St Vincent‘s Hospital Faculty of Medicine, University of New South Wales, NSW, Australia
| | - Christopher J Ormandy
- Cancer Research Program, Garvan Institute of Medical Research & The Kinghorn Cancer Centre, 384 Victoria Street, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
17
|
Bischof JM, Gillen AE, Song L, Gosalia N, London D, Furey TS, Crawford GE, Harris A. A genome-wide analysis of open chromatin in human epididymis epithelial cells reveals candidate regulatory elements for genes coordinating epididymal function. Biol Reprod 2013; 89:104. [PMID: 24006278 DOI: 10.1095/biolreprod.113.110403] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The epithelium lining the epididymis has a pivotal role in ensuring a luminal environment that can support normal sperm maturation. Many of the individual genes that encode proteins involved in establishing the epididymal luminal fluid are well characterized. They include ion channels, ion exchangers, transporters, and solute carriers. However, the molecular mechanisms that coordinate expression of these genes and modulate their activities in response to biological stimuli are less well understood. To identify cis-regulatory elements for genes expressed in human epididymis epithelial cells, we generated genome-wide maps of open chromatin by DNase-seq. This analysis identified 33,542 epididymis-selective DNase I hypersensitive sites (DHS), which were not evident in five cell types of different lineages. Identification of genes with epididymis-selective DHS at their promoters revealed gene pathways that are active in immature epididymis epithelial cells. These include processes correlating with epithelial function and also others with specific roles in the epididymis, including retinol metabolism and ascorbate and aldarate metabolism. Peaks of epididymis-selective chromatin were seen in the androgen receptor gene and the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which has a critical role in regulating ion transport across the epididymis epithelium. In silico prediction of transcription factor binding sites that were overrepresented in epididymis-selective DHS identified epithelial transcription factors, including ELF5 and ELF3, the androgen receptor, Pax2, and Sox9, as components of epididymis transcriptional networks. Active genes, which are targets of each transcription factor, reveal important biological processes in the epididymis epithelium.
Collapse
Affiliation(s)
- Jared M Bischof
- Human Molecular Genetics Program, Lurie Children's Research Center, and Department of Pediatrics, Northwestern University Feinberg School of Medicine Chicago, Illinois
| | | | | | | | | | | | | | | |
Collapse
|
18
|
ELF5 suppresses estrogen sensitivity and underpins the acquisition of antiestrogen resistance in luminal breast cancer. PLoS Biol 2012; 10:e1001461. [PMID: 23300383 PMCID: PMC3531499 DOI: 10.1371/journal.pbio.1001461] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 11/14/2012] [Indexed: 12/22/2022] Open
Abstract
The transcription factor ELF5 is responsible for gene expression patterning underlying molecular subtypes of breast cancer and may mediate acquired resistance to anti-estrogen therapy. We have previously shown that during pregnancy the E-twenty-six (ETS) transcription factor ELF5 directs the differentiation of mammary progenitor cells toward the estrogen receptor (ER)-negative and milk producing cell lineage, raising the possibility that ELF5 may suppress the estrogen sensitivity of breast cancers. To test this we constructed inducible models of ELF5 expression in ER positive luminal breast cancer cells and interrogated them using transcript profiling and chromatin immunoprecipitation of DNA followed by DNA sequencing (ChIP-Seq). ELF5 suppressed ER and FOXA1 expression and broadly suppressed ER-driven patterns of gene expression including sets of genes distinguishing the luminal molecular subtype. Direct transcriptional targets of ELF5, which included FOXA1, EGFR, and MYC, accurately classified a large cohort of breast cancers into their intrinsic molecular subtypes, predicted ER status with high precision, and defined groups with differential prognosis. Knockdown of ELF5 in basal breast cancer cell lines suppressed basal patterns of gene expression and produced a shift in molecular subtype toward the claudin-low and normal-like groups. Luminal breast cancer cells that acquired resistance to the antiestrogen Tamoxifen showed greatly elevated levels of ELF5 and its transcriptional signature, and became dependent on ELF5 for proliferation, compared to the parental cells. Thus ELF5 provides a key transcriptional determinant of breast cancer molecular subtype by suppression of estrogen sensitivity in luminal breast cancer cells and promotion of basal characteristics in basal breast cancer cells, an action that may be utilised to acquire antiestrogen resistance. The molecular subtypes of breast cancer are distinguished by their intrinsic patterns of gene expression and can be used to group patients with different prognoses and treatment options. Although molecular subtyping tests are currently under evaluation, some of them are already in use to better tailor therapy for patients; however, the molecular events that are responsible for these different patterns of gene expression in breast cancer are largely undefined. The elucidation of their mechanistic basis would improve our understanding of the disease process and enhance the chances of developing better predictive and prognostic markers, new therapies, and interventions to overcome resistance to existing therapies. Here, we show that the transcription factor ELF5 is responsible for much of the patterning of gene expression that distinguishes the breast cancer subtypes. Additionally, our data suggest that ELF5 may also be involved in the development of resistance to therapies designed to stop estrogen stimulation of breast cancer. These effects of ELF5 appear to represent a partial carryover into breast cancer of its normal role in the mammary gland, where it is responsible for the development of milk-producing structures during pregnancy.
Collapse
|
19
|
Chakrabarti R, Wei Y, Romano RA, DeCoste C, Kang Y, Sinha S. Elf5 regulates mammary gland stem/progenitor cell fate by influencing notch signaling. Stem Cells 2012; 30:1496-508. [PMID: 22523003 DOI: 10.1002/stem.1112] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The transcription factor E74-like factor 5 (Elf5) functions downstream of the prolactin receptor signaling pathway and plays an important role in mammary gland development. Using conditional mouse knockouts, we have previously shown that Elf5-null mammary glands exhibit a complete failure of alveologenesis during pregnancy. The Elf5-null developmental phenotype is mediated through alteration in the expression of several critical genes involved in alveologenesis, particularly those belonging to the JAK/STAT pathway. Here, we demonstrate that in addition to regulating terminal differentiation of alveolar cells, Elf5 also plays a critical role in determining cell fate and in regulating the stem/progenitor function of the mammary epithelium. Targeted deletion of Elf5 in the mammary glands leads to accumulation of cell types with dual luminal/basal properties such as coexpression of K8 and K14 and an increase in CD61(+) luminal progenitor population during pregnancy. Further interrogation suggests that the abnormal increase in K14(+) K8(+) cells may represent the CD61(+) luminal progenitors blocked in differentiation. Remarkably, Elf5 deficiency in mammary epithelium also triggers an increase of adult mammary stem activity as evidenced by the accumulation of mammary stem cell (MaSC)-enriched cell population in both pregnant and virgin mice and further confirmed by mammosphere and transplantation assays. Additional support for this phenotype comes from the enriched MaSC gene signature based on transcriptomic analysis of the Elf5-null mammary gland. Finally, our biochemical studies suggest that Elf5 loss leads to hyperactivation of the Notch signaling pathway, which might constitute in part, the underlying molecular mechanism for the altered cell lineage decisions in Elf5-null mammary epithelial cells.
Collapse
Affiliation(s)
- Rumela Chakrabarti
- Department of Molecular Biology, Princeton University, Princeton, NJ 08554, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Lee HJ, Ormandy CJ. Elf5, hormones and cell fate. Trends Endocrinol Metab 2012; 23:292-8. [PMID: 22464677 DOI: 10.1016/j.tem.2012.02.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 02/16/2012] [Accepted: 02/22/2012] [Indexed: 01/05/2023]
Abstract
Recent elucidation of the stem and progenitor cell hierarchies that operate during normal tissue and organ development has provided a foundation for the development of new insights into the disease process. These hierarchies are established by genetic mechanisms, which specify and determine cell fate and act as cell-clade gatekeepers, upon which all multicellular organisms depend for viability. Perturbation of this gatekeeper function characterizes developmentally based diseases, such as cancer. Here, the emerging gatekeeper and master regulatory roles of the ETS transcription factor Elf5 in several diverse developmental scenarios is reviewed, and how this function intersects with hormonal and growth factor mediated regulation of these processes is shown.
Collapse
Affiliation(s)
- Heather J Lee
- Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
| | | |
Collapse
|
21
|
Bischof JM, Ott CJ, Leir SH, Gosalia N, Song L, London D, Furey TS, Cotton CU, Crawford GE, Harris A. A genome-wide analysis of open chromatin in human tracheal epithelial cells reveals novel candidate regulatory elements for lung function. Thorax 2012; 67:385-91. [PMID: 22169360 PMCID: PMC3384740 DOI: 10.1136/thoraxjnl-2011-200880] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Distal cell-type-specific regulatory elements may be located at very large distances from the genes that they control and are often hidden within intergenic regions or in introns of other genes. The development of methods that enable mapping of regions of open chromatin genome wide has greatly advanced the identification and characterisation of these elements. METHODS Here we use DNase I hypersensitivity mapping followed by deep sequencing (DNase-seq) to generate a map of open chromatin in primary human tracheal epithelial (HTE) cells and use bioinformatic approaches to characterise the distribution of these sites within the genome and with respect to gene promoters, intronic and intergenic regions. RESULTS Genes with HTE-selective open chromatin at their promoters were associated with multiple pathways of epithelial function and differentiation. The data predict novel cell-type-specific regulatory elements for genes involved in HTE cell function, such as structural proteins and ion channels, and the transcription factors that may interact with them to control gene expression. Moreover, the map of open chromatin can identify the location of potentially critical regulatory elements in genome-wide association studies (GWAS) in which the strongest association is with single nucleotide polymorphisms in non-coding regions of the genome. We demonstrate its relevance to a recent GWAS that identifies modifiers of cystic fibrosis lung disease severity. CONCLUSION Since HTE cells have many functional similarities with bronchial epithelial cells and other differentiated cells in the respiratory epithelium, these data are of direct relevance to elucidating the molecular basis of normal lung function and lung disease.
Collapse
Affiliation(s)
- Jared M Bischof
- Human Molecular Genetics Program, Children's Memorial Research Center, Chicago, Illinois, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Christopher J Ott
- Human Molecular Genetics Program, Children's Memorial Research Center, Chicago, Illinois, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Shih-Hsing Leir
- Human Molecular Genetics Program, Children's Memorial Research Center, Chicago, Illinois, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nehal Gosalia
- Human Molecular Genetics Program, Children's Memorial Research Center, Chicago, Illinois, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lingyun Song
- Institute for Genome Science and Policy, Duke University, Durham, North Carolina, USA
| | - Darin London
- Institute for Genome Science and Policy, Duke University, Durham, North Carolina, USA
| | - Terrence S Furey
- Department of Genetics, Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biology, Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Calvin U Cotton
- Department of Pediatrics, Case Western University, School of Medicine, Cleveland, Ohio, USA
- Department of Physiology and Biophysics, Case Western University, School of Medicine, Cleveland, Ohio, USA
| | - Gregory E Crawford
- Institute for Genome Science and Policy, Duke University, Durham, North Carolina, USA
| | - Ann Harris
- Human Molecular Genetics Program, Children's Memorial Research Center, Chicago, Illinois, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
22
|
Pearton DJ, Broadhurst R, Donnison M, Pfeffer PL. Elf5 regulation in the Trophectoderm. Dev Biol 2011; 360:343-50. [DOI: 10.1016/j.ydbio.2011.10.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 09/28/2011] [Accepted: 10/04/2011] [Indexed: 10/16/2022]
|
23
|
Lapinskas EJ, Svobodova S, Davis ID, Cebon J, Hertzog PJ, Pritchard MA. The Ets transcription factor ELF5 functions as a tumor suppressor in the kidney. Twin Res Hum Genet 2011; 14:316-22. [PMID: 21787113 DOI: 10.1375/twin.14.4.316] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Renal cell carcinoma is an important clinical disease with poorly understood etiology. ELF5 is an epithelial-specific member of the Ets family of transcription factors, characterized by the 80 amino acid Ets domain that binds the purine-rich GGAA/T Ets motif found in the promoter regions of a variety of genes. Since ELF5 is highly expressed in kidney and has been postulated to function as a tumor suppressor, at least in the context of the breast, we investigated its role in kidney cancer. In renal cell carcinoma ELF5 expression was consistently decreased in tumor samples versus normal. ELF5 mRNA was decreased in 94% of lesions tested and ELF5 protein was undetectable in 40/40 kidney-derived carcinomas. Re-expression of the ELF5 gene in 786-O renal carcinoma cells suppressed their tumorigenic capacity in vitro and in vivo. This work is the first to suggest that ELF5 has tumor suppressor activity in the kidney.
Collapse
Affiliation(s)
- Erika J Lapinskas
- Centre for Functional Genomics and Human Disease, Monash Institute of Medical Research, Monash University, Australia
| | | | | | | | | | | |
Collapse
|
24
|
Sevilla LM, Bayo P, Latorre V, Sanchis A, Pérez P. Glucocorticoid receptor regulates overlapping and differential gene subsets in developing and adult skin. Mol Endocrinol 2010; 24:2166-78. [PMID: 20880987 DOI: 10.1210/me.2010-0183] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We have previously shown that the glucocorticoid receptor (GR) is required for skin homeostasis and epidermal barrier competence. To understand the transcriptional program by which GR regulates skin development, we performed a microarray analysis using the skin of GR(-/-) and GR(+/+) mice of embryonic d 18.5 and identified 442 differentially expressed genes. Functional clustering demonstrated overrepresentation of genes involved in ectoderm/epidermis development. We found strong repression of genes encoding proteins associated with the later stages of epidermal differentiation, such as several small proline-rich proteins (Sprrs) and corneodesmosin (Cdsn). This, together with the up-regulation of genes induced earlier during epidermal development, including the epithelial-specific gene transcripts E74-like factor 5 (Elf5) and keratin 77 (Krt77), fits with the phenotype of defective epidermal differentiation observed in the GR(-/-) mice. We also found down-regulation of the antimicrobial peptide defensin β 1 (Defb1) and FK506-binding protein 51 (Fkbp51). Skin developmental expression profiling of these genes and studies in cultured keratinocytes from GR(-/-) and wild type embryos demonstrated that gene regulation occurred in a cell-autonomous manner. To investigate the consequences of GR loss in adult epidermis, we generated mice with inducible inactivation of GR restricted to keratinocytes (K14-cre-ER(T2)//GR(loxP/loxP) mice). K14-cre-ER(T2)//GR(loxP/loxP) mice featured thickened skin with increased keratinocyte proliferation and impaired differentiation. Whereas Krt77 and Elf5 expression remained unaffected by loss of GR in adult epidermis, Fkbp51, Sprr2d, and Defb1 were strongly repressed. Importantly, we have identified both Fkbp51 and Defb1 as direct transcriptional targets of GR, and we have shown that GR-mediated regulation of these genes occurs in both developing and adult epidermis. We conclude that both overlapping and differential GR targets are regulated in developing vs. adult skin.
Collapse
Affiliation(s)
- Lisa M Sevilla
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Jaime Roig 11, E-46010-Valencia, Spain
| | | | | | | | | |
Collapse
|
25
|
Choi YS, Chakrabarti R, Escamilla-Hernandez R, Sinha S. Elf5 conditional knockout mice reveal its role as a master regulator in mammary alveolar development: failure of Stat5 activation and functional differentiation in the absence of Elf5. Dev Biol 2009; 329:227-41. [PMID: 19269284 DOI: 10.1016/j.ydbio.2009.02.032] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Revised: 02/20/2009] [Accepted: 02/20/2009] [Indexed: 01/21/2023]
Abstract
The transcription factor Elf5 plays an important role in mammary gland development. However, because of the embryonic lethality of Elf5 straight knockout mice, prior studies have been limited to experiments with Elf5 haploinsufficient animals, overexpression systems or transplants. Here, we have utilized K14-Cre to generate mammary-gland specific Elf5 conditional knockout mice. During pregnancy, Elf5-null mammary epithelium completely failed to initiate alveologenesis, and a characteristic of virgin ductal epithelial cells persisted postpartum. We demonstrate that the loss of Elf5 leads to the absence of alveolar secretory markers confirming previous published data. Interestingly, the developmental block due to a lack of Elf5 could not be restored by multiple gestations. Elf5-null mammary epithelial cells also display disorganized cell structures as evident by altered cell polarities, which might be the cause for collapsed lumina. We observe reduced levels of Stat5 and attenuated Stat5 activity as measured by p-Stat5 levels both in Elf5-null mammary glands as well as cultured mammary epithelial cells. This data suggests that the failure of alveolar and lactogenic differentiation due to the loss of Elf5 is mediated in part due to impaired Stat5 activity. In support of this hypothesis, we show by ChIP experiments that Stat5a promoter contains a conserved Elf5-binding site that is occupied by Elf5 in mammary glands. Mammary epithelia lacking Elf5 exhibited downregulation of several other critical genes involved in alveologenesis, suggesting Elf5 as a master regulator in alveolar development. We propose a model for Elf5-mediated alveolar development, in which Elf5 regulates the expression of key mediators of the PrlR/Jak2/Stat5 signaling pathway.
Collapse
Affiliation(s)
- Yeon Sook Choi
- Department of Biochemistry, State University of New York at Buffalo, Center of Excellence in Bioinformatics and Life Sciences, 701 Ellicott Street, Buffalo, NY 14203, USA
| | | | | | | |
Collapse
|
26
|
State-of-the-art technologies, current opinions and developments, and novel findings: news from the field of histochemistry and cell biology. Histochem Cell Biol 2008; 130:1205-51. [PMID: 18985372 DOI: 10.1007/s00418-008-0535-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2008] [Indexed: 10/25/2022]
Abstract
Investigations of cell and tissue structure and function using innovative methods and approaches have again yielded numerous exciting findings in recent months and have added important data to current knowledge, inspiring new ideas and hypotheses in various fields of modern life sciences. Topics and contents of comprehensive expert reviews covering different aspects in methodological advances, cell biology, tissue function and morphology, and novel findings reported in original papers are summarized in the present review.
Collapse
|
27
|
Metzger DE, Stahlman MT, Shannon JM. Misexpression of ELF5 disrupts lung branching and inhibits epithelial differentiation. Dev Biol 2008; 320:149-60. [PMID: 18544451 DOI: 10.1016/j.ydbio.2008.04.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Revised: 04/29/2008] [Accepted: 04/30/2008] [Indexed: 12/30/2022]
Abstract
ELF5, an Ets family transcription factor found exclusively in epithelial cells, is expressed in the distal lung epithelium during embryogenesis, then becomes restricted to proximal airways at the end of gestation and postnatally. To test the hypothesis that ELF5 represses distal epithelial differentiation, we generated a transgenic mouse model in which a doxycycline inducible HA-tagged mouse Elf5 transgene was placed under the control of the lung epithelium-specific human SFTPC promoter. We found that expressing high levels of ELF5 during early lung development disrupted branching morphogenesis and produced a dilated epithelium. The effects of ELF5 on morphogenesis were stage-dependent, since inducing the transgene on E16.5 had no effect on branching. ELF5 reduced expression of the distal lung epithelial differentiation markers Erm, Napsa and Sftpc, and type II cell ultrastructural differentiation was immature. ELF5 overexpression did not induce the proximal airway epithelial markers Ccsp and Foxj1, but did induce expression of p63, a marker of basal cells in the trachea and esophagus. High ELF5 levels also induced the expression of genes found in other endodermal epithelia but not normally associated with the lung. These results suggest that precise levels of ELF5 regulate the specification and differentiation of epithelial cells in the lung.
Collapse
Affiliation(s)
- David E Metzger
- Division of Pulmonary Biology, Cincinnati Children's Hosptial Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | | | | |
Collapse
|
28
|
Choi YS, Cheng J, Segre J, Sinha S. Generation and analysis of Elf5-LacZ mouse: unique and dynamic expression of Elf5 (ESE-2) in the inner root sheath of cycling hair follicles. Histochem Cell Biol 2007; 129:85-94. [DOI: 10.1007/s00418-007-0347-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2007] [Indexed: 01/20/2023]
|
29
|
Metzger DE, Xu Y, Shannon JM. Elf5 is an epithelium-specific, fibroblast growth factor-sensitive transcription factor in the embryonic lung. Dev Dyn 2007; 236:1175-92. [PMID: 17394208 DOI: 10.1002/dvdy.21133] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factor (FGF) signaling has been shown to be essential for many aspects of normal lung development. To determine epithelial targets of FGF signaling, we cultured embryonic day (E) 11.5 mouse lungs for 24 hr in the presence or absence of the FGF receptor antagonist SU5402, which inhibited branching morphogenesis. Affymetrix gene chip analysis of treated and control epithelia identified several genes regulated by FGF signaling, including Elf5, a member of the Epithelial-specific Ets family of transcription factors. SU5402 reduced Elf5 expression in mesenchyme-free cultures of E12.5 epithelium, demonstrating that the inhibition was direct. In situ hybridization revealed that Elf5 had a dynamic pattern of expression during lung development. We found that expression of Elf5 was induced by FGF7 and FGF10, ligands that primarily bind FGFR2b. To further define the pathways by which FGFs activate Elf5 expression, we cultured E11.5 lung tips in the presence of compounds to inhibit FGF receptors (SU5402), PI3-Kinase/Akt-mediated signaling (LY294002), and MAP Kinase/Erk-mediated signaling (U0126). We found that SU5402 and LY294002 significantly reduced Elf5 expression, whereas U0126 had no effect. LY294002 also reduced Elf5 expression in cultures of purified epithelium. Finally, pAkt was coexpressed with Elf5 in the proximal epithelial airways of E17.5 lungs. These results demonstrate that Elf5 is an FGF-sensitive transcription factor in the lung with a dynamic pattern of expression and that FGF regulation of Elf5 by means of FGFR2b occurs through the PI3-Kinase/Akt pathway.
Collapse
Affiliation(s)
- David E Metzger
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229-3039, USA.
| | | | | |
Collapse
|
30
|
Abstract
The mouse mammary gland is a complex tissue that proliferates and differentiates under the control of systemic hormones during puberty, pregnancy and lactation. Once a highly branched milk duct system has been established, during mid/late pregnancy, alveoli, little saccular outpouchings, sprout all over the ductal system and differentiate to become the sites of milk secretion. Here, we review the emerging network of the signaling pathways that connects hormonal stimuli with locally produced signaling molecules and the components of intracellular pathways that regulate alveologenesis and lactation. The powerful tools of mouse genetics have been instrumental in uncovering many of the signaling components involved in controlling alveolar and lactogenic differentiation.
Collapse
Affiliation(s)
- Cathrin Brisken
- National Center of Competence in Research (NCCR) Molecular Oncology, Swiss Institute for Experimental Cancer Research (ISREC), 155 Chemin des Boveresses, CH-1066, Epalinges, Lausanne, Switzerland.
| | | |
Collapse
|
31
|
Choi Y, Sinha S. Determination of the consensus DNA-binding sequence and a transcriptional activation domain for ESE-2. Biochem J 2006; 398:497-507. [PMID: 16704374 PMCID: PMC1559455 DOI: 10.1042/bj20060375] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The ESE (epithelium-specific Ets) subfamily of Ets transcription factors plays an important role in regulating gene expression in a variety of epithelial cell types. Although ESE proteins have been shown to bind to regulatory elements of some epithelial genes, the optimal DNA-binding sequence has not been experimentally ascertained for any member of the ESE subfamily of transcription factors. This has made the identification and validation of their targets difficult. We are studying ESE-2 (Elf5), which is highly expressed in epithelial cells of many tissues including skin keratinocytes. Here, we identify the preferred DNA-binding site of ESE-2 by performing CASTing (cyclic amplification and selection of targets) experiments. Our analysis shows that the optimal ESE-2 consensus motif consists of a GGA core and an AT-rich 5'- and 3'-flanking sequences. Mutational and competition experiments demonstrate that the flanking sequences that confer high DNA-binding affinity for ESE-2 show considerable differences from the known consensus DNA-binding sites of other Ets proteins, thus reinforcing the idea that the flanking sequences may impart recognition specificity for Ets proteins. In addition, we have identified a novel isoform of murine ESE-2, ESE-2L, that is generated by use of a hitherto unreported new exon and an alternate promoter. Interestingly, transient transfection assays with an optimal ESE-2 responsive reporter show that both ESE-2 and ESE-2L are weak transactivators. However, similar studies utilizing GAL4 chimaeras of ESE-2 demonstrate that while the DNA-binding ETS (E twenty-six) domain functions as a repressor, the PNT (pointed domain) of ESE-2 can act as a potent transcriptional activation domain. This novel transactivating property of PNT is also shared by ESE-3, another ESE family member. Identification of the ESE-2 consensus site and characterization of the transcriptional activation properties of ESE-2 shed new light on its potential as a regulator of target genes.
Collapse
Affiliation(s)
- Yeon Sook Choi
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY 14214, U.S.A
| | - Satrajit Sinha
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY 14214, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
32
|
Taatjes DJ, Roth J. Recent progress in histochemistry and cell biology: the state of the art 2005. Histochem Cell Biol 2005; 124:547-74. [PMID: 16283358 DOI: 10.1007/s00418-005-0110-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2005] [Indexed: 10/25/2022]
Abstract
Advances in the field of histochemistry, a multidisciplinary area including the detection, localization and functional characterization of molecules in single cells and complex tissues, often drives the attainment of new knowledge in the broadly defined discipline of cell biology. These two disciplines, histochemistry and cell biology, have been joined in this journal to facilitate the flow of information with celerity from technical advancement in histochemical procedures, to their utilization in experimental models. This review summarizes advancements in these fields during the past year.
Collapse
Affiliation(s)
- Douglas J Taatjes
- Microscopy Imaging Center, Department of Pathology, College of Medicine, University of Vermont, Burlington, VT 05405, USA.
| | | |
Collapse
|
33
|
Abstract
Unique developmental features during puberty, pregnancy, lactation and post-lactation make the mammary gland a prime object to explore genetic circuits that control the specification, proliferation, differentiation, survival and death of cells. Steroids and simple peptide hormones initiate and carry out complex developmental programmes, and reverse genetics has been used to define the underlying mechanistic connections.
Collapse
Affiliation(s)
- Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
34
|
Zhou J, Chehab R, Tkalcevic J, Naylor MJ, Harris J, Wilson TJ, Tsao S, Tellis I, Zavarsek S, Xu D, Lapinskas EJ, Visvader J, Lindeman GJ, Thomas R, Ormandy CJ, Hertzog PJ, Kola I, Pritchard MA. Elf5 is essential for early embryogenesis and mammary gland development during pregnancy and lactation. EMBO J 2005; 24:635-44. [PMID: 15650748 PMCID: PMC548648 DOI: 10.1038/sj.emboj.7600538] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2004] [Accepted: 12/08/2004] [Indexed: 11/09/2022] Open
Abstract
Elf5 is an epithelial-specific ETS factor. Embryos with a null mutation in the Elf5 gene died before embryonic day 7.5, indicating that Elf5 is essential during mouse embryogenesis. Elf5 is also required for proliferation and differentiation of mouse mammary alveolar epithelial cells during pregnancy and lactation. The loss of one functional allele led to complete developmental arrest of the mammary gland in pregnant Elf5 heterozygous mice. A quantitative mRNA expression study and Western blot analysis revealed that decreased expression of Elf5 correlated with the downregulation of milk proteins in Elf5(+/-) mammary glands. Mammary gland transplants into Rag(-/-) mice demonstrated that Elf5(+/-) mammary alveolar buds failed to develop in an Elf5(+/+) mammary fat pad during pregnancy, demonstrating an epithelial cell autonomous defect. Elf5 expression was reduced in Prolactin receptor (Prlr) heterozygous mammary glands, which phenocopy Elf5(+/-) glands, suggesting that Elf5 and Prlr are in the same pathway. Our data demonstrate that Elf5 is essential for developmental processes in the embryo and in the mammary gland during pregnancy.
Collapse
Affiliation(s)
- Jiong Zhou
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Renee Chehab
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Josephine Tkalcevic
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Matthew J Naylor
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Jessica Harris
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Trevor J Wilson
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Sue Tsao
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Irene Tellis
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Silva Zavarsek
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Dakang Xu
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Erika J Lapinskas
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Jane Visvader
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Geoffrey J Lindeman
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Ross Thomas
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | | | - Paul J Hertzog
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Ismail Kola
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
| | - Melanie A Pritchard
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, Melbourne, Victoria, Australia
- Centre for Functional Genomics and Human Disease, Monash Institute of Reproduction and Development, Monash University, 27-31 Wright Street, Clayton, Victoria 3168, Australia. Tel.: +61 3 9594 7208; Fax: +61 3 9594 7211; E-mail
| |
Collapse
|
35
|
Asan E, Drenckhahn D. News and views in Histochemistry and Cell Biology. Histochem Cell Biol 2004; 122:593-621. [PMID: 15614519 DOI: 10.1007/s00418-004-0735-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2004] [Indexed: 11/29/2022]
Abstract
Advances in histochemical methodology and ingenious applications of novel and improved methods continue to confirm the standing of morphological means and approaches in research efforts, and contribute significantly to increasing our knowledge about structures and functions in all areas of the life sciences from cell biology to pathology. Reports published during recent months documenting this progress are summarized in the present review.
Collapse
Affiliation(s)
- Esther Asan
- Department of Anatomy and Cell Biology, University of Wuerzburg, Koellikerstrasse 6, 97070 Wuerzburg, Germany.
| | | |
Collapse
|