1
|
Wei Z, Babkirk K, Chen S, Pei M. Epithelial-to-mesenchymal transition transcription factors: New strategies for mesenchymal tissue regeneration. Cytokine Growth Factor Rev 2025:S1359-6101(25)00032-2. [PMID: 40011185 DOI: 10.1016/j.cytogfr.2025.02.001] [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] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
The epithelial-mesenchymal transition transcription factors (EMT-TFs)-ZEB, SNAI, and TWIST families-have been extensively studied in embryonic development and tumor metastasis, providing valuable insight into their roles in cell behavior and transformation. These EMT-TFs have garnered increasing attention in the context of mesenchymal tissue regeneration, potentially contributing an approach for cell therapy. Given that dysregulated EMT-TF expression can impair cell survival and lineage differentiation, controlled regulation of their expression could offer significant advantages for tissue regeneration. However, there is a lack of comprehensive reviews to summarize the influence of the EMT-TFs on mesenchymal tissue regeneration and potential molecular mechanisms. This review explores the regulatory roles of ZEB, SNAI, and TWIST in the regeneration of bone, adipose, cartilage, muscle, and other mesenchymal tissues, with a focus on the underlying molecular signaling mechanisms. Gaining a deeper understanding of how EMT-TFs regulate cell proliferation, apoptosis, migration, and differentiation may offer new insights into the management of mesenchymal tissue repair and open novel avenues for enhancing tissue regeneration.
Collapse
Affiliation(s)
- Zhixin Wei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Kiya Babkirk
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
2
|
Voutsadakis IA. EMT Features in Claudin-Low versus Claudin-Non-Suppressed Breast Cancers and the Role of Epigenetic Modifications. Curr Issues Mol Biol 2023; 45:6040-6054. [PMID: 37504297 PMCID: PMC10378159 DOI: 10.3390/cimb45070381] [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: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Breast cancers are heterogeneous and are classified according to the expression of ER, PR and HER2 receptors to distinct groups with prognostic and therapeutic implications. Within the triple-negative group, with no expression of these three receptors, molecular heterogeneity exists but is currently not exploited in the clinic. The claudin-low phenotype is present in a subset of triple-negative breast cancers and constitutes together with basal-like cancers the most extensive groups within triple-negative breast cancers. Suppression of epithelial cell adhesion molecules in claudin-low cancers is also a hallmark of Epithelial Mesenchymal Transition (EMT). METHODS The groups of claudin-low and claudin-non-suppressed breast cancers from the extensive publicly available genomic cohorts of the METABRIC study were examined to delineate and compare their molecular landscape. Genetic and epigenetic alterations of key factors involved in EMT and potentially associated with the pathogenesis of the claudin-low phenotype were analyzed in the two groups. RESULTS Claudin-low cancers displayed up-regulation of several core transcription factors of EMT at the mRNA level, compared with claudin-non-suppressed breast cancers. Global promoter DNA methylation was increased in both groups of triple-negative cancers and in claudin-low ER-positive cancers compared with the rest of ER-positive cancers. Histone modifier enzymes, including methyltransferases, demethylases, acetyltransferases and deacetylases displayed amplifications more frequently in claudin-non-suppressed triple-negative cancers than in claudin-low counterparts and the expression of some of these enzymes differed significantly between the two groups. CONCLUSION Claudin-low and claudin-non-suppressed triple-negative breast cancers differ in their landscape of EMT core regulators and epigenetic regulators. These differences may be explored as targets for therapeutic interventions specific to the two groups of triple-negative breast cancers.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, 750 Great Northern Road, Sault Ste Marie, ON P6B 0A8, Canada
- Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
3
|
Epithelial Mesenchymal Transition and its transcription factors. Biosci Rep 2021; 42:230017. [PMID: 34708244 PMCID: PMC8703024 DOI: 10.1042/bsr20211754] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 11/17/2022] Open
Abstract
Epithelial–mesenchymal transition or EMT is an extremely dynamic process involved in conversion of epithelial cells into mesenchymal cells, stimulated by an ensemble of signaling pathways, leading to change in cellular morphology, suppression of epithelial characters and acquisition of properties such as enhanced cell motility and invasiveness, reduced cell death by apoptosis, resistance to chemotherapeutic drugs etc. Significantly, EMT has been found to play a crucial role during embryonic development, tissue fibrosis and would healing, as well as during cancer metastasis. Over the years, work from various laboratories have identified a rather large number of transcription factors (TFs) including the master regulators of EMT, with the ability to regulate the EMT process directly. In this review, we put together these EMT TFs and discussed their role in the process. We have also tried to focus on their mechanism of action, their interdependency, and the large regulatory network they form. Subsequently, it has become clear that the composition and structure of the transcriptional regulatory network behind EMT probably varies based upon various physiological and pathological contexts, or even in a cell/tissue type-dependent manner.
Collapse
|
4
|
Voutsadakis IA. Epithelial-Mesenchymal Transition (EMT) and Regulation of EMT Factors by Steroid Nuclear Receptors in Breast Cancer: A Review and in Silico Investigation. J Clin Med 2016; 5:E11. [PMID: 26797644 PMCID: PMC4730136 DOI: 10.3390/jcm5010011] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 12/23/2015] [Accepted: 12/30/2015] [Indexed: 12/20/2022] Open
Abstract
Steroid Nuclear Receptors (SNRs) are transcription factors of the nuclear receptor super-family. Estrogen Receptor (ERα) is the best-studied and has a seminal role in the clinic both as a prognostic marker but also as a predictor of response to anti-estrogenic therapies. Progesterone Receptor (PR) is also used in the clinic but with a more debatable prognostic role and the role of the four other SNRs, ERβ, Androgen Receptor (AR), Glucocorticoid Receptor (GR) and Mineralocorticoid Receptor (MR), is starting only to be appreciated. ERα, but also to a certain degree the other SNRs, have been reported to be involved in virtually every cancer-enabling process, both promoting and impeding carcinogenesis. Epithelial-Mesenchymal Transition (EMT) and the reverse Mesenchymal Epithelial Transition (MET) are such carcinogenesis-enabling processes with important roles in invasion and metastasis initiation but also establishment of tumor in the metastatic site. EMT is governed by several signal transduction pathways culminating in core transcription factors of the process, such as Snail, Slug, ZEB1 and ZEB2, and Twist, among others. This paper will discuss direct regulation of these core transcription factors by SNRs in breast cancer. Interrogation of publicly available databases for binding sites of SNRs on promoters of core EMT factors will also be included in an attempt to fill gaps where other experimental data are not available.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, Department of Internal Medicine, Sault Area Hospital, Sault Ste Marie, ON P6B 0A8, Canada.
- Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, QC P3E 2C6, Canada.
| |
Collapse
|
5
|
Gaur N, Gandhi J, Robertson ES, Verma SC, Kaul R. Epstein-Barr virus latent antigens EBNA3C and EBNA1 modulate epithelial to mesenchymal transition of cancer cells associated with tumor metastasis. Tumour Biol 2015; 36:3051-60. [PMID: 25501510 PMCID: PMC4793920 DOI: 10.1007/s13277-014-2941-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/04/2014] [Indexed: 10/24/2022] Open
Abstract
Epithelial-mesenchymal transition is an important mechanism in cancer invasiveness and metastasis. We had previously reported that cancer cells expressing Epstein-Barr virus (EBV) latent viral antigens EBV nuclear antigen EBNA3C and/ or EBNA1 showed higher motility and migration potential and had a propensity for increased metastases when tested in nude mice model. We now show that both EBNA3C and EBNA1 can modulate cellular pathways critical for epithelial to mesenchymal transition of cancer cells. Our data confirms that presence of EBNA3C or EBNA1 result in upregulation of transcriptional repressor Slug and Snail, upregulation of intermediate filament of mesenchymal origin vimentin, upregulation of transcription factor TCF8/ZEB1, downregulation as well as disruption of tight junction zona occludens protein ZO-1, downregulation of cell adhesion molecule E-cadherin, and nuclear translocation of β-catenin. We further show that the primary tumors as well as metastasized lesions derived from EBV antigen-expressing cancer cells in nude mice model display EMT markers expression pattern suggesting their greater propensity to mesenchymal transition.
Collapse
Affiliation(s)
- Nivedita Gaur
- Department of Microbiology, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Jaya Gandhi
- Department of Microbiology, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Erle S. Robertson
- Department of Microbiology and Tumour Virology Program, Abramson Cancer Centre, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Subhash C. Verma
- Department of Microbiology and Immunology, University of Nevada Reno, Reno, NV, USA
| | - Rajeev Kaul
- Department of Microbiology, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| |
Collapse
|
6
|
Lim W, Song G. Novel genes and hormonal regulation for gonadal development during embryogenesis in chickens. Gen Comp Endocrinol 2015; 211:20-7. [PMID: 25452029 DOI: 10.1016/j.ygcen.2014.11.009] [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: 04/01/2014] [Revised: 10/26/2014] [Accepted: 11/08/2014] [Indexed: 01/30/2023]
Abstract
Asymmetrical gonadal morphogenesis is well known in female chickens in contrast to males where both gonads develop symmetrically. However, only a few genes have been reported to determine differential morphology between female and male gonads in chicken and their mechanisms of action are unclear. Therefore, we focused on three genes (TOM1L1, TTR, and ZEB1) that are related to cellular proliferation and embryonic development based on previous study indicating up- or down-regulated transcripts in the asymmetric female gonads between embryonic day 6 (E6) and E9 by microarray analyses. To define the validity of the gene expression pattern discovered, q-PCR and in situ hybridization analyses were performed. In the left female gonad between E6 and E9 the expression of TOM1L1, TTR and ZEB1 increased at E9. On the other hand, TOM1L1 and TTR increased significantly in both male gonads between E6 and E9. In addition, recombinant FSH and LH stimulated proliferation of gonadal cells and influenced expression of selected genes in chickens. This suggests that hormonal regulation is involved in growth and development in the embryonic gonad of chickens. Collectively, the results show differential gene expression between the left and right gonads in chicken embryos and that of is regulated by gonadotropin. These results provide novel insights into candidate genes regulating gonad development and differentiation.
Collapse
Affiliation(s)
- Whasun Lim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 136-713, Republic of Korea.
| |
Collapse
|
7
|
Lim W, Song G. Identification of novel regulatory genes in development of the avian reproductive tracts. PLoS One 2014; 9:e96175. [PMID: 24763497 PMCID: PMC3999111 DOI: 10.1371/journal.pone.0096175] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 04/04/2014] [Indexed: 01/12/2023] Open
Abstract
The chicken reproductive system is unique in maintaining its functions including production of eggs or sperm, fertilization of the egg by sperm maintained in sperm nests, production of hormones regulating its growth, development and function, and reproduction. Development of the reproductive organs is a highly regulated process that results in differentiation and proliferation of germ cells in response to predominant regulatory factors such as hormones and transcription factors. However, only a few genes are known to determine morphogenesis of the chicken reproductive tract and their mechanisms are unknown. Therefore, in the present study, we investigated the expression patterns of four genes including SNCA, TOM1L1, TTR and ZEB1 in the gonads at embryonic days 14 and 18, and in immature (12-week-old) and mature (50-week-old) chickens, as well as the reproductive tract including ovary, oviduct and testes of the respective sexes by qRT-PCR, in situ hybridization and immunofluorescence analyses. The expression of SNCA, TOM1L1 and ZEB1 genes was higher in immature and mature female reproductive tracts than expression of TTR. In addition, different temporal and spatial patterns of expression of the four genes were observed during maturation of testis in chickens. Specifically, SNCA, TOM1L1 and TTR were highly expressed in testes of 12-week-old chickens. Moreover, several chicken specific microRNAs (miRs) were demonstrated to affect expression of target gene mRNAs by directly binding to the 3′-UTR of their target genes through actions at the post-transcriptional level as follows: miR-153 and miR-1643 for SNCA; miR-1680* for TTR; and miR-200b and miR-1786 for ZEB1. These results suggest that four-selected genes play an important role in development of the male and female reproductive tract in chickens and expression of most candidate genes is regulated at the post-transcriptional level through specific microRNAs.
Collapse
Affiliation(s)
- Whasun Lim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
8
|
Sánchez-Tilló E, Liu Y, de Barrios O, Siles L, Fanlo L, Cuatrecasas M, Darling DS, Dean DC, Castells A, Postigo A. EMT-activating transcription factors in cancer: beyond EMT and tumor invasiveness. Cell Mol Life Sci 2012; 69:3429-56. [PMID: 22945800 PMCID: PMC11115078 DOI: 10.1007/s00018-012-1122-2] [Citation(s) in RCA: 406] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 07/20/2012] [Accepted: 08/09/2012] [Indexed: 12/13/2022]
Abstract
Cancer is a complex multistep process involving genetic and epigenetic changes that eventually result in the activation of oncogenic pathways and/or inactivation of tumor suppressor signals. During cancer progression, cancer cells acquire a number of hallmarks that promote tumor growth and invasion. A crucial mechanism by which carcinoma cells enhance their invasive capacity is the dissolution of intercellular adhesions and the acquisition of a more motile mesenchymal phenotype as part of an epithelial-to-mesenchymal transition (EMT). Although many transcription factors can trigger it, the full molecular reprogramming occurring during an EMT is mainly orchestrated by three major groups of transcription factors: the ZEB, Snail and Twist families. Upregulated expression of these EMT-activating transcription factors (EMT-ATFs) promotes tumor invasiveness in cell lines and xenograft mice models and has been associated with poor clinical prognosis in human cancers. Evidence accumulated in the last few years indicates that EMT-ATFs also regulate an expanding set of cancer cell capabilities beyond tumor invasion. Thus, EMT-ATFs have been shown to cooperate in oncogenic transformation, regulate cancer cell stemness, override safeguard programs against cancer like apoptosis and senescence, determine resistance to chemotherapy and promote tumor angiogenesis. This article reviews the expanding portfolio of functions played by EMT-ATFs in cancer progression.
Collapse
Affiliation(s)
- Ester Sánchez-Tilló
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
| | - Yongqing Liu
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- Department of Ophthalmology and Birth Defects Center, Louisville Health Science Center, Louisville, KY 40202 USA
| | - Oriol de Barrios
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
| | - Laura Siles
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
| | - Lucia Fanlo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- Master Program in Biomedical Research, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Miriam Cuatrecasas
- Department of Pathology, Hospital Clinic and IDIBAPS’ Tumor Bank, 08036 Barcelona, Spain
| | - Douglas S. Darling
- Department of Oral Health and Rehabilitation, Center for Genetics and Molecular Medicine, University of Louisville, Louisville, KY 40202 USA
| | - Douglas C. Dean
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- Department of Ophthalmology and Birth Defects Center, Louisville Health Science Center, Louisville, KY 40202 USA
| | - Antoni Castells
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
- Institute of Digestive and Metabolic Diseases, Hospital Clinic, 08036 Barcelona, Spain
| | - Antonio Postigo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- ICREA, 08010 Barcelona, Spain
| |
Collapse
|
9
|
Kurima K, Hertzano R, Gavrilova O, Monahan K, Shpargel KB, Nadaraja G, Kawashima Y, Lee KY, Ito T, Higashi Y, Eisenman DJ, Strome SE, Griffith AJ. A noncoding point mutation of Zeb1 causes multiple developmental malformations and obesity in Twirler mice. PLoS Genet 2011; 7:e1002307. [PMID: 21980308 PMCID: PMC3183090 DOI: 10.1371/journal.pgen.1002307] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 07/30/2011] [Indexed: 01/05/2023] Open
Abstract
Heterozygous Twirler (Tw) mice develop obesity and circling behavior associated with malformations of the inner ear, whereas homozygous Tw mice have cleft palate and die shortly after birth. Zeb1 is a zinc finger protein that contributes to mesenchymal cell fate by repression of genes whose expression defines epithelial cell identity. This developmental pathway is disrupted in inner ears of Tw/Tw mice. The purpose of our study was to comprehensively characterize the Twirler phenotype and to identify the causative mutation. The Tw/+ inner ear phenotype includes irregularities of the semicircular canals, abnormal utricular otoconia, a shortened cochlear duct, and hearing loss, whereas Tw/Tw ears are severely malformed with barely recognizable anatomy. Tw/+ mice have obesity associated with insulin-resistance and have lymphoid organ hypoplasia. We identified a noncoding nucleotide substitution, c.58+181G>A, in the first intron of the Tw allele of Zeb1 (Zeb1Tw). A knockin mouse model of c.58+181G>A recapitulated the Tw phenotype, whereas a wild-type knockin control did not, confirming the mutation as pathogenic. c.58+181G>A does not affect splicing but disrupts a predicted site for Myb protein binding, which we confirmed in vitro. In comparison, homozygosity for a targeted deletion of exon 1 of mouse Zeb1, Zeb1ΔEx1, is associated with a subtle abnormality of the lateral semicircular canal that is different than those in Tw mice. Expression analyses of E13.5 Twirler and Zeb1ΔEx1 ears confirm that Zeb1ΔEx1 is a null allele, whereas Zeb1Tw RNA is expressed at increased levels in comparison to wild-type Zeb1. We conclude that a noncoding point mutation of Zeb1 acts via a gain-of-function to disrupt regulation of Zeb1Tw expression, epithelial-mesenchymal cell fate or interactions, and structural development of the inner ear in Twirler mice. This is a novel mechanism underlying disorders of hearing or balance. Twirler (Tw) mice have a combination of abnormalities that includes cleft palate, malformations of the inner ear, hearing loss, vestibular dysfunction, obesity, and lymphoid hypoplasia. In this study, we show that the underlying mutation affects the Zeb1 gene. Zeb1 was already known to encode a protein normally expressed in mesenchymal cells, where it represses expression of genes that are uniquely expressed in epithelial cells. The Tw mutation is a rare example of a single-nucleotide substitution in a region of a gene that does not encode protein, promoter, or splice sites, so we engineered a mouse model with the mutation that confirmed its causative role. The Tw mutation disrupts a consensus DNA binding site sequence for the Myb family of regulatory proteins. We conclude that this mutation leads to abnormal expression of Zeb1, structural malformations of the inner ear, and a loss of hearing and balance function. A similar mechanism may underlie other features of Twirler, such as obesity and cleft palate.
Collapse
Affiliation(s)
- Kiyoto Kurima
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, United States of America
| | - Ronna Hertzano
- Department of Otorhinolaryngology–Head and Neck Surgery, University of Maryland, Baltimore, Maryland, United States of America
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kelly Monahan
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, United States of America
| | - Karl B. Shpargel
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, United States of America
| | - Garani Nadaraja
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, United States of America
| | - Yoshiyuki Kawashima
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, United States of America
| | - Kyu Yup Lee
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, United States of America
| | - Taku Ito
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, United States of America
| | - Yujiro Higashi
- Department of Perinatology, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Japan
| | - David J. Eisenman
- Department of Otorhinolaryngology–Head and Neck Surgery, University of Maryland, Baltimore, Maryland, United States of America
| | - Scott E. Strome
- Department of Otorhinolaryngology–Head and Neck Surgery, University of Maryland, Baltimore, Maryland, United States of America
| | - Andrew J. Griffith
- Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
10
|
Riazuddin SA, McGlumphy EJ, Yeo WS, Wang J, Katsanis N, Gottsch JD. Replication of the TCF4 intronic variant in late-onset Fuchs corneal dystrophy and evidence of independence from the FCD2 locus. Invest Ophthalmol Vis Sci 2011; 52:2825-9. [PMID: 21245398 DOI: 10.1167/iovs.10-6497] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Fuchs corneal dystrophy (FCD) is an autosomal dominant disease of the corneal endothelium with variable penetrance and expressivity. Recently, rs613872, an intronic variation of TCF4 associated with late-onset FCD, was reported. The present study was undertaken to examine this association in our cohort of FCD patients, to assess the significance of this finding, and to investigate the candidacy of TCF4 in the context of the mapped FCD2 locus. METHODS The authors recruited 170 patients with late-onset FCD and 180 age-matched controls. Blood samples were collected, and genomic DNA was extracted. A panel of nine SNPs spanning the entire TCF4 locus was genotyped both on this cohort and on three previously reported FCD2-linked families. The association of an individual SNP with late-onset FCD was evaluated with the Fisher exact test, and the coding exons and exon-intron boundaries of TCF4 were sequenced in 96 affected persons. RESULTS The risk allele G of rs613872 is associated significantly with late-onset FCD (odds ratio, 4.2; P = 4.28 x 10⁻¹⁵) and was present in male and female affected persons without any sex bias, replicating recent findings, though the authors found no apparent correlation with the severity of the disease phenotype. Moreover, the risk allele did not cosegregate with the disease phenotype in any of the three FCD2-linked families. The authors did not identify any pathogenic variants in the coding region of TCF4. CONCLUSIONS The authors report the first independent replication of rs613872 conferring risk of late-onset FCD. Their data suggest that this risk factor is likely independent of the FCD2 locus, whose causality remains unknown.
Collapse
Affiliation(s)
- S Amer Riazuddin
- Wilmer Eye Institute, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|
11
|
Qiao L, Tasian GE, Zhang H, Cunha GR, Baskin L. ZEB1 is estrogen responsive in vitro in human foreskin cells and is over expressed in penile skin in patients with severe hypospadias. J Urol 2011; 185:1888-93. [PMID: 21421232 DOI: 10.1016/j.juro.2010.12.066] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Indexed: 11/29/2022]
Abstract
PURPOSE We determined the effect of estrogen on ZEB1 in vitro and tested the hypothesis that ZEB1 is over expressed in the penile skin of subjects with hypospadias. MATERIALS AND METHODS Hs68 cells, a fibroblast cell line derived from human foreskin, were exposed to 0, 1, 10 and 100 nM estrogen, and the expression level of ZEB1 was assessed using reverse transcription real-time polymerase chain reaction, Western blot and immunocytochemical analysis. Next, preputial skin was prospectively collected from case and control subjects at hypospadias repair (37 cases) and circumcision (11). Hypospadias was classified as severe (13 cases) or mild (24) based on the position of the urethral meatus. ZEB1 expression was quantified using reverse transcription real-time polymerase chain reaction, Western blot and immunohistochemical analysis. RESULTS Estrogen increased ZEB1 expression at the mRNA and protein levels in Hs68 cells in a concentration dependent fashion (p <0.01). Subjects with severe hypospadias had significantly higher ZEB1 mRNA levels and protein expression compared to controls or subjects with mild hypospadias (both p <0.01). Subjects with severe hypospadias had increased expression of ZEB1 in the basal layers of the preputial epidermis. CONCLUSIONS Estrogen increases ZEB1 expression in a human foreskin fibroblast cell line in vitro. Furthermore, ZEB1 is significantly over expressed in the penile skin of subjects with severe hypospadias. We propose that ZEB1 overexpression may contribute to development of hypospadias and may mediate the effect of estrogen on developing external male genitalia.
Collapse
Affiliation(s)
- Liang Qiao
- Frank Hinman, Jr. Urological Research Laboratory, Department of Urology, University of California, San Francisco, California 94143, USA
| | | | | | | | | |
Collapse
|
12
|
Anose BM, Sanders MM. Androgen Receptor Regulates Transcription of the ZEB1 Transcription Factor. Int J Endocrinol 2011; 2011:903918. [PMID: 22190929 PMCID: PMC3235469 DOI: 10.1155/2011/903918] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 09/15/2011] [Indexed: 11/17/2022] Open
Abstract
The zinc finger E-box binding protein 1 (ZEB1) transcription factor belongs to a two-member family of zinc-finger homeodomain proteins involved in physiological and pathological events mostly relating to cell migration and epithelial to mesenchymal transitions (EMTs). ZEB1 (also known as δEF1, zfhx1a, TCF8, and Zfhep) plays a key role in regulating such diverse processes as T-cell development, skeletal patterning, reproduction, and cancer cell metastasis. However, the factors that regulate its expression and consequently the signaling pathways in which ZEB1 participates are poorly defined. Because it is induced by estrogen and progesterone and is high in prostate cancer, we investigated whether tcf8, which encodes ZEB1, is regulated by androgen. Data herein demonstrate that tcf8 is induced by dihydrotestosterone (DHT) in the human PC-3/AR prostate cancer cell line and that this induction is mediated by two androgen response elements (AREs). These results demonstrate that ZEB1 is an intermediary in androgen signaling pathways.
Collapse
Affiliation(s)
- Bynthia M. Anose
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Chemistry, Bethel University, St. Paul, MN 55112, USA
| | - Michel M. Sanders
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- *Michel M. Sanders:
| |
Collapse
|
13
|
Voelzmann A, Bauer R. Ceramide synthases in mammalians, worms, and insects: emerging schemes. Biomol Concepts 2010; 1:411-22. [DOI: 10.1515/bmc.2010.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
AbstractThe ceramide synthase (CerS) gene family comprises a group of highly conserved transmembrane proteins, which are found in all studied eukaryotes. The key feature of the CerS proteins is their role in ceramide synthase activity. Therefore, their original name ‘longevity assurance gene (Lass) homologs’, after the founding member, the yeast longevity assurance gene lag1, was altered to ‘CerS’. All CerS have high sequence similarity in a domain called LAG1 motif and a subset of CerS proteins is predicted to contain a Homeobox (Hox) domain. These domains could be the key to the multiple roles CerS have. CerS proteins play a role in diverse biological processes such as proliferation, differentiation, apoptosis, stress response, cancer, and neurodegeneration. In this review, we focus on CerS structure and biological function with emphasis of biological functions in the widely used model systems Caenorhabditis elegans and Drosophila melanogaster. Also, we focus on the accumulating data suggesting a role for CerS in lipid homeostasis.
Collapse
Affiliation(s)
- André Voelzmann
- 1LIMES Institute, Program Unit Development and Genetics, Laboratory for Molecular Developmental Biology, University of Bonn, Carl-Troll-Str. 31, D-53115 Bonn, Germany
| | - Reinhard Bauer
- 1LIMES Institute, Program Unit Development and Genetics, Laboratory for Molecular Developmental Biology, University of Bonn, Carl-Troll-Str. 31, D-53115 Bonn, Germany
| |
Collapse
|
14
|
Saykally JN, Dogan S, Cleary MP, Sanders MM. The ZEB1 transcription factor is a novel repressor of adiposity in female mice. PLoS One 2009; 4:e8460. [PMID: 20041147 PMCID: PMC2794530 DOI: 10.1371/journal.pone.0008460] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Accepted: 10/15/2009] [Indexed: 12/31/2022] Open
Abstract
Background Four genome-wide association studies mapped an “obesity” gene to human chromosome 10p11–12. As the zinc finger E-box binding homeobox 1 (ZEB1) transcription factor is encoded by the TCF8 gene located in that region, and as it influences the differentiation of various mesodermal lineages, we hypothesized that ZEB1 might also modulate adiposity. The goal of these studies was to test that hypothesis in mice. Methodology/Principal Findings To ascertain whether fat accumulation affects ZEB1 expression, female C57BL/6 mice were fed a regular chow diet (RCD) ad libitum or a 25% calorie-restricted diet from 2.5 to 18.3 months of age. ZEB1 mRNA levels in parametrial fat were six to ten times higher in the obese mice. To determine directly whether ZEB1 affects adiposity, wild type (WT) mice and mice heterozygous for TCF8 (TCF8+/−) were fed an RCD or a high-fat diet (HFD) (60% calories from fat). By two months of age on an HFD and three months on an RCD, TCF8+/− mice were heavier than WT controls, which was attributed by Echo MRI to increased fat mass (at three months on an HFD: 0.517±0.081 total fat/lean mass versus 0.313±0.036; at three months on an RCD: 0.175±0.013 versus 0.124±0.012). No differences were observed in food uptake or physical activity, suggesting that the genotypes differ in some aspect of their metabolic activity. ZEB1 expression also increases during adipogenesis in cell culture. Conclusion/Significance These results show for the first time that the ZEB1 transcription factor regulates the accumulation of adipose tissue. Furthermore, they corroborate the genome-wide association studies that mapped an “obesity” gene at chromosome 10p11–12.
Collapse
Affiliation(s)
- Jessica N. Saykally
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Soner Dogan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Margot P. Cleary
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Michel M. Sanders
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
15
|
Reciprocal regulation of ZEB1 and AR in triple negative breast cancer cells. Breast Cancer Res Treat 2009; 123:139-47. [PMID: 19921427 DOI: 10.1007/s10549-009-0623-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Accepted: 10/28/2009] [Indexed: 10/20/2022]
Abstract
Zinc-finger enhancer binding protein (ZEB1) is a transcription factor involved in the progression of cancer primarily through promoting epithelial to mesenchymal transition (EMT). ZEB1 represses the expression of E-cadherin by binding to E-box sequences in the promoter, thus decreasing epithelial differentiation. We show that ZEB1 and androgen receptor (AR) cross-talk in triple negative breast cancer cell lines. Chromatin immunoprecipitation analysis demonstrates that ZEB1 binds directly to the E-box located in the AR promoter. ZEB1 suppression by stably transfecting shRNA in a triple negative breast cancer cell line resulted in a decrease of AR mRNA, protein, and AR downstream targets. ZEB1 knockdown in triple negative breast cancer cells sensitized the cells to bicalutamide by reducing migration compared to the control cells. Conversely, blockade of AR signaling with bicalutamide resulted in a suppression of ZEB1 protein expression in two triple negative breast cancer cell lines. Furthermore, using a breast cancer tissue microarray, a majority of triple negative breast cancers exhibit positive staining for both ZEB1 and AR. Taken together, these results indicate that ZEB1 and AR regulate each other to promote cell migration in triple negative breast cancer cells.
Collapse
|
16
|
Schmalhofer O, Brabletz S, Brabletz T. E-cadherin, beta-catenin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev 2009; 28:151-66. [PMID: 19153669 DOI: 10.1007/s10555-008-9179-y] [Citation(s) in RCA: 622] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The embryonic program 'epithelial-mesenchymal transition' (EMT) is activated during tumor invasion in disseminating cancer cells. Characteristic to these cells is a loss of E-cadherin expression, which can be mediated by EMT-inducing transcriptional repressors, e.g. ZEB1. Consequences of a loss of E-cadherin are an impairment of cell-cell adhesion, which allows detachment of cells, and nuclear localization of beta-catenin. In addition to an accumulation of cancer stem cells, nuclear beta-catenin induces a gene expression pattern favoring tumor invasion, and mounting evidence indicates multiple reciprocal interactions of E-cadherin and beta-catenin with EMT-inducing transcriptional repressors to stabilize an invasive mesenchymal phenotype of epithelial tumor cells.
Collapse
Affiliation(s)
- Otto Schmalhofer
- Department of Visceral Surgery, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | | | | |
Collapse
|
17
|
Dougherty DC, Park HM, Sanders MM. Interferon regulatory factors (IRFs) repress transcription of the chicken ovalbumin gene. Gene 2009; 439:63-70. [PMID: 19341784 PMCID: PMC2749989 DOI: 10.1016/j.gene.2009.03.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 03/14/2009] [Accepted: 03/18/2009] [Indexed: 01/03/2023]
Abstract
Although the ovalbumin (Ov) gene has served as a model to study tissue-specific, steroid hormone-induced gene expression in vertebrates for decades, the mechanisms responsible for regulating this gene remain elusive. Ov is repressed in non-oviduct tissue and in estrogen-deprived oviduct by a strong repressor site located from -130 to -100 and designated CAR for COUP-TF adjacent repressor. The goal of this study was to identify the CAR binding protein(s). A transcription factor database search revealed that a putative interferon-stimulated response element (ISRE), which binds interferon regulatory factors (IRFs), is located in this region. Gel mobility shift assays demonstrated that the protein(s) binding to the CAR site is recognized by an IRF antibody and that mutations in the ISRE abolish that binding. In hopes of identifying the IRF(s) responsible for the tissue-specific regulation of Ov, mRNA levels for IRFs-4, -8, and -10 were measured in seven tissues from chicks treated with or without estrogen. PCR experiments showed that both IRF-8 and -10 are expressed in all chick tissues tested whereas IRF-4 has a much more limited expression pattern. Transfection experiments with OvCAT (chloramphenicol acetyltransferase) reporter constructs demonstrated that both IRF-4 and IRF-10 are capable of repressing the Ov gene even in the presence of steroid hormones and that nucleotides in the ISRE are required for repression. These experiments indicate that the repressor activity associated with the CAR site is mediated by IRF family members and suggest that IRF members also repress Ov in non-oviduct tissues.
Collapse
|
18
|
Miller MM, Jarosinski KW, Schat KA. Negative modulation of the chicken infectious anemia virus promoter by COUP-TF1 and an E box-like element at the transcription start site binding δEF1. J Gen Virol 2008; 89:2998-3003. [DOI: 10.1099/vir.0.2008/003103-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Expression of enhanced green fluorescent protein (EGFP) under control of the promoter-enhancer of chicken infectious anemia virus (CAV) is increased in an oestrogen receptor-enhanced cell line when treated with oestrogen and the promoter-enhancer binds unidentified proteins that recognize a consensus oestrogen response element (ERE). Co-transfection assays with the CAV promoter and the nuclear receptor chicken ovalbumin upstream promoter transcription factor 1 (COUP-TF1) showed that expression of EGFP was decreased by 50 to 60 % in DF-1 and LMH cells. The CAV promoter that included sequences at and downstream of the transcription start point had less expression than a short promoter construct. Mutation of a putative E box at this site restored expression levels. Electromobility shift assays showed that the transcription regulator delta-EF1 (δEF1) binds to this E box region. These findings indicate that the CAV promoter activity can be affected directly or indirectly by COUP-TF1 andδEF1.
Collapse
Affiliation(s)
- Myrna M. Miller
- Unit of Avian Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Keith W. Jarosinski
- Unit of Avian Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Karel A. Schat
- Unit of Avian Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
19
|
Hurt EM, Saykally JN, Anose BM, Kalli KR, Sanders MM. Expression of the ZEB1 (deltaEF1) transcription factor in human: additional insights. Mol Cell Biochem 2008; 318:89-99. [PMID: 18622689 DOI: 10.1007/s11010-008-9860-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Accepted: 06/25/2008] [Indexed: 01/13/2023]
Abstract
The zinc finger E-box binding transcription factor ZEB1 (deltaEF1/Nil-2-a/AREB6/zfhx1a/TCF8/zfhep/BZP) is emerging as an important regulator of the epithelial to mesenchymal transitions (EMT) required for development and cancer metastasis. ZEB1 promotes EMT by repressing genes contributing to the epithelial phenotype while activating those associated with the mesenchymal phenotype. TCF8 (zfhx1a), the gene encoding ZEB1, is induced by several potentially oncogenic ligands including TGF-beta, estrogen, and progesterone. TGF-beta appears to activate EMT, at least in part, by inducing ZEB1. However, our understanding of how ZEB1 contributes to signaling pathways elicited by estrogen and progesterone is quite limited, as is our understanding of its functional roles in normal adult tissues. To begin to address these questions, a human tissue mRNA array analysis was done. In adults, the highest ZEB1 mRNA expression is in bladder and uterus, whereas in the fetus highest expression is in lung, thymus, and heart. To further investigate the regulation of TCF8 by estrogen, ZEB1 mRNA was measured in ten estrogen-responsive cell lines, but it is only induced in the OV266 ovarian carcinoma line. Although high expression of ZEB1 mRNA is estrogen-dependent in normal human ovarian and endometrial biopsies, high expression is estrogen-independent in late stage ovarian and endometrial carcinomas, raising the possibility that deregulated expression promotes cancer progression. In contrast, TCF8 is at least partially deleted in 4 of 5 well-differentiated, grade I endometrial carcinomas, which may contribute to their non-aggressive phenotype. These data support the contention that high ZEB1 encourages gynecologic carcinoma progression.
Collapse
Affiliation(s)
- Elaine M Hurt
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD 21702, USA
| | | | | | | | | |
Collapse
|
20
|
Liu Y, El-Naggar S, Darling DS, Higashi Y, Dean DC. Zeb1 links epithelial-mesenchymal transition and cellular senescence. Development 2008; 135:579-88. [PMID: 18192284 PMCID: PMC2507753 DOI: 10.1242/dev.007047] [Citation(s) in RCA: 244] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Overexpression of zinc finger E-box binding homeobox transcription factor 1 (Zeb1) in cancer leads to epithelial-to-mesenchymal transition (EMT) and increased metastasis. As opposed to overexpression, we show that mutation of Zeb1 in mice causes a mesenchymal-epithelial transition in gene expression characterized by ectopic expression of epithelial genes such as E-cadherin and loss of expression of mesenchymal genes such as vimentin. In contrast to rapid proliferation in cancer cells where Zeb1 is overexpressed, this mesenchymal-epithelial transition in mutant mice is associated with diminished proliferation of progenitor cells at sites of developmental defects, including the forming palate, skeleton and CNS. Zeb1 dosage-dependent deregulation of epithelial and mesenchymal genes extends to mouse embryonic fibroblasts (MEFs), and mutant MEFs also display diminished replicative capacity in culture, leading to premature senescence. Replicative senescence in MEFs is classically triggered by products of the Ink4a (Cdkn2a) gene. However, this Ink4a pathway is not activated during senescence of Zeb1 mutant MEFs. Instead, there is ectopic expression of two other cell cycle inhibitory cyclin-dependent kinase inhibitors, p15Ink4b (Cdkn2b) and p21Cdkn1a (Cdkn1a). We demonstrate that this ectopic expression of p15Ink4b extends in vivo to sites of diminished progenitor cell proliferation and developmental defects in Zeb1-null mice.
Collapse
Affiliation(s)
- Yongqing Liu
- James Graham Brown Cancer Center; Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - Shahenda El-Naggar
- James Graham Brown Cancer Center; Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202
| | - Douglas S. Darling
- Departments of Peiodontics, Endodontics and Dental Hygiene; Center for Oral Health and Systemic Disease, University of Louisville School of Dentistry, Louisville, KY 40292
| | - Yujiro Higashi
- Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Douglas C. Dean
- James Graham Brown Cancer Center; Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202
| |
Collapse
|
21
|
Liu Y, Costantino M, Montoya-Durango D, Higashi Y, Darling D, Dean D. The zinc finger transcription factor ZFHX1A is linked to cell proliferation by Rb-E2F1. Biochem J 2007; 408:79-85. [PMID: 17655524 PMCID: PMC2049079 DOI: 10.1042/bj20070344] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Revised: 07/16/2007] [Accepted: 07/27/2007] [Indexed: 11/17/2022]
Abstract
ZFHX1A is expressed in proliferating cells in the developing embryo, and in the present study we provide evidence that its expression is confined to proliferating cells through dependence on the Rb (retinoblastoma protein) family/E2F cell cycle pathway. Mutation of the Rb or E2F1 genes lead to induction of ZFHX1A mRNA, implying that the Rb-E2F1 repressor complex is important for repression of ZFHX1A. This repression is associated with recruitment of an E2F-Rb-histone deacetylase repressor complex to the promoter. A dominant-negative form of E2F1 inhibited ZFHX1A expression in p16INK4a- cells where Rb is constitutively hyperphosphorylated and inactive, suggesting that E2F can contribute to ZFHX1A transactivation in the absence of functional Rb. ZFHX1A is an E-box-binding transcription factor whose binding sites overlap with those bound by Snail1 and 2, and ZFHX1B/SIP1 (leading to at least partially overlapping function; for example, each of the proteins can repress E-cadherin expression). We found that expression of Snail1 and ZFHX1B/SIP1 is also regulated by E2Fs, but in contrast with ZFHX1A this regulation is Rb-family-independent. Snail2 expression was unaffected by either E2F or the Rb family. We propose that the differential effects of the Rb family/E2F pathway on expression of these E-box-binding proteins are important in maintaining their distinct patterns (and thus distinct functions) during embryogenesis.
Collapse
Key Words
- proliferation
- retinoblastoma protein
- zfhx1a
- zinc finger transcription factor
- cdk, cyclin-dependent kinase
- chip, chromatin immunoprecipitation
- cmv, cytomegalovirus
- cns, central nervous system
- ctbp, c-terminal-binding protein
- db, dna-binding domain
- gapdh, glyceraldehyde-3-phosphate dehydrogenase
- hdac, histone deacetylase
- iptg, isopropyl β-d-thiogalactoside
- mef, mouse embryonic fibroblast
- mer, mutant oestrogen receptor
- oht, tamoxifen
- rb, retinoblastoma protein
- rt, reverse transcriptase
- tgf-β, transforming growth factor-β
- tko, triple knockout
Collapse
Affiliation(s)
- Yongqing Liu
- *James Graham Brown Cancer Center, Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, U.S.A
| | - Mary E. Costantino
- †Departments of Peiodontics, Endodontics and Dental Hygiene, Center for Oral Health and Systemic Disease, University of Louisville School of Dentistry, Louisville, KY 40292, U.S.A
| | - Diego Montoya-Durango
- ‡Department of Biochemistry, University of Louisville Health Sciences Center, Louisville, KY 40202, U.S.A
| | - Yujiro Higashi
- §Developmental Biology Group, Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Douglas S. Darling
- †Departments of Peiodontics, Endodontics and Dental Hygiene, Center for Oral Health and Systemic Disease, University of Louisville School of Dentistry, Louisville, KY 40292, U.S.A
| | - Douglas C. Dean
- *James Graham Brown Cancer Center, Department of Ophthalmology and Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, U.S.A
| |
Collapse
|
22
|
Yang S, Du J, Wang Z, Yuan W, Qiao Y, Zhang M, Zhang J, Gao S, Yin J, Sun B, Zhu T. BMP-6 promotes E-cadherin expression through repressing deltaEF1 in breast cancer cells. BMC Cancer 2007; 7:211. [PMID: 17997862 PMCID: PMC2217560 DOI: 10.1186/1471-2407-7-211] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Accepted: 11/13/2007] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Bone morphogenetic protein-6 (BMP-6) is critically involved in many developmental processes. Recent studies indicate that BMP-6 is closely related to tumor differentiation and metastasis. METHODS Quantitative RT-PCR was used to determine the expression of BMP-6, E-cadherin, and deltaEF1 at the mRNA level in MCF-7 and MDA-MB-231 breast cancer cells, as well as in 16 breast cancer specimens. Immunoblot analysis was used to measure the expression of deltaEF1 at the protein level in deltaEF1-overexpressing and deltaEF1-interfered MDA-MB-231 cells. Luciferase assay was used to determine the rhBMP-6 or deltaEF1 driven transcriptional activity of the E-cadherin promoter in MDA-MB-231 cells. Quantitative CHIP assay was used to detect the direct association of deltaEF1 with the E-cadherin proximal promoter in MDA-MB-231 cells. RESULTS MCF-7 breast cancer cells, an ER+ cell line that expressed high levels of BMP-6 and E-cadherin exhibited very low levels of deltaEF1 transcript. In contrast, MDA-MB-231 cells, an ER- cell line had significantly reduced BMP-6 and E-cadherin mRNA levels, suggesting an inverse correlation between BMP-6/E-cadherin and deltaEF1. To determine if the same relationship exists in human tumors, we examined tissue samples of breast cancer from human subjects. In 16 breast cancer specimens, the inverse correlation between BMP-6/E-cadherin and deltaEF1 was observed in both ER+ cases (4 of 8 cases) and ER- cases (7 of 8 cases). Further, we found that BMP-6 inhibited deltaEF1 transcription, resulting in an up-regulation of E-cadherin mRNA expression. This is consistent with our analysis of the E-cadherin promoter demonstrating that BMP-6 was a potent transcriptional activator. Interestingly, ectopic expression of deltaEF1 was able to block BMP-6-induced transactivation of E-cadherin, whereas RNA interference-mediated down-regulation of endogenous deltaEF1 in breast cancer cells abolished E-cadherin transactivation by BMP-6. In addition to down-regulating the expression of deltaEF1, BMP-6 also physically dislodged deltaEF1 from E-cadherin promoter to allow the activation of E-cadherin transcription. CONCLUSION We conclude that repression of deltaEF1 plays a key role in mediating BMP-6-induced transcriptional activation of E-cadherin in breast cancer cells. Consistent with the fact that higher level of deltaEF1 expression is associated with more invasive phenotype of breast cancer cells, our collective data suggests that deltaEF1 is likely the switch through which BMP-6 restores E-cadherin-mediated cell-to-cell adhesion and prevents breast cancer metastasis.
Collapse
Affiliation(s)
- Shuang Yang
- Medical College of Nankai University, Tianjin, China
| | - Jun Du
- Medical College of Nankai University, Tianjin, China
| | - Zhaoqi Wang
- Medical College of Nankai University, Tianjin, China
| | - Wei Yuan
- Medical College of Nankai University, Tianjin, China
| | - Yuhuan Qiao
- Medical College of Nankai University, Tianjin, China
| | - Ming Zhang
- Medical College of Nankai University, Tianjin, China
| | - Jie Zhang
- Medical College of Nankai University, Tianjin, China
| | - Songyuan Gao
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jian Yin
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Baocun Sun
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Tianhui Zhu
- Medical College of Nankai University, Tianjin, China
| |
Collapse
|
23
|
Yang S, Zhao L, Yang J, Chai D, Zhang M, Zhang J, Ji X, Zhu T. deltaEF1 represses BMP-2-induced differentiation of C2C12 myoblasts into the osteoblast lineage. J Biomed Sci 2007; 14:663-79. [PMID: 17479358 DOI: 10.1007/s11373-007-9155-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Accepted: 02/21/2007] [Indexed: 10/23/2022] Open
Abstract
Osteoblasts, derived from pluripotent mesenchymal precursor cells, acquire their differentiated phenotypes under the control of a series of regulatory factors, the best known of which is BMP-2. Our recent preliminary studies suggest that expression of deltaEF1, a member of the zinc finger-homeodomain transcription factor family, is significantly down-regulated as human mesenchymal stem cells (MSCs) are subjected to osteoblastic differentiation in the presence of BMP-2. Here we demonstrate that overexpression of deltaEF1 in murine pre-myoblast C2C12 cells resulted in a decrease in the mRNA levels of early osteoblast marker genes induced by BMP-2 including osterix and collagen type I. This inhibitory effect was further confirmed by decreased alkaline phosphatase (ALP) activities. Neither of the zinc finger clusters of deltaEF1 is necessary for its repressive effect on BMP-2-induced osteoblastic differentiation of C2C12 cells. Immunoprecipitation results indicated that deltaEF1 did not physically associate with Smads proteins, suggesting that the inhibitory effect of deltaEF1 may be Smad-independent. deltaEF1 overexpression in C2C12 cells resulted in down-regulation of activating protein-1 (AP-1) activities promoted by BMP-2. Moreover, deltaEF1 exhibited transrepression on murine osteocalcin gene which effect is partially mediated through diminishing of AP-1 signaling. These results suggest that deltaEF1 acts as a potent inhibitor of BMP-2-induced osteogenesis in vitro, in part, by differentially regulating the AP-1 signaling pathway.
Collapse
Affiliation(s)
- Shuang Yang
- Medical College of Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Wang J, Scully K, Zhu X, Cai L, Zhang J, Prefontaine GG, Krones A, Ohgi KA, Zhu P, Garcia-Bassets I, Liu F, Taylor H, Lozach J, Jayes FL, Korach KS, Glass CK, Fu XD, Rosenfeld MG. Opposing LSD1 complexes function in developmental gene activation and repression programmes. Nature 2007; 446:882-7. [PMID: 17392792 DOI: 10.1038/nature05671] [Citation(s) in RCA: 441] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Accepted: 02/05/2007] [Indexed: 12/16/2022]
Abstract
Precise control of transcriptional programmes underlying metazoan development is modulated by enzymatically active co-regulatory complexes, coupled with epigenetic strategies. One thing that remains unclear is how specific members of histone modification enzyme families, such as histone methyltransferases and demethylases, are used in vivo to simultaneously orchestrate distinct developmental gene activation and repression programmes. Here, we report that the histone lysine demethylase, LSD1--a component of the CoREST-CtBP co-repressor complex--is required for late cell-lineage determination and differentiation during pituitary organogenesis. LSD1 seems to act primarily on target gene activation programmes, as well as in gene repression programmes, on the basis of recruitment of distinct LSD1-containing co-activator or co-repressor complexes. LSD1-dependent gene repression programmes can be extended late in development with the induced expression of ZEB1, a Krüppel-like repressor that can act as a molecular beacon for recruitment of the LSD1-containing CoREST-CtBP co-repressor complex, causing repression of an additional cohort of genes, such as Gh, which previously required LSD1 for activation. These findings suggest that temporal patterns of expression of specific components of LSD1 complexes modulate gene regulatory programmes in many mammalian organs.
Collapse
Affiliation(s)
- Jianxun Wang
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, 9500 Gilman Drive, Room 345, La Jolla, California 92093-0648, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Morshed M, Ando M, Yamamoto J, Hotta A, Kaneoka H, Kojima J, Nishijima KI, Kamihira M, Iijima S. YY1 binds to regulatory element of chicken lysozyme and ovalbumin promoters. Cytotechnology 2006; 52:159-70. [PMID: 19002874 DOI: 10.1007/s10616-006-9017-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Accepted: 07/24/2006] [Indexed: 11/25/2022] Open
Abstract
Chicken lysozyme is highly expressed in the oviduct. The 5' regulatory region of this gene contains a negative element that represses transcription. To assess the molecular basis underlying the regulation of lysozyme gene expression, we investigated the binding protein to this region. Sequence motif analysis suggested the existence of putative YY1 binding sites in this regulatory region. Electrophoretic mobility shift assay showed the specific binding of YY1 to the negative element. In addition, chromatin immunoprecipitation assay indicated that YY1 specifically bound to the negative element in oviduct cells but not in erythrocytes. It was suggested by electrophoretic mobility shift assay and chromatin immunoprecipitation assay that YY1 also bound to the negative regulatory region in the promoter of the ovalbumin gene which also shows oviduct-specific expression. Western blot analysis showed that YY1 was expressed in relatively high levels in the oviduct and nucleus fractionation experiments showed that YY1 was localized both in chromosome and nuclear matrix fractions. These results suggest that there are some specific roles in the negative regulatory regions of these genes in relation to the multifunctional transcription factor YY1.
Collapse
Affiliation(s)
- Mahboob Morshed
- Department of Biotechnology, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Park HM, Sanders MM, Suzuki T, Muramatsu T. An oviduct-specific and enhancer-like element resides at about -3000 in the chicken ovalbumin gene. Biochimie 2006; 88:1909-14. [PMID: 16916570 DOI: 10.1016/j.biochi.2006.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Accepted: 06/25/2006] [Indexed: 11/30/2022]
Abstract
The chicken ovalbumin (Ov) gene is one of the best models to study tissue-specific gene regulation because it is only expressed in the oviduct. In this paper, a tissue-specific element was characterized by 5'-flanking region deletion in combination with in vivo gene electroporation (EP). Plasmids with varying lengths of truncated Ov 5'-flanking region fused to the Renilla luciferase reporter gene were transfected in vivo into oviduct, muscle, and pancreas. A chicken oviduct-specific and enhancer-like region (designated COSE) was implicated between -3100 and -2800. The COSE showed up-regulation of gene expression in oviduct, but not in muscle or in pancreas. The COSE region was further characterized by gel mobility shift assays using nuclear extracts from oviduct, pancreas and liver. With the region from -2900 to -2851, designated T2, there were two distinct protein-DNA complexes: one found only in oviduct extract and the other detected only in pancreas and liver. These data suggest a model where the regulation of Ov gene expression in the oviduct and non-oviduct tissues is exerted at least in part by the presence of protein modulators that bind to the COSE element.
Collapse
Affiliation(s)
- Hyi-Man Park
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
27
|
Spoelstra NS, Manning NG, Higashi Y, Darling D, Singh M, Shroyer KR, Broaddus RR, Horwitz KB, Richer JK. The transcription factor ZEB1 is aberrantly expressed in aggressive uterine cancers. Cancer Res 2006; 66:3893-902. [PMID: 16585218 DOI: 10.1158/0008-5472.can-05-2881] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The transcription factor ZEB1 (deltaEF1 in mice) has been implicated in cellular processes during development and tumor progression including epithelial to mesenchymal transition. deltaEF1 null mice die at birth, but heterozygotes expressing a LacZ reporter inserted into the deltaEF1 gene live and reproduce. Using these mice, we observed ZEB1 promoter activity in the virgin myometrium, and stroma and myometrium of the pregnant uterus. ZEB1 protein is up-regulated in the myometrium and endometrial stroma after progesterone or estrogen treatment of ovariectomized mice. In the normal human uterus, ZEB1 protein is increased in the myometrium and stroma during the secretory stage of the menstrual cycle. ZEB1 is not expressed in the normal endometrial epithelium. In malignancies of the uterus, we find that ZEB1 (a) is overexpressed in malignant tumors derived from the myometrium (leiomyosarcomas), (b) is overexpressed in tumor-associated stroma of low-grade endometrioid adenocarcinomas, and (c) is aberrantly expressed in the tumor epithelial cells of aggressive endometrial cancers. Specifically, in grade 3 endometrioid adenocarcinomas and uterine papillary serous carcinomas, ZEB1 could be expressed in the epithelial-derived carcinoma cells as well as in the stroma. In malignant mixed Müllerian tumors, the sarcomatous component always expresses ZEB1, and the carcinomatous component can also be positive. In summary, ZEB1 is normally regulated by both estrogen and progesterone receptors, but in uterine cancers, it is likely no longer under control of steroid hormone receptors and becomes aberrantly expressed in epithelial-derived tumor cells, supporting a role for ZEB1 in epithelial to mesenchymal transitions associated with aggressive tumors.
Collapse
Affiliation(s)
- Nicole S Spoelstra
- Department of Medicine, Division of Endocrinology, University of Colorado Health Sciences Center at Fitzsimons, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Dougherty DC, Sanders MM. Estrogen action: revitalization of the chick oviduct model. Trends Endocrinol Metab 2005; 16:414-9. [PMID: 16202618 DOI: 10.1016/j.tem.2005.09.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 08/23/2005] [Accepted: 09/21/2005] [Indexed: 11/26/2022]
Abstract
Despite decades of investigation, the molecular pathways triggered by estrogen that lead to tissue-specific cell proliferation, differentiation and survival are only superficially understood. If we are to modulate the actions of estrogen selectively in these processes, continued investigation using biologically relevant models is essential. The chick oviduct emerged as an early model for investigating the mechanism of action of steroid hormones because of its exquisite responsiveness to them. Unfortunately, because of experimental limitations, this model has been neglected in the past decade. Reviving this model has become intellectually attractive and technically feasible.
Collapse
Affiliation(s)
- Dawne C Dougherty
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church St. SE, Minneapolis, MN 55455, USA
| | | |
Collapse
|
29
|
Frigeri C, Martin CC, Svitek CA, Oeser JK, Hutton JC, Gannon M, O'Brien RM. The proximal islet-specific glucose-6-phosphatase catalytic subunit-related protein autoantigen promoter is sufficient to initiate but not maintain transgene expression in mouse islets in vivo. Diabetes 2004; 53:1754-64. [PMID: 15220199 DOI: 10.2337/diabetes.53.7.1754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We have previously reported the discovery of an islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) that is predominantly expressed in islet beta-cells. IGRP has recently been identified as a major autoantigen in a mouse model of type 1 diabetes. The analysis of IGRP-chloramphenicol acetyltransferase (CAT) fusion gene expression in transiently transfected islet-derived hamster insulinoma tumor and betaTC-3 cells revealed that the promoter region located between -306 and +3 confers high-level reporter gene expression. To determine whether this same promoter region is sufficient to confer islet beta-cell-specific gene expression in vivo, it was ligated to a beta-galactosidase reporter gene, and transgenic mice expressing the resulting fusion gene were generated. In two independent founder lines, this -306 to +3 promoter region was sufficient to drive beta-galactosidase expression in newborn mouse islets, predominantly in beta-cells, which was initiated during the expected time in development, around embryonic day 12.5. However, unlike the endogenous IGRP gene, beta-galactosidase expression was also detected in the cerebellum. Moreover, beta-galactosidase expression was almost completely absent in adult mouse islets, suggesting that cis-acting elements elsewhere in the IGRP gene are required for determining appropriate IGRP tissue-specific expression and for the maintenance of IGRP gene expression in adult mice.
Collapse
Affiliation(s)
- Claudia Frigeri
- Department of Molecular Physiology and Biophysics, 761 PRB, Vanderbilt University Medical School, Nashville, TN 37232-0615, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Dillner NB, Sanders MM. Transcriptional Activation by the Zinc-Finger Homeodomain ProteinδEF1 in Estrogen Signaling Cascades. DNA Cell Biol 2004; 23:25-34. [PMID: 14965470 DOI: 10.1089/104454904322745907] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The transcription factor delta EF1 is a key player in estrogen-signaling cascades in vertebrates. In this pathway, estrogen induces the expression of the gene encoding delta EF1, and then delta EF1 activates transcription of downstream targets. Yet, the molecular mechanisms of transcriptional activation by delta EF1 have remained obscure. Furthermore, most work has concentrated on the capacity of delta EF1 to repress gene expression, rather than its ability to activate transcription. To investigate this activation potential in an endogenous signaling pathway, we characterized ovalbumin (Ov) gene induction by delta EF1. Gel mobility shift assays demonstrate that delta EF1 binds to the 5' flanking region of the Ov gene at two sites, one at -810 to -806 and one at -152 to -148 with respect to the start point of transcription. Correspondingly, these sites are required for induction by estrogen or by delta EF1 in transfection experiments. Furthermore, the activation potential of delta EF1 is not restricted to the chick homolog, as the human ZEB and the mouse delta EF1 homologs also induce Ov gene expression. To characterize the molecular mechanisms whereby delta EF1 activates gene expression, its C-terminal acidic domain was deleted and shown to be necessary for activation of transcription. Furthermore, the acidic domain has intrinsic activation potential, as it can induce the heterologous thymidine kinase promoter. These data establish delta EF1 as an activator of transcription whose action may be DNA-context and cell-type specific, but not species specific.
Collapse
Affiliation(s)
- Naomi B Dillner
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | | |
Collapse
|
31
|
van Grunsven LA, Michiels C, Van de Putte T, Nelles L, Wuytens G, Verschueren K, Huylebroeck D. Interaction between Smad-interacting protein-1 and the corepressor C-terminal binding protein is dispensable for transcriptional repression of E-cadherin. J Biol Chem 2003; 278:26135-45. [PMID: 12714599 DOI: 10.1074/jbc.m300597200] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
deltaEF1 and SIP1 (or Zfhx1a and Zfhx1b, respectively) are the only known members of the vertebrate Zfh1 family of homeodomain/zinc finger-containing proteins. Similar to other transcription factors, both Smad-interacting protein-1 (SIP1) and deltaEF1 are capable of repressing E-cadherin transcription through binding to the E2 boxes located in its promoter. In the case of deltaEF1, this repression has been proposed to occur via interaction with the corepressor C-terminal binding protein (CtBP). In this study, we show by coimmunoprecipitation that SIP1 and CtBP interact in vivo and that an isolated CtBP-binding SIP1 fragment depends on CtBP for transcriptional repression. However, and most importantly, full-length SIP1 and deltaEF1 proteins do not depend on their interaction with CtBP to repress transcription from the E-cadherin promoter. Furthermore, in E-cadherin-positive kidney epithelial cells, the conditional synthesis of mutant SIP1 that cannot bind to CtBP abrogates endogenous E-cadherin expression in a similar way as wild-type SIP1. Our results indicate that full-length SIP1 can repress E-cadherin in a CtBP-independent manner.
Collapse
Affiliation(s)
- Leo A van Grunsven
- Department of Developmental Biology (VIB7), Flanders Interuniversity Institute for Biotechnology (VIB) and Laboratory of Molecular Biology (Celgen), University of Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
32
|
Postigo AA. Opposing functions of ZEB proteins in the regulation of the TGFbeta/BMP signaling pathway. EMBO J 2003; 22:2443-52. [PMID: 12743038 PMCID: PMC155983 DOI: 10.1093/emboj/cdg225] [Citation(s) in RCA: 228] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Binding of TGFbeta/BMP factors to their receptors leads to translocation of Smad proteins to the nucleus where they activate transcription of target genes. The two-handed zinc finger proteins encoded by Zfhx1a and Zfhx1b, ZEB-1/deltaEF1 and ZEB-2/SIP1, respectively, regulate gene expression and differentiation programs in a number of tissues. Here I demonstrate that ZEB proteins are also crucial regulators of TGFbeta/BMP signaling with opposing effects on this pathway. Both ZEB proteins bind to Smads, but while ZEB-1/deltaEF1 synergizes with Smad proteins to activate transcription, promote osteoblastic differentiation and induce cell growth arrest, the highly related ZEB-2/SIP1 protein has the opposite effect. Finally, the ability of TGFbeta to mediate transcription of TGFbeta-dependent genes and induce growth arrest depends on the presence of endogenous ZEB-1/deltaEF1 protein.
Collapse
Affiliation(s)
- Antonio A Postigo
- Division of Molecular Oncology, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA.
| |
Collapse
|
33
|
Postigo AA, Depp JL, Taylor JJ, Kroll KL. Regulation of Smad signaling through a differential recruitment of coactivators and corepressors by ZEB proteins. EMBO J 2003; 22:2453-62. [PMID: 12743039 PMCID: PMC155984 DOI: 10.1093/emboj/cdg226] [Citation(s) in RCA: 293] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2002] [Revised: 03/18/2003] [Accepted: 03/18/2003] [Indexed: 11/14/2022] Open
Abstract
Balancing signals derived from the TGFbeta family is crucial for regulating cell proliferation and differentiation, and in establishing the embryonic axis during development. TGFbeta/BMP signaling leads to the activation and nuclear translocation of Smad proteins, which activate transcription of specific target genes by recruiting P/CAF and p300. The two members of the ZEB family of zinc finger factors (ZEB-1/deltaEF1 and ZEB-2/SIP1) regulate TGFbeta/BMP signaling in opposite ways: ZEB-1/deltaEF1 synergizes with Smad-mediated transcriptional activation, while ZEB-2/SIP1 represses it. Here we report that these antagonistic effects by the ZEB proteins arise from the differential recruitment of transcriptional coactivators (p300 and P/CAF) and corepressors (CtBP) to the Smads. Thus, while ZEB-1/deltaEF1 binds to p300 and promotes the formation of a p300-Smad transcriptional complex, ZEB-2/SIP1 acts as a repressor by recruiting CtBP. This model of regulation by ZEB proteins also functions in vivo, where they have opposing effects on the regulation of TGFbeta family-dependent genes during Xenopus development.
Collapse
Affiliation(s)
- Antonio A Postigo
- Division of Molecular Oncology, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | | | | | |
Collapse
|
34
|
Kraus RJ, Perrigoue JG, Mertz JE. ZEB negatively regulates the lytic-switch BZLF1 gene promoter of Epstein-Barr virus. J Virol 2003; 77:199-207. [PMID: 12477825 PMCID: PMC140584 DOI: 10.1128/jvi.77.1.199-207.2003] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epstein-Barr virus (EBV) is a human herpesvirus capable of establishing a latent state in B lymphocytes. The product of the immediate-early BZLF1 gene, Zta, is a transcriptional transactivator essential for viral DNA amplification and virion production. Previously, we identified a negative cis-acting element within the BZLF1 promoter termed ZV. ZV contains the sequence 5'-CAGGTA-3' located at nucleotides -17 to -12 relative to the transcription initiation site. It sequence specifically binds a cellular factor, ZVR. Based on sequence binding specificity, we postulated that ZVR may be zinc finger E-box binding factor (ZEB) or a related zinc finger/homeodomain family member. We show here by immunoshift assays that ZVR and human ZEB specifically cross-react with an antibody to deltaEF1, the chicken homolog of ZEB. Competition electrophoretic mobility shift assays confirmed that ZEB binds to the ZV element with the same binding specificity as ZVR. Overexpression of ZEB in either B-lymphocytic DG75 cells or mammary epithelial MCF-7 cells repressed Zta-induced activation of the BZLF1 promoter four- to fivefold via the ZV site. Thus, we conclude that the previously identified cellular repressor ZVR is, in fact, ZEB. We also present evidence that other cellular factors likely affect the transcriptional activity of ZEB. Lastly, we identify a ZEB-binding site within the promoter of the lytic BRLF1 gene of EBV. We postulate that ZEB likely plays an important role in regulating the life cycle of EBV.
Collapse
Affiliation(s)
- Richard J Kraus
- McArdle Laboratory for Cancer Research, University of Wisconsin Medical School, Madison 53706-1599, USA
| | | | | |
Collapse
|
35
|
Dillner NB, Sanders MM. Upstream stimulatory factor (USF) is recruited into a steroid hormone-triggered regulatory circuit by the estrogen-inducible transcription factor delta EF1. J Biol Chem 2002; 277:33890-4. [PMID: 12107170 DOI: 10.1074/jbc.m204399200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In the past decade, investigation into steroid hormone signaling has focused on the mechanisms of steroid hormone receptors as they act as signaling molecules and transcription factors in cells. However, the majority of hormone-responsive genes are not directly regulated by hormone receptors. These genes are termed secondary response genes. To explore the molecular mechanisms by which the steroid hormone estrogen regulates secondary response genes, the ovalbumin (Ov) gene was analyzed. Three protein-protein complexes (Chirp-I, -II, -III), which do not contain the estrogen receptor, are induced by estrogen to bind to the 5'-flanking region of the Ov gene. The Chirp-III DNA binding site, which is required for estrogen induction, binds a complex of proteins that contains the estrogen-inducible transcription factor deltaEF1. Experiments undertaken to identify proteins complexed with deltaEF1 led to the elucidation of a novel mechanism of action of upstream stimulatory factor-1 (USF-1), which involves its tethering to the Ov gene 5'-flanking region by deltaEF1. Gel mobility shift assays and co-immunoprecipitation experiments identify USF-1 as a component of Chirp-III. However, USF-1 is not able to bind to the Chirp-III site independently. In addition, USF-1 overexpression is able to induce Ov gene promoter activity in transfection experiments. USF-1 can also potentiate the induction of the Ov gene by the transcription factor deltaEF1. Moreover, mutating the deltaEF1 binding sites in the 5'-flanking region of the Ov gene abrogates induction of the gene by USF-1. These data begin to establish a molecular mechanism by which hormone-inducible transcription factors and ubiquitous transcription factors cooperate to regulate estrogen-induced secondary response gene expression.
Collapse
Affiliation(s)
- Naomi B Dillner
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street, Minneapolis, MN 55455, USA
| | | |
Collapse
|
36
|
Costantino ME, Stearman RP, Smith GE, Darling DS. Cell-specific phosphorylation of Zfhep transcription factor. Biochem Biophys Res Commun 2002; 296:368-73. [PMID: 12163027 PMCID: PMC3682420 DOI: 10.1016/s0006-291x(02)00880-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Zinc finger homeodomain enhancer-binding protein (Zfhep/Zfhx1a) is a transcription factor essential for immune system development, skeletal patterning, and life. Regulation of the interleukin-2 gene in T cells has been suggested to depend on post-translational processing of Zfhep, however, no modifications of Zfhep are known. Here we demonstrate that Zfhep is present in both hyperphosphorylated and hypophosphorylated forms. Western blot analysis demonstrates two forms of Zfhep with different mobilities. Differences in phosphorylation are sufficient to explain the difference in mobilities. Zfhep is primarily phosphorylated on Ser and Thr residues since PP2A dephosphorylates the slower mobility band. Treatment of nuclear extract with O-GlcNAcase did not detect O-linked sugar. Importantly, post-translational processing is cell-specific. Doublets of Zfhep were detected in five cell lines, whereas 6 cell lines contain only, or predominantly, non-phosphorylated Zfhep, and Saos-2 cells contain predominantly the phosphorylated form. These data provide the first demonstration that Zfhep is post-translationally modified.
Collapse
Affiliation(s)
- Mary E. Costantino
- Biochemistry and Molecular Biology, University of Louisville Health Sciences Center, Louisville, KY 40292
| | - Randi P. Stearman
- Periodontics, Endodontics and Dental Hygiene, University of Louisville Health Sciences Center, Louisville, KY 40292
| | - Gregory E. Smith
- Biochemistry and Molecular Biology, University of Louisville Health Sciences Center, Louisville, KY 40292
| | - Douglas S. Darling
- Biochemistry and Molecular Biology, University of Louisville Health Sciences Center, Louisville, KY 40292
- Periodontics, Endodontics and Dental Hygiene, University of Louisville Health Sciences Center, Louisville, KY 40292
- To whom correspondence should be addressed at University of Louisville School of Dentistry 501 South Preston St., Room 315 Louisville, KY 40292, Tel: (502) 852-5508, FAX: (502) 852-1317,
| |
Collapse
|
37
|
Dillner NB, Sanders MM. The zinc finger/homeodomain protein deltaEF1 mediates estrogen-specific induction of the ovalbumin gene. Mol Cell Endocrinol 2002; 192:85-91. [PMID: 12088870 DOI: 10.1016/s0303-7207(02)00088-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Regulation of the ovalbumin (Ov) gene is strictly controlled by precise developmental, tissue-specific, and hormonal cues. The Ov gene is transcriptionally activated by four classes of steroid hormones: estrogens, androgens, glucocorticoids, and progestins. Although it has served as a model to study multi-hormone gene regulation for the past 30 years, the pathways that relay each hormone signal to the Ov gene are largely unclear. Extensive linker-scanner and point mutation analysis has revealed elements necessary for its induction by estrogen, androgen, progesterone, or glucocorticoid but has failed to identify any elements that are specific to the action of any one steroid hormone. These observations in conjunction with the observation that the Ov gene is indirectly regulated by steroid hormones suggest that these signals may all induce the same transcription factor. However, here we have identified two cis-acting DNA elements in the 5' flanking region of the Ov gene that are required for induction by estrogen, but not by androgen or progesterone. These elements span -152 to -146 and -810 to -806 with respect to the start point of transcription. This implies that estrogen induces the Ov gene by a separate pathway than do androgens or progestins. Gel mobility shift assays demonstrate that the estrogen-specific sequences bind the estrogen inducible transcription factor deltaEF1, suggesting that deltaEF1 plays a distinct role in mediating the estrogen signal to the Ov gene.
Collapse
Affiliation(s)
- Naomi B Dillner
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA
| | | |
Collapse
|
38
|
Kraus RJ, Mirocha SJ, Stephany HM, Puchalski JR, Mertz JE. Identification of a novel element involved in regulation of the lytic switch BZLF1 gene promoter of Epstein-Barr virus. J Virol 2001; 75:867-77. [PMID: 11134300 PMCID: PMC113983 DOI: 10.1128/jvi.75.2.867-877.2001] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Epstein-Barr virus (EBV) is a human herpesvirus capable of establishing a latent state in B lymphocytes. EBV's BZLF1 gene product plays a central role in regulating the switch from latency to productive infection. Here, we identify a sequence element, 5'-CAGGTA-3', called ZV, located at nucleotides -17 to -12 relative to the transcription initiation site of the BZLF1 promoter. ZV sequence-specifically binds a cellular nuclear factor(s), ZVR. ZVR DNA-binding activity was present in the EBV-negative B-lymphocytic cell line DG75, the EBV-positive B-lymphocytic cell lines GG68 and 721, the cervical cell line C33A, and the kidney cell line CV-1 but not in the breast carcinoma cell line MCF-7. Mutations in ZV that relieve binding of ZVR lead to a two- to fourfold increase in basal expression of the BZLF1 promoter in DG75, C33A, and CV-1 cells. The same mutants exhibited a 40- to 180-fold increase in tetradecanoyl phorbol acetate-ionomycin-induced expression in DG75 cells and a 22-fold increase in C33A cells. Thus, ZVR functions as a regulator of the BZLF1 promoter, repressing transcription when bound to the ZV site in the absence of inducers. No differences in basal or induced transcription between wild-type and ZV mutant BZLF1 promoters were observed in ZVR-negative MCF-7 cells. ZVR failed to bind any of the previously identified negative regulatory elements within the BZLF1 promoter. We conclude that ZV functions as an important regulatory element of the BZLF1 promoter, with ZVR likely playing important roles in the maintenance of latency and reactivation of EBV.
Collapse
Affiliation(s)
- R J Kraus
- McArdle Laboratory for Cancer Research, University of Wisconsin Medical School, Madison, Wisconsin 53706-1599, USA
| | | | | | | | | |
Collapse
|
39
|
Dean DM, Jones PS, Sanders MM. Alterations in chromatin structure are implicated in the activation of the steroid hormone response unit of the ovalbumin gene. DNA Cell Biol 2001; 20:27-39. [PMID: 11242541 DOI: 10.1089/10445490150504675] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Hormone-responsive genes rely on complex regulatory elements known as hormone response units to integrate various regulatory signals. Characterization of the steroid-dependent regulatory element (SDRE) in the check ovalbumin gene (--892 to --796) suggests that it functions as a hormone response unit. Previous studies using gel mobility shift assays and several types of footprinting analyses demonstrated that proteins bind to this entire element in vitro even in the absence of steroid hormones. However, the genomic footprinting experiments described herein indicate that the binding of three different proteins or protein complexes to the SDRE requires estrogen and corticosterone, suggesting that the chromatin structure of this site is restricted in vivo. Transfection experiments using linker scanning and point mutations support the contention that the binding of these three complexes is essential for induction of the ovalbumin gene by steroid hormones. In addition, functional analyses suggest that a fourth complex is also necessary for maximal induction. These and other data suggest that the SDRE functions as a hormone response unit to coordinate signals generated by two steroid hormones.
Collapse
Affiliation(s)
- D M Dean
- Department of Chemistry, St. Joseph's College, West Hartford, Connecticut, USA
| | | | | |
Collapse
|
40
|
Hagen SG, Monroe DG, Dean DM, Sanders MM. Repression of chick multidrug resistance-associated protein 1 (chMRP1) gene expression by estrogen. Gene 2000; 257:243-9. [PMID: 11080590 DOI: 10.1016/s0378-1119(00)00403-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Although a number of genes have been identified whose transcriptional activities are stimulated by estrogen, relatively few have been discovered that are repressed. In an effort to determine whether estrogen can directly repress gene expression, attempts were made to identify genes that are direct targets of the estrogen receptor and whose activities are repressed by it. Because the development and differentiation of the chick oviduct are exquisitely dependent upon estrogen, this seemed an appropriate model system for testing this hypothesis. RNA was isolated from estrogen-treated and estrogen-withdrawn chick oviducts and was subjected to differential display analysis. Surprisingly, one of the products repressed by estrogen encoded the chick homolog of the multidrug resistance-associated protein 1 (MRP1) gene. Further cloning resulted in a chick MRP1 (chMRP1) cDNA clone that is 72% identical with human MRP1. Translation of the chMRP1 sequence indicates a 77% amino acid identity with both the human and mouse MRP1 proteins. Treatment of estrogen-withdrawn chicks with 17beta-estradiol decreased chMRP1 mRNA levels to 50% within 30 min and to 70% by 1h, which is comparable to the level observed with chronic repression by estrogen. ChMRP1 mRNA is present in many other tissues, including the heart, lung, brain, kidney, skeletal muscle, and intestine, but is undetectable in the liver. This study indicates that in estrogen-responsive tissues such as chick oviduct, the regulation of chMRP1 gene expression is controlled by estrogen.
Collapse
Affiliation(s)
- S G Hagen
- University of Minnesota, Department of Biochemistry, Molecular Biology and Biophysics, 6-155 Jackson Hall 321 Church St., SE Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
41
|
Monroe DG, Jin DF, Sanders MM. Estrogen opposes the apoptotic effects of bone morphogenetic protein 7 on tissue remodeling. Mol Cell Biol 2000; 20:4626-34. [PMID: 10848589 PMCID: PMC85866 DOI: 10.1128/mcb.20.13.4626-4634.2000] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interactions between estrogen and growth factor signaling pathways at the level of gene expression play important roles in the function of reproductive tissues. For example, estrogen regulates transforming growth factor beta (TGFbeta) in the uterus during the proliferative phase of the mammalian reproductive cycle. Bone morphogenetic protein 7 (BMP-7), a member of the TGFbeta superfamily, is also involved in the development and function of reproductive tissues. However, relatively few studies have addressed the expression of BMP-7 in reproductive tissues, and the role of BMP-7 remains unclear. As part of an ongoing effort to understand how estrogen represses gene expression and to study its interactions with other signaling pathways, chick BMP-7 (cBMP-7) was cloned. cBMP-7 mRNA levels are repressed threefold within 8 h following estrogen treatment in the chick oviduct, an extremely estrogen-responsive reproductive tissue. This regulation occurs at the transcriptional level. Estrogen has a protective role in many tissues, and withdrawal from estrogen often leads to tissue regression; however, the mechanisms mediating regression of the oviduct remain unknown. Terminal transferase-mediated end-labeling and DNA laddering assays demonstrated that regression of the oviduct during estrogen withdrawal involves apoptosis, which is a novel observation. cBMP-7 mRNA levels during estrogen withdrawal increase concurrently with the apoptotic index of the oviduct. Furthermore, addition of purified BMP-7 induces apoptosis in primary oviduct cells. This report demonstrates that the function of BMP-7 in the oviduct involves the induction of apoptosis and that estrogen plays an important role in opposing this function.
Collapse
Affiliation(s)
- D G Monroe
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
42
|
Berger RR, Sanders MM. Estrogen modulates HNF-3beta mRNA levels in the developing chick oviduct. DNA Cell Biol 2000; 19:103-12. [PMID: 10701776 DOI: 10.1089/104454900314618] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Steroid hormones are involved in many physiological processes, including tissue-specific gene expression, homeostasis, and development. The chick oviduct represents an excellent system in which to study many of these events, as it is highly steroid responsive. Here, we report the cloning of chick HNF-3beta from an oviduct cDNA library and its expression pattern in adult tissues and in the developing oviduct in response to estrogen treatment. Overall, cHNF-3beta was expressed at high levels in the immature chick oviduct and lung and, to a lesser extent, in the liver, kidney, and muscle. This expression pattern is divergent from that of mammalian HNF-3beta, which is not expressed in kidney or muscle. Furthermore, several lengths of cHNF-3beta mRNA transcripts were detected that were expressed tissue specifically. Interestingly, cHNF-3beta mRNA levels were differentially influenced by estrogen as a result of a post-transcriptional effect on the cHNF-3beta message in some tissues. Finally, a role for cHNF-3beta is proposed in the estrogen-stimulated differentiation and development of the oviduct, as cHNF-3beta mRNA expression is induced in the early stages of oviduct development and declines as the animal becomes sexually mature.
Collapse
Affiliation(s)
- R R Berger
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis 55455, USA
| | | |
Collapse
|