1
|
Chen SY, Liu FC. The Fgf9-Nolz1-Wnt2 axis regulates morphogenesis of the lung. Development 2023; 150:dev201827. [PMID: 37497597 DOI: 10.1242/dev.201827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023]
Abstract
Morphological development of the lung requires complex signal crosstalk between the mesenchymal and epithelial progenitors. Elucidating the genetic cascades underlying signal crosstalk is essential to understanding lung morphogenesis. Here, we identified Nolz1 as a mesenchymal lineage-specific transcriptional regulator that plays a key role in lung morphogenesis. Nolz1 null mutation resulted in a severe hypoplasia phenotype, including a decreased proliferation of mesenchymal cells, aberrant differentiation of epithelial cells and defective growth of epithelial branches. Nolz1 deletion also downregulated Wnt2, Lef1, Fgf10, Gli3 and Bmp4 mRNAs. Mechanistically, Nolz1 regulates lung morphogenesis primarily through Wnt2 signaling. Loss-of-function and overexpression studies demonstrated that Nolz1 transcriptionally activated Wnt2 and downstream β-catenin signaling to control mesenchymal cell proliferation and epithelial branching. Exogenous Wnt2 could rescue defective proliferation and epithelial branching in Nolz1 knockout lungs. Finally, we identified Fgf9 as an upstream regulator of Nolz1. Collectively, Fgf9-Nolz1-Wnt2 signaling represents a novel axis in the control of lung morphogenesis. These findings are relevant to lung tumorigenesis, in which a pathological function of Nolz1 is implicated.
Collapse
Affiliation(s)
- Shih-Yun Chen
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Fu-Chin Liu
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
2
|
Shan H, Ren K, Liu J, Rehman SU, Yan X, Ma X, Zheng Y, Feng T, Wang X, Li Z, Zhou W, Chuang C, Liang M, Zheng J, Liu Q. Comprehensive Transcriptome Sequencing Analysis of Hirudinaria manillensis in Different Growth Periods. Front Physiol 2022; 13:897458. [PMID: 35694407 PMCID: PMC9174698 DOI: 10.3389/fphys.2022.897458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/22/2022] [Indexed: 02/04/2023] Open
Abstract
Medical leeches are widely been used in biochemical and clinical medical studies, helping to restore blood circulation to grafted or severely injured tissue. Mostly, adult leeches are being used in the traditional pharmacopeia, but the gene expression profiling of leeches in different growth periods is not well-reported. So, in this study, we used transcriptome analysis to analyze the comparative gene expression patterns of Hirudinaria manillensis (H. manillensis) in different growth periods, including larval, young, and adult stages. We constructed 24 cDNA libraries from H. manillensis larval, young, and adult stages, and about 54,639,118 sequences were generated, 18,106 mRNA transcripts of which 958 novel mRNAs and 491 lncRNAs were also assembled as well. Furthermore, the results of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that the differentially upregulated genes from the larval to adult stages were enriched in pathways such as cilium, myofibril, contractile fiber, cytoskeleton proteins, dilated cardiomyopathy, adrenergic signaling in cardiomyocytes, etc. Moreover, in the adult stages, a significant increase in the expression of the Hirudin-HM (HIRM2) genes was detected. In addition, our comparative transcriptome profiling data from different growth stages of H. manillensis also identified a large number of DEGs and DElncRNAs which were tentatively found to be associated with the growth of H. manillensis; as it grew, the muscle-related gene expression increased, while the lipid metabolism and need for stimulation and nutrition-related genes decreased. Similarly, the higher expression of HIRM2 might attribute to the high expression of protein disulfide isomerase gene family (PDI) family genes in adulthood, which provides an important clue that why adult leeches rather than young leeches are widely used in clinical therapeutics and traditional Chinese medicine.
Collapse
Affiliation(s)
- Huiquan Shan
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Ke Ren
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Jiasheng Liu
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Saif ur Rehman
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Xiuying Yan
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Xiaocong Ma
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Yalin Zheng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Tong Feng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Xiaobo Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Zhipeng Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro- Bioresources, Guangxi University, Nanning, China
| | - Weiguan Zhou
- THAI Natural Hirudin Co., Ltd., Bangkok, Thailand
| | - Chen Chuang
- Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mingkun Liang
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Jinghui Zheng
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
- *Correspondence: Jinghui Zheng, ; Qingyou Liu,
| | - Qingyou Liu
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan, China
- *Correspondence: Jinghui Zheng, ; Qingyou Liu,
| |
Collapse
|
3
|
Quan H, Arsala D, Lynch JA. Transcriptomic and functional analysis of the oosome, a unique form of germ plasm in the wasp Nasonia vitripennis. BMC Biol 2019; 17:78. [PMID: 31601213 PMCID: PMC6785909 DOI: 10.1186/s12915-019-0696-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/30/2019] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND The oosome is the germline determinant in the wasp Nasonia vitripennis and is homologous to the polar granules of Drosophila. Despite a common evolutionary origin and developmental role, the oosome is morphologically quite distinct from polar granules. It is a solid sphere that migrates within the cytoplasm before budding out and forming pole cells. RESULTS To gain an understanding of both the molecular basis of oosome development and the conserved essential features of germ plasm, we quantified and compared transcript levels between embryo fragments that contained the oosome and those that did not. The identity of the differentially localized transcripts indicated that Nasonia uses a distinct set of molecules to carry out conserved germ plasm functions. In addition, functional testing of a sample of localized transcripts revealed potentially novel mechanisms of ribonucleoprotein assembly and pole cell cellularization in the wasp. CONCLUSIONS Our results demonstrate that the composition of germ plasm varies significantly within Holometabola, as very few mRNAs share localization to the oosome and polar granules. Some of this variability appears to be related to the unique properties of the oosome relative to the polar granules in Drosophila, and some may be related to differences in pole formation between species. This work will serve as the basis for further investigation into the patterns of germline determinant evolution among insects, the molecular basis of the unique properties of the oosome, and the incorporation of novel components into developmental networks.
Collapse
Affiliation(s)
- Honghu Quan
- Department of Pathology and Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Deanna Arsala
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607 USA
| | - Jeremy A. Lynch
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607 USA
| |
Collapse
|
4
|
Abstract
Znf703 is an RAR- and Wnt-inducible transcription factor that exhibits a complex expression pattern in the developing embryo: Znf703 mRNA is found in the early circumblastoporal ring, then later throughout the neural plate and its border, and subsequently in the mid/hindbrain and somites. We show that Znf703 has a different and separable function in early mesoderm versus neural crest and placode development. Independent of its early knockdown phenotype on Gdf3 and Wnt8, Znf703 disrupts patterning of distinct neural crest migratory streams normally delineated by Sox10, Twist, and Foxd3 and inhibits otocyst formation and otic expression of Sox10 and Eya1. Furthermore, Znf703 promotes massive overgrowth of SOX2+ cells, disrupting the SoxB1 balance at the neural plate border. Despite prominent expression in other neural plate border-derived cranial and sensory domains, Znf703 is selectively absent from the otocyst, suggesting that Znf703 must be specifically cleared or down-regulated for proper otic development. We show that mutation of the putative Groucho-repression domain does not ameliorate Znf703 effects on mesoderm, neural crest, and placodes. We instead provide evidence that Znf703 requires the Buttonhead domain for transcriptional repression.
Collapse
|
5
|
ZNF703 is Overexpressed in Papillary Thyroid Carcinoma Tissues and Mediates K1 Cell Proliferation. Pathol Oncol Res 2018; 26:355-364. [DOI: 10.1007/s12253-018-0494-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022]
|
6
|
Li K, Wang J, Han J, Lan Y, Xie C, Pan S, Liu L. Overexpression of ZNF703 facilitates tumorigenesis and predicts unfavorable prognosis in patients with cholangiocarcinoma. Oncotarget 2018; 7:76108-76117. [PMID: 27764785 PMCID: PMC5342799 DOI: 10.18632/oncotarget.12627] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/27/2016] [Indexed: 01/03/2023] Open
Abstract
Background NET (NocA/Nlz, Elbow, Tlp-1) family members have recently emerged as important players in the development of human cancers. Zinc finger protein 703 (ZNF703), locating on chromosome 8 (8p11.23), a member of the NET/Nlz family of zinc finger transcription factors, had been demonstrated to be a much novel oncogene of several malignancies. This study aimed to investigate the expression of ZNF703 in cholangiocarcinoma (CCA) and attempted to elucidate its biological effects in CCA progression. Methods The correlation between ZNF703 expression and clinicopathological characteristics of CCA was evaluated through analyzing 85 cases. The biological effects of ZNF703 were investigated both in vitro and in vivo in which proliferation, migration, and invasive potential were mainly explored. Statistical software SPSS 16.0 was used for statistical analyses. Results ZNF703 was overexpressed in CCA tissues with subcellular localizations mainly in the nucleus and partly in the cytoplasm or membrane. High expression of ZNF703 was related to tumor location (P=0.002), pathological grading (P=0.024), depth of invasion (P=0.002), distant metastasis (P=0. 011) and AJCC stage (P=0.008). Both in vitro and in vivo studies demonstrated that ZNF703 could potently promote proliferation, migration and invasion throughout the progression of CCA. Conclusion ZNF703 can potently facilitate tumor growth and metastasis in many respects throughout the progression of CCA, which may act as an oncogene in CCA and can be considered as a novel potential therapeutic target.
Collapse
Affiliation(s)
- Keyu Li
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Jiabei Wang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Jihua Han
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Yaliang Lan
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Changming Xie
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Shangha Pan
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| | - Lianxin Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, 15001, China
| |
Collapse
|
7
|
Hong CS, Saint-Jeannet JP. Znf703, a novel target of Pax3 and Zic1, regulates hindbrain and neural crest development in Xenopus. Genesis 2017; 55. [PMID: 29086464 DOI: 10.1002/dvg.23082] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/27/2017] [Accepted: 10/27/2017] [Indexed: 12/31/2022]
Abstract
The transcription factors Pax3 and Zic1 are critical to specify the neural plate border and to promote neural crest formation. In a microarray screen designed to identify genes regulated by Pax3 and Zic1 in Xenopus we isolated Znf703/Nlz1 a transcriptional repressor member of the NET (NocA/Nlz, Elbow, and TLP-1) protein family. At early neurula stage znf703 is expressed in the dorsal ectoderm, spanning the neural plate and neural plate border, with an anterior boundary of expression corresponding to rhombomeres 3 and 4 (r3/r4) in the prospective hindbrain. As a bonafide target of Pax3 and Zic1, znf703 is activated by neural plate border inducing signals, and its expression depends on Pax3 and Zic1 function in the embryo. Znf703 morpholino-mediated knockdown expanded several posterior hindbrain genes, while Znf703 overexpression completely obliterated the expression of these segmental genes, signifying that the transcriptional repressor activity of Znf703 is critical to pattern the hindbrain. Furthermore, snai2 and sox10 expression was severely impaired upon manipulation of Znf703 expression levels in the embryo suggesting that Znf703 participates in neural crest formation downstream of Pax3 and Zic1 in Xenopus.
Collapse
Affiliation(s)
- Chang-Soo Hong
- Department of Biological Sciences, College of Natural Sciences, Daegu University, Gyeongsan, Republic of Korea.,Department of Basic Science & Craniofacial Biology, College of Dentistry, New York University, New York
| | - Jean-Pierre Saint-Jeannet
- Department of Basic Science & Craniofacial Biology, College of Dentistry, New York University, New York
| |
Collapse
|
8
|
Liao X, Lu Y, Yang J, Kuang T, Jiang L, Wang Y, Kang H, Jiang B, Zhou X, He S. Transcription factor Sp1 is necessary and functional in regulating expression of oncogene ZNF703. Genes Genomics 2017. [DOI: 10.1007/s13258-017-0577-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
9
|
Lonsdale Z, Lee K, Kiriakidu M, Amarasinghe H, Nathanael D, O’Connor CJ, Mallon EB. Allele specific expression and methylation in the bumblebee, Bombus terrestris. PeerJ 2017; 5:e3798. [PMID: 28929021 PMCID: PMC5600721 DOI: 10.7717/peerj.3798] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/21/2017] [Indexed: 12/29/2022] Open
Abstract
The social hymenoptera are emerging as models for epigenetics. DNA methylation, the addition of a methyl group, is a common epigenetic marker. In mammals and flowering plants methylation affects allele specific expression. There is contradictory evidence for the role of methylation on allele specific expression in social insects. The aim of this paper is to investigate allele specific expression and monoallelic methylation in the bumblebee, Bombus terrestris. We found nineteen genes that were both monoallelically methylated and monoallelically expressed in a single bee. Fourteen of these genes express the hypermethylated allele, while the other five express the hypomethylated allele. We also searched for allele specific expression in twenty-nine published RNA-seq libraries. We found 555 loci with allele-specific expression. We discuss our results with reference to the functional role of methylation in gene expression in insects and in the as yet unquantified role of genetic cis effects in insect allele specific methylation and expression.
Collapse
Affiliation(s)
- Zoë Lonsdale
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Kate Lee
- Bioinformatics and Biostatistics Support Hub (B/BASH), University of Leicester, Leicester, United Kingdom
| | - Maria Kiriakidu
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Harindra Amarasinghe
- Academic Unit of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Despina Nathanael
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | | | - Eamonn B. Mallon
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
10
|
ZNF503/ Zpo2 drives aggressive breast cancer progression by down-regulation of GATA3 expression. Proc Natl Acad Sci U S A 2017; 114:3169-3174. [PMID: 28258171 DOI: 10.1073/pnas.1701690114] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The transcription factor GATA3 is the master regulator that drives mammary luminal epithelial cell differentiation and maintains mammary gland homeostasis. Loss of GATA3 is associated with aggressive breast cancer development. We have identified ZNF503/ZEPPO2 zinc-finger elbow-related proline domain protein 2 (ZPO2) as a transcriptional repressor of GATA3 expression and transcriptional activity that induces mammary epithelial cell proliferation and breast cancer development. We show that ZPO2 is recruited to GATA3 promoter in association with ZBTB32 (Repressor of GATA, ROG) and that ZBTB32 is essential for down-regulation of GATA3 via ZPO2. Through this modulation of GATA3 activity, ZPO2 promotes aggressive breast cancer development. Our data provide insight into a mechanism of GATA3 regulation, and identify ZPO2 as a possible candidate gene for future diagnostic and therapeutic strategies.
Collapse
|
11
|
Evolution of the NET (NocA, Nlz, Elbow, TLP-1) protein family in metazoans: insights from expression data and phylogenetic analysis. Sci Rep 2016; 6:38383. [PMID: 27929068 PMCID: PMC5144077 DOI: 10.1038/srep38383] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 10/24/2016] [Indexed: 02/03/2023] Open
Abstract
The NET (for NocA, Nlz, Elbow, TLP-1) protein family is a group of conserved zinc finger proteins linked to embryonic development and recently associated with breast cancer. The members of this family act as transcriptional repressors interacting with both class I histone deacetylases and Groucho/TLE co-repressors. In Drosophila, the NET family members Elbow and NocA are vital for the development of tracheae, eyes, wings and legs, whereas in vertebrates ZNF703 and ZNF503 are important for the development of the nervous system, eyes and limbs. Despite the relevance of this protein family in embryogenesis and cancer, many aspects of its origin and evolution remain unknown. Here, we show that NET family members are present and expressed in multiple metazoan lineages, from cnidarians to vertebrates. We identified several protein domains conserved in all metazoan species or in specific taxonomic groups. Our phylogenetic analysis suggests that the NET family emerged in the last common ancestor of cnidarians and bilaterians and that several rounds of independent events of gene duplication occurred throughout evolution. Overall, we provide novel data on the expression and evolutionary history of the NET family that can be relevant to understanding its biological role in both normal conditions and disease.
Collapse
|
12
|
Dubois L, Frendo JL, Chanut-Delalande H, Crozatier M, Vincent A. Genetic dissection of the Transcription Factor code controlling serial specification of muscle identities in Drosophila. eLife 2016; 5. [PMID: 27438571 PMCID: PMC4954755 DOI: 10.7554/elife.14979] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/11/2016] [Indexed: 12/30/2022] Open
Abstract
Each Drosophila muscle is seeded by one Founder Cell issued from terminal division of a Progenitor Cell (PC). Muscle identity reflects the expression by each PC of a specific combination of identity Transcription Factors (iTFs). Sequential emergence of several PCs at the same position raised the question of how developmental time controlled muscle identity. Here, we identified roles of Anterior Open and ETS domain lacking in controlling PC birth time and Eyes absent, No Ocelli, and Sine oculis in specifying PC identity. The windows of transcription of these and other TFs in wild type and mutant embryos, revealed a cascade of regulation integrating time and space, feed-forward loops and use of alternative transcription start sites. These data provide a dynamic view of the transcriptional control of muscle identity in Drosophila and an extended framework for studying interactions between general myogenic factors and iTFs in evolutionary diversification of muscle shapes. DOI:http://dx.doi.org/10.7554/eLife.14979.001 Animals have many different muscles of various shapes and sizes that are suited to specific tasks and behaviors. The fruit fly known as Drosophila has a fairly simple musculature, which makes it an ideal model animal to investigate how different muscles form. In fruit fly embryos, cells called progenitor cells divide to produce the cells that will go on to form the different muscles. Proteins called identity Transcription Factors are present in progenitor cells. Different combinations of identity Transcription Factors can switch certain genes on or off to control the muscle shapes in specific areas of an embryo. However, progenitor cells born in the same area but at different times display different patterns of identity Transcription Factors; this suggests that timing also influences the orientation, shape and size of a developing muscle, also known as muscle identity. Dubois et al. used a genetic screen to look for identity Transcription Factors and the roles these proteins play in muscle formation in fruit flies. Tracking the activity of these proteins revealed a precise timeline for specifying muscle identity. This timeline involves cascades of different identity Transcription Factors accumulating in the cells, which act to make sure that distinct muscle shapes are made. In flies with specific mutations, the timing of these events is disrupted, which results in muscles forming with different shapes to those seen in normal flies. The findings of Dubois et al. suggest that the timing of when particular progenitor cells form, as well as their location in the embryo, contribute to determine the shapes of muscles. The next step following on from this work is to use video-microscopy to track identity Transcription Factors when the final muscle shapes emerge. Further experiments will investigate how identity Transcription Factors work together with proteins that are directly involved in muscle development. DOI:http://dx.doi.org/10.7554/eLife.14979.002
Collapse
Affiliation(s)
- Laurence Dubois
- Centre de Biologie du Développement (CBD), CNRS and Université de Toulouse, Toulouse, France.,Centre de Biologie Intégrative (CBI), CNRS and Université de Toulouse, Toulouse, France
| | - Jean-Louis Frendo
- Centre de Biologie du Développement (CBD), CNRS and Université de Toulouse, Toulouse, France.,Centre de Biologie Intégrative (CBI), CNRS and Université de Toulouse, Toulouse, France
| | - Hélène Chanut-Delalande
- Centre de Biologie du Développement (CBD), CNRS and Université de Toulouse, Toulouse, France.,Centre de Biologie Intégrative (CBI), CNRS and Université de Toulouse, Toulouse, France
| | - Michèle Crozatier
- Centre de Biologie du Développement (CBD), CNRS and Université de Toulouse, Toulouse, France.,Centre de Biologie Intégrative (CBI), CNRS and Université de Toulouse, Toulouse, France
| | - Alain Vincent
- Centre de Biologie du Développement (CBD), CNRS and Université de Toulouse, Toulouse, France.,Centre de Biologie Intégrative (CBI), CNRS and Université de Toulouse, Toulouse, France
| |
Collapse
|
13
|
Elevated ZNF703 Protein Expression Is an Independent Unfavorable Prognostic Factor for Survival of the Patients with Head and Neck Squamous Cell Carcinoma. DISEASE MARKERS 2015; 2015:640263. [PMID: 26063961 PMCID: PMC4429199 DOI: 10.1155/2015/640263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/15/2015] [Indexed: 01/26/2023]
Abstract
AIM Data from The Cancer Genome Atlas (TCGA) show that the ZNF703 gene amplifies and overexpresses in head and neck squamous cell carcinomas (HNSCC). However, the clinical relevance of this observation in HNSCC is unclear. The purpose of this study was to clarify the expression of ZNF703 protein and its prognostic effect on HNSCC. METHODS Two hundred ten HNSCC patients from Sun Yat-Sen University Cancer Center with complete survival follow-up were included in this study. Tumor samples from primary sites were collected. The expression of the ZNF703 protein was tested by immunohistochemistry (IHC). RESULTS The high expression of ZNF703 in HNSCC tumor tissues was significantly higher than that of the matched noncancerous tissues (48.6% versus 11.6%, P < 0.001). ZNF703 overexpression was correlated with tumor position (laryngeal carcinoma) and recurrence (all P < 0.05). Multivariate analysis revealed that ZNF703 protein overexpression was an independent prognostic factor (P = 0.022, hazard ratio = 1.635, 95% CI 1.073-2.493) in HNSCC patients. CONCLUSION ZNF703 overexpression is associated with adverse prognosis in HNSCC, which might be a novel biomarker of HNSCC.
Collapse
|
14
|
Shahi P, Slorach EM, Wang CY, Chou J, Lu A, Ruderisch A, Werb Z. The Transcriptional Repressor ZNF503/Zeppo2 Promotes Mammary Epithelial Cell Proliferation and Enhances Cell Invasion. J Biol Chem 2014; 290:3803-13. [PMID: 25538248 DOI: 10.1074/jbc.m114.611202] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The NET (nocA, Nlz, elB, TLP-1) subfamily of zinc finger proteins is an important mediator during developmental processes. The evolutionary conserved zinc finger protein ZNF503/Zeppo2 (zinc finger elbow-related proline domain protein 2, Zpo2) plays critical roles during embryogenesis. We found that Zpo2 is expressed in adult tissue and examined its function. We found that ZPO2 is a nuclearly targeted transcriptional repressor that is expressed in mammary epithelial cells. Elevated Zpo2 levels increase mammary epithelial cell proliferation. Zpo2 promotes cellular invasion through down-regulation of E-cadherin and regulates the invasive phenotype in a RAC1-dependent manner. We detect elevated Zpo2 expression during breast cancer progression in a MMTV-PyMT transgenic mouse model. Tumor transplant experiments indicated that overexpression of Zpo2 in MMTV-PyMT mammary tumor cell lines enhances lung metastasis. Our findings suggest that Zpo2 plays a significant role in mammary gland homeostasis and that deregulation of Zpo2 may promote breast cancer development.
Collapse
Affiliation(s)
- Payam Shahi
- From the Department of Anatomy, University of California, San Francisco, California 94143-0452 and
| | - Euan M Slorach
- From the Department of Anatomy, University of California, San Francisco, California 94143-0452 and
| | - Chih-Yang Wang
- the Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan 70101
| | - Jonathan Chou
- From the Department of Anatomy, University of California, San Francisco, California 94143-0452 and
| | - Angela Lu
- From the Department of Anatomy, University of California, San Francisco, California 94143-0452 and
| | - Aline Ruderisch
- From the Department of Anatomy, University of California, San Francisco, California 94143-0452 and
| | - Zena Werb
- From the Department of Anatomy, University of California, San Francisco, California 94143-0452 and
| |
Collapse
|
15
|
Genetic dissection of photoreceptor subtype specification by the Drosophila melanogaster zinc finger proteins elbow and no ocelli. PLoS Genet 2014; 10:e1004210. [PMID: 24625735 PMCID: PMC3953069 DOI: 10.1371/journal.pgen.1004210] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/15/2014] [Indexed: 12/28/2022] Open
Abstract
The elbow/no ocelli (elb/noc) complex of Drosophila melanogaster encodes two paralogs of the evolutionarily conserved NET family of zinc finger proteins. These transcriptional repressors share a conserved domain structure, including a single atypical C2H2 zinc finger. In flies, Elb and Noc are important for the development of legs, eyes and tracheae. Vertebrate NET proteins play an important role in the developing nervous system, and mutations in the homolog ZNF703 human promote luminal breast cancer. However, their interaction with transcriptional regulators is incompletely understood. Here we show that loss of both Elb and Noc causes mis-specification of polarization-sensitive photoreceptors in the 'dorsal rim area' (DRA) of the fly retina. This phenotype is identical to the loss of the homeodomain transcription factor Homothorax (Hth)/dMeis. Development of DRA ommatidia and expression of Hth are induced by the Wingless/Wnt pathway. Our data suggest that Elb/Noc genetically interact with Hth, and we identify two conserved domains crucial for this function. Furthermore, we show that Elb/Noc specifically interact with the transcription factor Orthodenticle (Otd)/Otx, a crucial regulator of rhodopsin gene transcription. Interestingly, different Elb/Noc domains are required to antagonize Otd functions in transcriptional activation, versus transcriptional repression. We propose that similar interactions between vertebrate NET proteins and Meis and Otx factors might play a role in development and disease.
Collapse
|
16
|
YANG GONGLI, MA FENG, ZHONG MUXIAO, FANG LIN, PENG YAO, XIN XIAOMING, ZHONG JIETAO, YUAN FANGFANG, GU HONGXIANG, ZHU WEI, ZHANG YALI. ZNF703 acts as an oncogene that promotes progression in gastric cancer. Oncol Rep 2014; 31:1877-82. [DOI: 10.3892/or.2014.2997] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 01/08/2014] [Indexed: 11/05/2022] Open
|
17
|
Ectopic expression of nolz-1 in neural progenitors promotes cell cycle exit/premature neuronal differentiation accompanying with abnormal apoptosis in the developing mouse telencephalon. PLoS One 2013; 8:e74975. [PMID: 24073229 PMCID: PMC3779228 DOI: 10.1371/journal.pone.0074975] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 08/13/2013] [Indexed: 01/01/2023] Open
Abstract
Nolz-1, as a murine member of the NET zinc-finger protein family, is expressed in post-mitotic differentiating neurons of striatum during development. To explore the function of Nolz-1 in regulating the neurogenesis of forebrain, we studied the effects of ectopic expression of Nolz-1 in neural progenitors. We generated the Cre-loxP dependent conditional transgenic mice in which Nolz-1 was ectopically expressed in proliferative neural progenitors. Ectopic expression of Nolz-1 in neural progenitors by intercrossing the Nolz-1 conditional transgenic mice with the nestin-Cre mice resulted in hypoplasia of telencephalon in double transgenic mice. Decreased proliferation of neural progenitor cells were found in the telencephalon, as evidenced by the reduction of BrdU-, Ki67- and phospho-histone 3-positive cells in E11.5-12.5 germinal zone of telencephalon. Transgenic Nolz-1 also promoted cell cycle exit and as a consequence might facilitate premature differentiation of progenitors, because TuJ1-positive neurons were ectopically found in the ventricular zone and there was a general increase of TuJ1 immunoreactivity in the telencephalon. Moreover, clusters of strong TuJ1-expressing neurons were present in E12.5 germinal zone. Some of these strong TuJ1-positive clusters, however, contained apoptotic condensed DNA, suggesting that inappropriate premature differentiation may lead to abnormal apoptosis in some progenitor cells. Consistent with the transgenic mouse analysis in vivo, similar effects of Nozl-1 over-expression in induction of apoptosis, inhibition of cell proliferation and promotion of neuronal differentiation were also observed in three different N18, ST14A and N2A neural cell lines in vitro. Taken together, our study indicates that ectopic expression of Nolz-1 in neural progenitors promotes cell cycle exit/premature neuronal differentiation and induces abnormal apoptosis in the developing telencephalon.
Collapse
|
18
|
Pereira-Castro I, Costa AMS, Oliveira MJ, Barbosa I, Rocha AS, Azevedo L, da Costa LT. Characterization of human NLZ1/ZNF703 identifies conserved domains essential for proper subcellular localization and transcriptional repression. J Cell Biochem 2013; 114:120-33. [PMID: 22886885 DOI: 10.1002/jcb.24309] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 07/26/2012] [Indexed: 11/06/2022]
Abstract
NET family members have recently emerged as important players in the development of multiple structures, from the trachea of fly larvae to the vertebrate eye and human breast cancers. However, their mechanisms of action are still poorly understood, and we lack a detailed characterization of their functional domains, as well as gene expression patterns-particularly in adult mammals. Here, we present a characterization of human NLZ1/ZNF703 (NocA-like zinc finger 1/Zinc finger 703), one of the two human NET family member genes. We show that the gene is ubiquitously expressed in adult human and mouse tissues, that three mRNA species with the same coding sequence are generated by alternative polyadenylation, and that the encoded protein contains six evolutionarily conserved domains, three of which are specific to NET proteins. Finally, we present functional evidence that these domains are necessary for proper subcellular distribution of and transcription repression by the NLZ1 protein, but not for its interaction with Groucho family co-repressors.
Collapse
Affiliation(s)
- Isabel Pereira-Castro
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | | | | | | | | | | | | |
Collapse
|
19
|
Identification of two evolutionarily conserved 5' cis-elements involved in regulating spatiotemporal expression of Nolz-1 during mouse embryogenesis. PLoS One 2013; 8:e54485. [PMID: 23349903 PMCID: PMC3551757 DOI: 10.1371/journal.pone.0054485] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 12/12/2012] [Indexed: 01/13/2023] Open
Abstract
Proper development of vertebrate embryos depends not only on the crucial funtions of key evolutionarily conserved transcriptional regulators, but also on the precisely spatiotemporal expression of these transcriptional regulators. The mouse Nolz-1/Znf503/Zfp503 gene is a mammalian member of the conserved zinc-finger containing NET family. The expression pattern of Nolz-1 in mouse embryos is highly correlated with that of its homologues in different species. To study the spatiotemporal regulation of Nolz-1, we first identified two evolutionarily conserved cis-elements, UREA and UREB, in 5' upstream regions of mouse Nolz-1 locus. We then generated UREA-LacZ and UREB-LacZ transgenic reporter mice to characterize the putative enhancer activity of UREA and UREB. The results indicated that both UREA and UREB contained tissue-specific enhancer activity for directing LacZ expression in selective tissue organs during mouse embryogensis. UREA directed LacZ expression preferentially in selective regions of developing central nervous system, including the forebrain, hindbrain and spinal cord, whereas UREB directed LacZ expression mainly in other developing tissue organs such as the Nolz-1 expressing branchial arches and its derivatives, the apical ectodermal ridge of limb buds and the urogenital tissues. Both UREA and UREB directed strong LacZ expression in the lateral plate mesoderm where endogenous Nolz-1 was also expressed. Despite that the LacZ expression pattern did not full recapitulated the endogenous Nolz-1 expression and some mismatched expression patterns were observed, co-expression of LacZ and Nolz-1 did occur in many cells of selective tissue organs, such as in the ventrolateral cortex and ventral spinal cord of UREA-LacZ embryos, and the urogenital tubes of UREB-LacZ embryos. Taken together, our study suggests that UREA and UREB may function as evolutionarily conserved cis-regulatory elements that coordinate with other cis-elements to regulate spatiotemporal expression of Nolz-1 in different tissue organs during mouse embryogenesis.
Collapse
|
20
|
Marden JH, Fescemyer HW, Schilder RJ, Doerfler WR, Vera JC, Wheat CW. GENETIC VARIATION IN HIF SIGNALING UNDERLIES QUANTITATIVE VARIATION IN PHYSIOLOGICAL AND LIFE-HISTORY TRAITS WITHIN LOWLAND BUTTERFLY POPULATIONS. Evolution 2012; 67:1105-15. [DOI: 10.1111/evo.12004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Trachealess (Trh) regulates all tracheal genes during Drosophila embryogenesis. Dev Biol 2011; 360:160-72. [PMID: 21963537 DOI: 10.1016/j.ydbio.2011.09.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Revised: 08/08/2011] [Accepted: 09/14/2011] [Indexed: 02/03/2023]
Abstract
The Drosophila trachea is a branched tubular epithelia that transports oxygen and other gases. trachealess (trh), which encodes a bHLH-PAS transcription factor, is among the first genes to be expressed in the cells that will form the trachea. In the absence of trh, tracheal cells fail to invaginate to form tubes and remain on the embryo surface. Expression of many tracheal-specific genes depends on trh, but all of the known targets have relatively minor phenotypes compared to loss of trh, suggesting that there are additional targets. To identify uncharacterized transcriptional targets of Trh and to further understand the role of Trh in embryonic tracheal formation, we performed an in situ hybridization screen using a library of ~100 tracheal-expressed genes identified by the Berkeley Drosophila Genome Project (BDGP). Surprisingly, expression of every tracheal gene we tested was dependent on Trh, suggesting a major role for Trh in activation and maintenance of tracheal gene expression. A re-examination of the interdependence of the known early-expressed transcription factors, including trh, ventral veinless (vvl) and knirps/knirps-related (kni/knrl), suggests a new model for how gene expression is controlled in the trachea, with trh regulating expression of vvl and kni, but not vice versa. A pilot screen for the targets of Vvl and Kni/Knrl revealed that Vvl and Kni have only minor roles compared to Trh. Finally, genome-wide microarray experiments identified additional Trh targets and revealed that a variety of biological processes are affected by the loss of trh.
Collapse
|
22
|
Chang SLY, Yan YT, Shi YL, Liu YC, Takahashi H, Liu FC. Region- and cell type-selective expression of the evolutionarily conserved Nolz-1/zfp503 gene in the developing mouse hindbrain. Gene Expr Patterns 2011; 11:525-32. [PMID: 21945624 DOI: 10.1016/j.gep.2011.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Revised: 09/01/2011] [Accepted: 09/06/2011] [Indexed: 11/30/2022]
Abstract
Nolz-1/Zfp503, a zinc finger-containing gene, is a mammalian member of the SP1-related nocA/elb/tlp-1 gene family. Previous studies have shown that Nolz-1 homologs are important for patterning the rhombomeres in zebrafish hindbrain. We therefore studied the expression pattern of Nolz-1 in the developing mouse hindbrain. Nolz-1 mRNA expression was detected in the prospective rhombomere 3, 5 and caudal regions as early as E8.75. After E11.5, Nolz-1-positive cells were organized as distinct cell clusters, and they were largely non-overlapped with either Pax2-positive or Phox2b-positive domains. Most interestingly, we found that Nolz-1 was specifically expressed by Phox2b-negative/Isl1/2-positive somatic motor neurons, but not by Phox2b-positive/Isl1/2-positive branchial and visceral motor neurons, suggesting that Nolz-1 may regulate development of somatic motor neurons in the hindbrain. In addition to be expressed in differentiating post-mitotic neurons, Nolz-1 was also expressed by progenitor cells in the ventricular zone located in the dorsal part of aqueduct and the alar plates of hindbrain, which suggests a regulatory role of Nolz-1 in the germinal zone. Taken together, based on its domain- and cell type-selective pattern, Nolz-1 may involve in regulation of various developmental processes, including regional patterning and cell-type specification and differentiation in the developing mouse hindbrain.
Collapse
Affiliation(s)
- Sunny Li-Yun Chang
- Institute of Neuroscience, National Yang-Ming University, Taipei 112, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
23
|
Holland DG, Burleigh A, Git A, Goldgraben MA, Perez-Mancera PA, Chin SF, Hurtado A, Bruna A, Ali HR, Greenwood W, Dunning MJ, Samarajiwa S, Menon S, Rueda OM, Lynch AG, McKinney S, Ellis IO, Eaves CJ, Carroll JS, Curtis C, Aparicio S, Caldas C. ZNF703 is a common Luminal B breast cancer oncogene that differentially regulates luminal and basal progenitors in human mammary epithelium. EMBO Mol Med 2011; 3:167-80. [PMID: 21337521 PMCID: PMC3395113 DOI: 10.1002/emmm.201100122] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 01/16/2011] [Accepted: 01/18/2011] [Indexed: 01/01/2023] Open
Abstract
The telomeric amplicon at 8p12 is common in oestrogen receptor-positive (ER+) breast cancers. Array-CGH and expression analyses of 1172 primary breast tumours revealed that ZNF703 was the single gene within the minimal amplicon and was amplified predominantly in the Luminal B subtype. Amplification was shown to correlate with increased gene and protein expression and was associated with a distinct expression signature and poor clinical outcome. ZNF703 transformed NIH 3T3 fibroblasts, behaving as a classical oncogene, and regulated proliferation in human luminal breast cancer cell lines and immortalized human mammary epithelial cells. Manipulation of ZNF703 expression in the luminal MCF7 cell line modified the effects of TGFβ on proliferation. Overexpression of ZNF703 in normal human breast epithelial cells enhanced the frequency of in vitro colony-forming cells from luminal progenitors. Taken together, these data strongly point to ZNF703 as a novel oncogene in Luminal B breast cancer.
Collapse
Affiliation(s)
- Daniel G Holland
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Oncology, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Angela Burleigh
- Department of Pathology, University of British ColumbiaVancouver, Canada
- BC Cancer AgencyVancouver, Canada
| | - Anna Git
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Oncology, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Mae A Goldgraben
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Oncology, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Pedro A Perez-Mancera
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Oncology, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Suet-Feung Chin
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
| | - Antonio Hurtado
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
| | - Alejandra Bruna
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Oncology, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - H Raza Ali
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Oncology, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Wendy Greenwood
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Oncology, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Mark J Dunning
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
| | | | - Suraj Menon
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
| | - Oscar M Rueda
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Oncology, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Andy G Lynch
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
| | - Steven McKinney
- Department of Pathology, University of British ColumbiaVancouver, Canada
- BC Cancer AgencyVancouver, Canada
| | - Ian O Ellis
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham City HospitalNottingham, UK
| | - Connie J Eaves
- Department of Pathology, University of British ColumbiaVancouver, Canada
- BC Cancer AgencyVancouver, Canada
| | - Jason S Carroll
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Oncology, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Christina Curtis
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Preventive Medicine, Keck School of Medicine, University of Southern CaliforniaLos Angles, California
| | - Samuel Aparicio
- Department of Pathology, University of British ColumbiaVancouver, Canada
- BC Cancer AgencyVancouver, Canada
| | - Carlos Caldas
- Cancer Research UK, Cambridge Research InstituteCambridge, UK
- Department of Oncology, University of Cambridge, Li Ka Shing CentreCambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation TrustCambridge, UK
- Cambridge Experimental Cancer Medicine Centre, Cambridge University Hospitals NHS Foundation TrustCambridge, UK
| |
Collapse
|
24
|
Slorach EM, Chou J, Werb Z. Zeppo1 is a novel metastasis promoter that represses E-cadherin expression and regulates p120-catenin isoform expression and localization. Genes Dev 2011; 25:471-84. [PMID: 21317240 DOI: 10.1101/gad.1998111] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Amplification of 8p11-12 in human breast cancers is associated with increased proliferation and tumor grade and reduced metastasis-free patient survival. We identified Zeppo1 (zinc finger elbow-related proline domain protein 1) (FLJ14299/ZNF703) within this amplicon as a regulator of cell adhesion, migration, and proliferation in mammary epithelial cells. Overexpression of Zeppo1 reduces cell-cell adhesion and stimulates migration and proliferation. Knockdown of Zeppo1 induces adhesion and lumen formation. Zeppo1 regulates transcription, complexing with Groucho and repressing E-cadherin expression and Wnt and TGFβ reporter expression. Zeppo1 promotes expression of metastasis-associated p120-catenin isoform 1 and alters p120-catenin localization upon cell contact with the extracellular matrix. Significantly, Zeppo1 overexpression in a mouse breast cancer model increases lung metastases, while reducing Zeppo1 expression reduces both tumor size and the number of lung metastases. These results indicate that Zeppo1 is a key regulator of breast cancer progression.
Collapse
Affiliation(s)
- Euan M Slorach
- Department of Anatomy, University of California at San Francisco, San Francisco, California 94143-0452, USA
| | | | | |
Collapse
|
25
|
Urbán N, Martín-Ibáñez R, Herranz C, Esgleas M, Crespo E, Pardo M, Crespo-Enríquez I, Méndez-Gómez HR, Waclaw R, Chatzi C, Alvarez S, Alvarez R, Duester G, Campbell K, de Lera AR, Vicario-Abejón C, Martinez S, Alberch J, Canals JM. Nolz1 promotes striatal neurogenesis through the regulation of retinoic acid signaling. Neural Dev 2010; 5:21. [PMID: 20735826 PMCID: PMC2939507 DOI: 10.1186/1749-8104-5-21] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 08/24/2010] [Indexed: 12/18/2022] Open
Abstract
Background Nolz1 is a zinc finger transcription factor whose expression is enriched in the lateral ganglionic eminence (LGE), although its function is still unknown. Results Here we analyze the role of Nolz1 during LGE development. We show that Nolz1 expression is high in proliferating neural progenitor cells (NPCs) of the LGE subventricular zone. In addition, low levels of Nolz1 are detected in the mantle zone, as well as in the adult striatum. Similarly, Nolz1 is highly expressed in proliferating LGE-derived NPC cultures, but its levels rapidly decrease upon cell differentiation, pointing to a role of Nolz1 in the control of NPC proliferation and/or differentiation. In agreement with this hypothesis, we find that Nolz1 over-expression promotes cell cycle exit of NPCs in neurosphere cultures and negatively regulates proliferation in telencephalic organotypic cultures. Within LGE primary cultures, Nolz1 over-expression promotes the acquisition of a neuronal phenotype, since it increases the number of β-III tubulin (Tuj1)- and microtubule-associated protein (MAP)2-positive neurons, and inhibits astrocyte generation and/or differentiation. Retinoic acid (RA) is one of the most important morphogens involved in striatal neurogenesis, and regulates Nolz1 expression in different systems. Here we show that Nolz1 also responds to this morphogen in E12.5 LGE-derived cell cultures. However, Nolz1 expression is not regulated by RA in E14.5 LGE-derived cell cultures, nor is it affected during LGE development in mouse models that present decreased RA levels. Interestingly, we find that Gsx2, which is necessary for normal RA signaling during LGE development, is also required for Nolz1 expression, which is lost in Gsx2 knockout mice. These findings suggest that Nolz1 might act downstream of Gsx2 to regulate RA-induced neurogenesis. Keeping with this hypothesis, we show that Nolz1 induces the selective expression of the RA receptor (RAR)β without altering RARα or RARγ. In addition, Nozl1 over-expression increases RA signaling since it stimulates the RA response element. This RA signaling is essential for Nolz1-induced neurogenesis, which is impaired in a RA-free environment or in the presence of a RAR inverse agonist. It has been proposed that Drosophila Gsx2 and Nolz1 homologues could cooperate with the transcriptional co-repressors Groucho-TLE to regulate cell proliferation. In agreement with this view, we show that Nolz1 could act in collaboration with TLE-4, as they are expressed at the same time in NPC cultures and during mouse development. Conclusions Nolz1 promotes RA signaling in the LGE, contributing to the striatal neurogenesis during development.
Collapse
Affiliation(s)
- Noelia Urbán
- Department of Cell Biology, Immunology and Neuroscience, Faculty of Medicine, IDIBAPS, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhong L, Hwang RY, Tracey WD. Pickpocket is a DEG/ENaC protein required for mechanical nociception in Drosophila larvae. Curr Biol 2010; 20:429-34. [PMID: 20171104 PMCID: PMC2995491 DOI: 10.1016/j.cub.2009.12.057] [Citation(s) in RCA: 200] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 12/29/2009] [Accepted: 12/30/2009] [Indexed: 11/17/2022]
Abstract
Highly branched class IV multidendritic sensory neurons of the Drosophila larva function as polymodal nociceptors that are necessary for behavioral responses to noxious heat (>39 degrees C) or noxious mechanical (>30 mN) stimuli. However, the molecular mechanisms that allow these cells to detect both heat and force are unknown. Here, we report that the pickpocket (ppk) gene, which encodes a Degenerin/Epithelial Sodium Channel (DEG/ENaC) subunit, is required for mechanical nociception but not thermal nociception in these sensory cells. Larvae mutant for pickpocket show greatly reduced nociception behaviors in response to harsh mechanical stimuli. However, pickpocket mutants display normal behavioral responses to gentle touch. Tissue-specific knockdown of pickpocket in nociceptors phenocopies the mechanical nociception impairment without causing defects in thermal nociception behavior. Finally, optogenetically triggered nociception behavior is unaffected by pickpocket RNAi, which indicates that ppk is not generally required for the excitability of the nociceptors. Interestingly, DEG/ENaCs are known to play a critical role in detecting gentle touch stimuli in Caenorhabditis elegans and have also been implicated in some aspects of harsh touch sensation in mammals. Our results suggest that neurons that detect harsh touch in Drosophila utilize similar mechanosensory molecules.
Collapse
Affiliation(s)
- Lixian Zhong
- Pharmacology Science Training Program, Duke University Medical Center, Durham, North Carolina, 27710
| | - Richard Y. Hwang
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, 27710
| | - W. Daniel Tracey
- Pharmacology Science Training Program, Duke University Medical Center, Durham, North Carolina, 27710
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, 27710
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, 27710
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, 27710
| |
Collapse
|
27
|
Dubruille R, Murad A, Rosbash M, Emery P. A constant light-genetic screen identifies KISMET as a regulator of circadian photoresponses. PLoS Genet 2009; 5:e1000787. [PMID: 20041201 PMCID: PMC2789323 DOI: 10.1371/journal.pgen.1000787] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Accepted: 11/24/2009] [Indexed: 12/28/2022] Open
Abstract
Circadian pacemakers are essential to synchronize animal physiology and behavior with the day∶night cycle. They are self-sustained, but the phase of their oscillations is determined by environmental cues, particularly light intensity and temperature cycles. In Drosophila, light is primarily detected by a dedicated blue-light photoreceptor: CRYPTOCHROME (CRY). Upon light activation, CRY binds to the pacemaker protein TIMELESS (TIM) and triggers its proteasomal degradation, thus resetting the circadian pacemaker. To understand further the CRY input pathway, we conducted a misexpression screen under constant light based on the observation that flies with a disruption in the CRY input pathway remain robustly rhythmic instead of becoming behaviorally arrhythmic. We report the identification of more than 20 potential regulators of CRY-dependent light responses. We demonstrate that one of them, the chromatin-remodeling enzyme KISMET (KIS), is necessary for normal circadian photoresponses, but does not affect the circadian pacemaker. KIS genetically interacts with CRY and functions in PDF-negative circadian neurons, which play an important role in circadian light responses. It also affects daily CRY-dependent TIM oscillations in a peripheral tissue: the eyes. We therefore conclude that KIS is a key transcriptional regulator of genes that function in the CRY signaling cascade, and thus it plays an important role in the synchronization of circadian rhythms with the day∶night cycle. In most organisms, intracellular molecular pacemakers called circadian clocks coordinate metabolic, physiological, and behavioral processes during the course of the day. For example, they determine when animals are active or resting. Circadian clocks are self-sustained oscillators, but their free-running period does not exactly match day length. Thus, they have to be reset by environmental inputs to stay properly phased with the day∶night cycle. The fruit fly Drosophila melanogaster relies primarily on CRYPTOCHROME (CRY)—a cell-autonomous blue-light photoreceptor—to synchronize its circadian clocks with the light∶dark cycle. With a genetic screen, we identified over 20 candidate genes that might regulate CRY function. kismet (kis) is among them: it encodes a chromatin remodeling factor essential for the development of Drosophila. We show that, in adult flies, KIS is expressed and functions in brain neurons that control daily behavioral rhythms. KIS determines how Drosophila circadian behavior responds to light, but not its free-running period. Moreover, manipulating simultaneously kis and cry activity demonstrates that these two genes interact to control molecular and behavioral circadian photoresponses. Our work therefore reveals that KIS regulates CRY signaling and thus determines how circadian clocks respond to light input.
Collapse
Affiliation(s)
- Raphaëlle Dubruille
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Alejandro Murad
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Neuroscience, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Michael Rosbash
- Howard Hughes Medical Institute, National Center for Behavioral Genetics and Department of Biology, Waltham, Massachusetts, United States of America
| | - Patrick Emery
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Neuroscience, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
28
|
van de Hoef DL, Hughes J, Livne-Bar I, Garza D, Konsolaki M, Boulianne GL. Identifying genes that interact with Drosophila presenilin and amyloid precursor protein. Genesis 2009; 47:246-60. [PMID: 19241393 DOI: 10.1002/dvg.20485] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The gamma-secretase complex is involved in cleaving transmembrane proteins such as Notch and one of the genes targeted in Alzheimer's disease known as amyloid precursor protein (APP). Presenilins function within the catalytic core of gamma-secretase, and mutated forms of presenilins were identified as causative factors in familial Alzheimer's disease. Recent studies show that in addition to Notch and APP, numerous signal transduction pathways are modulated by presenilins, including intracellular calcium signaling. Thus, presenilins appear to have diverse roles. To further understand presenilin function, we searched for Presenilin-interacting genes in Drosophila by performing a genetic modifier screen for enhancers and suppressors of Presenilin-dependent Notch-related phenotypes. We identified 177 modifiers, including known members of the Notch pathway and genes involved in intracellular calcium homeostasis. We further demonstrate that 53 of these modifiers genetically interacted with APP. Characterization of these genes may provide valuable insights into Presenilin function in development and disease.
Collapse
Affiliation(s)
- Diana L van de Hoef
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
29
|
Hou HY, Heffer A, Anderson WR, Liu J, Bowler T, Pick L. Stripy Ftz target genes are coordinately regulated by Ftz-F1. Dev Biol 2009; 335:442-53. [PMID: 19679121 DOI: 10.1016/j.ydbio.2009.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 07/26/2009] [Accepted: 08/03/2009] [Indexed: 01/12/2023]
Abstract
During development, cascades of regulatory genes act in a hierarchical fashion to subdivide the embryo into increasingly specified body regions. This has been best characterized in Drosophila, where genes encoding regulatory transcription factors form a network to direct the development of the basic segmented body plan. The pair-rule genes are pivotal in this process as they are responsible for the first subdivision of the embryo into repeated metameric units. The Drosophila pair-rule gene fushi tarazu (ftz) is a derived Hox gene expressed in and required for the development of alternate parasegments. Previous studies suggested that Ftz achieves its distinct regulatory specificity as a segmentation protein by interacting with a ubiquitously expressed cofactor, the nuclear receptor Ftz-F1. However, the downstream target genes regulated by Ftz and other pair-rule genes to direct segment formation are not known. In this study, we selected candidate Ftz targets by virtue of their early expression in Ftz-like stripes. This identified two new Ftz target genes, drumstick (drm) and no ocelli (noc), and confirmed that Ftz regulates a serotonin receptor (5-HT2). These are the earliest Ftz targets identified to date and all are coordinately regulated by Ftz-F1. Engrailed (En), the best-characterized Ftz/Ftz-F1 downstream target, is not an intermediate in regulation. The drm genomic region harbors two separate seven-stripe enhancers, identified by virtue of predicted Ftz-F1 binding sites, and these sites are necessary for stripe expression in vivo. We propose that pair-rule genes, exemplified by Ftz/Ftz-F1, promote segmentation by acting at different hierarchical levels, regulating first, other segmentation genes; second, other regulatory genes that in turn control specific cellular processes such as tissue differentiation; and, third, 'segmentation realizator genes' that are directly involved in morphogenesis.
Collapse
Affiliation(s)
- Hui Ying Hou
- Department of Entomology, University of Maryland, College Park, 20742, USA
| | | | | | | | | | | |
Collapse
|
30
|
Nakamura M, Choe SK, Runko AP, Gardner PD, Sagerström CG. Nlz1/Znf703 acts as a repressor of transcription. BMC DEVELOPMENTAL BIOLOGY 2008; 8:108. [PMID: 19014486 PMCID: PMC2588584 DOI: 10.1186/1471-213x-8-108] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Accepted: 11/12/2008] [Indexed: 11/13/2022]
Abstract
Background Members of the NET subfamily of zinc-finger proteins are related to the Sp-family of transcription factors and are required during embryogenesis. In particular, Nlz1/Znf703 and Nlz2/Znf503 are required for formation of rhombomere 4 of the vertebrate hindbrain. While NET family proteins have been hypothesized to regulate transcription, it remains unclear if they function as activators or repressors of transcription. Results Here we demonstrate that Nlz proteins repress transcription both in cell lines and in developing zebrafish embryos. We first use standard cell culture-based reporter assays to demonstrate that Nlz1/Znf703 represses transcription of a luciferase reporter in four different cell lines. Structure-function analyses and pharmacological inhibition further reveal that Nlz1-mediated repression requires histone deacetylase activity. We next generate a stable transgenic zebrafish reporter line to demonstrate that Nlz1 promotes histone deacetylation at the transgenic promoter and repression of transgene expression during embryogenesis. Lastly, taking a genetic approach we find that endogenous Nlz proteins are required for formation of hindbrain rhombomere 4 during zebrafish embryogenesis by repressing expression of non-rhombomere 4 genes. Conclusion We conclude that Nlz1/Znf703 acts as a repressor of transcription and hypothesize that other NET family members function in a similar manner.
Collapse
Affiliation(s)
- Mako Nakamura
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, USA.
| | | | | | | | | |
Collapse
|
31
|
McGlinn E, Richman JM, Metzis V, Town L, Butterfield NC, Wainwright BJ, Wicking C. Expression of the NET family member Zfp503 is regulated by hedgehog and BMP signaling in the limb. Dev Dyn 2008; 237:1172-82. [PMID: 18351672 DOI: 10.1002/dvdy.21508] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The NET/Nlz family of zinc finger transcription factors contribute to aspects of developmental growth and patterning across evolutionarily diverse species. To date, however, these molecules remain largely uncharacterized in mouse and chick. We previously reported that limb bud expression of Zfp503, the mouse orthologue of zebrafish nlz2/znf503, is dependent on Gli3. Here, we show that Zfp503/Znf503 is expressed in a restricted pattern during mouse and chick embryogenesis, with particularly dynamic expression in the developing limbs, face, somites, and brain. We also add to our previous data on Gli3 regulation by showing that the anterior domain of Zfp503 expression in the mouse limb is responsive to genetic and nongenetic manipulation of hedgehog signaling. Finally, we demonstrate that posterior expression of Znf503 in the chick limb is responsive to bone morphogenetic protein (BMP) signaling, indicating that Zfp503/Znf503 may act at the nexus of multiple signaling pathways in development.
Collapse
Affiliation(s)
- Edwina McGlinn
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | | | | | | | | | | | | |
Collapse
|
32
|
Engström PG, Ho Sui SJ, Drivenes O, Becker TS, Lenhard B. Genomic regulatory blocks underlie extensive microsynteny conservation in insects. Genome Res 2007; 17:1898-908. [PMID: 17989259 DOI: 10.1101/gr.6669607] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Insect genomes contain larger blocks of conserved gene order (microsynteny) than would be expected under a random breakage model of chromosome evolution. We present evidence that microsynteny has been retained to keep large arrays of highly conserved noncoding elements (HCNEs) intact. These arrays span key developmental regulatory genes, forming genomic regulatory blocks (GRBs). We recently described GRBs in vertebrates, where most HCNEs function as enhancers and HCNE arrays specify complex expression programs of their target genes. Here we present a comparison of five Drosophila genomes showing that HCNE density peaks centrally in large synteny blocks containing multiple genes. Besides developmental regulators that are likely targets of HCNE enhancers, HCNE arrays often span unrelated neighboring genes. We describe differences in core promoters between the target genes and the unrelated genes that offer an explanation for the differences in their responsiveness to enhancers. We show examples of a striking correspondence between boundaries of synteny blocks, HCNE arrays, and Polycomb binding regions, confirming that the synteny blocks correspond to regulatory domains. Although few noncoding elements are highly conserved between Drosophila and the malaria mosquito Anopheles gambiae, we find that A. gambiae regions orthologous to Drosophila GRBs contain an equivalent distribution of noncoding elements highly conserved in the yellow fever mosquito Aëdes aegypti and coincide with regions of ancient microsynteny between Drosophila and mosquitoes. The structural and functional equivalence between insect and vertebrate GRBs marks them as an ancient feature of metazoan genomes and as a key to future studies of development and gene regulation.
Collapse
Affiliation(s)
- Pär G Engström
- Computational Biology Unit, Bergen Center for Computational Science, University of Bergen, Bergen 5008, Norway
| | | | | | | | | |
Collapse
|
33
|
Luque CM, Milán M. Growth control in the proliferative region of the Drosophila eye–head primordium: The elbow–noc gene complex. Dev Biol 2007; 301:327-39. [PMID: 17014842 DOI: 10.1016/j.ydbio.2006.07.050] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2006] [Revised: 07/27/2006] [Accepted: 07/28/2006] [Indexed: 11/25/2022]
Abstract
Notch signaling is involved in cell differentiation and patterning, as well as in the regulation of growth and cell survival. Notch activation at the dorsal-ventral boundary of the Drosophila eye-head primordium leads to the expression of the secreted protein Unpaired, a ligand of the JAK-STAT pathway that induces cell proliferation in the undifferentiated tissue. The zinc finger proteins encoded by elbow and no ocelli are expressed in the highly proliferative region of the eye-head primordium. Loss of elbow and no ocelli activities induces overgrowths of the head capsule, without inducing Upd expression de novo. These overgrowths depend on Notch activity suggesting that elbow and noc repress a Upd independent role of Notch in driving cell proliferation. When the size of the overgrown tissue is increased, ectopic antenna and eye structures can be found. Thus, tight regulation of the size of the eye-head primordium by elbow and no ocelli is crucial for proper fate specification and generation of the adult structures.
Collapse
Affiliation(s)
- Carlos M Luque
- ICREA and Institute for Research in Biomedicine (IRB), Parc Científic de Barcelona, Josep Samitier, 1-5, 08028 Barcelona, Spain
| | | |
Collapse
|
34
|
Nègre N, Hennetin J, Sun LV, Lavrov S, Bellis M, White KP, Cavalli G. Chromosomal distribution of PcG proteins during Drosophila development. PLoS Biol 2006; 4:e170. [PMID: 16613483 PMCID: PMC1440717 DOI: 10.1371/journal.pbio.0040170] [Citation(s) in RCA: 199] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2005] [Accepted: 03/23/2006] [Indexed: 11/18/2022] Open
Abstract
Polycomb group (PcG) proteins are able to maintain the memory of silent transcriptional states of homeotic genes throughout development. In Drosophila, they form multimeric complexes that bind to specific DNA regulatory elements named PcG response elements (PREs). To date, few PREs have been identified and the chromosomal distribution of PcG proteins during development is unknown. We used chromatin immunoprecipitation (ChIP) with genomic tiling path microarrays to analyze the binding profile of the PcG proteins Polycomb (PC) and Polyhomeotic (PH) across 10 Mb of euchromatin. We also analyzed the distribution of GAGA factor (GAF), a sequence-specific DNA binding protein that is found at most previously identified PREs. Our data show that PC and PH often bind to clustered regions within large loci that encode transcription factors which play multiple roles in developmental patterning and in the regulation of cell proliferation. GAF co-localizes with PC and PH to a limited extent, suggesting that GAF is not a necessary component of chromatin at PREs. Finally, the chromosome-association profile of PC and PH changes during development, suggesting that the function of these proteins in the regulation of some of their target genes might be more dynamic than previously anticipated.
Collapse
Affiliation(s)
- Nicolas Nègre
- 1Institute of Human Genetics, Centre national de la recherche scientifique (CNRS), Montpellier Cedex, France
| | - Jérôme Hennetin
- 2Centre de Recherche en Biochimie Macromoléculaire, CNRS, Montpellier, France
| | - Ling V Sun
- 3Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sergey Lavrov
- 1Institute of Human Genetics, Centre national de la recherche scientifique (CNRS), Montpellier Cedex, France
| | - Michel Bellis
- 2Centre de Recherche en Biochimie Macromoléculaire, CNRS, Montpellier, France
| | - Kevin P White
- 3Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Giacomo Cavalli
- 1Institute of Human Genetics, Centre national de la recherche scientifique (CNRS), Montpellier Cedex, France
| |
Collapse
|
35
|
Nakamura M, Runko AP, Sagerström CG. A novel subfamily of zinc finger genes involved in embryonic development. J Cell Biochem 2005; 93:887-95. [PMID: 15449319 DOI: 10.1002/jcb.20255] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
C2H2 zinc finger proteins make up one of the largest protein families in eukaryotic organisms. Recent study in several different systems has identified a set of novel zinc finger proteins that appear to form a distinct subfamily that we have named the NET family. Members of the NET family (Noc, Nlz, Elbow, and Tlp-1) share two protein motifs--a buttonhead box and an Sp motif--with zinc finger proteins from the Sp family. However, the NET family is uniquely characterized by a single atypical C2H2 zinc finger, in contrast to the Sp family that contains three tandem C2H2 fingers. Here, we review current information about the biochemical function and in vivo role for members of this subfamily. In general, NET family proteins are required during embryonic development. They appear to act by regulating transcription, most likely as repressors, although they are unlikely to bind DNA directly. In the future, it will be important to directly test if NET family proteins control transcription of specific target genes, perhaps via interactions with DNA-binding transcription factors, as well as to further explore their function in vivo.
Collapse
Affiliation(s)
- Mako Nakamura
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605-2324, USA
| | | | | |
Collapse
|
36
|
Jacobs HT, Fernández-Ayala DJM, Manjiry S, Kemppainen E, Toivonen JM, O'Dell KMC. Mitochondrial disease in flies. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2005; 1659:190-6. [PMID: 15576051 DOI: 10.1016/j.bbabio.2004.07.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2004] [Accepted: 07/13/2004] [Indexed: 10/26/2022]
Abstract
The Drosophila mutant technical knockout (tko), affecting the mitochondrial protein synthetic apparatus, exhibits respiratory chain deficiency and a phenotype resembling various features of mitochondrial disease in humans (paralytic seizures, deafness, developmental retardation). We are using this mutant to analyse the cellular and genomic targets of mitochondrial dysfunction, and to identify ways in which the phenotype can be alleviated. Transgenic expression of wild-type tko in different patterns in the mutant background reveals critical times and cell-types for production of components of the mitochondrial disease-like phenotype. Mitochondrial bioenergy deficit during the period of maximal growth, as well as in specific parts of the nervous system, appears to be most deleterious. Inbreeding of tko mutant lines results in a systematic improvement in all phenotypic parameters tested. The resulting sub-lines can be used for genetic mapping and transcriptomic analysis, revealing clues as to the genes and pathways that can modify mitochondrial disease-like phenotypes in a model metazoan.
Collapse
Affiliation(s)
- Howard T Jacobs
- Institute of Medical Technology and Tampere University Hospital, University of Tampere, FI-33014, Finland.
| | | | | | | | | | | |
Collapse
|
37
|
Hoyle J, Tang YP, Wiellette EL, Wardle FC, Sive H. nlz gene family is required for hindbrain patterning in the zebrafish. Dev Dyn 2004; 229:835-46. [PMID: 15042707 DOI: 10.1002/dvdy.20001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This study describes the conserved nlz gene family whose members encode unusual zinc finger proteins. In the zebrafish neurectoderm, both nlz1 and the newly isolated nlz2 are expressed in the presumptive hindbrain and midbrain/hindbrain boundary, where expression of nlz1 is dependent on pax2a. In addition, nlz2 is uniquely expressed more anteriorly, in the presumptive midbrain and diencephalon. Overexpression of Nlz proteins during gastrula stages inhibits hindbrain development. In particular, ectopically expressed Nlz1 inhibits formation of future rhombomeres 2 and 3 (r2, r3), whereas neighboring r1 and r4 are not affected. Conversely, simultaneous reduction of Nlz1 and Nlz2 protein function by expression of antisense morpholino-modified oligomers leads to expansion of future r3 and r5, with associated loss of r4. These data indicate that one function of the nlz gene family is to specify or maintain r4 identity, and to limit r3 and r5 during hindbrain formation.
Collapse
Affiliation(s)
- Jacqueline Hoyle
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, Massachusetts, USA
| | | | | | | | | |
Collapse
|
38
|
Runko AP, Sagerström CG. Isolation of nlz2 and characterization of essential domains in Nlz family proteins. J Biol Chem 2004; 279:11917-25. [PMID: 14709556 DOI: 10.1074/jbc.m310076200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In this study, we first cloned nlz2, a second zebrafish member of the nlz-related zinc-finger gene family. nlz2 was expressed together with nlz1 in a broad posterior domain during gastrula stages as well as at the midbrain-hindbrain boundary and in the hindbrain caudal to rhombomere 4 during segmentation. nlz2 was also expressed in regions distinct from nlz1, notably in the forebrain, midbrain, and trunk. Misexpression of nlz2 in zebrafish embryos disrupted gene expression in the rostral hindbrain, similar to the effect of misexpressing nlz1. We next compared the nlz1 and nlz2 sequences to identify and characterize domains conserved within this family. We found a C-terminal domain required for nuclear localization and two conserved domains (the Sp motif and a putative C(2)H(2) zinc finger) required for nlz1 function. We also demonstrate that Nlz1 self-associated via its C terminus, interacted with Nlz2, and bound to histone deacetylases. Last, we found two forms of Nlz1 generated from alternative translation initiation sites in vivo. These forms have distinct activities, apparently depending on the function of the N-terminal Sp motif. Our data demonstrate that nlz2 functions similarly to nlz1 and define conserved domains essential for nuclear localization, self-association, and corepressor binding in this novel family of zinc-finger genes.
Collapse
Affiliation(s)
- Alexander P Runko
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| | | |
Collapse
|
39
|
Runko AP, Sagerström CG. Nlz belongs to a family of zinc-finger-containing repressors and controls segmental gene expression in the zebrafish hindbrain. Dev Biol 2003; 262:254-67. [PMID: 14550789 DOI: 10.1016/s0012-1606(03)00388-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The zebrafish nlz gene has a rostral expression limit at the presumptive rhombomere (r) 3/r4 boundary during gastrula stages, and its expression progressively expands rostrally to encompass both r3 and r2 by segmentation stages, suggesting a role for nlz in hindbrain development. We find that Nlz is a nuclear protein that associates with the corepressor Groucho, suggesting that Nlz acts to repress transcription. Consistent with a role as a repressor, misexpression of nlz causes a loss of gene expression in the rostral hindbrain, likely due to ectopic nlz acting prematurely in this domain, and this repression is accompanied by a partial expansion in the expression domains of r4-specific genes. To interfere with endogenous nlz function, we generated a form of nlz that lacks the Groucho binding site and demonstrate that this construct has a dominant negative effect. We find that interfering with endogenous Nlz function promotes the expansion of r5 and, to a lesser extent, r3 gene expression into r4, leading to a reduction in the size of r4. We conclude that Nlz is a transcriptional repressor that controls segmental gene expression in the hindbrain. Lastly, we identify additional nlz-related genes, suggesting that Nlz belongs to a family of zinc-finger proteins.
Collapse
Affiliation(s)
- Alexander P Runko
- Department of Biochemistry and Molecular Pharmacology, and Program in Neuroscience, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | |
Collapse
|
40
|
Abstract
Many organs including the mammalian lung and vascular system consist of branched tubular networks that transport essential gases or fluids, but the genetic programs that control the development of these complex three-dimensional structures are not well understood. The Drosophila melanogaster tracheal (respiratory) system is a network of interconnected epithelial tubes that transports oxygen and other gases in the body and provides a paradigm of branching morphogenesis. It develops by sequential sprouting of primary, secondary, and terminal branches from an epithelial sac of approximately 80 cells in each body segment of the embryo. Mapping of the cell movements and shape changes during the sprouting process has revealed that distinct mechanisms of epithelial migration and tube formation are used at each stage of branching. Genetic dissection of the process has identified a general program in which a fibroblast growth factor (FGF) and fibroblast growth factor receptor (FGFR) are used repeatedly to control branch budding and outgrowth. At each stage of branching, the mechanisms controlling FGF expression and the downstream signal transduction pathway change, altering the pattern and structure of the branches that form. During terminal branching, FGF expression is regulated by hypoxia, ensuring that tracheal structure matches cellular oxygen need. A branch diversification program operates in parallel to the general budding program: Regional signals locally modify the general program, conferring specific structural features and other properties on individual branches, such as their substrate outgrowth preferences, differences in tube size and shape, and the ability to fuse to other branches to interconnect the network.
Collapse
Affiliation(s)
- Amin Ghabrial
- Howard Hughes Medical Institute, Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305-5307, USA
| | | | | | | |
Collapse
|