301
|
Evolution of the mechanisms and molecular control of endoderm formation. Mech Dev 2007; 124:253-78. [PMID: 17307341 DOI: 10.1016/j.mod.2007.01.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2006] [Revised: 12/24/2006] [Accepted: 01/03/2007] [Indexed: 01/13/2023]
Abstract
Endoderm differentiation and movements are of fundamental importance not only for subsequent morphogenesis of the digestive tract but also to enable normal patterning and differentiation of mesoderm- and ectoderm-derived organs. This review defines the tissues that have been called endoderm in different species, their cellular origin and their movements. We take a comparative approach to ask how signaling pathways leading to embryonic and extraembryonic endoderm differentiation have emerged in different organisms, how they became integrated and point to specific gaps in our knowledge that would be worth filling. Lastly, we address whether the gastrulation movements that lead to endoderm internalization are coupled with its differentiation.
Collapse
|
302
|
Foxb1-drivenCre expression in somites and the neuroepithelium of diencephalon, brainstem, and spinal cord. Genesis 2007; 45:781-7. [DOI: 10.1002/dvg.20356] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
303
|
Cirillo LA, Zaret KS. Specific interactions of the wing domains of FOXA1 transcription factor with DNA. J Mol Biol 2006; 366:720-4. [PMID: 17189638 PMCID: PMC1793999 DOI: 10.1016/j.jmb.2006.11.087] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Revised: 11/28/2006] [Accepted: 11/28/2006] [Indexed: 11/27/2022]
Abstract
FOX (forkhead box) transcription factors have diverse regulatory roles in development, signaling, and longevity, as well as being able to bind stably to target sites in silent chromatin. Crystal structure analysis showed that the FOXA DNA binding domain folds into a helix-turn-helix (HTH) motif flanked on either side by "wings" of polypeptide chain. The wings have the potential to interact with the DNA minor groove along the long axis of the DNA helix, flanking the HTH interactions with the major groove. Diverse FOX family homologs exist, and structural studies with certain DNA target sites suggest that neither of the wing regions are well ordered or provide a stable contribution to DNA target site binding. However, FOXA1 binds certain DNA target sites with high affinity, and as a monomer. To determine whether the wing domains contribute to stable DNA binding, we assessed complexes of FOXA with high and lower affinity DNA target sites by hydroxyl radical footprinting and site-directed mutagenesis. The data revealed clear protections predicted for wing interactions at the high affinity target, but less so at the lower affinity target, indicating that the wing domains stably interact with high affinity DNA sites for FOXA proteins.
Collapse
Affiliation(s)
- Lisa A Cirillo
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | |
Collapse
|
304
|
Tu Q, Brown CT, Davidson EH, Oliveri P. Sea urchin Forkhead gene family: Phylogeny and embryonic expression. Dev Biol 2006; 300:49-62. [PMID: 17081512 DOI: 10.1016/j.ydbio.2006.09.031] [Citation(s) in RCA: 162] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Revised: 09/14/2006] [Accepted: 09/15/2006] [Indexed: 11/21/2022]
Abstract
Transcription factors of the Forkhead (Fox) family have been identified in many metazoans, and play important roles in diverse biological processes. Here we define the set of fox genes present in the sea urchin genome, and survey their usage during development. This genome includes 22 fox genes, only three of which were previously known. Of the 23 fox gene subclasses identified in vertebrate genomes, the Strongylocentrotus purpuratus genome has orthologues of all but four (E, H, R and S). Phylogenetic analysis suggests that one S. purpuratus fox gene is equally related to foxA and foxB of vertebrates; this gene defines a new class. Two other genes appear to be specific to the sea urchin, with respect to the genomes so far sequenced. Fox genes orthologous with those of vertebrates but lacking in arthropod or nematode genomes may be deuterostome-specific (subclasses I, J1, J2, L1, M and Q1), while the majority are pan-bilaterian. All but one of the S. purpuratus fox genes (SpfoxQ1) are expressed during embryogenesis, most in a very specific temporal and spatial manner. The sea urchin fox genes clearly execute many different regulatory functions, and almost all of them participate in the process of embryonic development.
Collapse
Affiliation(s)
- Qiang Tu
- Division of Biology 156-29, California Institute of Technology, Pasadena, CA 91125, USA
| | | | | | | |
Collapse
|
305
|
Kerman BE, Cheshire AM, Andrew DJ. From fate to function: the Drosophila trachea and salivary gland as models for tubulogenesis. Differentiation 2006; 74:326-48. [PMID: 16916373 PMCID: PMC2827874 DOI: 10.1111/j.1432-0436.2006.00095.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Tube formation is a ubiquitous process required to sustain life in multicellular organisms. The tubular organs of adult mammals include the lungs, vasculature, digestive and excretory systems, as well as secretory organs such as the pancreas, salivary, prostate, and mammary glands. Other tissues, including the embryonic heart and neural tube, have requisite stages of tubular organization early in development. To learn the molecular and cellular basis of how epithelial cells are organized into tubular organs of various shapes and sizes, investigators have focused on the Drosophila trachea and salivary gland as model genetic systems for branched and unbranched tubes, respectively. Both organs begin as polarized epithelial placodes, which through coordinated cell shape changes, cell rearrangement, and cell migration form elongated tubes. Here, we discuss what has been discovered regarding the details of cell fate specification and tube formation in the two organs; these discoveries reveal significant conservation in the cellular and molecular events of tubulogenesis.
Collapse
Affiliation(s)
- Bilal E Kerman
- Department of Cell Biology, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205-2196, USA
| | | | | |
Collapse
|
306
|
Nakada C, Satoh S, Tabata Y, Arai KI, Watanabe S. Transcriptional repressor foxl1 regulates central nervous system development by suppressing shh expression in zebra fish. Mol Cell Biol 2006; 26:7246-57. [PMID: 16980626 PMCID: PMC1592895 DOI: 10.1128/mcb.00429-06] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We identified zebra fish forkhead transcription factor l1 (zfoxl1) as a gene strongly expressed in neural tissues such as midbrain, hindbrain, and the otic vesicle at the early embryonic stage. Loss of the function of zfoxl1 effected by morpholino antisense oligonucleotide resulted in defects in midbrain and eye development, and in that of formation of the pectoral fins. Interestingly, ectopic expression of shh in the midbrain and elevated pax2a expression in the optic stalk were observed in foxl1 MO-injected embryos. In contrast, expression of pax6a, which is negatively regulated by shh, was suppressed in the thalamus and pretectum regions, supporting the idea of augmentation of the shh signaling pathway by suppression of foxl1. Expression of zfoxl1-EnR (repressing) rather than zfoxl1-VP16 (activating) resulted in a phenotype similar to that induced by foxl1-mRNA, suggesting that foxl1 may act as a transcriptional repressor of shh in zebra fish embryos. Supporting this notion, foxl1 suppressed isolated 2.7-kb shh promoter activity in PC12 cells, and the minimal region of foxl1 required for its transcriptional repressor activity showed strong homology with the groucho binding motif, which is found in genes encoding various homeodomain proteins. In view of all of our data taken together, we propose zfoxl1 to be a novel regulator of neural development that acts by suppressing shh expression.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Biomarkers
- Brain/cytology
- Brain/embryology
- Brain/metabolism
- Cells, Cultured
- Embryo, Nonmammalian/cytology
- Embryo, Nonmammalian/embryology
- Fibroblasts/metabolism
- Forkhead Transcription Factors/chemistry
- Forkhead Transcription Factors/isolation & purification
- Forkhead Transcription Factors/metabolism
- Gastrula/metabolism
- Gene Expression Regulation, Developmental
- Hedgehog Proteins
- Mice
- Molecular Sequence Data
- NIH 3T3 Cells
- Oligonucleotides, Antisense/metabolism
- PC12 Cells
- Promoter Regions, Genetic/genetics
- Protein Binding
- Protein Structure, Tertiary
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Repressor Proteins/chemistry
- Repressor Proteins/isolation & purification
- Repressor Proteins/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription, Genetic
- Zebrafish/embryology
- Zebrafish/metabolism
- Zebrafish Proteins/chemistry
- Zebrafish Proteins/isolation & purification
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Chisako Nakada
- Department of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | |
Collapse
|
307
|
Kelly LE, Nekkalapudi S, El-Hodiri HM. Expression of the forkhead transcription factor FoxN4 in progenitor cells in the developing Xenopus laevis retina and brain. Gene Expr Patterns 2006; 7:233-8. [PMID: 17110173 PMCID: PMC1986660 DOI: 10.1016/j.modgep.2006.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Revised: 09/14/2006] [Accepted: 10/02/2006] [Indexed: 01/07/2023]
Abstract
Forkhead proteins are involved in gene regulation in a large variety of developmental situations. Several forkhead gene products are expressed in the developing eye and brain. Here we characterize the expression of FoxN4 during Xenopus development. We report that FoxN4 is expressed in the eye from the earliest stages of specification through retinal maturation. FoxN4 is also expressed in the pallium, optic tectum, isthmus, reticular formation, and in cells lining the ventricle of the tadpole brain.
Collapse
Affiliation(s)
- Lisa E Kelly
- Center for Molecular and Human Genetics, Columbus Children's Research Institute, Columbus, OH 43205, USA
| | | | | |
Collapse
|
308
|
Abrams EW, Mihoulides WK, Andrew DJ. Fork head and Sage maintain a uniform and patent salivary gland lumen through regulation of two downstream target genes, PH4alphaSG1 and PH4alphaSG2. Development 2006; 133:3517-27. [PMID: 16914497 DOI: 10.1242/dev.02525] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
(Fkh) is required to block salivary gland apoptosis, internalize salivary gland precursors, prevent expression of duct genes in secretory cells and maintain expression of CrebA, which is required for elevated secretory function. Here, we characterize two new Fkh-dependent genes: PH4alphaSG1 and PH4alphaSG2. We show through in vitro DNA-binding studies and in vivo expression assays that Fkh cooperates with the salivary gland-specific bHLH protein Sage to directly regulate expression of PH4alphaSG2, as well as sage itself, and to indirectly regulate expression of PH4alphaSG1. PH4alphaSG1 and PH4alphaSG2 encode alpha-subunits of resident ER enzymes that hydroxylate prolines in collagen and other secreted proteins. We demonstrate that salivary gland secretions are altered in embryos missing function of PH4alphaSG1 and PH4alphaSG2; secretory content is reduced and shows increased electron density by TEM. Interestingly, the altered secretory content results in regions of tube dilation and constriction, with intermittent tube closure. The regulation studies and phenotypic characterization of PH4alphaSG1 and PH4alphaSG2 link Fkh, which initiates tube formation, to the maintenance of an open and uniformly sized secretory tube.
Collapse
Affiliation(s)
- Elliott W Abrams
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205-2196, USA
| | | | | |
Collapse
|
309
|
Wijchers PJEC, Burbach JPH, Smidt MP. In control of biology: of mice, men and Foxes. Biochem J 2006; 397:233-46. [PMID: 16792526 PMCID: PMC1513289 DOI: 10.1042/bj20060387] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 05/04/2006] [Accepted: 05/05/2006] [Indexed: 12/11/2022]
Abstract
Forkhead proteins comprise a highly conserved family of transcription factors, named after the original forkhead gene in Drosophila. To date, over 100 forkhead genes have been identified in a large variety of species, all sharing the evolutionary conserved 'forkhead' DNA-binding domain, and the cloning and characterization of forkhead genes have continued in recent years. Forkhead transcription factors regulate the expression of countless genes downstream of important signalling pathways in most, if not all, tissues and cell types. Recent work has provided novel insights into the mechanisms that contribute to their functional diversity, including functional protein domains and interactions of forkheads with other transcription factors. Studies using loss- and gain-of-function models have elucidated the role of forkhead factors in developmental biology and cellular functions such as metabolism, cell division and cell survival. The importance of forkhead transcription factors is underlined by the developmental defects observed in mutant model organisms, and multiple human disorders and cancers which can be attributed to mutations within members of the forkhead gene family. This review provides a comprehensive overview of current knowledge on forkhead transcription factors, from structural organization and regulatory mechanisms to cellular and developmental functions in mice and humans. Finally, we will discuss how novel insights gained from involvement of 'Foxes' in the mechanisms underlying human pathology may create new opportunities for treatment strategies.
Collapse
Key Words
- cell cycle
- development
- forkhead
- fox
- immunoregulation
- transcription factor
- cbp, creb (camp-response-element-binding protein)-binding protein
- ccnb, cyclin b
- cdk, cyclin-dependent kinase
- cki, cdk inhibitor
- dyrk1a, dual-specificity tyrosine-phosphorylated and -regulated kinase 1a
- er, oestrogen receptor
- fha, forkhead-associated domain
- fm, foxh1 motif
- fox, forkhead box
- gadd45a, growth arrest and dna-damage-inducible protein 45α
- hdac, histone deacetylase
- iκb, inhibitory κb
- ikkβ, iκb kinase β
- mh domain, mothers against decapentaplegic homology domain
- nf-κb, nuclear factor κb
- nls, nuclear localization signal
- pkb, protein kinase b
- plk-1, polo-like kinase 1
- scf, skp2/cullin/f-box
- sgk, serum- and glucocorticoid-induced protein kinase
- smad, similar to mothers against decapentaplegic
- sid, smad-interaction domain
- sim, smad-interaction motif
- tgfβ, transforming growth factor β
Collapse
Affiliation(s)
- Patrick J E C Wijchers
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands.
| | | | | |
Collapse
|
310
|
Morán E, Jiménez G. The tailless nuclear receptor acts as a dedicated repressor in the early Drosophila embryo. Mol Cell Biol 2006; 26:3446-54. [PMID: 16611987 PMCID: PMC1447415 DOI: 10.1128/mcb.26.9.3446-3454.2006] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tailless is an orphan nuclear receptor that controls terminal body patterning in Drosophila. Genetic analyses have revealed both positive and negative regulatory interactions of Tailless with various target genes, leading to the idea that, like many other nuclear receptors, Tailless mediates both activation and repression of transcription. In this paper, we have examined the consequences of converting Tailless into an obligate repressor and compared the activities of the resulting protein with those of wild-type Tailless. We find that this repressor form of Tailless behaves like the intact protein in gain- and loss-of-function experiments, being sufficient to support normal embryonic development and establish accurate patterns of gene expression even for positive Tailless targets such as hunchback and brachyenteron. This suggests that Tailless functions exclusively as a transcriptional repressor in the embryo and that the observed positive interactions of Tailless with specific targets are secondary effects involving repression of repressors. We provide evidence that knirps is one such repressor gene acting between Tailless and its indirect positive targets. Finally, our results indicate that Tailless exerts an active mechanism of repression via its ligand-binding domain and that this activity is largely independent of the activation function 2 (AF2) motif characteristic of most nuclear receptors.
Collapse
Affiliation(s)
- Erica Morán
- Institut de Biologia Molecular de Barcelona-CSIC, Parc Científic de Barcelona, Josep Samitier 1-5, Barcelona 08028, Spain
| | | |
Collapse
|
311
|
Kurth I, Hentschke M, Hentschke S, Borgmeyer U, Gal A, Hübner C. The forkhead transcription factor Foxi1 directly activates the AE4 promoter. Biochem J 2006; 393:277-83. [PMID: 16159312 PMCID: PMC1383686 DOI: 10.1042/bj20051094] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Intercalated cells are highly specialized cells within the renal collecting duct epithelium and play an important role in systemic acid-base homoeostasis. Whereas type A intercalated cells secrete protons via an apically localized H+-ATPase, type B intercalated cells secrete HCO3-. Type B intercalated cells specifically express the HCO3-/Cl- exchanger AE4 (anion exchanger 4), encoded by Slc4a9. Mice with a targeted disruption of the gene for the forkhead transcription factor Foxi1 display renal tubular acidosis due to an intercalated cell-differentiation defect. Collecting duct cells in these mice are characterized by the absence of inter-calated cell markers including AE4. To test whether Slc4a9 is a direct target gene of Foxi1, an AE4 promoter construct was generated for a cell-based reporter gene assay. Co-transfection with the Foxi1 cDNA resulted in an approx. 100-fold activation of the AE4 promoter construct. By truncating the AE4 promoter at the 5'-end, we demonstrate that a fragment of approx. 462 bp upstream of the transcription start point is sufficient to mediate activation by Foxi1. Sequence analysis of this region revealed at least eight potential binding sites for Foxi1 in both sense and antisense orientation. Only one element was bound by recombinant Foxi1 protein in bandshift assays. Mutation of this site abolished both binding in bandshift assays and transcriptional activation by co-transfection of Foxi1 in the reporter gene assay. We thus identify the AE4 promoter as a direct target of Foxi1.
Collapse
Affiliation(s)
- Ingo Kurth
- *Institut für Humangenetik, Universitätsklinikum Hamburg-Eppendorf, Butenfeld 42, 22529 Hamburg, Germany
| | - Moritz Hentschke
- *Institut für Humangenetik, Universitätsklinikum Hamburg-Eppendorf, Butenfeld 42, 22529 Hamburg, Germany
| | - Suna Hentschke
- *Institut für Humangenetik, Universitätsklinikum Hamburg-Eppendorf, Butenfeld 42, 22529 Hamburg, Germany
| | - Uwe Borgmeyer
- †Zentrum für Molekulare Neurobiologie Hamburg (ZMNH), Universität Hamburg, Falkenried 94, 20246 Hamburg, Germany
| | - Andreas Gal
- *Institut für Humangenetik, Universitätsklinikum Hamburg-Eppendorf, Butenfeld 42, 22529 Hamburg, Germany
| | - Christian A. Hübner
- *Institut für Humangenetik, Universitätsklinikum Hamburg-Eppendorf, Butenfeld 42, 22529 Hamburg, Germany
- To whom correspondence should be addressed (email )
| |
Collapse
|
312
|
Kraus Y, Technau U. Gastrulation in the sea anemone Nematostella vectensis occurs by invagination and immigration: an ultrastructural study. Dev Genes Evol 2006; 216:119-32. [PMID: 16416137 DOI: 10.1007/s00427-005-0038-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2005] [Accepted: 10/17/2005] [Indexed: 12/28/2022]
Abstract
The sea anemone Nematostella vectensis has recently been established as a new model system for the understanding of the evolution of developmental processes. In particular, the evolutionary origin of gastrulation and its molecular regulation are the subject of intense investigation. However, while molecular data are rapidly accumulating, no detailed morphological data exist describing the process of gastrulation. Here, we carried out an ultrastructural study of different stages of gastrulation in Nematostella using transmission electron microscope and scanning electron microscopy techniques. We show that presumptive endodermal cells undergo a change in cell shape, reminiscent of the bottle cells known from vertebrates and several invertebrates. Presumptive endodermal cells organize into a field, the pre-endodermal plate, which undergoes invagination. In parallel, the endodermal cells decrease their apical cell contacts but remain loosely attached to each other. Hence, during early gastrulation they display an incomplete epithelial-mesenchymal transition (EMT). At a late stage of gastrulation, the cells eventually detach and fill the interior of the blastocoel as mesenchymal cells. This shows that gastrulation in Nematostella occurs by a combination of invagination and late immigration involving EMT. The comparison with molecular expression studies suggests that cells expressing snailA undergo EMT and become endodermal, whereas forkhead/brachyury expressing cells at the ectodermal margin of the blastopore retain their epithelial integrity throughout gastrulation.
Collapse
Affiliation(s)
- Yulia Kraus
- Department of Evolutionary Biology, Biological Faculty, Moscow State University, 199992 Moscow, Russia.
| | | |
Collapse
|
313
|
Bagheri-Fam S, Barrionuevo F, Dohrmann U, Günther T, Schüle R, Kemler R, Mallo M, Kanzler B, Scherer G. Long-range upstream and downstream enhancers control distinct subsets of the complex spatiotemporal Sox9 expression pattern. Dev Biol 2006; 291:382-97. [PMID: 16458883 DOI: 10.1016/j.ydbio.2005.11.013] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2005] [Revised: 11/09/2005] [Accepted: 08/29/2005] [Indexed: 11/20/2022]
Abstract
SOX9 is an evolutionary conserved transcription factor that is expressed in a variety of tissues, with essential functions in cartilage, testis, heart, glial cell, inner ear and neural crest development. By comparing human and pufferfish genomic sequences, we previously identified eight highly conserved sequence elements between 290 kb 5' and 450 kb 3' to human SOX9. In this study, we assayed the regulatory potential of elements E1 to E7 in transgenic mice using a lacZ reporter gene driven by a 529 bp minimal mouse Sox9 promoter. We found that three of these elements and the Sox9 promoter control distinct subsets of the tissue-specific expression pattern of Sox9. E3, located 251 kb 5' to SOX9, directs lacZ expression to cranial neural crest cells and to the inner ear. E1 is located 28 kb 5' to SOX9 and controls expression in the node, notochord, gut, bronchial epithelium and pancreas. Transgene expression in the neuroectoderm is mediated by E7, located 95 kb 3' to SOX9, which regulates expression in the telencephalon and midbrain, and by the Sox9 minimal promoter which controls expression in the ventral spinal cord and hindbrain. We show that E3-directed reporter gene expression in neural crest cells of the first but not of the second and third pharyngeal arch is dependent on beta-catenin, revealing a complex regulation of Sox9 in cranial neural crest cells. Moreover, we identify and discuss highly conserved transcription factor binding sites within enhancer E3 that are in good agreement with current models for neural crest and inner ear development. Finally, we identify enhancer E1 as a cis-regulatory element conserved between vertebrates and invertebrates, indicating that some cis-regulatory sequences that control developmental genes in vertebrates might be phylogenetically ancient.
Collapse
Affiliation(s)
- Stefan Bagheri-Fam
- Institute of Human Genetics and Anthropology, University of Freiburg, Breisacherstr. 33, D-79106 Freiburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
314
|
Lee HC, Huang HY, Lin CY, Chen YH, Tsai HJ. Foxd3 mediates zebrafish myf5 expression during early somitogenesis. Dev Biol 2006; 290:359-72. [PMID: 16386728 DOI: 10.1016/j.ydbio.2005.11.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2005] [Revised: 11/17/2005] [Accepted: 11/22/2005] [Indexed: 10/25/2022]
Abstract
Myf5, one of the basic helix-loop-helix transcription factors, controls muscle differentiation and is expressed in somites during early embryogenesis. However, the transcription factors bound to the cis-elements of myf5 are poorly understood. In this study, we used the yeast one-hybrid assay and found that Forkhead box d3 (Foxd3) interacted specifically with the -82/-62 cassette, a key element directing somite-specific expression of myf5. The dual-luciferase assay revealed that the expression of Foxd3 potently transactivated the myf5 promoter. Knocking down foxd3 with morpholino oligonucleotide (MO) resulted in a dramatic down-regulation of myf5 in somites and adaxial cells but not in the presomitic mesoderm. On the other hand, myod expression remained unchanged in foxd3 morphants. Foxd3 mediation of myf5 expression is stage-dependent, maintaining myf5 expression in the somites and adaxial cells during the 7- to 18-somite stage. Furthermore, in the pax3 morphant, the expression of foxd3 was down-regulated greatly and the expression of myf5 was similar to that of the foxd3 morphant. Co-injection of foxd3 mRNA and pax3-MO1 greatly restored the expression of myf5 in the somites and adaxial cells, suggesting that pax3 induces foxd3 expression, which then induces the expression of myf5. This report is the first study to show that Foxd3, a well-known regulator in neural crest development, is also involved in myf5 regulation.
Collapse
Affiliation(s)
- Hung-Chieh Lee
- Institute of Molecular and Cellular Biology, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 106, Taiwan
| | | | | | | | | |
Collapse
|
315
|
Wijchers PJEC, Hoekman MFM, Burbach JPH, Smidt MP. Identification of forkhead transcription factors in cortical and dopaminergic areas of the adult murine brain. Brain Res 2006; 1068:23-33. [PMID: 16376864 DOI: 10.1016/j.brainres.2005.11.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Revised: 11/02/2005] [Accepted: 11/10/2005] [Indexed: 11/30/2022]
Abstract
The murine forkhead family of transcription factors consists of over 30 members, the vast majority of which is important in embryonic development. Implicated in processes such as proliferation, differentiation and survival, forkhead factors show highly restricted expression patterns. In search for forkhead genes expressed in specific neural systems, we identified multiple family members. We performed a detailed expression analysis for Foxj2, Foxk1 and the murine orthologue of the human ILF1 gene, which show a remarkable preference for complex cortical structures. In addition, a comprehensive examination of forkhead gene expression in dopamine neurons of the ventral tegmental area and substantia nigra pars compacta, revealed Ilf1 as a novel transcriptional regulator in midbrain dopamine neurons. These forkhead transcription factors may play a role in maintenance and survival of developing and adult neurons.
Collapse
Affiliation(s)
- Patrick J E C Wijchers
- Rudolf Magnus Institute of Neuroscience, Department of Pharmacology and Anatomy, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | | | | | | |
Collapse
|
316
|
Yan J, Xu L, Crawford G, Wang Z, Burgess SM. The forkhead transcription factor FoxI1 remains bound to condensed mitotic chromosomes and stably remodels chromatin structure. Mol Cell Biol 2006; 26:155-68. [PMID: 16354687 PMCID: PMC1317626 DOI: 10.1128/mcb.26.1.155-168.2006] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2005] [Revised: 08/04/2005] [Accepted: 10/03/2005] [Indexed: 11/20/2022] Open
Abstract
All forkhead (Fox) proteins contain a highly conserved DNA binding domain whose structure is remarkably similar to the winged-helix structures of histones H1 and H5. Little is known about Fox protein binding in the context of higher-order chromatin structure in living cells. We created a stable cell line expressing FoxI1-green fluorescent protein (GFP) or FoxI1-V5 fusion proteins under control of the reverse tetracycline-controlled transactivator doxycycline inducible system and found that unlike most transcription factors, FoxI1 remains bound to the condensed chromosomes during mitosis. To isolate DNA fragments directly bound by the FoxI1 protein within living cells, we performed chromatin immunoprecipitation assays (ChIPs) with antibodies to either enhanced GFP or the V5 epitope and subcloned the FoxI1-enriched DNA fragments. Sequence analyses indicated that 88% (106/121) of ChIP sequences contain the consensus binding sites for all Fox proteins. Testing ChIP sequences with a quantitative DNase I hypersensitivity assay showed that FoxI1 created stable DNase I sensitivity changes in condensed chromosomes. The majority of ChIP targets and random targets increased in resistance to DNase I in FoxI1-expressing cells, but a small number of targets became more accessible to DNase I. Consistently, the accessibility of micrococcal nuclease to chromatin was generally inhibited. Micrococcal nuclease partial digestion generated a ladder in which all oligonucleosomes were slightly longer than those observed with the controls. On the basis of these findings, we propose that FoxI1 is capable of remodeling chromatin higher-order structure and can stably create site-specific changes in chromatin to either stably create or remove DNase I hypersensitive sites.
Collapse
Affiliation(s)
- Jizhou Yan
- National Human Genome Research Institute, Genome Technology Branch, NIH, Bldg. 50, Rm. 5537, 50 South Dr., Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
317
|
Kolesnikov T, Beckendorf SK. NETRIN and SLIT guide salivary gland migration. Dev Biol 2005; 284:102-11. [PMID: 15950216 DOI: 10.1016/j.ydbio.2005.04.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Revised: 04/26/2005] [Accepted: 04/29/2005] [Indexed: 11/18/2022]
Abstract
Directed migration is pivotal for the proper placement and function of nearly all organs. The majority of known guidance molecules involved in directed migration have been identified from studies of migrating axons during nervous system development. Here, we show that at least two of these axon guidance molecules, NETRIN and SLIT, act through their canonical receptors, to guide Drosophila embryonic salivary glands. NETRIN serves as a chemo-attractant while SLIT functions antagonistically to NETRIN as a chemo-repellent during salivary gland migration. CNS midline expression of both NETRIN and SLIT directs the glands to move unswervingly parallel to the CNS. NETRIN expression is also required in the visceral mesoderm, along which the glands move during their migration. We propose that analogous to axon guidance, a balance between chemo-attractants and chemo-repellents is required for the proper migratory path of the developing salivary glands.
Collapse
Affiliation(s)
- Tereza Kolesnikov
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
318
|
Scott KL, Plon SE. CHES1/FOXN3 interacts with Ski-interacting protein and acts as a transcriptional repressor. Gene 2005; 359:119-26. [PMID: 16102918 DOI: 10.1016/j.gene.2005.06.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2005] [Revised: 05/26/2005] [Accepted: 06/03/2005] [Indexed: 11/29/2022]
Abstract
Checkpoint Suppressor 1 (CHES1; FOXN3) encodes a member of the forkhead/winged-helix transcription factor family. The human CHES1 cDNA was originally identified by its ability to function as a high-copy suppressor of multiple checkpoint mutants of Saccharomyces cerevisiae. Accumulating expression profile data suggest that CHES1 plays a role in tumorigenicity and responses to cancer treatments, though nothing is known regarding the transcriptional function of CHES1 or other FOXN proteins in human cells. In this report, we find that the carboxyl terminus of CHES1 fused to a heterologous DNA binding domain consistently represses reporter gene transcription in cell lines derived from tumor tissues. Using a cytoplasmic two-hybrid screening approach, we find that this portion of CHES1 interacts with Ski-interacting protein (SKIP; NCoA-62), which is a transcriptional co-regulator known to associate with repressor complexes. We verify this interaction through co-immunoprecipitation experiments performed in mammalian cells. Further analysis of the CHES1/SKIP interaction indicates that CHES1 binds to a region within the final 66 hydrophobic residues of SKIP thus defining a new protein-protein interaction domain of SKIP. These data suggest that CHES1 recruits SKIP to repress genes important for tumorigenesis and the response to cancer treatments.
Collapse
Affiliation(s)
- Kenneth L Scott
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | | |
Collapse
|
319
|
Abstract
Epithelial and endothelial tubes come in various shapes and sizes and form the basic units of many tubular organs. During embryonic development, single unbranched tubes as well as highly branched networks of tubes form from simple sheets of cells by several morphogenic movements. Studies of tube formation in the Drosophila embryo have greatly advanced our understanding of the cellular and molecular mechanisms by which tubes are formed. This review highlights recent progress on formation of the hindgut, Malpighian tubules, proventriculus, salivary gland, and trachea of the Drosophila embryo, focusing on the cellular events that form each tube and their genetic requirements.
Collapse
Affiliation(s)
- Monn Monn Myat
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, New York 10021, USA.
| |
Collapse
|
320
|
Heglind M, Cederberg A, Aquino J, Lucas G, Ernfors P, Enerbäck S. Lack of the central nervous system- and neural crest-expressed forkhead gene Foxs1 affects motor function and body weight. Mol Cell Biol 2005; 25:5616-25. [PMID: 15964817 PMCID: PMC1157007 DOI: 10.1128/mcb.25.13.5616-5625.2005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To gain insight into the expression pattern and functional importance of the forkhead transcription factor Foxs1, we constructed a Foxs1-beta-galactosidase reporter gene "knock-in" (Foxs1beta-gal/beta-gal) mouse, in which the wild-type (wt) Foxs1 allele has been inactivated and replaced by a beta-galactosidase reporter gene. Staining for beta-galactosidase activity reveals an expression pattern encompassing neural crest-derived cells, e.g., cranial and dorsal root ganglia as well as several other cell populations in the central nervous system (CNS), most prominently the internal granule layer of cerebellum. Other sites of expression include the lachrymal gland, outer nuclear layer of retina, enteric ganglion neurons, and a subset of thalamic and hypothalamic nuclei. In the CNS, blood vessel-associated smooth muscle cells and pericytes stain positive for Foxs1. Foxs1beta-gal/beta-gal mice perform significantly better (P < 0.01) on a rotating rod than do wt littermates. We have also noted a lower body weight gain (P < 0.05) in Foxs1beta-gal/lbeta-gal males on a high-fat diet, and we speculate that dorsomedial hypothalamic neurons, expressing Foxs1, could play a role in regulating body weight via regulation of sympathetic outflow. In support of this, we observed increased levels of uncoupling protein 1 mRNA in Foxs1beta-gal/beta-gal mice. This points toward a role for Foxs1 in the integration and processing of neuronal signals of importance for energy turnover and motor function.
Collapse
Affiliation(s)
- Mikael Heglind
- Medical Genetics, Department of Medical Biochemistry, Göteborg University, Medicinareg. 9A, Box 440, SE 405 30 Göteborg, Sweden
| | | | | | | | | | | |
Collapse
|
321
|
Abstract
The discovery of homeobox gene clusters led us to realize that the mechanisms for body patterning and other developmental programs are evolutionally-conserved in vertebrates and invertebrates. The endoderm contributes to the lining of the gut and associated organs such as the liver and pancreas, which are critical for physiological functions. Our knowledge of endoderm development is limited; however, recent studies suggest that cooperation between the HNF3/Fork head and GATA transcription factors is crucial for endoderm specification. It is necessary to further understand the mechanism through which cells become functionally organized. Molecular genetic analyses of the Drosophila endoderm would provide insights into this issue. During proventriculus morphogenesis, a simple epithelial tube is folded into a functional multilayered structure, while two functions of midgut copper cells (i.e. copper absorption and acid secretion) can be easily visualized. The homeobox gene defective proventriculus (dve) plays key roles in these functional specifications.
Collapse
Affiliation(s)
- Hideki Nakagoshi
- Graduate School of Natural and Science Technology, Okayama University, 3-1-1 Tsushima-naka, Okayama 700-8530, Japan.
| |
Collapse
|
322
|
Streubel B, Vinatzer U, Lamprecht A, Raderer M, Chott A. T(3;14)(p14.1;q32) involving IGH and FOXP1 is a novel recurrent chromosomal aberration in MALT lymphoma. Leukemia 2005; 19:652-8. [PMID: 15703784 DOI: 10.1038/sj.leu.2403644] [Citation(s) in RCA: 242] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The three chromosomal translocations t(11;18)(q21;q21), t(14;18)(q32;q21), and t(1;14)(p22;q32) are associated with MALT lymphoma. In a case of MALT lymphoma of the thyroid, we observed t(3;14)(p14.1;q32) by cytogenetic analysis. Fluorescence in situ hybridization studies showed that the immunoglobulin heavy chain locus (IGH) was rearranged on chromosome 14. Long-distance inverse polymerase chain reaction identified FOXP1 as the partner gene on chromosome 3. To determine the frequency of the t(3;14)(p14.1;q32), two fluorescence in situ hybridization assays were established to screen 91 MALT lymphomas, all of which were negative for the above-mentioned three translocations, and eight splenic and six nodal marginal zone lymphomas. Overall, nine MALT lymphomas (10%) harbored t(3;14)(p14.1;q32) comprising tumors of the thyroid (three of six), ocular adnexa (four of 20), and skin (two of 20), whereas those of the stomach (n = 20), salivary gland (n = 20), and lung (n = 5) were negative as well as the splenic and nodal marginal zone lymphomas. Most t(3;14)(p14.1;q32) + MALT lymphomas harbored additional genetic abnormalities, such as trisomy 3. Further studies revealed that the three known translocations and t(3;14)(p14.1;q32) are mutually exclusive. Real-time quantitative reverse transcriptase polymerase chain reaction showed upregulation of FOXP1 in cases with t(3;14)(p14.1;q32) or trisomy 3. This study identifies FOXP1 as a new translocation partner of IGH in a site-dependent subset of MALT lymphomas.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Aged
- Aged, 80 and over
- B-Cell CLL-Lymphoma 10 Protein
- Caspases
- Chromosomes, Human, Pair 14
- Chromosomes, Human, Pair 3
- Cloning, Molecular
- Female
- Forkhead Transcription Factors
- Humans
- Immunoglobulin Heavy Chains/genetics
- In Situ Hybridization, Fluorescence
- Lymphoma, B-Cell, Marginal Zone/genetics
- Male
- Middle Aged
- Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein
- Neoplasm Proteins/genetics
- Repressor Proteins/genetics
- Translocation, Genetic
- Trisomy
Collapse
Affiliation(s)
- B Streubel
- Department of Pathology, Vienna General Hospital, Medical University of Vienna, Vienna, Austria.
| | | | | | | | | |
Collapse
|
323
|
Descipio C, Schneider L, Young TL, Wasserman N, Yaeger D, Lu F, Wheeler PG, Williams MS, Bason L, Jukofsky L, Menon A, Geschwindt R, Chudley AE, Saraiva J, Schinzel AAGL, Guichet A, Dobyns WE, Toutain A, Spinner NB, Krantz ID. Subtelomeric deletions of chromosome 6p: molecular and cytogenetic characterization of three new cases with phenotypic overlap with Ritscher-Schinzel (3C) syndrome. Am J Med Genet A 2005; 134A:3-11. [PMID: 15704124 DOI: 10.1002/ajmg.a.30573] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We have identified six children in three families with subtelomeric deletions of 6p25 and a recognizable phenotype consisting of ptosis, posterior embryotoxon, optic nerve abnormalities, mild glaucoma, Dandy-Walker malformation, hydrocephalus, atrial septal defect, patent ductus arteriosus, and mild mental retardation. There is considerable clinical overlap between these children and individuals with the Ritscher-Schinzel (or cranio-cerebello-cardiac (3C)) syndrome (OMIM #220210). Clinical features of 3C syndrome include craniofacial anomalies (macrocephaly, prominent forehead and occiput, foramina parietalia, hypertelorism, down-slanting palpebral fissures, ocular colobomas, depressed nasal bridge, narrow or cleft palate, and low-set ears), cerebellar malformations (variable manifestations of a Dandy-Walker malformation with moderate mental retardation), and cardiac defects (primarily septal defects). Since the original report, over 25 patients with 3C syndrome have been reported. Recessive inheritance has been postulated based on recurrence in siblings born to unaffected parents and parental consanguinity in two familial cases. Molecular and cytogenetic mapping of the 6p deletions in these three families with subtelomeric deletions of chromosome 6p have defined a 1.3 Mb minimally deleted critical region. To determine if 6p deletions are common in 3C syndrome, we analyzed seven unrelated individuals with 3C syndrome for deletions of this region. Three forkhead genes (FOXF1 and FOXQ1 from within the critical region, and FOXC1 proximal to this region) were evaluated as potential candidate disease genes for this disorder. No deletions or disease-causing mutations were identified.
Collapse
MESH Headings
- Abnormalities, Multiple/genetics
- Abnormalities, Multiple/pathology
- Cerebellum/abnormalities
- Child
- Child, Preschool
- Chromosome Banding
- Chromosome Deletion
- Chromosomes, Human, Pair 16/genetics
- Chromosomes, Human, Pair 6/genetics
- Craniofacial Abnormalities/pathology
- Diagnosis, Differential
- Family Health
- Fatal Outcome
- Female
- Fetal Death
- Heart Defects, Congenital/pathology
- Humans
- In Situ Hybridization, Fluorescence
- Karyotyping
- Male
- Phenotype
- Syndrome
- Telomere/genetics
- Translocation, Genetic
Collapse
Affiliation(s)
- Cheryl Descipio
- Division of Human Genetics and Molecular Biology, The Children's Hospital of Philadelphia, and The University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
324
|
Affiliation(s)
- Roong Zhao
- Department of Cell Biology Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | |
Collapse
|
325
|
Okumura T, Matsumoto A, Tanimura T, Murakami R. An endoderm-specific GATA factor gene, dGATAe, is required for the terminal differentiation of the Drosophila endoderm. Dev Biol 2005; 278:576-86. [PMID: 15680371 DOI: 10.1016/j.ydbio.2004.11.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2004] [Revised: 11/16/2004] [Accepted: 11/16/2004] [Indexed: 11/24/2022]
Abstract
GATA factors play an essential role in endodermal specification in both protostomes and deuterostomes. In Drosophila, the GATA factor gene serpent (srp) is critical for differentiation of the endoderm. However, the expression of srp disappears around stage 11, which is much earlier than overt differentiation occurs in the midgut, an entirely endodermal organ. We have identified another endoderm-specific Drosophila GATA factor gene, dGATAe. Expression of dGATAe is first detected at stage 8 in the endoderm, and its expression continues in the endodermal midgut throughout the life cycle. srp is required for expression of dGATAe, and misexpression of srp resulted in ectopic dGATAe expression. Embryos that either lacked dGATAe or were injected with double-stranded RNA (dsRNA) corresponding to dGATAe failed to express marker genes that are characteristic of differentiated midgut. Conversely, overexpression of dGATAe induced ectopic expression of endodermal markers even in the absence of srp activity. Transfection of the dGATAe cDNA also induced endodermal markers in Drosophila S2 cells. These studies provide an outline of the genetic pathway that establishes the endoderm in Drosophila. This pathway is triggered by sequential signaling through the maternal torso gene, a terminal gap gene, huckebein (hkb), and finally, two GATA factor genes, srp and dGATAe.
Collapse
Affiliation(s)
- Takashi Okumura
- Department of Physics, Biology, and Informatics, Yamaguchi University, Yamaguchi 753-8512, Japan
| | | | | | | |
Collapse
|
326
|
Abstract
The forkhead gene family, named after the founding gene member in Drosophila, is characterized by a unique DNA-binding domain. This so-called forkhead box encodes a winged-helix DNA-binding motif, the name of which describes the structure of the domain when bound to DNA. The three Fox (forkhead box) group A genes, Foxa1, Foxa2 and Foxa3, are expressed in embryonic endoderm, the germ layer that gives rise to the digestive system, and contribute to the specification of the pancreas and the regulation of glucose homoeostasis. Deletion of the Foxa2 gene in pancreatic beta-cells in mice results in a phenotype resembling PHHI (persistent hyperinsulinaemic hypoglycaemia of infancy). Molecular analyses have demonstrated that Foxa2 is an important regulator of the genes encoding Sur1, Kir6.2 and Schad (short chain L-3-hydroxyacyl-CoA dehydrogenase), mutation of which causes PHHI in humans. Foxa1 was shown to be an essential activator of glucagon gene expression in vivo. An additional winged-helix protein, Foxo1, contributes to pancreatic beta-cell function by regulating the Pdx1 gene, which is required for pancreatic development in cooperation with Foxa2.
Collapse
Affiliation(s)
- Kristen A Lantz
- Department of Genetics, University of Pennsylvania Medical School, 560 Clinical Research Building, 415 Curie Blvd, Philadelphia, PA 19104, USA
| | | |
Collapse
|
327
|
Lesné S, Gabriel C, Nelson DA, White E, Mackenzie ET, Vivien D, Buisson A. Akt-dependent expression of NAIP-1 protects neurons against amyloid-{beta} toxicity. J Biol Chem 2005; 280:24941-7. [PMID: 15797869 DOI: 10.1074/jbc.m413495200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neurotrophins are a family of growth factors that attenuate several forms of pathological neuronal cell death and may represent a putative therapeutic approach to neurodegenerative diseases. In Alzheimer disease, amyloid-beta (Abeta) is thought to play a central role in the neuronal death occurring in brains of patients. In the present study, we evaluate the ability of neurotrophin-3 (NT-3) to protect neurons against the toxicity induced by aggregated Abeta. We showed that in primary cultures of cortical neurons, NT-3 reduces Abeta-induced apoptosis by limiting caspase-8, caspase-9, and caspase-3 cleavage. This neuroprotective effect of NT-3 was concomitant to an increased level of Akt phosphorylation and was abolished by an inhibitor of the phosphatidylinositol-3 kinase (PI-3K), LY294002. In parallel, NT-3 treatment reduced Abeta induced caspase-3 processing to control levels. In an attempt to link PI-3K/Akt to caspase inhibition, we evaluated the influence of the PI-3K/Akt axis on the expression of a member of the inhibitors of apoptosis proteins (IAPs), the neuronal apoptosis inhibitory protein-1. We demonstrated that NT-3 induces an up-regulation of neuronal apoptosis inhibitory protein-1 expression in neurons that promotes the inhibition of Abeta-induced neuronal apoptosis. Together, these findings demonstrate that NT-3 signaling counters Abeta-dependent neuronal cell death and may represent an innovative therapeutic intervention to limit neuronal death in Alzheimer disease.
Collapse
Affiliation(s)
- Sylvain Lesné
- UMR CNRS 6185, Université de Caen, Bd. H. Becquerel BP5229, 14074 Caen, France and Howard Hughes Medical Institute, Rutgers University, Piscataway, New Jersey 08854, USA
| | | | | | | | | | | | | |
Collapse
|
328
|
Abstract
Recent models of craniofacial development suggest the existence of a common pan-placodal domain lying next to the neural plate, from which all sensory placodes will arise. In support of this idea, several genes are expressed in the surface ectoderm of the head adjacent to the neural plate, before the appearance of genes in specific cranial placodes. In this study, we examine the expression patterns of the mouse Foxi class genes from embryonic day 6.5 to 10.5. Foxi2 is expressed throughout the cranial ectoderm adjacent to the neural plate from the 4-somite stage, later becoming excluded from the otic placode. Foxi3 is expressed in a broad region of the pan-placodal ectoderm adjacent to the neural plate from embryonic day (E) 6.75 to the first somite stage. Its expression becomes restricted to the ectoderm and the endoderm of the branchial pouches at E10.5. Foxi1 expression is first detected in the endolymphatic duct in the otic vesicle at E10.5. These results suggest that the mouse Foxi class genes may play important roles, both during cranial placode specification and in later development of individual cranial sensory structures and other organs derived from the cranial ectoderm.
Collapse
Affiliation(s)
- Takahiro Ohyama
- Gonda Department of Cell and Molecular Biology, House Ear Institute, 2100 West 3rd Street, Los Angeles, CA 90057, USA
| | | |
Collapse
|
329
|
Mirosevich J, Gao N, Matusik RJ. Expression of Foxa transcription factors in the developing and adult murine prostate. Prostate 2005; 62:339-52. [PMID: 15389796 DOI: 10.1002/pros.20131] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND The Foxa family (a1, a2, and a3) of proteins are transcription factors that are central to endodermal development. Recently, Foxa1 has been shown to regulate the transcription of several murine and human prostate specific genes involved in differentiated function by interacting with DNA promoter sequences and androgen receptors. Currently, the developmental expression pattern of Foxa proteins in the murine prostate is unknown. METHODS Male CD-1 mice (embryonic, prepubertal, pubertal, and adult) were used for immunohistochemical analysis of Foxa1, a2, and a3. Immunofluorescence was also performed for androgen receptor and cytokeratin 14 expression. Prostate tissue from pre-pubertal, pubertal, and adult mice were analyzed by Western blot and RT-PCR analysis for Foxa1, a2, and a3 expression. RESULTS Strong Foxa1 immunoreactivity was observed in epithelial cells throughout prostate development, growth, and adult differentiation. Prominent Foxa2 protein expression was only observed in the early stages of prostate development and was exclusively localized to epithelial cells of the forming buds. RT-PCR analysis identified low Foxa2 mRNA expression levels in the ventral and dorsolateral lobes of the adult prostate, with Foxa2 epithelial cell expression being localized to periurethral regions of the murine adult prostatic complex. Foxa3 expression was not observed in the murine prostate. CONCLUSIONS Foxa proteins represent epithelial cell markers in the murine prostate gland. The early expression of Foxa1 and a2 proteins in prostate formation suggests that these proteins play an important role in normal prostate development, in addition to differentiated secretory function.
Collapse
Affiliation(s)
- Janni Mirosevich
- Vanderbilt Prostate Cancer Center, Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2765, USA
| | | | | |
Collapse
|
330
|
Fritzenwanker JH, Saina M, Technau U. Analysis of forkhead and snail expression reveals epithelial-mesenchymal transitions during embryonic and larval development of Nematostella vectensis. Dev Biol 2005; 275:389-402. [PMID: 15501226 DOI: 10.1016/j.ydbio.2004.08.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2004] [Revised: 08/10/2004] [Accepted: 08/12/2004] [Indexed: 01/15/2023]
Abstract
The winged helix transcription factor Forkhead and the zinc finger transcription factor Snail are crucially involved in germ layer formation in Bilateria. Here, we isolated and characterized a homolog of forkhead/HNF3 (FoxA/group 1) and of snail from a diploblast, the sea anemone Nematostella vectensis. We show that Nematostella forkhead expression starts during late Blastula stage in a ring of cells that demarcate the blastopore margin during early gastrulation, thereby marking the boundary between ectodermal and endodermal tissue. snail, by contrast, is expressed in a complementary pattern in the center of forkhead-expressing cells marking the presumptive endodermal cells fated to ingress during gastrulation. In a significant portion of early gastrulating embryos, forkhead is expressed asymmetrically around the blastopore. While snail-expressing cells form the endodermal cell mass, forkhead marks the pharynx anlage throughout embryonic and larval development. In the primary polyp, forkhead remains expressed in the pharynx. The detailed analysis of forkhead and snail expression during Nematostella embryonic and larval development further suggests that endoderm formation results from epithelial invagination, mesenchymal immigration, and reorganization of the endodermal epithelial layer, that is, by epithelial-mesenchymal transitions (EMT) in combination with extensive morphogenetic movements. snail also governs EMT at different processes during embryonic development in Bilateria. Our data indicate that the function of snail in Diploblasts is to regulate motility and cell adhesion, supporting that the triggering of changes in cell behavior is the ancestral role of snail in Metazoa.
Collapse
Affiliation(s)
- Jens H Fritzenwanker
- Molecular Cell Biology, Institute for Zoology, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | | | | |
Collapse
|
331
|
Park KW, Kim DH, You HJ, Sir JJ, Jeon SI, Youn SW, Yang HM, Skurk C, Park YB, Walsh K, Kim HS. Activated forkhead transcription factor inhibits neointimal hyperplasia after angioplasty through induction of p27. Arterioscler Thromb Vasc Biol 2005; 25:742-7. [PMID: 15662024 DOI: 10.1161/01.atv.0000156288.70849.26] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We examined the effects of FKHRL1 (forkhead transcription factor in rhabdomyosarcoma like-1) overexpression on vascular smooth muscle cell (VSMC) proliferation, apoptosis, and cell cycle, in vitro, and the role of FKHRL1 and p27 in the pathophysiology of neointimal growth after balloon angioplasty, in vivo. Furthermore, we tested whether FKHRL1 overexpression can inhibit neointimal hyperplasia in a rat carotid artery model. METHODS AND RESULTS Adenovirus expressing the constitutively active FKHRL1 (FKHRL1-TM; triple mutant) with 3 Akt phosphorylation sites mutated was transfected to subconfluent VSMCs. FKHRL1 overexpression in cultured VSMCs increased p27 expression, leading to G1 phase cell-cycle arrest and increased apoptosis. In vivo, the phosphorylation of FKHRL1 increased significantly 3 hours after balloon injury and decreased thereafter, with the subsequent downregulation of p27. Although the phosphorylation of FKHRL1 was greatest at 3 hours, the downregulation of p27 showed a temporal delay, only slightly starting to decrease after 3 hours and reaching a nadir at 72 hours after balloon injury. Gene transfer of FKHRL1-TM increased p27, decreased proliferation, and increased apoptosis of VSMCs, which resulted in a marked reduction in neointima formation (intima-to-media ratio: 0.31+/-0.13 versus 1.17+/-0.28, for FKHRL1-TM versus Adv-GFP; P<0.001). CONCLUSIONS Balloon angioplasty leads to the phosphorylation of FKHRL1 and decreased expression of p27, thereby promoting a proliferative phenotype in VSMCs in vitro and in vivo. This study reveals the importance of FKHRL1 in proliferation and viability of VSMCs and suggests that it may serve as a molecular target for interventions to reduce neointima formation after angioplasty.
Collapse
MESH Headings
- Angioplasty, Balloon/adverse effects
- Animals
- Aorta, Thoracic/injuries
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiology
- Apoptosis/physiology
- Cell Cycle Proteins/metabolism
- Cell Survival/physiology
- Cells, Cultured
- Cyclin-Dependent Kinase Inhibitor p27
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Forkhead Box Protein O1
- Forkhead Box Protein O3
- Forkhead Transcription Factors
- Gene Expression
- Gene Transfer Techniques
- Humans
- Hyperplasia
- Male
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiology
- Nerve Tissue Proteins
- Phosphorylation
- Rats
- Rats, Sprague-Dawley
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Tumor Suppressor Proteins/metabolism
- Tunica Intima/pathology
Collapse
Affiliation(s)
- Kyung-Woo Park
- Cardiovascular Laboratory, Clinical Research Institute, Seoul National University Hospital, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
332
|
Morphogens of hydra Hydra sp. J EVOL BIOCHEM PHYS+ 2005. [DOI: 10.1007/s10893-005-0028-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
333
|
Pohl BS, Knöchel W. Of Fox and Frogs: Fox (fork head/winged helix) transcription factors in Xenopus development. Gene 2005; 344:21-32. [PMID: 15656969 DOI: 10.1016/j.gene.2004.09.037] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2004] [Revised: 09/09/2004] [Accepted: 09/28/2004] [Indexed: 01/03/2023]
Abstract
Transcription factors of the Fox (fork head box) family have been found in all metazoan organisms. They are characterised by an evolutionary conserved DNA-binding domain of winged helix structure. In the South African clawed frog, Xenopus laevis, more than 30 Fox genes have been found so far. This review summarises our present knowledge regarding the general structure and common features of the fork head box and will then characterise Fox genes that have been described in Xenopus. Special attention was paid to the temporal and spatial expression patterns during early embryonic development. For some of these genes, the molecular mechanisms leading to their regulation after the onset of zygotic transcription are known. We also report on functional aspects including target gene regulation, cell or tissue specification and interference with the cell cycle. Finally, Fox proteins serve as mediators of signalling pathways and they might function as checkpoint molecules for the cross-regulatory interactions of different intracellular signal transduction chains.
Collapse
Affiliation(s)
- Barbara S Pohl
- Abteilung Biochemie, Universität Ulm, Albert-Einstein-Allee 11, Ulm D-89081, Germany
| | | |
Collapse
|
334
|
Coffer PJ, Burgering BMT. Forkhead-box transcription factors and their role in the immune system. Nat Rev Immunol 2004; 4:889-99. [PMID: 15516968 DOI: 10.1038/nri1488] [Citation(s) in RCA: 299] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
It is more than a decade since the discovery of the first forkhead-box (FOX) transcription factor in the fruit fly Drosophila melanogaster. In the intervening time, there has been an explosion in the identification and characterization of members of this family of proteins. Importantly, in the past few years, it has become clear that members of the FOX family have crucial roles in various aspects of immune regulation, from lymphocyte survival to thymic development. This review focuses on FOXP3, FOXN1, FOXJ1 and members of the FOXO subfamily and their function in the immune system.
Collapse
Affiliation(s)
- Paul J Coffer
- Department of Pulmonary Diseases, University Medical Centre, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands.
| | | |
Collapse
|
335
|
Govoroun MS, Pannetier M, Pailhoux E, Cocquet J, Brillard JP, Couty I, Batellier F, Cotinot C. Isolation of chicken homolog of theFOXL2gene and comparison of its expression patterns with those of aromatase during ovarian development. Dev Dyn 2004; 231:859-70. [PMID: 15517586 DOI: 10.1002/dvdy.20189] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Mutations in the forkhead transcription factor gene FOXL2 are involved in ovarian failure, which occurs in human BPES syndrome. This syndrome presents a sexually dimorphic expression, specific to the ovary in several vertebrates. We cloned the open reading frame of chicken FOXL2 (cFoxL2) and studied cFoxL2 expression in developing gonads and during adulthood to examine the role of FOXL2 in ovarian differentiation and function in birds. The spatial and temporal dynamics of cFoxL2 and aromatase expression were analyzed in parallel by using real-time quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry in attempt to investigate the possible role of cFoxL2 in the regulation of aromatase. The expression patterns of cFoxL2 and aromatase transcripts were highly correlated during the sex-differentiation period (4.7-12.7 days of incubation). Aromatase and cFoxL2 proteins were colocalized in the medullar part of female gonads on embryonic day 14. Fourteen days after hatching, cFoxL2 protein was mainly detected in granulosa cells of developing follicles. In adult ovary follicular envelopes, apart from granulosa cells, cFoxL2 transcript and protein were detected at lower levels in theca cells where aromatase was present. A high level of cFoxL2 transcription was also observed in maturing and ovulated oocytes. Our results confirm that FoxL2 is an early regulator of ovarian development in birds and may be involved in aromatase transcription regulation.
Collapse
|
336
|
De Velasco B, Shen J, Go S, Hartenstein V. Embryonic development of the Drosophila corpus cardiacum, a neuroendocrine gland with similarity to the vertebrate pituitary, is controlled by sine oculis and glass. Dev Biol 2004; 274:280-94. [PMID: 15385159 DOI: 10.1016/j.ydbio.2004.07.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2004] [Revised: 07/14/2004] [Accepted: 07/19/2004] [Indexed: 11/16/2022]
Abstract
We have investigated the development of the Drosophila neuroendocrine gland, the corpus cardiacum (CC), and identified the role of regulatory genes and signaling pathways in CC morphogenesis. CC progenitors segregate from the blastoderm as part of the anterior lip of the ventral furrow. Among the early genetic determinants expressed and required in this domain are the genes giant (gt) and sine oculis (so). During the extended germ band stage, CC progenitor cells form a paired cluster of 6-8 cells sandwiched in between the inner surface of the protocerebrum and the foregut. While flanking the protocerebrum, CC progenitors are in direct contact with the neural precursors that give rise to the pars intercerebralis, the part of the brain whose neurons later innervate the CC. At this stage, the CC progenitors turn on the homeobox gene glass (gl), which is essential for the differentiation of the CC. During germ band retraction, CC progenitors increase in number and migrate posteriorly, passing underneath the brain commissure and attaching themselves to the primordia of the corpora allata (CA). During dorsal closure, the CC and CA move around the anterior aorta to become the ring gland. Signaling pathways that shape the determination and morphogenesis of the CC are decapentaplegic (dpp) and its antagonist short gastrulation (sog), as well as hedgehog (hh) and heartless (htl; a Drosophila FGFR homolog). Sog is expressed in the midventral domain from where CC progenitors originate, and these cells are completely absent in sog mutants. Dpp and hh are expressed in the anterior visceral head mesoderm and the foregut, respectively; both of these tissues flank the CC. Loss of hh and dpp results in defects in CC proliferation and migration. Htl appears in the somatic mesoderm of the head and trunk. Although mutations of htl do not cause direct effects on the early development of the CC, the later formation of the ring gland is highly abnormal due to the absence of the aorta in these mutants. Defects in the CC are also caused by mutations that severely reduce the protocerebrum, including tailless (tll), suggesting that additional signaling events exist between brain and CC progenitors. We discuss the parallels between neuroendocrine development in Drosophila and vertebrates.
Collapse
Affiliation(s)
- Begona De Velasco
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095-1606, USA
| | | | | | | |
Collapse
|
337
|
Gaudet J, Muttumu S, Horner M, Mango SE. Whole-genome analysis of temporal gene expression during foregut development. PLoS Biol 2004; 2:e352. [PMID: 15492775 PMCID: PMC523228 DOI: 10.1371/journal.pbio.0020352] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2004] [Accepted: 08/13/2004] [Indexed: 12/05/2022] Open
Abstract
We have investigated the cis-regulatory network that mediates temporal gene expression during organogenesis. Previous studies demonstrated that the organ selector gene pha-4/FoxA is critical to establish the onset of transcription of Caenorhabditis elegans foregut (pharynx) genes. Here, we discover additional cis-regulatory elements that function in combination with PHA-4. We use a computational approach to identify candidate cis-regulatory sites for genes activated either early or late during pharyngeal development. Analysis of natural or synthetic promoters reveals that six of these sites function in vivo. The newly discovered temporal elements, together with predicted PHA-4 sites, account for the onset of expression of roughly half of the pharyngeal genes examined. Moreover, combinations of temporal elements and PHA-4 sites can be used in genome-wide searches to predict pharyngeal genes, with more than 85% accuracy for their onset of expression. These findings suggest a regulatory code for temporal gene expression during foregut development and provide a means to predict gene expression patterns based solely on genomic sequence.
Collapse
Affiliation(s)
- Jeb Gaudet
- 1Huntsman Cancer Institute, University of UtahSalt Lake City, UtahUnited States of America
| | - Srikanth Muttumu
- 1Huntsman Cancer Institute, University of UtahSalt Lake City, UtahUnited States of America
| | - Michael Horner
- 1Huntsman Cancer Institute, University of UtahSalt Lake City, UtahUnited States of America
| | - Susan E Mango
- 1Huntsman Cancer Institute, University of UtahSalt Lake City, UtahUnited States of America
| |
Collapse
|
338
|
Lee HH, Frasch M. Survey of forkhead domain encoding genes in the Drosophila genome: Classification and embryonic expression patterns. Dev Dyn 2004; 229:357-66. [PMID: 14745961 DOI: 10.1002/dvdy.10443] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Genetic approaches in Drosophila led to the identification of Forkhead, the prototype of forkhead domain transcription factors that are now known to comprise an evolutionarily conserved family of proteins with essential roles in development and differentiation. Sequence analysis of the recently published genomic scaffold sequence from Drosophila melanogaster has allowed us to determine the presumably full complement of forkhead domain encoding genes in this species. We show herein that the Drosophila genome contains 17 forkhead domain encoding genes; 13 of these genes have orthologs in chordate species, and their products can be assigned to 10 of the 17 forkhead domain subclasses known from chordates. One Drosophila forkhead domain gene only has a Caenorhabditis elegans ortholog and may represent a subclass that is absent in chordates, while the remaining three cannot be classified. We present the mRNA expression patterns of seven previously uncharacterized members of this gene family and show that they are expressed in tissues from all three germ layers, including central and peripheral nervous system, epidermis, salivary gland primordia, endoderm, somatic mesoderm, and hemocyte progenitors. Furthermore, the expression patterns of two of these genes, fd19B and fd102C, suggest a role for them as gap genes during early embryonic head segmentation.
Collapse
Affiliation(s)
- Hsiu-Hsiang Lee
- Mount Sinai School of Medicine, Brookdale Department of Molecular, Cell and Developmental Biology, New York, New York, USA
| | | |
Collapse
|
339
|
Devany M, Kotharu NP, Matsuo H. Solution NMR structure of the C-terminal domain of the human protein DEK. Protein Sci 2004; 13:2252-9. [PMID: 15238633 PMCID: PMC2279821 DOI: 10.1110/ps.04797104] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Revised: 05/03/2004] [Accepted: 05/03/2004] [Indexed: 10/26/2022]
Abstract
The chromatin-associated protein DEK was first identified as a fusion protein in patients with a subtype of acute myelogenous leukemia. It has since become associated with diverse human ailments ranging from cancers to autoimmune diseases. Despite much research effort, the biochemical basis for these clinical connections has yet to be explained. We have identified a structural domain in the C-terminal region of DEK [DEK(309-375)]. DEK(309-375) implies clinical importance because it can reverse the characteristic abnormal DNA-mutagen sensitivity in fibroblasts from ataxia-telangiectasia (A-T) patients. We determined the solution structure of DEK(309-375) by nuclear magnetic resonance spectroscopy, and found it to be structurally homologous to the E2F/DP transcription factor family. On the basis of this homology, we tested whether DEK(309-375) could bind DNA and identified the DNA-interacting surface. DEK presents a hydrophobic surface on the side opposite the DNA-interacting surface. The structure of the C-terminal region of DEK provides insights into the protein function of DEK.
Collapse
Affiliation(s)
- Matthew Devany
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
340
|
Pisarska MD, Bae J, Klein C, Hsueh AJW. Forkhead l2 is expressed in the ovary and represses the promoter activity of the steroidogenic acute regulatory gene. Endocrinology 2004; 145:3424-33. [PMID: 15059956 DOI: 10.1210/en.2003-1141] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Premature ovarian failure in a subgroup of women with blepharophimosis-ptosis-epicanthus inversus type 1 syndrome has been associated with nonsense mutations in the gene encoding a Forkhead transcription factor, Forkhead L2 (FOXL2). However, the exact function of FOXL2 in the ovary is unclear. We investigated the expression of FOXL2 in the mouse ovary during follicular development and maturation by RT-PCR and in situ hybridization. The FOXL2 mRNA is expressed in ovaries throughout development and adulthood and is localized to the undifferentiated granulosa cells in small and medium follicles as well as cumulus cells of preovulatory follicles. FOXL2 belongs to a group of transcription factors capable of interacting with specific DNA sequences in diverse gene promoters. With the presence of multiple putative forkhead DNA consensus sites, the promoter of the human steroidogenic acute regulatory (StAR) gene was used to test for regulation by FOXL2. Cotransfection studies revealed that wild-type FOXL2 represses the activity of the StAR promoter, and the first 95 bp upstream of the transcriptional start site of the StAR gene is sufficient for FOXL2 repression. EMSAs confirmed that FOXL2 interacts directly with this region. Analyses using FOXL2 mutants also demonstrated the importance of the entire alanine/proline-rich carboxyl terminus of FOXL2 for transcriptional repression. Furthermore, these mutations produce a protein with a dominant-negative effect that disables the transcriptional repressor activity of wild-type FOXL2. Dominant-negative mutations of FOXL2 could increase expression of StAR and other follicle differentiation genes in small and medium follicles to accelerate follicle development, resulting in increased initial recruitment of dormant follicles and thus the premature ovarian failure phenotype.
Collapse
Affiliation(s)
- Margareta D Pisarska
- Division of Reproductive Biology, Department of Obstetrics and Gynecology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305-5317, USA
| | | | | | | |
Collapse
|
341
|
Castillejo-López C, Arias WM, Baumgartner S. The fat-like Gene of Drosophila Is the True Orthologue of Vertebrate Fat Cadherins and Is Involved in the Formation of Tubular Organs. J Biol Chem 2004; 279:24034-43. [PMID: 15047711 DOI: 10.1074/jbc.m313878200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Fat cadherins constitute a subclass of the large cadherin family characterized by the presence of 34 cadherin motifs. To date, three mammalian Fat cadherins have been described; however, only limited information is known about the function of these molecules. In this paper, we describe the second fat cadherin in Drosophila, fat-like (ftl). We show that ftl is the true orthologue of vertebrate fat-like genes, whereas the previously characterized tumor suppressor cadherin, fat, is more distantly related as compared with ftl. Ftl is a large molecule of 4705 amino acids. It is expressed apically in luminal tissues such as trachea, salivary glands, proventriculus, and hindgut. Silencing of ftl results in the collapse of tracheal epithelia giving rise to breaks, deletions, and sac-like structures. Other tubular organs such as proventriculus, salivary glands, and hindgut are also malformed or missing. These data suggest that Ftl is required for morphogenesis and maintenance of tubular structures of ectodermal origin and underline its similarity in function to a reported lethal mouse knock-out of fat1 where glomerular epithelial processes collapse. Based on our results, we propose a model where Ftl acts as a spacer to keep tubular epithelia apart rather than the previously described adhesive properties of the cadherin superfamily.
Collapse
|
342
|
Reed BH, Wilk R, Schöck F, Lipshitz HD. Integrin-dependent apposition of Drosophila extraembryonic membranes promotes morphogenesis and prevents anoikis. Curr Biol 2004; 14:372-80. [PMID: 15028211 DOI: 10.1016/j.cub.2004.02.029] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2003] [Revised: 02/04/2004] [Accepted: 02/06/2004] [Indexed: 12/28/2022]
Abstract
BACKGROUND Two extraembryonic tissues form early in Drosophila development. One, the amnioserosa, has been implicated in the morphogenetic processes of germ band retraction and dorsal closure. The developmental role of the other, the yolk sac, is obscure. RESULTS By using live-imaging techniques, we report intimate interactions between the amnioserosa and the yolk sac during germ band retraction and dorsal closure. These tissue interactions fail in a subset of myospheroid (mys: betaPS integrin) mutant embryos, leading to failure of germ band retraction and dorsal closure. The Drosophila homolog of mammalian basigin (EMMPRIN, CD147)-an integrin-associated transmembrane glycoprotein-is highly enriched in the extraembryonic tissues. Strong dominant genetic interactions between basigin and mys mutations cause severe defects in dorsal closure, consistent with basigin functioning together with betaPS integrin in extraembryonic membrane apposition. During normal development, JNK signaling is upregulated in the amnioserosa, as midgut closure disrupts contact with the yolk sac. Subsequently, the amnioserosal epithelium degenerates in a process that is independent of the reaper, hid, and grim cell death genes. In mys mutants that fail to establish contact between the extraembryonic membranes, the amnioserosa undergoes premature disintegration and death. CONCLUSIONS Intimate apposition of the amnioserosa and yolk sac prevents anoikis of the amnioserosa. Survival of the amnioserosa is essential for germ band retraction and dorsal closure. We hypothesize that during normal development, loss of integrin-dependent contact between the extraembryonic tissues results in JNK-dependent amnioserosal disintegration and death, thus representing an example of developmentally programmed anoikis.
Collapse
Affiliation(s)
- Bruce H Reed
- Program in Developmental Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada.
| | | | | | | |
Collapse
|
343
|
Martindale MQ, Pang K, Finnerty JR. Investigating the origins of triploblasty: `mesodermal' gene expression in a diploblastic animal, the sea anemone Nematostella vectensis(phylum, Cnidaria; class, Anthozoa). Development 2004; 131:2463-74. [PMID: 15128674 DOI: 10.1242/dev.01119] [Citation(s) in RCA: 241] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mesoderm played a crucial role in the radiation of the triploblastic Bilateria, permitting the evolution of larger and more complex body plans than in the diploblastic, non-bilaterian animals. The sea anemone Nematostella is a non-bilaterian animal, a member of the phylum Cnidaria. The phylum Cnidaria (sea anemones, corals, hydras and jellyfish) is the likely sister group of the triploblastic Bilateria. Cnidarians are generally regarded as diploblastic animals, possessing endoderm and ectoderm,but lacking mesoderm. To investigate the origin of triploblasty, we studied the developmental expression of seven genes from Nematostella whose bilaterian homologs are implicated in mesodermal specification and the differentiation of mesodermal cell types (twist, snailA, snailB, forkhead,mef2, a GATA transcription factor and a LIMtranscription factor). Except for mef2, the expression of these genes is largely restricted to the endodermal layer, the gastrodermis. mef2is restricted to the ectoderm. The temporal and spatial expression of these`mesoderm' genes suggests that they may play a role in germ layer specification. Furthermore, the predominantly endodermal expression of these genes reinforces the hypothesis that the mesoderm and endoderm of triploblastic animals could be derived from the endoderm of a diploblastic ancestor. Alternatively, we consider the possibility that the diploblastic condition of cnidarians is a secondary simplification, derived from an ancestral condition of triploblasty.
Collapse
Affiliation(s)
- Mark Q Martindale
- Kewalo Marine Laboratory, Pacific Biomedical Research Center, University of Hawaii, 41 Ahui Street, Honolulu, HI 96813, USA.
| | | | | |
Collapse
|
344
|
Glass AS, Dahm R. The Zebrafish as a Model Organism for Eye Development. Ophthalmic Res 2004; 36:4-24. [PMID: 15007235 DOI: 10.1159/000076105] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2003] [Accepted: 12/09/2003] [Indexed: 11/19/2022]
Abstract
In recent years, the zebrafish has become a favourite model organism for biologists studying developmental processes in vertebrates. Its rapid embryonic development, the transparency of its embryos, the large number of offspring together with several other advantages make it ideal for discovering and understanding the genes that regulate embryonic development as well as the physiology of the adult organism. Zebrafish are very visually orientated, and their retina and lens show much the same morphology as other vertebrates including humans. For this reason, they are well suited for examining ocular development, function and disease. This review describes the advantages of the zebrafish as a model organism as well as giving an overview of eye development in this species. It has a particular focus on morphological as well as molecular aspects of the development of the lens.
Collapse
Affiliation(s)
- Anne S Glass
- Medizinische Genetik, Eberhard-Karls-Universität Tübingen, Germany
| | | |
Collapse
|
345
|
Müller WEG, Wiens M, Adell T, Gamulin V, Schröder HC, Müller IM. Bauplan of Urmetazoa: Basis for Genetic Complexity of Metazoa. INTERNATIONAL REVIEW OF CYTOLOGY 2004; 235:53-92. [PMID: 15219781 DOI: 10.1016/s0074-7696(04)35002-3] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Sponges were first grouped to the animal-plants or plant-animals then to the Zoophyta or Mesozoa and finally to the Parazoa. Only after the application of molecular biological techniques was it possible to place the Porifera monophyletically with the other metazoan phyla, justifying a unification of all multicellular animals to only one kingdom, the Metazoa. The first strong support came from the discovery that cell-cell and cell-matrix adhesion molecules that were cloned from sponges and were subsequently expressed share a high DNA sequence and protein function similarity with the corresponding molecules of other metazoans. Besides these evolutionary novelties for Metazoa, sponges also have morphogens and transcription factors in common with other metazoan phyla. Surprisingly, even those elements exist in Porifera, which are characteristic for pattern and axis formation. Recent studies showed that epithelial layers of sponges are sealed against the extracellular milieu through tight-junction proteins. The cell culture system from sponges, the primmorphs, was suitable for understanding morphogenetic events. Finally, stem cell marker genes were isolated, which underscored that sponge cells have the capacity to differentiate. In the relatively short period of time, approximately 200 million years, the basic pathways had to be established that allowed the transition for multicellular organisms to a colonial system through the formation of adhesion molecules; based on the development of a complex immune system and the apoptotic machinery of an integrated system, the Urmetazoa, which evolved approximately 800 million years ago, could be reached. Hence, the Bauplan of the hypothetical Urmetazoa can now be constructed according to genomic regulatory systems similar to those found in higher Metazoa. These data caused a paradigmatic change; the Porifera are complex and simple but by far not primitive.
Collapse
Affiliation(s)
- Werner E G Müller
- Institut für Physiologische Chemie, Abteilung Angewandte Molekularbiologie, Universität, D-55099 Mainz, Germany
| | | | | | | | | | | |
Collapse
|
346
|
Takiya S, Gazi M, Mach V. The DNA binding of insect Fork head factors is strongly influenced by the negative cooperation of neighbouring bases. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2003; 33:1145-1154. [PMID: 14563365 DOI: 10.1016/s0965-1748(03)00134-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The Drosophila melanogaster Fork head and Bombyx mori SGF1/Fork head proteins are key regulators of tissue specific gene expression in the modified larval labial glands. Here we use the competitive electrophoretic mobility shift assay to create a detailed Fork head binding matrix and we investigate some unusual features of the Fork head interaction with DNA. We found that the Fork head-DNA interaction is context dependent--the binding specificity of the protein is partly determined by specific combinations of neighbouring bases. Although the total number of the sub-optimal dinucleotide steps is not high, the negative cooperation of neighbouring bases significantly contributes to the overall binding site specificity. Our results allow efficient recognition of insect Fork head binding sites and we show that the putative Fork head cognate elements preferentially accumulate in the near upstream region of genes abundantly expressed in the labial gland.
Collapse
Affiliation(s)
- Shigeharu Takiya
- Center for Advanced Science and Technology, and Division of Bioscience, Graduate School of Environmental Earth Science, Hokkaido University, Kita-ku, Sapporo 060-0810, Japan
| | | | | |
Collapse
|
347
|
Abstract
The mature eye is a complex organ that develops through a highly organized process during embryogenesis. Alterations in its genetic programming can lead to severe disorders that become apparent at birth or shortly afterwards; for example, one-half of the cases of blindness in children have a genetic cause. This review outlines the genetic basis of eye development, as determined by mutation analysis in patients and in model organisms. A better understanding of how this intricate organ develops at the genetic and cellular level is central to our understanding of the pathologies that afflict it.
Collapse
Affiliation(s)
- Jochen Graw
- GSF-National Research Center for Environment and Health, Institute of Developmental Genetics, D-85764 Neuherberg, Germany.
| |
Collapse
|
348
|
Chandrasekaran V, Beckendorf SK. senseless is necessary for the survival of embryonic salivary glands in Drosophila. Development 2003; 130:4719-28. [PMID: 12925597 DOI: 10.1242/dev.00677] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Apoptosis in developing Drosophila embryos is rare and confined to specific groups of cells. We explain how one organ, salivary glands, of Drosophila embryos avoids apoptosis. senseless (sens), a Zn-finger transcription factor, is expressed in the salivary primordium and later in the differentiated salivary glands. The regulation of sens expression in the salivary placodes is more complex than observed in the embryonic PNS. We have shown that sens expression is initiated in the salivary placodes by fork head (fkh), a winged helix transcription factor. The expression of sens is maintained in the salivary glands by fkh and by daughterless (da), a bHLH family member. In this study, we have identified sage, a salivary-specific bHLH protein as a new heterodimeric partner for da protein in the salivary glands. In addition, our data suggest that sage RNAi embryos have a phenotype similar to sens and that sage is necessary to maintain expression of sens in the embryonic salivary glands. Furthermore, we show that in the salivary glands, sens acts as an anti-apoptotic protein by repressing reaper and possibly hid.
Collapse
Affiliation(s)
- Vidya Chandrasekaran
- Department of Molecular and Cell Biology, 401 Barker Hall, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
349
|
Dhawan S, Gopinathan KP. Spatio-temporal expression of wnt-1 during embryonic-, wing- and silkgland development in Bombyx mori. Gene Expr Patterns 2003; 3:559-70. [PMID: 12971988 DOI: 10.1016/s1567-133x(03)00122-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
A homologue of the segment polarity gene wnt-1 from Bombyx mori (Bmwnt-1) has been characterized. The segmentally reiterated pattern of Bmwnt-1 transcrip9t distribution in B. mori embryos suggested its segment polarity function. Maximal levels of Bmwnt-1 RNA during embryonic development were reached by stage 21A. In the larval stages, Bmwnt-1 was expressed in the fore- and hindwing discs, ovaries, testes and gut, reminiscent of the expression domains in Drosophila. Bmwnt-1 was expressed in the wing-margin area of both the fore- and hindwing discs. The pattern of wnt-1 expression in the hindwing discs was similar to that in the butterfly Precis coenia but subtle differences existed in forewing discs of the two species, which correlated well with the absence of proximal bands of pigmentation in the adult Bombyx wings. In addition, Bmwnt-1 was expressed in the silkglands and the expression was confined to the anterior sub-compartment within the middle silkglands throughout development from the embryonic to late larval stages. This domain of Bmwnt-1 expression overlapped with those of Cubitus interruptus (BmCi) and sericin-2 but excluded the Engrailed expression domain viz. the middle and posterior sub-compartments of middle silkglands. Bmwnt-1 expression was detected only during the intermoults and not in the moulting periods.
Collapse
Affiliation(s)
- S Dhawan
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | | |
Collapse
|
350
|
Teufel A, Wong EA, Mukhopadhyay M, Malik N, Westphal H. FoxP4, a novel forkhead transcription factor. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1627:147-52. [PMID: 12818433 DOI: 10.1016/s0167-4781(03)00074-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Forkhead proteins have been demonstrated to play key roles in embryonic development, cell cycle regulation, and oncogenesis. We report the characterization of a new forkhead transcription factor, which is a member of the FoxP subfamily. In adult tissues FoxP4 is expressed in heart, brain, lung, liver, kidney, and testis. By Northern hybridization, very low levels of FoxP4 expression were found as early as E7 during embryonic development. Embryonic expression was highest at E11 and subsequently decreased at E15 and E17. In situ hybridization revealed expression of FoxP4 in the developing lung and gut, suggesting a role for FoxP4 during the development of these organs. In addition, FoxP4 was found to be significantly reduced in patients with kidney tumors. Lastly, FoxP4 matches an uncharacterized human EST that has previously been shown to be down-regulated in larynx carcinoma.
Collapse
Affiliation(s)
- Andreas Teufel
- Laboratory of Mammalian Genes and Development, National Institute of Child Health and Human Development, National Institutes of Health, Building 6B, Room 413, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|