1
|
Löbel U, Catala M, D'Arco F, Lequin MH, Pasquariello R, Ilves P, Loorits D, Tähepõld A, Pezzetti G, Craven I, Severino M, Rossi A. Pituitary Gland Duplication Syndrome: An International Imaging Analysis. AJNR Am J Neuroradiol 2025; 46:808-814. [PMID: 39393841 PMCID: PMC11979832 DOI: 10.3174/ajnr.a8534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/09/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND AND PURPOSE Duplication of the pituitary gland is a rare developmental anomaly. Multiple associated craniofacial malformations have previously been reported with the largest series to date consisting of 5 patients. In this multi-institutional series of 10 patients, we present a detailed review of the imaging features and discuss a possible overarching pathogenesis that would explain most of the detected malformations. MATERIALS AND METHODS Inclusion criteria for this retrospective imaging review were the presence of a pituitary stalk and gland duplication and the characteristic appearance of the hypothalamic ventral midline. In addition to the clinical presentation, we recorded the imaging findings of 10 patients (9 girls) through onsite and online reviews. Genetic analysis was available for 6 patients. RESULTS The duplicated pituitary stalk and gland showed normal imaging appearances in all patients. Mammillary bodies were clearly identified lateral to the characteristic prominence of the hypothalamic ventral midline. Strands of tissue extending to the anterior dura ("limited ventral myeloschisis") were noted at the medulla oblongata in 10, and at the cervical spinal cord in 7 patients. The medulla oblongata showed a "butterfly" appearance on axial images in 9 patients. Ten patients had cervical segmentation anomalies ("zipperlike"), 9 had anterior-posterior brainstem patterning defects (small pons, elongated medulla), and corpus callosum measurements were abnormal in all patients. Three patients each presented with diencephalic-mesencephalic junction abnormalities and 4 with an anterior mesencephalic "cap." An oropharyngeal teratoma was present in 4 patients. Genetics was normal in 3 of the 6 patients studied; the remainder were found to have mutations in EFNB1 and a gene variant of GIT1, 2 copies of 7 and 8 exon of SMN1 gene, and 2.126 megabase duplication at bands q11.1 and q11.2 of 1 chromosome 15, respectively. CONCLUSIONS Duplication of the pituitary gland presents as well-defined craniofacial and cervical spine malformation phenotype. Axial mesoderm duplication generating an excess of Sonic Hedgehog may be the primary embryologic driver leading to this condition.
Collapse
Affiliation(s)
- Ulrike Löbel
- From the Department of Radiology (U.L., F.D.), Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Martin Catala
- Laboratory of Developmental Biology (M.C.), CNRS, Sorbonne-University, IPBS, Paris, France
| | - Felice D'Arco
- From the Department of Radiology (U.L., F.D.), Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Maarten H Lequin
- Department of Radiology (M.H.L.), Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Rosa Pasquariello
- Department of Developmental Neuroscience (R.P.), IRCCS Stella Maris Foundation, Pisa, Italy
| | - Pilvi Ilves
- Department of Radiology (P.I., D.L., A.T.), Tartu University Hospital, Tartu, Estonia
- Department of Radiology (P.I.), The University of Tartu, Tartu, Estonia
| | - Dagmar Loorits
- Department of Radiology (P.I., D.L., A.T.), Tartu University Hospital, Tartu, Estonia
| | - Annika Tähepõld
- Department of Radiology (P.I., D.L., A.T.), Tartu University Hospital, Tartu, Estonia
| | - Giulio Pezzetti
- UOC Neuroradiologia (G.P.), ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Ian Craven
- Department of Radiology (I.C.), Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | | | - Andrea Rossi
- Neuroradiology Unit (M.S., A.R.), IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Health Sciences (DISSAL)(A.R.), University of Genoa, Genoa, Italy
| |
Collapse
|
2
|
Wang Y, Liu J, Du LY, Wyss JL, Farrell JA, Schier AF. Gene module reconstruction identifies cellular differentiation processes and the regulatory logic of specialized secretion in zebrafish. Dev Cell 2025; 60:581-598.e9. [PMID: 39591963 PMCID: PMC12097649 DOI: 10.1016/j.devcel.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/30/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024]
Abstract
During differentiation, cells become structurally and functionally specialized, but comprehensive views of the underlying remodeling processes are elusive. Here, we leverage single-cell RNA sequencing (scRNA-seq) developmental trajectories to reconstruct differentiation using two secretory tissues as models-the zebrafish notochord and hatching gland. First, we integrated expression and functional similarities to identify gene modules, revealing dozens of modules representing known and newly associated differentiation processes and their dynamics. Second, we focused on the unfolded protein response (UPR) transducer module to study how general versus cell-type-specific secretory functions are regulated. Profiling loss- and gain-of-function embryos identified that the UPR transcription factors creb3l1, creb3l2, and xbp1 are master regulators of a general secretion program. creb3l1/creb3l2 additionally activate an extracellular matrix secretion program, while xbp1 partners with bhlha15 to activate a gland-like secretion program. Our study presents module identification via multi-source integration for reconstructing differentiation (MIMIR) and illustrates how transcription factors confer general and specialized cellular functions.
Collapse
Affiliation(s)
- Yiqun Wang
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Biozentrum, University of Basel, Basel 4056, Switzerland
| | - Jialin Liu
- Biozentrum, University of Basel, Basel 4056, Switzerland; Allen Discovery Center for Cell Lineage Tracing, University of Washington, Seattle, WA 98195, USA
| | - Lucia Y Du
- Biozentrum, University of Basel, Basel 4056, Switzerland; Allen Discovery Center for Cell Lineage Tracing, University of Washington, Seattle, WA 98195, USA
| | - Jannik L Wyss
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Jeffrey A Farrell
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA.
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Biozentrum, University of Basel, Basel 4056, Switzerland; Allen Discovery Center for Cell Lineage Tracing, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
3
|
Paladino O, Moranda A, Falugi C. Spatiotemporal role of muscarinic signaling in early chick development: exposure to cholinomimetic agents by a mathematical model. Cell Biol Toxicol 2023; 39:1453-1469. [PMID: 36098822 PMCID: PMC10425487 DOI: 10.1007/s10565-022-09770-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 08/26/2022] [Indexed: 11/02/2022]
Abstract
Awareness is growing that, besides several neurotoxic effects, cholinomimetic drugs able to interfere the cholinergic neurotransmitter system may exert a teratogen effect in developing embryos of vertebrate and invertebrate organisms. Cholinomimetic substances exert their toxic activity on organisms as they inhibit the functionality of the cholinergic system by completely or partially replacing the ACh molecule both at the level of the AChE active site and at the level of acetylcholine receptors. In this work, we focused the attention on the effects of muscarinic antagonist (atropine) and agonist (carbachol) drugs during the early development and ontogenesis of chick embryos. An unsteady-state mathematical model of the drug release and fate was developed, to synchronize exposure to a gradient of drug concentrations with the different developmental events. Since concentration measures in time and space cannot be taken without damaging the embryo itself, the diffusion model was the only way to establish at each time-step the exact concentration of drug at the different points of the embryo body (considered two-dimensional up to the 50 h stage). This concentration depends on the distance and position of the embryo with respect to the releasing source. The exposure to carbachol generally enhanced dimensions and stages of the embryos, while atropine mainly caused delay in development and small size of the embryos. Both the drugs were able to cause developmental anomalies, depending on the moment of development, in a time- and dose-dependent way, regardless the expression of genes driving each event. 1. Early chick embryos were exposed to muscarinic drugs in a spatial-temporal context. 2. Effects were stage-(time) dependent, according to distance and position of the source. 3. Atropine inhibited growth, mainly interfering with the cephalic process formation and heart differentiation; carbachol increased growth reducing differentiation. 4. Interferences may be exerted by alteration of calcium responses to naturally occurring morphogen-driven mechanisms.
Collapse
Affiliation(s)
- Ombretta Paladino
- Department of Civil, Chemical and Environmental Engineering (DICCA), University of Genoa, Via Opera Pia 15, 16145 Genoa, Italy
| | - Arianna Moranda
- Department of Civil, Chemical and Environmental Engineering (DICCA), University of Genoa, Via Opera Pia 15, 16145 Genoa, Italy
| | - Carla Falugi
- Department of Earth, Environmental and Life Sciences (DISTAV), University of Genoa, Corso Europa 26, 16132 Genoa, Italy
| |
Collapse
|
4
|
Abstract
The EPH receptor tyrosine kinases and their signaling partners, the EPHRINS, comprise a large class of cell signaling molecules that plays diverse roles in development. As cell membrane-anchored signaling molecules, they regulate cellular organization by modulating the strength of cellular contacts, usually by impacting the actin cytoskeleton or cell adhesion programs. Through these cellular functions, EPH/EPHRIN signaling often regulates tissue shape. Indeed, recent evidence indicates that this signaling family is ancient and associated with the origin of multicellularity. Though extensively studied, our understanding of the signaling mechanisms employed by this large family of signaling proteins remains patchwork, and a truly "canonical" EPH/EPHRIN signal transduction pathway is not known and may not exist. Instead, several foundational evolutionarily conserved mechanisms are overlaid by a myriad of tissue -specific functions, though common themes emerge from these as well. Here, I review recent advances and the related contexts that have provided new understanding of the conserved and varied molecular and cellular mechanisms employed by EPH/EPHRIN signaling during development.
Collapse
Affiliation(s)
- Jeffrey O Bush
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, United States; Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, United States; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, United States; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
5
|
Abstract
During early development, the hindbrain is sub-divided into rhombomeres that underlie the organisation of neurons and adjacent craniofacial tissues. A gene regulatory network of signals and transcription factors establish and pattern segments with a distinct anteroposterior identity. Initially, the borders of segmental gene expression are imprecise, but then become sharply defined, and specialised boundary cells form. In this Review, we summarise key aspects of the conserved regulatory cascade that underlies the formation of hindbrain segments. We describe how the pattern is sharpened and stabilised through the dynamic regulation of cell identity, acting in parallel with cell segregation. Finally, we discuss evidence that boundary cells have roles in local patterning, and act as a site of neurogenesis within the hindbrain.
Collapse
Affiliation(s)
- Robb Krumlauf
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA.,Dept of Anatomy and Cell Biology, Kansas University Medical School, Kansas City, KS 66160, USA
| | | |
Collapse
|
6
|
|
7
|
Cayuso J, Xu Q, Addison M, Wilkinson DG. Actomyosin regulation by Eph receptor signaling couples boundary cell formation to border sharpness. eLife 2019; 8:49696. [PMID: 31502954 PMCID: PMC6739871 DOI: 10.7554/elife.49696] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023] Open
Abstract
The segregation of cells with distinct regional identity underlies formation of a sharp border, which in some tissues serves to organise a boundary signaling centre. It is unclear whether or how border sharpness is coordinated with induction of boundary-specific gene expression. We show that forward signaling of EphA4 is required for border sharpening and induction of boundary cells in the zebrafish hindbrain, which we find both require kinase-dependent signaling, with a lesser input of PDZ domain-dependent signaling. We find that boundary-specific gene expression is regulated by myosin II phosphorylation, which increases actomyosin contraction downstream of EphA4 signaling. Myosin phosphorylation leads to nuclear translocation of Taz, which together with Tead1a is required for boundary marker expression. Since actomyosin contraction maintains sharp borders, there is direct coupling of border sharpness to boundary cell induction that ensures correct organisation of signaling centres.
Collapse
Affiliation(s)
- Jordi Cayuso
- The Francis Crick Institute, London, United Kingdom
| | - Qiling Xu
- The Francis Crick Institute, London, United Kingdom
| | | | | |
Collapse
|
8
|
Jung YH, Lee HJ, Kim JS, Lee SJ, Han HJ. EphB2 signaling-mediated Sirt3 expression reduces MSC senescence by maintaining mitochondrial ROS homeostasis. Free Radic Biol Med 2017; 110:368-380. [PMID: 28687409 DOI: 10.1016/j.freeradbiomed.2017.07.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/12/2017] [Accepted: 07/02/2017] [Indexed: 02/07/2023]
Abstract
Disruption of mitochondrial reactive oxygen species (mtROS) homeostasis is a key factor inducing UCB-MSC senescence. Accordingly, preventing mtROS accumulation will help in suppressing the UCB-MSC senescence. In this study, we observed that the expressions of EphrinB2 and EphB2 were inversely regulated by UCB-MSC passage-dependent manner. EphB2 signaling induced mitochondrial translocation of Sirt3. The knockdown of SIRT3 inhibited the effect of EphB2 signaling in UCB-MSCs. Subsequently, EphrinB2-Fc induced the nuclear translocation of Nrf-2 via c-Src phosphorylation dependent manner, and Sirt3 expression was regulated by Nrf-2. Among Sirt3 target genes, EphB2 signaling increased MnSOD and reduced the mtROS level in UCB-MSCs. Furthermore, the deacetylase effect of Sirt3 enhanced the MnSOD activity by deacetylation at the lysine 68 residue and therapeutic effect of UCB-MSCs on skin-wound healing was increased by EphB2 activation. In conclusion, the EphB2 can serve as a novel target for the optimizing the therapeutic use of UCB-MSCs in wound repair by MnSOD-mediated mtROS scavenging through EphB2/c-Src signaling pathway and Nrf-2-dependent Sirt3 expression.
Collapse
Affiliation(s)
- Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Jun Sung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Republic of Korea
| | - Sei-Jung Lee
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
9
|
McGuigan AP, Javaherian S. Tissue Patterning: Translating Design Principles from In Vivo to In Vitro. Annu Rev Biomed Eng 2016; 18:1-24. [DOI: 10.1146/annurev-bioeng-083115-032943] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Alison P. McGuigan
- Department of Chemical Engineering and Applied Chemistry and
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3E5, Canada;
| | | |
Collapse
|
10
|
Abstract
The subdivision of tissues into sharply demarcated regions with distinct and homogenous identity is an essential aspect of embryonic development. Along the anteroposterior axis of the vertebrate nervous system, this involves signaling which induces spatially restricted expression of transcription factors that specify regional identity. The spatial expression of such transcription factors is initially imprecise, with overlapping expression of genes that specify distinct identities, and a ragged border at the interface of adjacent regions. This pattern becomes sharpened by establishment of mutually exclusive expression of transcription factors, and by cell segregation that underlies formation of a straight border. In this review, we discuss studies of the vertebrate hindbrain which have revealed how discrete regional identity is established, the roles of Eph-ephrin signaling in cell segregation and border sharpening, and how cell identity and cell segregation are coupled.
Collapse
|
11
|
Ferrari L, Pistocchi A, Libera L, Boari N, Mortini P, Bellipanni G, Giordano A, Cotelli F, Riva P. FAS/FASL are dysregulated in chordoma and their loss-of-function impairs zebrafish notochord formation. Oncotarget 2015; 5:5712-24. [PMID: 25071022 PMCID: PMC4170636 DOI: 10.18632/oncotarget.2145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Chordoma is a rare malignant tumor that recapitulates the notochord phenotype and is thought to derive from notochord remnants not correctly regressed during development. Apoptosis is necessary for the proper notochord development in vertebrates, and the apoptotic pathway mediated by Fas and Fasl has been demonstrated to be involved in notochord cells regression. This study was conducted to investigate the expression of FAS/FASL pathway in a cohort of skull base chordomas and to analyze the role of fas/fasl homologs in zebrafish notochord formation. FAS/FASL expression was found to be dysregulated in chordoma leading to inactivation of the downstream Caspases in the samples analyzed. Both fas and fasl were specifically expressed in zebrafish notochord sorted cells. fas and fasl loss-of-function mainly resulted in larvae with notochord multi-cell-layer jumps organization, larger vacuolated notochord cells, defects in the peri-notochordal sheath structure and in vertebral mineralization. Interestingly, we observed the persistent expression of ntla and col2a1a, the zebrafish homologs of the human T gene and COL2A1 respectively, which are specifically up-regulated in chordoma. These results demonstrate for the first time the dysregulation of FAS/FASL in chordoma and their role in notochord formation in the zebrafish model, suggesting their possible implication in chordoma onset.
Collapse
Affiliation(s)
- Luca Ferrari
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy; These authors contribute equally in this study
| | - Anna Pistocchi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy; Dipartimento di Bioscienze, Università Degli Studi di Milano, Via Celoria 26 20133 Milan, Italy; These authors contribute equally in this study
| | - Laura Libera
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy
| | - Nicola Boari
- Dipartimento di Neurochirurgia, Università Vita-Salute IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132 Milan, Italy
| | - Pietro Mortini
- Dipartimento di Neurochirurgia, Università Vita-Salute IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132 Milan, Italy
| | - Gianfranco Bellipanni
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA; Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Antonio Giordano
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA; Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Franco Cotelli
- Dipartimento di Bioscienze, Università Degli Studi di Milano, Via Celoria 26 20133 Milan, Italy
| | - Paola Riva
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy
| |
Collapse
|
12
|
Hermkens DMA, van Impel A, Urasaki A, Bussmann J, Duckers HJ, Schulte-Merker S. Sox7 controls arterial specification in conjunction with hey2 and efnb2 function. Development 2015; 142:1695-704. [PMID: 25834021 DOI: 10.1242/dev.117275] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 03/06/2015] [Indexed: 12/22/2022]
Abstract
SoxF family members have been linked to arterio-venous specification events and human pathological conditions, but in contrast to Sox17 and Sox18, a detailed in vivo analysis of a Sox7 mutant model is still lacking. In this study we generated zebrafish sox7 mutants to understand the role of Sox7 during vascular development. By in vivo imaging of transgenic zebrafish lines we show that sox7 mutants display a short circulatory loop around the heart as a result of aberrant connections between the lateral dorsal aorta (LDA) and either the venous primary head sinus (PHS) or the common cardinal vein (CCV). In situ hybridization and live observations in flt4:mCitrine transgenic embryos revealed increased expression levels of flt4 in arterial endothelial cells at the exact location of the aberrant vascular connections in sox7 mutants. An identical circulatory short loop could also be observed in newly generated mutants for hey2 and efnb2. By genetically modulating levels of sox7, hey2 and efnb2 we demonstrate a genetic interaction of sox7 with hey2 and efnb2. The specific spatially confined effect of loss of Sox7 function can be rescued by overexpressing the Notch intracellular domain (NICD) in arterial cells of sox7 mutants, placing Sox7 upstream of Notch in this aspect of arterial development. Hence, sox7 levels are crucial in arterial specification in conjunction with hey2 and efnb2 function, with mutants in all three genes displaying shunt formation and an arterial block.
Collapse
Affiliation(s)
- Dorien M A Hermkens
- Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences and University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands Erasmus MC Rotterdam, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | - Andreas van Impel
- Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences and University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Akihiro Urasaki
- Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences and University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Jeroen Bussmann
- Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences and University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Henricus J Duckers
- Erasmus MC Rotterdam, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | - Stefan Schulte-Merker
- Hubrecht Institute - Royal Netherlands Academy of Arts and Sciences and University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, 48149 Münster, Germany Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Westfälische Wilhelms-Universität Münster (WWU), Mendelstrasse 7, 48149 Münster, Germany
| |
Collapse
|
13
|
Umetsu D, Dunst S, Dahmann C. An RNA interference screen for genes required to shape the anteroposterior compartment boundary in Drosophila identifies the Eph receptor. PLoS One 2014; 9:e114340. [PMID: 25473846 PMCID: PMC4256218 DOI: 10.1371/journal.pone.0114340] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 11/07/2014] [Indexed: 12/01/2022] Open
Abstract
The formation of straight compartment boundaries separating groups of cells with distinct fates and functions is an evolutionarily conserved strategy during animal development. The physical mechanisms that shape compartment boundaries have recently been further elucidated, however, the molecular mechanisms that underlie compartment boundary formation and maintenance remain poorly understood. Here, we report on the outcome of an RNA interference screen aimed at identifying novel genes involved in maintaining the straight shape of the anteroposterior compartment boundary in Drosophila wing imaginal discs. Out of screening 3114 transgenic RNA interference lines targeting a total of 2863 genes, we identified a single novel candidate that interfered with the formation of a straight anteroposterior compartment boundary. Interestingly, the targeted gene encodes for the Eph receptor tyrosine kinase, an evolutionarily conserved family of signal transducers that has previously been shown to be important for maintaining straight compartment boundaries in vertebrate embryos. Our results identify a hitherto unknown role of the Eph receptor tyrosine kinase in Drosophila and suggest that Eph receptors have important functions in shaping compartment boundaries in both vertebrate and insect development.
Collapse
Affiliation(s)
- Daiki Umetsu
- Institute of Genetics, Technische Universität Dresden, Dresden, Germany
| | - Sebastian Dunst
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Christian Dahmann
- Institute of Genetics, Technische Universität Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
14
|
Cayuso J, Xu Q, Wilkinson DG. Mechanisms of boundary formation by Eph receptor and ephrin signaling. Dev Biol 2014; 401:122-31. [PMID: 25448699 DOI: 10.1016/j.ydbio.2014.11.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 12/21/2022]
Abstract
The formation of sharp borders, across which cell intermingling is restricted, has a crucial role in the establishment and maintenance of organized tissues. Signaling of Eph receptors and ephrins underlies formation of a number of boundaries between and within tissues during vertebrate development. Eph-ephrin signaling can regulate several types of cell response-adhesion, repulsion and tension-that can in principle underlie the segregation of cells and formation of sharp borders. Recent studies have implicated each of these cell responses as having important roles at different boundaries: repulsion at the mesoderm-ectoderm border, decreased adhesion at the notochord-presomitic mesoderm border, and tension at boundaries within the hindbrain and forebrain. These distinct responses to Eph receptor and ephrin activation may in part be due to the adhesive properties of the tissue.
Collapse
Affiliation(s)
- Jordi Cayuso
- Division of Developmental Neurobiology, MRC National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Qiling Xu
- Division of Developmental Neurobiology, MRC National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - David G Wilkinson
- Division of Developmental Neurobiology, MRC National Institute for Medical Research, London NW7 1AA, United Kingdom.
| |
Collapse
|
15
|
LU YUCHENG, HENG XUEYUAN, YU JIXU, SU QUANPING, GUAN XIANGHONG, YOU CUIPING, WANG LONG, CHE FENGYUAN. miR-137 regulates the migration of human umbilical vein endothelial cells by targeting ephrin-type A receptor 7. Mol Med Rep 2014; 10:1475-80. [DOI: 10.3892/mmr.2014.2319] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 03/04/2014] [Indexed: 11/06/2022] Open
|
16
|
Du TT, Xu PF, Dong ZW, Fan HB, Jin Y, Dong M, Chen Y, Pan WJ, Ren RB, Liu TX, Deng M, Huang QH. Setdb2 controls convergence and extension movements during zebrafish gastrulation by transcriptional regulation of dvr1. Dev Biol 2014; 392:233-44. [PMID: 24892953 DOI: 10.1016/j.ydbio.2014.05.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 05/15/2014] [Accepted: 05/24/2014] [Indexed: 12/15/2022]
Abstract
As the primary driving forces of gastrulation, convergence and extension (C&E) movements lead to a medio-lateral narrowing and an anterior-posterior elongation of the embryonic body axis. Histone methylation as a post-translational modification plays a critical role in early embryonic development, but its functions in C&E movements remain largely unknown. Here, we show that the setdb2-dvr1 transcriptional cascade plays a critical role in C&E movements during zebrafish gastrulation. Knockdown of Setdb2, a SET domain-containing protein possessing a potential histone H3K9 methyltransferase activity, induced abnormal C&E movements, resulting in anterior-posterior shortening and medio-lateral expansion of the embryonic axis, as well as abnormal notochord cell polarity. Furthermore, we found that Setdb2 functions through fine-tuning the expression of dvr1, a ligand of the TGF-β superfamily, to an appropriate level to ensure proper C&E movements in a non-cell-autonomous manner. In addition, both overexpression and knockdown of Dvr1 at the one-cell stage resulted in defects at epiboly and C&E. These data demonstrate that Setdb2 is a novel regulator for C&E movements and acts by modulating the expression level of dvr1, suggesting that Dvr1 acts as a direct and essential mediator for C&E cell movements.
Collapse
Affiliation(s)
- Ting-Ting Du
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng-Fei Xu
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Zhi-Wei Dong
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hong-Bo Fan
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Jin
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mei Dong
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Chen
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Jun Pan
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui-Bao Ren
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting-Xi Liu
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Min Deng
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Qiu-Hua Huang
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, RuiJin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
17
|
Kawasaki J, Aegerter S, Fevurly RD, Mammoto A, Mammoto T, Sahin M, Mably JD, Fishman SJ, Chan J. RASA1 functions in EPHB4 signaling pathway to suppress endothelial mTORC1 activity. J Clin Invest 2014; 124:2774-84. [PMID: 24837431 DOI: 10.1172/jci67084] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/27/2014] [Indexed: 11/17/2022] Open
Abstract
Vascular malformations are linked to mutations in RAS p21 protein activator 1 (RASA1, also known as p120RasGAP); however, due to the global expression of this gene, it is unclear how these mutations specifically affect the vasculature. Here, we tested the hypothesis that RASA1 performs a critical effector function downstream of the endothelial receptor EPHB4. In zebrafish models, we found that either RASA1 or EPHB4 deficiency induced strikingly similar abnormalities in blood vessel formation and function. Expression of WT EPHB4 receptor or engineered receptors with altered RASA1 binding revealed that the ability of EPHB4 to recruit RASA1 is required to restore blood flow in EPHB4-deficient animals. Analysis of EPHB4-deficient zebrafish tissue lysates revealed that mTORC1 is robustly overactivated, and pharmacological inhibition of mTORC1 in these animals rescued both vessel structure and function. Furthermore, overexpression of mTORC1 in endothelial cells exacerbated vascular phenotypes in animals with reduced EPHB4 or RASA1, suggesting a functional EPHB4/RASA1/mTORC1 signaling axis in endothelial cells. Tissue samples from patients with arteriovenous malformations displayed strong endothelial phospho-S6 staining, indicating increased mTORC1 activity. These results indicate that deregulation of EPHB4/RASA1/mTORC1 signaling in endothelial cells promotes vascular malformation and suggest that mTORC1 inhibitors, many of which are approved for the treatment of certain cancers, should be further explored as a potential strategy to treat patients with vascular malformations.
Collapse
|
18
|
Fagotto F, Winklbauer R, Rohani N. Ephrin-Eph signaling in embryonic tissue separation. Cell Adh Migr 2014; 8:308-26. [PMID: 25482630 PMCID: PMC4594459 DOI: 10.4161/19336918.2014.970028] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/19/2014] [Accepted: 08/25/2014] [Indexed: 01/19/2023] Open
Abstract
The physical separation of the embryonic regions that give rise to the tissues and organs of multicellular organisms is a fundamental aspect of morphogenesis. Pioneer experiments by Holtfreter had shown that embryonic cells can sort based on "tissue affinities," which have long been considered to rely on differences in cell-cell adhesion. However, vertebrate embryonic tissues also express a variety of cell surface cues, in particular ephrins and Eph receptors, and there is now firm evidence that these molecules are systematically used to induce local repulsion at contacts between different cell types, efficiently preventing mixing of adjacent cell populations.
Collapse
Affiliation(s)
| | - Rudolf Winklbauer
- Dpt. of Cell and Systems Biology; University of Toronto; Toronto, Canada
| | - Nazanin Rohani
- Dpt. of Biology; McGill University; Montreal, Quebec, Canada
| |
Collapse
|
19
|
Jayashankar V, Nguyen MJ, Carr BW, Zheng DC, Rosales JB, Rosales JB, Weiser DC. Protein phosphatase 1 β paralogs encode the zebrafish myosin phosphatase catalytic subunit. PLoS One 2013; 8:e75766. [PMID: 24040418 PMCID: PMC3770619 DOI: 10.1371/journal.pone.0075766] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 08/19/2013] [Indexed: 12/21/2022] Open
Abstract
Background The myosin phosphatase is a highly conserved regulator of actomyosin contractility. Zebrafish has emerged as an ideal model system to study the invivo role of myosin phosphatase in controlling cell contractility, cell movement and epithelial biology. Most work in zebrafish has focused on the regulatory subunit of the myosin phosphatase called Mypt1. In this work, we examined the critical role of Protein Phosphatase 1, PP1, the catalytic subunit of the myosin phosphatase. Methodology/Principal Findings We observed that in zebrafish two paralogous genes encoding PP1β, called ppp1cba and ppp1cbb, are both broadly expressed during early development. Furthermore, we found that both gene products interact with Mypt1 and assemble an active myosin phosphatase complex. In addition, expression of this complex results in dephosphorylation of the myosin regulatory light chain and large scale rearrangements of the actin cytoskeleton. Morpholino knock-down of ppp1cba and ppp1cbb results in severe defects in morphogenetic cell movements during gastrulation through loss of myosin phosphatase function. Conclusions/Significance Our work demonstrates that zebrafish have two genes encoding PP1β, both of which can interact with Mypt1 and assemble an active myosin phosphatase. In addition, both genes are required for convergence and extension during gastrulation and correct dosage of the protein products is required.
Collapse
Affiliation(s)
- Vaishali Jayashankar
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Michael J. Nguyen
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Brandon W. Carr
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Dale C. Zheng
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Joseph B. Rosales
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Joshua B. Rosales
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
| | - Douglas C. Weiser
- Department of Biological Sciences, University of the Pacific, Stockton, California, United States of America
- * E-mail:
| |
Collapse
|
20
|
Xu Q, Wilkinson DG. Boundary formation in the development of the vertebrate hindbrain. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2013; 2:735-45. [PMID: 24014457 DOI: 10.1002/wdev.106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The formation of a sharp interface of adjacent subdivisions is important for establishing the precision of tissue organization, and at specific borders it serves to organize key signaling centers. We discuss studies of vertebrate hindbrain development that have given important insights into mechanisms that underlie the formation and maintenance of sharp borders. The hindbrain is subdivided into a series of segments with distinct anteroposterior identity that underlies the specification of distinct neuronal cell types. During early stages of segmentation, cell identity switching contributes to the refinement of borders and enables homogenous territories to be maintained despite intermingling of cells between segments. At later stages, there is a specific restriction to cell intermingling between segments that is mediated by Eph receptor and ephrin signaling. Eph-ephrin signaling can restrict cell intermingling and sharpen borders through multiple mechanisms, including the regulation of cell adhesion and contact inhibition of cell migration.
Collapse
Affiliation(s)
- Qiling Xu
- Division of Developmental Neurobiology, MRC National Institute for Medical Research, London, UK
| | | |
Collapse
|
21
|
Allen-Sharpley MR, Cramer KS. Coordinated Eph-ephrin signaling guides migration and axon targeting in the avian auditory system. Neural Dev 2012; 7:29. [PMID: 22908944 PMCID: PMC3515360 DOI: 10.1186/1749-8104-7-29] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 07/26/2012] [Indexed: 12/21/2022] Open
Abstract
Background In the avian sound localization circuit, nucleus magnocellularis (NM) projects bilaterally to nucleus laminaris (NL), with ipsilateral and contralateral NM axon branches directed to dorsal and ventral NL dendrites, respectively. We previously showed that the Eph receptor EphB2 is expressed in NL neuropil and NM axons during development. Here we tested whether EphB2 contributes to NM-NL circuit formation. Results We found that misexpression of EphB2 in embryonic NM precursors significantly increased the number of axon targeting errors from NM to contralateral NL in a cell-autonomous manner when forward signaling was impaired. We also tested the effects of inhibiting forward signaling of different Eph receptor subclasses by injecting soluble unclustered Fc-fusion proteins at stages when NM axons are approaching their NL target. Again we found an increase in axon targeting errors compared to controls when forward signaling was impaired, an effect that was significantly increased when both Eph receptor subclasses were inhibited together. In addition to axon targeting errors, we also observed morphological abnormalities of the auditory nuclei when EphB2 forward signaling was increased by E2 transfection, and when Eph-ephrin forward signaling was inhibited by E6-E8 injection of Eph receptor fusion proteins. Conclusions These data suggest that EphB signaling has distinct functions in axon guidance and morphogenesis. The results provide evidence that multiple Eph receptors work synergistically in the formation of precise auditory circuitry.
Collapse
Affiliation(s)
- Michelle R Allen-Sharpley
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697-4550, USA
| | | |
Collapse
|
22
|
Terriente J, Gerety SS, Watanabe-Asaka T, Gonzalez-Quevedo R, Wilkinson DG. Signalling from hindbrain boundaries regulates neuronal clustering that patterns neurogenesis. Development 2012; 139:2978-87. [PMID: 22764046 DOI: 10.1242/dev.080135] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
During central nervous system development, neural progenitors are patterned to form discrete neurogenic and non-neurogenic zones. In the zebrafish hindbrain, neurogenesis is organised by Fgf20a emanating from neurons located at each segment centre that inhibits neuronal differentiation in adjacent progenitors. Here, we have identified a molecular mechanism that clusters fgf20a-expressing neurons in segment centres and uncovered a requirement for this positioning in the regulation of neurogenesis. Disruption of hindbrain boundary cell formation alters the organisation of fgf20a-expressing neurons, consistent with a role of chemorepulsion from boundaries. The semaphorins Sema3fb and Sema3gb, which are expressed by boundary cells, and their receptor Nrp2a are required for clustering of fgf20a-expressing neurons at segment centres. The dispersal of fgf20a-expressing neurons that occurs following the disruption of boundaries or of Sema3fb/Sema3gb signalling leads to reduced FGF target gene expression in progenitors and an increased number of differentiating neurons. Sema3 signalling from boundaries thus links hindbrain segmentation to the positioning of fgf20a-expressing neurons that regulates neurogenesis.
Collapse
Affiliation(s)
- Javier Terriente
- Division of Developmental Neurobiology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | | | | | | | | |
Collapse
|
23
|
van Eekelen M, Runtuwene V, Masselink W, den Hertog J. Pair-wise regulation of convergence and extension cell movements by four phosphatases via RhoA. PLoS One 2012; 7:e35913. [PMID: 22545146 PMCID: PMC3335823 DOI: 10.1371/journal.pone.0035913] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 03/26/2012] [Indexed: 11/18/2022] Open
Abstract
Various signaling pathways regulate shaping of the main body axis during early vertebrate development. Here, we focused on the role of protein-tyrosine phosphatase signaling in convergence and extension cell movements. We identified Ptpn20 as a structural paralogue of PTP-BL and both phosphatases were required for normal gastrulation cell movements. Interestingly, knockdowns of PTP-BL and Ptpn20 evoked similar developmental defects as knockdown of RPTPα and PTPε. Co-knockdown of RPTPα and PTP-BL, but not Ptpn20, had synergistic effects and conversely, PTPε and Ptpn20, but not PTP-BL, cooperated, demonstrating the specificity of our approach. RPTPα and PTPε knockdowns were rescued by constitutively active RhoA, whereas PTP-BL and Ptpn20 knockdowns were rescued by dominant negative RhoA. Consistently, RPTPα and PTP-BL had opposite effects on RhoA activation, both in a PTP-dependent manner. Downstream of the PTPs, we identified NGEF and Arhgap29, regulating RhoA activation and inactivation, respectively, in convergence and extension cell movements. We propose a model in which two phosphatases activate RhoA and two phosphatases inhibit RhoA, resulting in proper cell polarization and normal convergence and extension cell movements.
Collapse
Affiliation(s)
- Mark van Eekelen
- Hubrecht Institute and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Vincent Runtuwene
- Hubrecht Institute and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wouter Masselink
- Hubrecht Institute and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jeroen den Hertog
- Hubrecht Institute and University Medical Center Utrecht, Utrecht, the Netherlands
- Institute of Biology, Leiden, the Netherlands
- * E-mail:
| |
Collapse
|
24
|
Batlle E, Wilkinson DG. Molecular mechanisms of cell segregation and boundary formation in development and tumorigenesis. Cold Spring Harb Perspect Biol 2012; 4:a008227. [PMID: 22214769 PMCID: PMC3249626 DOI: 10.1101/cshperspect.a008227] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The establishment and maintenance of precisely organized tissues requires the formation of sharp borders between distinct cell populations. The maintenance of segregated cell populations is also required for tissue homeostasis in the adult, and deficiencies in segregation underlie the metastatic spreading of tumor cells. Three classes of mechanisms that underlie cell segregation and border formation have been uncovered. The first involves differences in cadherin-mediated cell-cell adhesion that establishes interfacial tension at the border between distinct cell populations. A second mechanism involves the induction of actomyosin-mediated contraction by intercellular signaling, such that cortical tension is generated at the border. Third, activation of Eph receptors and ephrins can lead to both decreased adhesion by triggering cleavage of E-cadherin, and to repulsion of cells by regulation of the actin cytoskeleton, thus preventing intermingling between cell populations. These mechanisms play crucial roles at distinct boundaries during development, and alterations in cadherin or Eph/ephrin expression have been implicated in tumor metastasis.
Collapse
Affiliation(s)
- Eduard Batlle
- Oncology Program and ICREA, Institute for Research in Biomedicine, Josep Samitier 1-5, 08028 Barcelona, Spain
| | | |
Collapse
|
25
|
Liu Z, Lin X, Cai Z, Zhang Z, Han C, Jia S, Meng A, Wang Q. Global identification of SMAD2 target genes reveals a role for multiple co-regulatory factors in zebrafish early gastrulas. J Biol Chem 2011; 286:28520-32. [PMID: 21669877 PMCID: PMC3151094 DOI: 10.1074/jbc.m111.236307] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Nodal and Smad2/3 signals play pivotal roles in mesendoderm induction and axis determination during late blastulation and early gastrulation in vertebrate embryos. However, Smad2/3 direct target genes during those critical developmental stages have not been systematically identified. Here, through ChIP-chip assay, we show that the promoter/enhancer regions of 679 genes are bound by Smad2 in the zebrafish early gastrulas. Expression analyses confirm that a significant proportion of Smad2 targets are indeed subjected to Nodal/Smad2 regulation at the onset of gastrulation. The co-existence of DNA-binding sites of other transcription factors in the Smad2-bound regions allows the identification of well known Smad2-binding partners, such as FoxH1 and Lef1/β-catenin, as well as many previously unknown Smad2 partners, including Oct1 and Gata6, during embryogenesis. We demonstrate that Oct1 physically associates with and enhances the transcription and mesendodermal induction activity of Smad2, whereas Gata6 exerts an inhibitory role in Smad2 signaling and mesendodermal induction. Thus, our study systemically uncovers a large number of Smad2 targets in early gastrulas and suggests cooperative roles of Smad2 and other transcription factors in controlling target gene transcription, which will be valuable for studying regulatory cascades during germ layer formation and patterning of vertebrate embryos.
Collapse
Affiliation(s)
- Zhaoting Liu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Biology, Institute of Zoology, Chinese Academy of Sciences, 100101 Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
The involvement of Eph–Ephrin signaling in tissue separation and convergence during Xenopus gastrulation movements. Dev Biol 2011; 350:441-50. [DOI: 10.1016/j.ydbio.2010.12.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 12/03/2010] [Accepted: 12/03/2010] [Indexed: 11/21/2022]
|
27
|
Oda-Ishii I, Ishii Y, Mikawa T. Eph regulates dorsoventral asymmetry of the notochord plate and convergent extension-mediated notochord formation. PLoS One 2010; 5:e13689. [PMID: 21060822 PMCID: PMC2966392 DOI: 10.1371/journal.pone.0013689] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 10/01/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The notochord is a signaling center required for the patterning of the vertebrate embryonic midline, however, the molecular and cellular mechanisms involved in the formation of this essential embryonic tissue remain unclear. The urochordate Ciona intestinalis develops a simple notochord from 40 specific postmitotic mesodermal cells. The precursors intercalate mediolaterally and establish a single array of disk-shaped notochord cells along the midline. However, the role that notochord precursor polarization, particularly along the dorsoventral axis, plays in this morphogenetic process remains poorly understood. METHODOLOGY/PRINCIPAL FINDINGS Here we show that the notochord preferentially accumulates an apical cell polarity marker, aPKC, ventrally and a basement membrane marker, laminin, dorsally. This asymmetric accumulation of apicobasal cell polarity markers along the embryonic dorsoventral axis was sustained in notochord precursors during convergence and extension. Further, of several members of the Eph gene family implicated in cellular and tissue morphogenesis, only Ci-Eph4 was predominantly expressed in the notochord throughout cell intercalation. Introduction of a dominant-negative Ci-Eph4 to notochord precursors diminished asymmetric accumulation of apicobasal cell polarity markers, leading to defective intercalation. In contrast, misexpression of a dominant-negative mutant of a planar cell polarity gene Dishevelled preserved asymmetric accumulation of aPKC and laminin in notochord precursors, although their intercalation was incomplete. CONCLUSIONS/SIGNIFICANCE Our data support a model in which in ascidian embryos Eph-dependent dorsoventral polarity of notochord precursors plays a crucial role in mediolateral cell intercalation and is required for proper notochord morphogenesis.
Collapse
Affiliation(s)
- Izumi Oda-Ishii
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Yasuo Ishii
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| | - Takashi Mikawa
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
28
|
Wiens KM, Lee HL, Shimada H, Metcalf AE, Chao MY, Lien CL. Platelet-derived growth factor receptor beta is critical for zebrafish intersegmental vessel formation. PLoS One 2010; 5:e11324. [PMID: 20593033 PMCID: PMC2892519 DOI: 10.1371/journal.pone.0011324] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 05/24/2010] [Indexed: 12/14/2022] Open
Abstract
Background Platelet-derived growth factor receptor β (PDGFRβ) is a tyrosine kinase receptor known to affect vascular development. The zebrafish is an excellent model to study specific regulators of vascular development, yet the role of PDGF signaling has not been determined in early zebrafish embryos. Furthermore, vascular mural cells, in which PDGFRβ functions cell autonomously in other systems, have not been identified in zebrafish embryos younger than 72 hours post fertilization. Methodology/Principal Findings In order to investigate the role of PDGFRβ in zebrafish vascular development, we cloned the highly conserved zebrafish homolog of PDGFRβ. We found that pdgfrβ is expressed in the hypochord, a developmental structure that is immediately dorsal to the dorsal aorta and potentially regulates blood vessel development in the zebrafish. Using a PDGFR tyrosine kinase inhibitor, a morpholino oligonucleotide specific to PDGFRβ, and a dominant negative PDGFRβ transgenic line, we found that PDGFRβ is necessary for angiogenesis of the intersegmental vessels. Significance/Conclusion Our data provide the first evidence that PDGFRβ signaling is required for zebrafish angiogenesis. We propose a novel mechanism for zebrafish PDGFRβ signaling that regulates vascular angiogenesis in the absence of mural cells.
Collapse
Affiliation(s)
- Katie M. Wiens
- Department of Surgery, Keck School of Medicine, University of Southern California and The Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - Hyuna L. Lee
- Department of Surgery, Keck School of Medicine, University of Southern California and The Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - Hiroyuki Shimada
- Department of Pathology, Keck School of Medicine, University of Southern California and The Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California, United States of America
| | - Anthony E. Metcalf
- Department of Biology, California State University San Bernardino, San Bernardino, California, United States of America
| | - Michael Y. Chao
- Department of Biology, California State University San Bernardino, San Bernardino, California, United States of America
| | - Ching-Ling Lien
- Department of Surgery, Keck School of Medicine, University of Southern California and The Saban Research Institute of Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
29
|
Liu R, Linardopoulou EV, Osborn GE, Parkhurst SM. Formins in development: orchestrating body plan origami. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1803:207-25. [PMID: 18996154 PMCID: PMC2838992 DOI: 10.1016/j.bbamcr.2008.09.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2008] [Revised: 08/21/2008] [Accepted: 09/26/2008] [Indexed: 01/21/2023]
Abstract
Formins, proteins defined by the presence of an FH2 domain and their ability to nucleate linear F-actin de novo, play a key role in the regulation of the cytoskeleton. Initially thought to primarily regulate actin, recent studies have highlighted a role for formins in the regulation of microtubule dynamics, and most recently have uncovered the ability of some formins to coordinate the organization of both the microtubule and actin cytoskeletons. While biochemical analyses of this family of proteins have yielded many insights into how formins regulate diverse cytoskeletal reorganizations, we are only beginning to appreciate how and when these functional properties are relevant to biological processes in a developmental or organismal context. Developmental genetic studies in fungi, Dictyostelium, vertebrates, plants and other model organisms have revealed conserved roles for formins in cell polarity, actin cable assembly and cytokinesis. However, roles have also been discovered for formins that are specific to particular organisms. Thus, formins perform both global and specific functions, with some of these roles concurring with previous biochemical data and others exposing new properties of formins. While not all family members have been examined across all organisms, the analyses to date highlight the significance of the flexibility within the formin family to regulate a broad spectrum of diverse cytoskeletal processes during development.
Collapse
Affiliation(s)
- Raymond Liu
- Division of Basic Sciences Fred Hutchinson Cancer Research Center 1100 Fairview Avenue North Seattle, WA 98109 USA
| | - Elena V. Linardopoulou
- Division of Basic Sciences Fred Hutchinson Cancer Research Center 1100 Fairview Avenue North Seattle, WA 98109 USA
| | - Gregory E. Osborn
- Division of Basic Sciences Fred Hutchinson Cancer Research Center 1100 Fairview Avenue North Seattle, WA 98109 USA
| | - Susan M. Parkhurst
- Division of Basic Sciences Fred Hutchinson Cancer Research Center 1100 Fairview Avenue North Seattle, WA 98109 USA
| |
Collapse
|
30
|
Shinbrot T, Chun Y, Caicedo-Carvajal C, Foty R. Cellular morphogenesis in silico. Biophys J 2009; 97:958-67. [PMID: 19686642 PMCID: PMC2726306 DOI: 10.1016/j.bpj.2009.05.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Revised: 05/09/2009] [Accepted: 05/13/2009] [Indexed: 10/20/2022] Open
Abstract
We describe a model that simulates spherical cells of different types that can migrate and interact either attractively or repulsively. We find that both expected morphologies and previously unreported patterns spontaneously self-assemble. Among the newly discovered patterns are a segmented state of alternating discs, and a "shish-kebab" state, in which one cell type forms a ring around a second type. We show that these unique states result from cellular attraction that increases with distance (e.g., as membranes stretch viscoelastically), and would not be seen in traditional, e.g., molecular, potentials that diminish with distance. Most of the states found computationally have been observed in vitro, and it remains to be established what role these self-assembled states may play in in vivo morphogenesis.
Collapse
Affiliation(s)
- Troy Shinbrot
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA.
| | | | | | | |
Collapse
|
31
|
Nikolaou N, Watanabe-Asaka T, Gerety S, Distel M, Köster RW, Wilkinson DG. Lunatic fringe promotes the lateral inhibition of neurogenesis. Development 2009; 136:2523-33. [PMID: 19553285 DOI: 10.1242/dev.034736] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Previous studies have identified roles of the modulation of Notch activation by Fringe homologues in boundary formation and in regulating the differentiation of vertebrate thymocytes and Drosophila glial cells. We have investigated the role of Lunatic fringe (Lfng) expression during neurogenesis in the vertebrate neural tube. We find that in the zebrafish hindbrain, Lfng is expressed by progenitors in neurogenic regions and downregulated in cells that have initiated neuronal differentiation. Lfng is required cell autonomously in neural epithelial cells to limit the amount of neurogenesis and to maintain progenitors. By contrast, Lfng is not required for the role of Notch in interneuronal fate choice, which we show is mediated by Notch1a. The expression of Lfng does not require Notch activity, but rather is regulated downstream of proneural genes that are widely expressed by neural progenitors. These findings suggest that Lfng acts in a feedback loop downstream of proneural genes, which, by promoting Notch activation, maintains the sensitivity of progenitors to lateral inhibition and thus limits further proneural upregulation.
Collapse
Affiliation(s)
- Nikolas Nikolaou
- Division of Developmental Neurobiology, MRC National Institute for Medical Research, The Ridgeway, London, UK
| | | | | | | | | | | |
Collapse
|
32
|
Affiliation(s)
- Masazumi Tada
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Masatake Kai
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
33
|
Kemp HA, Cooke JE, Moens CB. EphA4 and EfnB2a maintain rhombomere coherence by independently regulating intercalation of progenitor cells in the zebrafish neural keel. Dev Biol 2008; 327:313-26. [PMID: 19135438 DOI: 10.1016/j.ydbio.2008.12.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 12/06/2008] [Accepted: 12/10/2008] [Indexed: 10/21/2022]
Abstract
During vertebrate development, the hindbrain is transiently segmented into 7 distinct rhombomeres (r). Hindbrain segmentation takes place within the context of the complex morphogenesis required for neurulation, which in zebrafish involves a characteristic cross-midline division that distributes progenitor cells bilaterally in the forming neural tube. The Eph receptor tyrosine kinase EphA4 and the membrane-bound Ephrin (Efn) ligand EfnB2a, which are expressed in complementary segments in the early hindbrain, are required for rhombomere boundary formation. We showed previously that EphA4 promotes cell-cell affinity within r3 and r5, and proposed that preferential adhesion within rhombomeres contributes to boundary formation. Here we show that EfnB2a is similarly required in r4 for normal cell affinity and that EphA4 and EfnB2a regulate cell affinity independently within their respective rhombomeres. Live imaging of cell sorting in mosaic embryos shows that both proteins function during cross-midline cell divisions in the hindbrain neural keel. Consistent with this, mosaic EfnB2a over-expression causes widespread cell sorting and disrupts hindbrain organization, but only if induced at or before neural keel stage. We propose a model in which Eph and Efn-dependent cell affinity within rhombomeres serve to maintain rhombomere organization during the potentially disruptive process of teleost neurulation.
Collapse
Affiliation(s)
- Hilary A Kemp
- Howard Hughes Medical Institute and Division of Basic Science, Fred Hutchinson Cancer Research Center, B2-152, 1100 Fairview Ave. N., Seattle, WA 98109, P.O. Box 19024, USA
| | | | | |
Collapse
|
34
|
Okamoto H, Sato T, Aizawa H. Transgenic technology for visualization and manipulation of the neural circuits controlling behavior in zebrafish. Dev Growth Differ 2008; 50 Suppl 1:S167-75. [DOI: 10.1111/j.1440-169x.2008.01003.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
35
|
Lan F, Bayliss PE, Rinn JL, Whetstine JR, Wang JK, Chen S, Iwase S, Alpatov R, Issaeva I, Canaani E, Roberts TM, Chang HY, Shi Y. A histone H3 lysine 27 demethylase regulates animal posterior development. Nature 2007; 449:689-94. [PMID: 17851529 DOI: 10.1038/nature06192] [Citation(s) in RCA: 618] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Accepted: 08/23/2007] [Indexed: 12/12/2022]
Abstract
The recent discovery of a large number of histone demethylases suggests a central role for these enzymes in regulating histone methylation dynamics. Histone H3K27 trimethylation (H3K27me3) has been linked to polycomb-group-protein-mediated suppression of Hox genes and animal body patterning, X-chromosome inactivation and possibly maintenance of embryonic stem cell (ESC) identity. An imbalance of H3K27 methylation owing to overexpression of the methylase EZH2 has been implicated in metastatic prostate and aggressive breast cancers. Here we show that the JmjC-domain-containing related proteins UTX and JMJD3 catalyse demethylation of H3K27me3/2. UTX is enriched around the transcription start sites of many HOX genes in primary human fibroblasts, in which HOX genes are differentially expressed, but is selectively excluded from the HOX loci in ESCs, in which HOX genes are largely silent. Consistently, RNA interference inhibition of UTX led to increased H3K27me3 levels at some HOX gene promoters. Importantly, morpholino oligonucleotide inhibition of a zebrafish UTX homologue resulted in mis-regulation of hox genes and a striking posterior developmental defect, which was partially rescued by wild-type, but not by catalytically inactive, human UTX. Taken together, these findings identify a small family of H3K27 demethylases with important, evolutionarily conserved roles in H3K27 methylation regulation and in animal anterior-posterior development.
Collapse
Affiliation(s)
- Fei Lan
- Department of Pathology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Shaut CA, Saneyoshi C, Morgan EA, Knosp WM, Sexton DR, Stadler HS. HOXA13 directly regulates EphA6 and EphA7 expression in the genital tubercle vascular endothelia. Dev Dyn 2007; 236:951-60. [PMID: 17304517 DOI: 10.1002/dvdy.21077] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Hypospadias, a common defect affecting the growth and closure of the external genitalia, is often accompanied by gross enlargements of the genital tubercle (GT) vasculature. Because Hoxa13 homozygous mutant mice also exhibit hypospadias and GT vessel expansion, we examined whether genes playing a role in angiogenesis exhibit reduced expression in the GT. From this analysis, reductions in EphA6 and EphA7 were detected. Characterization of EphA6 and EphA7 expression in the GT confirmed colocalization with HOXA13 in the GT vascular endothelia. Analysis of the EphA6 and EphA7 promoter regions revealed a series of highly conserved cis-regulatory elements bound by HOXA13 with high affinity. GT chromatin immunoprecipitation confirmed that HOXA13 binds these gene-regulatory elements in vivo. In vitro, HOXA13 activates gene expression through the EphA6 and EphA7 gene-regulatory elements. Together these findings indicate that HOXA13 directly regulates EphA6 and EphA7 in the developing GT and identifies the GT vascular endothelia as a novel site for HOXA13-dependent expression of EphA6 and EphA7.
Collapse
MESH Headings
- Animals
- Base Sequence
- Binding Sites
- Cells, Cultured
- Endothelium, Vascular/embryology
- Endothelium, Vascular/metabolism
- Gene Expression Regulation, Developmental
- Genitalia/blood supply
- Genitalia/embryology
- Genitalia/metabolism
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Homeodomain Proteins/physiology
- Mice
- Mice, Mutant Strains
- Molecular Sequence Data
- Promoter Regions, Genetic
- Receptor, EphA6/genetics
- Receptor, EphA6/metabolism
- Receptor, EphA7/genetics
- Receptor, EphA7/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Sequence Homology, Nucleic Acid
- Transfection
Collapse
Affiliation(s)
- Carley A Shaut
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon
| | | | | | | | | | | |
Collapse
|
37
|
Brunet I, Di Nardo AA, Sonnier L, Beurdeley M, Prochiantz A. The topological role of homeoproteins in the developing central nervous system. Trends Neurosci 2007; 30:260-7. [PMID: 17418905 DOI: 10.1016/j.tins.2007.03.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2006] [Revised: 02/13/2007] [Accepted: 03/29/2007] [Indexed: 11/24/2022]
Abstract
Homeogenes encode homeoprotein transcription factors that have fundamental roles in development. They are key players in genetic networks that lay out the body plan and also determine morphology and physiology at the cellular and multicellular level. However, homeoproteins share activities that extend beyond transcription, including translation regulation and signalling. For example, homeoproteins participate in the definition of territories in the neuroepithelium and also have a function in axonal guidance. Based on these examples, we propose that homeoproteins are not only morphogenetic transcription factors, but also morphogens themselves.
Collapse
Affiliation(s)
- Isabelle Brunet
- Unité Mixte de Recherche 8542, Development and Evolution of the Nervous System (Development and Neuropharmacology Group), Ecole normale supérieure, 46 rue d'Ulm, 75005 Paris, France
| | | | | | | | | |
Collapse
|
38
|
Sato T, Hamaoka T, Aizawa H, Hosoya T, Okamoto H. Genetic single-cell mosaic analysis implicates ephrinB2 reverse signaling in projections from the posterior tectum to the hindbrain in zebrafish. J Neurosci 2007; 27:5271-9. [PMID: 17507550 PMCID: PMC6672335 DOI: 10.1523/jneurosci.0883-07.2007] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The optic tectum is a visual center in vertebrates. It receives topographically ordered visual inputs from the retina in the superficial layers and then sends motor outputs from the deeper layers to the premotor reticulospinal system in the hindbrain. Although the topographic patterns of the retinotectal projection are well known, it is not yet well understood how tectal efferents in the tectobulbar tract project to the hindbrain. The retinotectal and the tectobulbar projections were visualized in a zebrafish stable transgenic line Tg(brn3a-hsp70:GFP). Using a single-neuron labeling system in combination with the cre/loxP and Gal4/UAS systems, we showed that the tectal neurons that projected to rhombomeres 2 and 6 were distributed with distinctive patterns along the anterior-posterior axis. Furthermore, we found that ephrinB2a was critically involved in increasing the probability of neurons projecting to rhombomere 2 through a reverse signaling mechanism. These results may provide a neuroanatomical and molecular basis for the motor command map in the tectum.
Collapse
Affiliation(s)
- Tomomi Sato
- Laboratory for Developmental Gene Regulation and
| | | | | | - Toshihiko Hosoya
- Hosoya Research Unit, Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
| | | |
Collapse
|
39
|
Kida YS, Sato T, Miyasaka KY, Suto A, Ogura T. Daam1 regulates the endocytosis of EphB during the convergent extension of the zebrafish notochord. Proc Natl Acad Sci U S A 2007; 104:6708-13. [PMID: 17412835 PMCID: PMC1871850 DOI: 10.1073/pnas.0608946104] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2006] [Indexed: 11/18/2022] Open
Abstract
Convergent extension (CE) movement of cells is one of the fundamental processes that control the organized morphogenesis of tissues and organs. The molecular events connecting the noncanonical Wnt pathway and CE movement, however, are not well understood. We show that subcellular localization of Daam1, an essential component of noncanonical Wnt signaling, changes dynamically during notochord formation. In the early phases, Daam1 complexes with EphB receptors and Disheveled 2. This complex is incorporated into endocytic vesicles in a dynamin-dependent manner, thereby resulting in the removal of EphB from the cell surface with subsequent switching of cell adhesiveness. In the next step, Daam1 colocalizes with the actin cytoskeleton to induce morphological extension of cells. We elucidate the molecular mechanism underlying the CE movement of notochord cells with Daam1 as a dynamic coordinator of endocytosis and cytoskeletal remodeling.
Collapse
Affiliation(s)
- Yasuyuki S Kida
- Department of Developmental Neurobiology, Institute of Development, Aging, and Cancer, Tohoku University, 4-1, Seiryo, Aoba, Sendai, Miyagi 980-8575, Japan
| | | | | | | | | |
Collapse
|
40
|
Iwase S, Lan F, Bayliss P, de la Torre-Ubieta L, Huarte M, Qi HH, Whetstine JR, Bonni A, Roberts TM, Shi Y. The X-linked mental retardation gene SMCX/JARID1C defines a family of histone H3 lysine 4 demethylases. Cell 2007; 128:1077-88. [PMID: 17320160 DOI: 10.1016/j.cell.2007.02.017] [Citation(s) in RCA: 535] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2006] [Revised: 01/16/2007] [Accepted: 02/09/2007] [Indexed: 12/13/2022]
Abstract
Histone methylation regulates chromatin structure and transcription. The recently identified histone demethylase lysine-specific demethylase 1 (LSD1) is chemically restricted to demethylation of only mono- and di- but not trimethylated histone H3 lysine 4 (H3K4me3). We show that the X-linked mental retardation (XLMR) gene SMCX (JARID1C), which encodes a JmjC-domain protein, reversed H3K4me3 to di- and mono- but not unmethylated products. Other SMCX family members, including SMCY, RBP2, and PLU-1, also demethylated H3K4me3. SMCX bound H3K9me3 via its N-terminal PHD (plant homeodomain) finger, which may help coordinate H3K4 demethylation and H3K9 methylation in transcriptional repression. Significantly, several XLMR-patient point mutations reduced SMCX demethylase activity and binding to H3K9me3 peptides, respectively. Importantly, studies in zebrafish and primary mammalian neurons demonstrated a role for SMCX in neuronal survival and dendritic development and a link to the demethylase activity. Our findings thus identify a family of H3K4me3 demethylases and uncover a critical link between histone modifications and XLMR.
Collapse
Affiliation(s)
- Shigeki Iwase
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Rohde LA, Heisenberg CP. Zebrafish Gastrulation: Cell Movements, Signals, and Mechanisms. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 261:159-92. [PMID: 17560282 DOI: 10.1016/s0074-7696(07)61004-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gastrulation is a morphogenetic process that results in the formation of the embryonic germ layers. Here we detail the major cell movements that occur during zebrafish gastrulation: epiboly, internalization, and convergent extension. Although gastrulation is known to be regulated by signaling pathways such as the Wnt/planar cell polarity pathway, many questions remain about the underlying molecular and cellular mechanisms. Key factors that may play a role in gastrulation cell movements are cell adhesion and cytoskeletal rearrangement. In addition, some of the driving force for gastrulation may derive from tissue interactions such as those described between the enveloping layer and the yolk syncytial layer. Future exploration of gastrulation mechanisms relies on the development of sensitive and quantitative techniques to characterize embryonic germ-layer properties.
Collapse
Affiliation(s)
- Laurel A Rohde
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | |
Collapse
|
42
|
Zhong H, Wu X, Huang H, Fan Q, Zhu Z, Lin S. Vertebrate MAX-1 is required for vascular patterning in zebrafish. Proc Natl Acad Sci U S A 2006; 103:16800-5. [PMID: 17065323 PMCID: PMC1636535 DOI: 10.1073/pnas.0603959103] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During embryogenesis, stereotypic vascular patterning requires guidance cues from neighboring tissues. However, key molecules involved in this process still remain largely elusive. Here, we report molecular cloning, expression, and functional studies of zebrafish max-1, a homolog of Caenorhabditis elegans max-1 that has been implicated in motor neuron axon guidance. During early embryonic development, zebrafish max-1 is specifically expressed in subsets of neuronal tissues, epithelial cells, and developing somites through which vascular endothelial cells migrate from large ventral axial vessels to form stereotypic intersegmental blood vessels (ISV). Blocking zebrafish max-1 mRNA splicing by morpholino injection led to aberrant ISV patterning, which could be rescued by injection of either C. elegans or zebrafish max-1 mRNA. Analysis of motor neurons in the same region showed normal neuronal axon pathfinding. Further studies suggested that the ISV defect caused by max-1 knockdown could be partially rescued by overexpression of ephrinb3 and that max-1 was involved in mediating membrane localization of ephrin proteins, which have been shown to provide guidance cues for endothelial cell migration. Our findings therefore suggest that max-1, acting upstream of the ephrin pathway, is critically required in vascular patterning in vertebrate species.
Collapse
Affiliation(s)
- Hanbing Zhong
- *College of Life Science, Peking University, Beijing 100871, China; and
| | - Xinrong Wu
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA 90095
| | - Haigen Huang
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA 90095
| | - Qichang Fan
- *College of Life Science, Peking University, Beijing 100871, China; and
| | - Zuoyan Zhu
- *College of Life Science, Peking University, Beijing 100871, China; and
| | - Shuo Lin
- *College of Life Science, Peking University, Beijing 100871, China; and
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA 90095
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
43
|
Jevince AR, Kadison SR, Pittman AJ, Chien CB, Kaprielian Z. Distribution of EphB receptors and ephrin-B1 in the developing vertebrate spinal cord. J Comp Neurol 2006; 497:734-50. [PMID: 16786562 PMCID: PMC2637817 DOI: 10.1002/cne.21001] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Contact-dependent interactions between EphB receptors and ephrin-B ligands mediate a variety of cell-cell communication events in the developing and mature central nervous system (CNS). These predominantly repulsive interactions occur at the interface between what are considered to be mutually exclusive EphB and ephrin-B expression domains. We previously used receptor and ligand affinity probes to show that ephrin-B ligands are expressed in the floor plate and within a dorsal region of the embryonic mouse spinal cord, while EphB receptors are present on decussated segments of commissural axons that navigate between these ephrin-B domains. Here we present the generation and characterization of two new monoclonal antibodies, mAb EfB1-3, which recognizes EphB1, EphB2, and EphB3, and mAb efrnB1, which is specific for ephrin-B1. We use these reagents and polyclonal antibodies specific for EphB1, EphB2, EphB3, or ephrin-B1 to describe the spatiotemporal expression patterns of EphB receptors and ephrin-B1 in the vertebrate spinal cord. Consistent with affinity probe binding, we show that EphB1, EphB2, and EphB3 are each preferentially expressed on decussated segments of commissural axons in vivo and in vitro, and that ephrin-B1 is expressed in a dorsal domain of the spinal cord that includes the roof plate. In contrast to affinity probe binding profiles, we show here that EphB1, EphB2, and EphB3 are present on the ventral commissure, and that EphB1 and EphB3 are expressed on axons that compose the dorsal funiculus. In addition, we unexpectedly find that mesenchymal cells, which surround the spinal cord and dorsal root ganglion, express ephrin-B1.
Collapse
Affiliation(s)
- Angela R Jevince
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | |
Collapse
|
44
|
Chittenden TW, Claes F, Lanahan AA, Autiero M, Palac RT, Tkachenko EV, Elfenbein A, Ruiz de Almodovar C, Dedkov E, Tomanek R, Li W, Westmore M, Singh JP, Horowitz A, Mulligan-Kehoe MJ, Moodie KL, Zhuang ZW, Carmeliet P, Simons M. Selective regulation of arterial branching morphogenesis by synectin. Dev Cell 2006; 10:783-95. [PMID: 16740480 DOI: 10.1016/j.devcel.2006.03.012] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Revised: 02/16/2006] [Accepted: 03/21/2006] [Indexed: 12/22/2022]
Abstract
Branching morphogenesis is a key process in the formation of vascular networks. To date, little is known regarding the molecular events regulating this process. We investigated the involvement of synectin in this process. In zebrafish embryos, synectin knockdown resulted in a hypoplastic dorsal aorta and hypobranched, stunted, and thin intersomitic vessels due to impaired migration and proliferation of angioblasts and arterial endothelial cells while not affecting venous development. Synectin(-/-) mice demonstrated decreased body and organ size, reduced numbers of arteries, and an altered pattern of arterial branching in multiple vascular beds while the venous system remained normal. Murine synectin(-/-) primary arterial, but not venous, endothelial cells showed decreased in vitro tube formation, migration, and proliferation and impaired polarization due to abnormal localization of activated Rac1. We conclude that synectin is involved in selective regulation of arterial, but not venous, growth and branching morphogenesis and that Rac1 plays an important role in this process.
Collapse
Affiliation(s)
- Thomas W Chittenden
- Angiogenesis Research Center, Section of Cardiology, Department of Medicine, Dartmouth Medical School, Hanover, New Hampshire 03755, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ho DM, Chan J, Bayliss P, Whitman M. Inhibitor-resistant type I receptors reveal specific requirements for TGF-beta signaling in vivo. Dev Biol 2006; 295:730-42. [PMID: 16684517 DOI: 10.1016/j.ydbio.2006.03.050] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Revised: 03/29/2006] [Accepted: 03/31/2006] [Indexed: 11/22/2022]
Abstract
Activin/nodal-like TGF-beta superfamily ligands signal through the type I receptors Alk4, Alk5, and Alk7, and are responsible for mediating a number of essential processes in development. SB-431542, a chemical inhibitor of activin/nodal signaling, acts by specifically interfering with type I receptors. Here, we use inhibitor-resistant mutant receptors to examine the efficacy and specificity of SB-431542 in Xenopus and zebrafish embryos. Treatment with SB-431542 eliminates Smad2 phosphorylation in vivo and generates a phenotype very similar to those observed in genetic mutants in the nodal signaling pathway. Inhibitor-resistant Alk4 efficiently rescues Smad2 signaling, developmental phenotype, and marker gene expression after inhibitor treatment. This system was used to examine type I receptor specificity for several activin/nodal ligands. We find that Alk4 can efficiently rescue signaling by a wide range of ligands, while Alk7 can only weakly rescue signaling by the same ligands. In whole embryos, nodal signaling during gastrulation can be rescued with Alk4, but not Alk7, while Alk5 can only mediate signaling by ligands expressed later in development. The combination of the ALK inhibitor SB-431542 with inhibitor-resistant ALKs provides a powerful set of tools for examining nodal/activin signaling during embryogenesis.
Collapse
Affiliation(s)
- Diana M Ho
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
46
|
Bayliss PE, Bellavance KL, Whitehead GG, Abrams JM, Aegerter S, Robbins HS, Cowan DB, Keating MT, O'Reilly T, Wood JM, Roberts TM, Chan J. Chemical modulation of receptor signaling inhibits regenerative angiogenesis in adult zebrafish. Nat Chem Biol 2006; 2:265-73. [PMID: 16565716 PMCID: PMC1534118 DOI: 10.1038/nchembio778] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Accepted: 02/21/2006] [Indexed: 12/20/2022]
Abstract
We examined the role of angiogenesis and the need for receptor signaling using chemical inhibition of the vascular endothelial growth factor receptor in the adult zebrafish tail fin. Using a small-molecule inhibitor, we were able to exert precise control over blood vessel regeneration. An angiogenic limit to tissue regeneration was determined, as avascular tissue containing skin, pigment, neuronal axons and bone precursors could regenerate up to about 1 mm. This indicates that tissues can regenerate without direct interaction with endothelial cells and at a distance from blood supply. We also investigated whether the effects of chemical inhibition could be enhanced in zebrafish vascular mutants. We found that adult zebrafish, heterozygous for a mutation in the critical receptor effector phospholipase Cgamma1, show a greater sensitivity to chemical inhibition. This study illustrates the utility of the adult zebrafish as a new model system for receptor signaling and chemical biology.
Collapse
Affiliation(s)
- Peter E Bayliss
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
The basic vertebrate body plan of the zebrafish embryo is established in the first 10 hours of development. This period is characterized by the formation of the anterior-posterior and dorsal-ventral axes, the development of the three germ layers, the specification of organ progenitors, and the complex morphogenetic movements of cells. During the past 10 years a combination of genetic, embryological, and molecular analyses has provided detailed insights into the mechanisms underlying this process. Maternal determinants control the expression of transcription factors and the location of signaling centers that pattern the blastula and gastrula. Bmp, Nodal, FGF, canonical Wnt, and retinoic acid signals generate positional information that leads to the restricted expression of transcription factors that control cell type specification. Noncanonical Wnt signaling is required for the morphogenetic movements during gastrulation. We review how the coordinated interplay of these molecules determines the fate and movement of embryonic cells.
Collapse
Affiliation(s)
- Alexander F Schier
- Developmental Genetics Program, Skirball Institute of Biomolecular Medicine, Department of Cell Biology, New York University School of Medicine, New York, NY 10016-6497, USA.
| | | |
Collapse
|
48
|
Jin SW, Beis D, Mitchell T, Chen JN, Stainier DYR. Cellular and molecular analyses of vascular tube and lumen formation in zebrafish. Development 2005; 132:5199-209. [PMID: 16251212 DOI: 10.1242/dev.02087] [Citation(s) in RCA: 653] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Tube and lumen formation are essential steps in forming a functional vasculature. Despite their significance, our understanding of these processes remains limited, especially at the cellular and molecular levels. In this study, we analyze mechanisms of angioblast coalescence in the zebrafish embryonic midline and subsequent vascular tube formation. To facilitate these studies, we generated a transgenic line where EGFP expression is controlled by the zebrafish flk1 promoter. We find that angioblasts migrate as individual cells to form a vascular cord at the midline. This transient structure is stabilized by endothelial cell-cell junctions, and subsequently undergoes lumen formation to form a fully patent vessel. Downregulating the VEGF signaling pathway, while affecting the number of angioblasts, does not appear to affect their migratory behavior. Our studies also indicate that the endoderm, a tissue previously implicated in vascular development, provides a substratum for endothelial cell migration and is involved in regulating the timing of this process, but that it is not essential for the direction of migration. In addition, the endothelial cells in endodermless embryos form properly lumenized vessels, contrary to what has been previously reported in Xenopus and avian embryos. These studies provide the tools and a cellular framework for the investigation of mutations affecting vasculogenesis in zebrafish.
Collapse
Affiliation(s)
- Suk-Won Jin
- Department of Biochemistry and Biophysics and Cardiovascular Research Institute, Program in Developmental Biology, University of California San Francisco, 1550 Fourth street, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
49
|
Honjo Y, Eisen JS. Slow muscle regulates the pattern of trunk neural crest migration in zebrafish. Development 2005; 132:4461-70. [PMID: 16162652 DOI: 10.1242/dev.02026] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In avians and mice, trunk neural crest migration is restricted to the anterior half of each somite. Sclerotome has been shown to play an essential role in this restriction; the potential role of other somite components in specifying neural crest migration is currently unclear. By contrast, in zebrafish trunk neural crest, migration on the medial pathway is restricted to the middle of the medial surface of each somite. Sclerotome comprises only a minor part of zebrafish somites, and the pattern of neural crest migration is established before crest cells contact sclerotome cells, suggesting other somite components regulate the pattern of zebrafish neural crest migration. Here, we use mutants to investigate which components regulate the pattern of zebrafish trunk neural crest migration on the medial pathway. The pattern of trunk neural crest migration is aberrant in spadetail mutants that have very reduced somitic mesoderm, in no tail mutants injected with spadetail morpholino antisense oligonucleotides that entirely lack somitic mesoderm and in somite segmentation mutants that have normal somite components but disrupted segment borders. Fast muscle cells appear dispensable for patterning trunk neural crest migration. However, migration is abnormal in Hedgehog signaling mutants that lack slow muscle cells, providing evidence that slow muscle cells regulate the pattern of trunk neural crest migration. Consistent with this idea, surgical removal of adaxial cells, which are slow muscle precursors, results in abnormal patterning of neural crest migration;normal patterning can be restored by replacing the ablated adaxial cells with ones transplanted from wild-type embryos.
Collapse
Affiliation(s)
- Yasuko Honjo
- Institute of Neuroscience, 1254 University of Oregon, Eugene, OR 97403, USA.
| | | |
Collapse
|
50
|
Barresi MJF, Hutson LD, Chien CB, Karlstrom RO. Hedgehog regulated Slit expression determines commissure and glial cell position in the zebrafish forebrain. Development 2005; 132:3643-56. [PMID: 16033800 DOI: 10.1242/dev.01929] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Three major axon pathways cross the midline of the vertebrate forebrain early in embryonic development: the postoptic commissure (POC), the anterior commissure (AC) and the optic nerve. We show that a small population of Gfap+ astroglia spans the midline of the zebrafish forebrain in the position of, and prior to, commissural and retinal axon crossing. These glial ;bridges' form in regions devoid of the guidance molecules slit2 and slit3, although a subset of these glial cells express slit1a. We show that Hh signaling is required for commissure formation, glial bridge formation, and the restricted expression of the guidance molecules slit1a, slit2, slit3 and sema3d, but that Hh does not appear to play a direct role in commissural and retinal axon guidance. Reducing Slit2 and/or Slit3 function expanded the glial bridges and caused defasciculation of the POC, consistent with a ;channeling' role for these repellent molecules. By contrast, reducing Slit1a function led to reduced midline axon crossing, suggesting a distinct role for Slit1a in midline axon guidance. Blocking Slit2 and Slit3, but not Slit1a, function in the Hh pathway mutant yot (gli2DR) dramatically rescued POC axon crossing and glial bridge formation at the midline, indicating that expanded Slit2 and Slit3 repellent function is largely responsible for the lack of midline crossing in these mutants. This analysis shows that Hh signaling helps to pattern the expression of Slit guidance molecules that then help to regulate glial cell position and axon guidance across the midline of the forebrain.
Collapse
|