Keep Your Friends Close: Cell–Cell Contact and Skeletal Myogenesis

Total Page:16

File Type:pdf, Size:1020Kb

Keep Your Friends Close: Cell–Cell Contact and Skeletal Myogenesis Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press Keep Your Friends Close: Cell–Cell Contact and Skeletal Myogenesis Robert S. Krauss, Giselle A. Joseph, and Aviva J. Goel Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029 Correspondence: [email protected] Development of skeletal muscle is a multistage process that includes lineage commitment of multipotent progenitor cells, differentiation and fusion of myoblasts into multinucleated myofibers, and maturation of myofibers into distinct types. Lineage-specific transcriptional regulation lies at the core of this process, but myogenesis is also regulated by extracellular cues. Some of these cues are initiated by direct cell–cell contact between muscle precursor cells themselves or between muscle precursors and cells of other lineages. Examples of the latter include interaction of migrating neural crest cells with multipotent muscle progenitor cells, muscle interstitial cells with myoblasts, and neurons with myofibers. Among the sig- naling factors involved are Notch ligands and receptors, cadherins, Ig superfamily members, and Ephrins and Eph receptors. In this article we describe recent progress in this area and highlight open questions raised by the findings. keletal muscle is the most abundant tissue in based on expression of distinct myosin heavy Sthe vertebrate body and necessary for breath- chain (MyHC) isoforms and metabolic capabil- ing, metabolic homeostasis, and locomotion. ities (Schiaffino and Reggiani 2011; Talbot and Development of skeletal muscle occurs in a se- Maves 2016). quential, multistage process that includes spec- Myogenic specification and differentiation ification of multipotent progenitors to lineage- are coordinated by the myogenic basic helix– committed myoblasts, differentiation of myo- loop–helix (bHLH) transcription factors Myf5, blasts into multinucleated myofibers, and mat- MyoD, myogenin, and MRF4. Uncommitted uration of myofibers by innervation and bun- progenitor cells are specified to become line- dling into functional muscles (Biressi et al. age-committed myoblasts through the com- 2007; Comai and Tajbakhsh 2014; Tintignac bined actions of Myf5, MRF4, and MyoD, et al. 2015). Myoblast differentiation is itself a whereas differentiation of myoblasts is regulated complex process that involves both induction of by myogenin, MyoD, and MRF4 (Tapscott2005; the muscle-specific transcriptome and fusion of Biressi et al. 2007; Comai and Tajbakhsh 2014). myoblasts into an elongated syncytium. Simi- Expression of these factors, particularly MyoD, larly, maturation is multifaceted. Mature myo- in many nonmuscle cell types converts such cells fibers are classified as “slow” or “fast” types, to the skeletal muscle program, revealing their Editors: Carien M. Niessen and Alpha S. Yap Additional Perspectives on Cell–Cell Junctions available at www.cshperspectives.org Copyright # 2017 Cold Spring Harbor Laboratory Press; all rights reserved Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 1 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press R.S. Krauss et al. ability to drive tissue-specific transcription and dermomyotome, which has epithelial character cell differentiation (Tapscott 2005; Fong and and gives rise to skeletal muscle and dermis, and Tapscott2013). The ability of MyoD to function ventrally into the sclerotome, which has mesen- in this manner occurs in conjunction with non- chymal character and gives rise to the bones and muscle-specific factors such as its heterodimeric cartilage of the axial skeleton (Brand-Saberi and partners, the E proteins; members of the Mef2 Christ 2000). Some dermomyotomal progenitor family of transcription factors; transcriptional cells undergo an epithelial-mesenchymal transi- coactivators; and chromatin remodeling factors tion (EMT), become committed to the skeletal (Tapscott 2005; Biressi et al. 2007; Sartorelli and muscle lineage, and migrate between the dermo- Juan 2011; Fong and Tapscott 2013; Comai and myotome and sclerotome to form the myotome, Tajbakhsh 2014). These and additional tran- a set of differentiated embryonic muscle cells. scription factors orchestrate development of Subsequent embryonic, fetal, and postnatal the skeletal muscle lineage. stages of myogenesis involve additional muscle Although the importance of lineage-specific progenitorsthat originally migrate from the der- transcriptional regulation in skeletal myogene- momyotome and ultimately establish the trunk sis has long been appreciated, myogenesis is also and limb musculature (Biressi et al. 2007; Comai regulated by extracellular cues that initiate in- and Tajbakhsh 2014). tracellular signaling (Guasconi and Puri 2009). Signals from the adjacent dorsal neural tube Some of these extracellular cues are secreted fac- and surface ectoderm play important roles in tors, such as fibroblast growth factor (Fgf ), Wnt maturation of the dorsal somite (Munsterberg family ligands, and Sonic hedgehog (Shh). A and Lassar 1995; Stern and Hauschka 1995). growing body of evidence indicates that signal- These tissues secrete Wnt1 and Wnt3a, which ing initiated by direct cell–cell contact also pro- signal via the “canonical” pathway to stimu- vides key regulatory information during devel- late b-catenin/T-cell factor (TCF)–dependent opment of the skeletal muscle lineage. In adult transcription. These Wnt ligands promote myo- skeletal muscle, stable cell–cell junctions (ex- genesis of explanted epithelial somites in vitro emplified by epithelial, cadherin-based adhe- (Munsterberg et al. 1995; Stern et al. 1995; Taj- rens junctions) do not appear to exist between bakhsh et al. 1998; Borello et al. 2006). Further- myofibers, which are the unit cells of this tissue. more, an enhancer that regulates Myf5 expres- (Cell–cell junctions do exist between myofibers sion in the dorsal somite harbors TCF binding and skeletal muscle stem cells known as satellite sites that are critical for Wnt responsiveness and cells; this is discussed below.) Nevertheless, enhancer function (Borello et al. 2006) (N.B.: cell–cell adhesion, and signaling that derives Myf5 is the first myogenic bHLH factor ex- from cell–cell contact, occurs between various pressed during development, and its role is muscle precursor cells and between these cells solely in lineage determination.) These results and nonmuscle cell types during myogenesis. argued that canonical Wnt ligands, along with Furthermore, such interactions are important Shh and soluble bone morphogenetic protein for muscle development. In this review, we dis- (BMP) inhibitors (Munsterberg et al. 1995; Re- cuss the roles and mechanisms whereby cell– shef et al. 1998; Borycki et al. 1999), induce cell contact regulates skeletal myogenesis. muscle lineage specification of dermomyotomal progenitor cells via b-catenin/TCF-dependent expression of Myf5. PROGENITOR CELL COMMITMENT TO THE b-catenin is well known for having two dis- SKELETAL MUSCLE CELL LINEAGE tinct cellular functions: (1) as a transcriptional In vertebrates, skeletal muscles of the trunk and regulator that associates with the DNA-binding limbs develop from somites, transiently existing TCF proteins in a Wnt signaling-responsive blocks of columnar epithelial cells that form in manner; and (2) as an adaptor protein that an anterior-to-posterior manner from paraxial binds to the cytoplasmic tail of cadherins and mesoderm. Somites mature dorsally into the promotes linkage to the cytoskeleton (van 2 Advanced Online Article. Cite this article as Cold Spring Harb Perspect Biol doi: 10.1101/cshperspect.a029298 Downloaded from http://cshperspectives.cshlp.org/ on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press Cell–Cell Contact and Skeletal Myogenesis Amerongen and Nusse 2009; Niessen et al. of the dermomyotome express Notch receptors 2011). Cells of the epithelial dermomyotome (Rios et al. 2011). Transient cell–cell contact express the cell–cell adhesion molecule N-cad- between migrating, Delta1þ neural crest cells herin with an apical localization; its adhesive and Notchþ muscle progenitors in the dermo- junction partner, b-catenin, is localized there myotome leads to Notch signaling in the latter. as well (Cinnamon et al. 2006). Studies with This is, in turn, required for Myf5 induction and chick embryos suggest that changes in N-cad- adoption of the skeletal muscle cell fate (Rios herin expression and subcellular localization et al. 2011). This “kiss and run” mode of cell– play important roles in specification of multi- cell contact-dependent signaling strongly sug- potent dermomyotomal cells to the muscle lin- gests that the concurrent timing of neural crest eage. As these cells divide, N-cadherin is asym- cell migration and myotome formation is mech- metrically localized in cells that are fated to anistically linked. become skeletal muscle cells of the myotome; Canonical Notch signaling involves cleavage conversely, daughter cells that lack N-cadherin of its intracellular domain (NICD), which take on features of early dermal cells (Cinna- translocates to the nucleus and binds RBP-J to mon et al. 2006). Consistent with these obser- stimulate pathway-responsive gene expression vations, electroporation of wild-type or domi- (Fortini 2009). In exploring how Notch signal-
Recommended publications
  • Supplemental Tables4.Pdf
    Yano_Supplemental_Table_S4 Gene ontology – Biological process 1 of 9 Fold List Pop Pop GO Term Count % PValue Bonferroni Benjamini FDR Genes Total Hits Total Enrichment DLC1, CADM1, NELL2, CLSTN1, PCDHGA8, CTNNB1, NRCAM, APP, CNTNAP2, FERT2, RAPGEF1, PTPRM, MPDZ, SDK1, PCDH9, PTPRS, VEZT, NRXN1, MYH9, GO:0007155~cell CTNNA2, NCAM1, NCAM2, DDR1, LSAMP, CNTN1, 50 5.61 2.14E-08 510 311 7436 2.34 4.50E-05 4.50E-05 3.70E-05 adhesion ROR2, VCAN, DST, LIMS1, TNC, ASTN1, CTNND2, CTNND1, CDH2, NEO1, CDH4, CD24A, FAT3, PVRL3, TRO, TTYH1, MLLT4, LPP, NLGN1, PCDH19, LAMA1, ITGA9, CDH13, CDON, PSPC1 DLC1, CADM1, NELL2, CLSTN1, PCDHGA8, CTNNB1, NRCAM, APP, CNTNAP2, FERT2, RAPGEF1, PTPRM, MPDZ, SDK1, PCDH9, PTPRS, VEZT, NRXN1, MYH9, GO:0022610~biological CTNNA2, NCAM1, NCAM2, DDR1, LSAMP, CNTN1, 50 5.61 2.14E-08 510 311 7436 2.34 4.50E-05 4.50E-05 3.70E-05 adhesion ROR2, VCAN, DST, LIMS1, TNC, ASTN1, CTNND2, CTNND1, CDH2, NEO1, CDH4, CD24A, FAT3, PVRL3, TRO, TTYH1, MLLT4, LPP, NLGN1, PCDH19, LAMA1, ITGA9, CDH13, CDON, PSPC1 DCC, ENAH, PLXNA2, CAPZA2, ATP5B, ASTN1, PAX6, ZEB2, CDH2, CDH4, GLI3, CD24A, EPHB1, NRCAM, GO:0006928~cell CTTNBP2, EDNRB, APP, PTK2, ETV1, CLASP2, STRBP, 36 4.04 3.46E-07 510 205 7436 2.56 7.28E-04 3.64E-04 5.98E-04 motion NRG1, DCLK1, PLAT, SGPL1, TGFBR1, EVL, MYH9, YWHAE, NCKAP1, CTNNA2, SEMA6A, EPHA4, NDEL1, FYN, LRP6 PLXNA2, ADCY5, PAX6, GLI3, CTNNB1, LPHN2, EDNRB, LPHN3, APP, CSNK2A1, GPR45, NRG1, RAPGEF1, WWOX, SGPL1, TLE4, SPEN, NCAM1, DDR1, GRB10, GRM3, GNAQ, HIPK1, GNB1, HIPK2, PYGO1, GO:0007166~cell RNF138, ROR2, CNTN1,
    [Show full text]
  • Hedgehog Signaling, Epithelial-To-Mesenchymal Transition and Mirna (Review)
    271-275 1/8/08 15:18 Page 271 INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE 22: 271-275, 2008 271 Hedgehog signaling, epithelial-to-mesenchymal transition and miRNA (Review) YURIKO KATOH1 and MASARU KATOH2 1M&M Medical Bioinformatics, Hongo 113-0033; 2Genetics and Cell Biology Section, National Cancer Center, Tokyo 104-0045, Japan Received May 19, 2008; Accepted June 26, 2008 DOI: 10.3892/ijmm_00000019 Abstract. SHH, IHH, and DHH are lipid-modified secreted strictly controlled before clinical application of synthetic proteins binding to Patched receptors, and CDON, BOC or miRNA. Peptide mimetic and RNA aptamer could also be GAS1 co-receptors. In the absence of Hedgehog signaling, utilized as Hedgehog signaling inhibitors or EMT suppressors. GLI1 is transcriptionally repressed, GLI2 is phosphorylated by GSK3 and CK1 for the FBXW11 (ßTRCP2)-mediated degradation, and GLI3 is processed to a cleaved repressor. In Contents the presence of Hedgehog signaling, Smoothened is relieved from Patched-mediated suppression due to the Hedgehog- 1. Introduction dependent internalization of Patched, which leads to 2. Hedgehog signaling pathway MAP3K10 (MST) activation and SUFU inactivation for the 3. Epithelial-to-mesenchymal transition (EMT) stabilization and nuclear accumulation of GLI family 4. Hedgehog and EMT members. GLI activators then upregulate CCND1, CCND2 5. MicroRNA (miRNA) for cell cycle acceleration, FOXA2, FOXC2, FOXE1, 6. Perspectives FOXF1, FOXL1, FOXP3, POU3F1, RUNX2, SOX13, TBX2 for cell fate determination, JAG2, INHBC, and INHBE for 1. Introduction stem cell signaling regulation. Hedgehog signals also up- regulate SFRP1 in mesenchymal cells for WNT signaling Hedgehog family members are key regulators of embryo- regulation. Epithelial-to-mesenchymal transition (EMT) during genesis, adult tissue homeostasis, and carcinogenesis (1-5).
    [Show full text]
  • Mutations in CDON, Encoding a Hedgehog Receptor, Result in Holoprosencephaly and Defective Interactions with Other Hedgehog Receptors
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector ARTICLE Mutations in CDON, Encoding a Hedgehog Receptor, Result in Holoprosencephaly and Defective Interactions with Other Hedgehog Receptors Gyu-Un Bae,1,2,4,6 Sabina Domene´,3,4,7 Erich Roessler,3 Karen Schachter,1,8 Jong-Sun Kang,2,5,* Maximilian Muenke,3,5,* and Robert S. Krauss1,5 Holoprosencephaly (HPE), a common human congenital anomaly defined by a failure to delineate the midline of the forebrain and/or midface, is associated with diminished Sonic hedgehog (SHH)-pathway activity in development of these structures. SHH signaling is regulated by a network of ligand-binding factors, including the primary receptor PTCH1 and the putative coreceptors, CDON (also called CDO), BOC, and GAS1. Although binding of SHH to these receptors promotes pathway activity, it is not known whether interactions between these receptors are important. We report here identification of missense CDON mutations in human HPE. These mutations diminish CDON’s ability to support SHH-dependent gene expression in cell-based signaling assays. The mutations occur outside the SHH-binding domain of CDON, and the encoded variant CDON proteins do not display defects in binding to SHH. In contrast, wild-type CDON associates with PTCH1 and GAS1, but the variants do so inefficiently, in a manner that parallels their activity in cell-based assays. Our findings argue that CDON must associate with both ligand and other hedgehog-receptor components, particularly PTCH1, for signaling to occur and that disruption of the latter interactions is a mechanism of HPE.
    [Show full text]
  • Role of Cytonemes in Wnt Transport Eliana Stanganello and Steffen Scholpp*
    © 2016. Published by The Company of Biologists Ltd | Journal of Cell Science (2016) 129, 665-672 doi:10.1242/jcs.182469 COMMENTARY Role of cytonemes in Wnt transport Eliana Stanganello and Steffen Scholpp* ABSTRACT dependent or canonical Wnt signaling (reviewed in Logan and Wnt signaling regulates a broad variety of processes during Nusse, 2004). Paracrine signaling activity is fundamental to the embryonic development and disease. A hallmark of the Wnt morphogenetic function of Wnt proteins in tissue patterning. signaling pathway is the formation of concentration gradients by However, the extracellular transport mechanism of this morphogen Wnt proteins across responsive tissues, which determines cell fate from the signal-releasing cell to the recipient cell is still debated. in invertebrates and vertebrates. To fulfill its paracrine function, Recent data suggest that Wnt proteins are distributed on long trafficking of the Wnt morphogen from an origin cell to a recipient cell signaling filopodia known as cytonemes, which allow contact- must be tightly regulated. A variety of models have been proposed to dependent, juxtacrine signaling over a considerable distance. We explain the extracellular transport of these lipid-modified signaling have recently shown that, in zebrafish, Wnt8a is transported on proteins in the aqueous extracellular space; however, there is still actin-containing cytonemes to cells, where it activates the signaling considerable debate with regard to which mechanisms allow the required for the specification of neural plate cells (Stanganello et al., precise distribution of ligand in order to generate a morphogenetic 2015). Concomitantly, the Wnt receptor Frz was identified on gradient within growing tissue. Recent evidence suggests that Wnt cytonemes to enable the retrograde transport of Wnt proteins on proteins are distributed along signaling filopodia during vertebrate these protrusions during flight muscle formation in Drosophila and invertebrate embryogenesis.
    [Show full text]
  • PRMT7 Ablation in Cardiomyocytes Causes Cardiac Hypertrophy and Fibrosis Through Β-Catenin Dysregulation
    PRMT7 Ablation in Cardiomyocytes Causes Cardiac Hypertrophy and Fibrosis Through β-Catenin Dysregulation Byeong-Yun Ahn Sungkyunkwan University School of Medicine Myong-Ho Jeong Sungkyunkwan University School of Medicine Jung-Hoon Pyun Sungkyunkwan University School of Medicine Hyeon-Ju Jeong Sungkyunkwan University School of Medicine Tuan Anh Vuong Sungkyunkwan University School of Medicine Ju-Hyeon Bae Sungkyunkwan University School of Medicine Subin An Sungkyunkwan University School of Medicine Su Woo Kim Sungkyunkwan University School of Medicine Yong Kee Kim Sookmyung Women's University Dongryeol Ryu Sungkyunkwan University School of Medicine Hyun-Ji Kim Sungkyunkwan University School of Medicine Hana Cho Sungkyunkwan University School of Medicine Gyu-Un Bae ( [email protected] ) Sookmyung Women's University College of Pharmacy https://orcid.org/0000-0003-4262-1895 Jong-Sun Kang Sungkyunkwan University School of Medicine Research Article Page 1/24 Keywords: PRMT7, cardiomyopathy, Wnt, β-Catenin Posted Date: August 19th, 2021 DOI: https://doi.org/10.21203/rs.3.rs-798227/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 2/24 Abstract Angiotensin II (AngII) has potent cardiac hypertrophic effects mediated through activation of hypertrophic signaling like Wnt/b-Catenin signaling. In the current study, we examined the role of protein arginine methyltransferase 7 (PRMT7) in cardiac function. PRMT7 was greatly decreased in hypertrophic hearts chronically infused with AngII and cardiomyocytes treated with AngII. PRMT7 depletion in rat cardiomyocytes resulted in hypertrophic responses. Consistently, mice lacking PRMT7 exhibited displayed the cardiac hypertrophy and brosis. PRMT7 overexpression abrogated the cellular hypertrophy elicited by AngII, while PRMT7 depletion exacerbated the hypertrophic response caused by AngII.
    [Show full text]
  • Sex-Specific Transcriptome Differences in Human Adipose
    G C A T T A C G G C A T genes Article Sex-Specific Transcriptome Differences in Human Adipose Mesenchymal Stem Cells 1, 2, 3 1,3 Eva Bianconi y, Raffaella Casadei y , Flavia Frabetti , Carlo Ventura , Federica Facchin 1,3,* and Silvia Canaider 1,3 1 National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)—Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy; [email protected] (E.B.); [email protected] (C.V.); [email protected] (S.C.) 2 Department for Life Quality Studies (QuVi), University of Bologna, Corso D’Augusto 237, 47921 Rimini, Italy; [email protected] 3 Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; fl[email protected] * Correspondence: [email protected]; Tel.: +39-051-2094114 These authors contributed equally to this work. y Received: 1 July 2020; Accepted: 6 August 2020; Published: 8 August 2020 Abstract: In humans, sexual dimorphism can manifest in many ways and it is widely studied in several knowledge fields. It is increasing the evidence that also cells differ according to sex, a correlation still little studied and poorly considered when cells are used in scientific research. Specifically, our interest is on the sex-related dimorphism on the human mesenchymal stem cells (hMSCs) transcriptome. A systematic meta-analysis of hMSC microarrays was performed by using the Transcriptome Mapper (TRAM) software. This bioinformatic tool was used to integrate and normalize datasets from multiple sources and allowed us to highlight chromosomal segments and genes differently expressed in hMSCs derived from adipose tissue (hADSCs) of male and female donors.
    [Show full text]
  • Deregulation of Cell Adhesion Molecules Is Associated with Progression and Poor Outcomes in Endometrial Cancer: Analysis of the Cancer Genome Atlas Data
    1906 ONCOLOGY LETTERS 19: 1906-1914, 2020 Deregulation of cell adhesion molecules is associated with progression and poor outcomes in endometrial cancer: Analysis of The Cancer Genome Atlas data XIANGJUN HE1, SHU LEI1,2, QI ZHANG1, LIPING MA1, NA LI1 and JIANLIU WANG2 1Central Laboratory and Institute of Clinical Molecular Biology; 2Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, P.R. China Received June 18, 2019; Accepted November 15, 2019 DOI: 10.3892/ol.2020.11295 Abstract. Cell adhesion molecules (CAMs) determine the 11 times of that in those without elevation of these 6 CAMs. behavior of cancer cells during metastasis. Although some Similar results were obtained when relapse-free survival (RFS) CAMs are dysregulated in certain types of cancer and time was used during the analysis. Prognostic reliability of the are associated with cancer progression, to the best of our six-gene model was validated using data of an independent knowledge, a comprehensive study of CAMs has not been cohort from the International Cancer Genome Consortium. In undertaken, particularly in endometrial cancer (EC). In the conclusion, a combination of CAM alterations contributed to present study the expression of 225 CAMs in EC patients progression and aggressiveness of EC. The six-gene signature with various clinicopathological phenotypes were evaluated was effective for predicting worse OS and RFS in patients by statistical analysis using publicly available data from The with EC and could be complementary to the present clinical Cancer Genome Atlas database. The Kaplan-Meier method, prognostic criteria. and univariate and multivariate Cox proportional hazards regression models were used for survival analyses.
    [Show full text]
  • Cdon Acts As a Hedgehog Decoy Receptor During Proximal-Distal Patterning of the Optic Vesicle
    ARTICLE Received 10 Dec 2013 | Accepted 26 May 2014 | Published 8 Jul 2014 DOI: 10.1038/ncomms5272 OPEN Cdon acts as a Hedgehog decoy receptor during proximal-distal patterning of the optic vesicle Marcos Julia´n Cardozo1,2, Luisa Sa´nchez-Arrones1,2,A´frica Sandonis1,2, Cristina Sa´nchez-Camacho2,w, Gaia Gestri3, Stephen W. Wilson3, Isabel Guerrero1 & Paola Bovolenta1,2 Patterning of the vertebrate optic vesicle into proximal/optic stalk and distal/neural retina involves midline-derived Hedgehog (Hh) signalling, which promotes stalk specification. In the absence of Hh signalling, the stalks are not specified, causing cyclopia. Recent studies showed that the cell adhesion molecule Cdon forms a heteromeric complex with the Hh receptor Patched 1 (Ptc1). This receptor complex binds Hh and enhances signalling activation, indicating that Cdon positively regulates the pathway. Here we show that in the developing zebrafish and chick optic vesicle, in which cdon and ptc1 are expressed with a complementary pattern, Cdon acts as a negative Hh signalling regulator. Cdon predominantly localizes to the basolateral side of neuroepithelial cells, promotes the enlargement of the neuroepithelial basal end-foot and traps Hh protein, thereby limiting its dispersion. This Ptc-independent function protects the retinal primordium from Hh activity, defines the stalk/retina boundary and thus the correct proximo-distal patterning of the eye. 1 Centro de Biologı´a Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientı´ficas—Universidad Auto´noma de Madrid, 28049 Madrid Spain. 2 Ciber de Enfermedades Raras (CIBERER), ISCIII, c/ Nicolas Cabrera 1, 28049 Madrid Spain. 3 Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
    [Show full text]
  • Cdon Mutation and Fetal Alcohol Converge on Nodal Signaling in a Mouse Model of Holoprosencephaly
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.30.070870; this version posted June 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Cdon mutation and fetal alcohol converge on Nodal signaling in a 2 mouse model of holoprosencephaly 3 4 Mingi Hong1, Annabel Christ2, Anna Christa2, Thomas E. Willnow2, and Robert S. Krauss1,* 5 6 7 1Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at 8 Mount Sinai, New York, NY 10029, USA; 2Max-Delbruck-Center for Molecular Medicine, 13125 9 Berlin, Germany 10 11 12 13 *Corresponding Author: 14 Robert S. Krauss, Department of Cell, Developmental, and Regenerative Biology, Icahn School 15 of Medicine at Mount Sinai, New York, NY 10029. Phone: 212-241-2177; Fax: 212-860-9279; 16 e-mail: [email protected] 17 18 19 20 21 22 23 24 25 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.30.070870; this version posted June 24, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 26 Abstract 27 Holoprosencephaly (HPE), a defect in midline patterning of the forebrain and midface, arises ~1 28 in 250 conceptions. It is associated with predisposing mutations in the Nodal and Hedgehog 29 (HH) pathways, with penetrance and expressivity graded by genetic and environmental 30 modifiers, via poorly understood mechanisms. CDON is a multifunctional co-receptor, including 31 for the HH pathway.
    [Show full text]
  • Developmental Enhancers Revealed by Extensive DNA Methylome Maps of Zebrafish Early Embryos
    ARTICLE Received 24 Oct 2014 | Accepted 19 Jan 2015 | Published 20 Feb 2015 DOI: 10.1038/ncomms7315 Developmental enhancers revealed by extensive DNA methylome maps of zebrafish early embryos Hyung Joo Lee1,2, Rebecca F. Lowdon1,2, Brett Maricque1,2, Bo Zhang1,2, Michael Stevens1,2, Daofeng Li1,2, Stephen L. Johnson1 & Ting Wang1,2 DNA methylation undergoes dynamic changes during development and cell differentiation. Recent genome-wide studies discovered that tissue-specific differentially methylated regions (DMRs) often overlap tissue-specific distal cis-regulatory elements. However, developmental DNA methylation dynamics of the majority of the genomic CpGs outside gene promoters and CpG islands has not been extensively characterized. Here, we generate and compare comprehensive DNA methylome maps of zebrafish developing embryos. From these maps, we identify thousands of developmental stage-specific DMRs (dsDMRs) across zebrafish developmental stages. The dsDMRs contain evolutionarily conserved sequences, are associated with developmental genes and are marked with active enhancer histone posttranslational modifications. Their methylation pattern correlates much stronger than promoter methylation with expression of putative target genes. When tested in vivo using a transgenic zebrafish assay, 20 out of 20 selected candidate dsDMRs exhibit functional enhancer activities. Our data suggest that developmental enhancers are a major target of DNA methylation changes during embryogenesis. 1 Department of Genetics, Washington University School of Medicine, St Louis, Missouri 63108, USA. 2 Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, Missouri 63108, USA. Correspondence and requests for materials should be addressed to T.W. (email: [email protected]). NATURE COMMUNICATIONS | 6:6315 | DOI: 10.1038/ncomms7315 | www.nature.com/naturecommunications 1 & 2015 Macmillan Publishers Limited.
    [Show full text]
  • Overweight in Mice and Enhanced Adipogenesis in Vitro Are Associated with Lack of the Hedgehog Coreceptor Boc
    2092 Diabetes Volume 64, June 2015 Hye-Jin Lee,1 Shin-Bum Jo,1 Anthony I. Romer,2,3 Hyo-Jeong Lim,1 Min-Jung Kim,4 Seung-Hoi Koo,4 Robert S. Krauss,2,3 and Jong-Sun Kang1 Overweight in Mice and Enhanced Adipogenesis In Vitro Are Associated With Lack of the Hedgehog Coreceptor Boc Diabetes 2015;64:2092–2103 | DOI: 10.2337/db14-1017 Obesity arises from a combination of genetic, environ- environmental, and behavioral factors that influence en- mental, and behavioral factors. However, the processes ergy balance. The principal feature of obesity is excessive that regulate white adipose tissue (WAT) expansion at accumulation of white adipose tissue (WAT) (1,3–5). Al- the level of the adipocyte are not well understood. The though obesity is understood to be a centrally regulated Hedgehog (HH) pathway plays a conserved role in consequence of overnutrition and/or reduced expenditure adipogenesis, inhibiting fat formation in vivo and in vitro, of energy, the processes that regulate WAT expansion at but it has not been shown that mice with reduced HH the level of the adipocyte are not well characterized. pathway activity have enhanced adiposity. We report WAT expansion occurs through adipocyte hypertrophy that mice lacking the HH coreceptor BOC displayed and hyperplasia, so such processes presumably engage at age-related overweight and excess WAT. They also some level the process of adipogenesis (3,4). Adipogenesis displayed alterations in some metabolic parameters has been analyzed with preadipocyte cell lines such as but normal food intake. Furthermore, they had an 3T3-L1 and various mutant mouse lines (6).
    [Show full text]
  • A Grainyhead-Like 2/Ovo-Like 2 Pathway Regulates Renal Epithelial Barrier Function and Lumen Expansion
    BASIC RESEARCH www.jasn.org A Grainyhead-Like 2/Ovo-Like 2 Pathway Regulates Renal Epithelial Barrier Function and Lumen Expansion † ‡ | Annekatrin Aue,* Christian Hinze,* Katharina Walentin,* Janett Ruffert,* Yesim Yurtdas,*§ | Max Werth,* Wei Chen,* Anja Rabien,§ Ergin Kilic,¶ Jörg-Dieter Schulzke,** †‡ Michael Schumann,** and Kai M. Schmidt-Ott* *Max Delbrueck Center for Molecular Medicine, Berlin, Germany; †Experimental and Clinical Research Center, and Departments of ‡Nephrology, §Urology, ¶Pathology, and **Gastroenterology, Charité Medical University, Berlin, Germany; and |Berlin Institute of Urologic Research, Berlin, Germany ABSTRACT Grainyhead transcription factors control epithelial barriers, tissue morphogenesis, and differentiation, but their role in the kidney is poorly understood. Here, we report that nephric duct, ureteric bud, and collecting duct epithelia express high levels of grainyhead-like homolog 2 (Grhl2) and that nephric duct lumen expansion is defective in Grhl2-deficient mice. In collecting duct epithelial cells, Grhl2 inactivation impaired epithelial barrier formation and inhibited lumen expansion. Molecular analyses showed that GRHL2 acts as a transcrip- tional activator and strongly associates with histone H3 lysine 4 trimethylation. Integrating genome-wide GRHL2 binding as well as H3 lysine 4 trimethylation chromatin immunoprecipitation sequencing and gene expression data allowed us to derive a high-confidence GRHL2 target set. GRHL2 transactivated a group of genes including Ovol2, encoding the ovo-like 2 zinc finger transcription factor, as well as E-cadherin, claudin 4 (Cldn4), and the small GTPase Rab25. Ovol2 induction alone was sufficient to bypass the requirement of Grhl2 for E-cadherin, Cldn4,andRab25 expression. Re-expression of either Ovol2 or a combination of Cldn4 and Rab25 was sufficient to rescue lumen expansion and barrier formation in Grhl2-deficient collecting duct cells.
    [Show full text]