Evolutionarily Conserved Tbx5–Wnt2/2B Pathway Orchestrates Cardiopulmonary Development

Total Page:16

File Type:pdf, Size:1020Kb

Evolutionarily Conserved Tbx5–Wnt2/2B Pathway Orchestrates Cardiopulmonary Development Evolutionarily conserved Tbx5–Wnt2/2b pathway orchestrates cardiopulmonary development Jeffrey D. Steimlea,b,c, Scott A. Rankind,e,f,g, Christopher E. Slagleh,i,j,k, Jenna Bekenya,b,c, Ariel B. Rydeena,b,c, Sunny Sun-Kin Chanl,m, Junghun Kweona,b,c, Xinan H. Yanga,b,c, Kohta Ikegamia,b,c, Rangarajan D. Nadadura,b,c, Megan Rowtona,b,c, Andrew D. Hoffmanna,b,c, Sonja Lazarevica,b,c, William Thomasn,o, Erin A. T. Boyle Andersonp, Marko E. Horbn,o, Luis Luna-Zuritaq,r, Robert K. Hom, Michael Kybal,m, Bjarke Jensens, Aaron M. Zornd,e,f,g, Frank L. Conlonh,i,j,k, and Ivan P. Moskowitza,b,c,1 aDepartment of Pediatrics, University of Chicago, Chicago, IL 60637; bDepartment of Pathology, University of Chicago, Chicago, IL 60637; cDepartment of Human Genetics, University of Chicago, Chicago, IL 60637; dCenter for Stem Cell and Organoid Medicine, Cincinnati Children’s Research Foundation, Cincinnati, OH 45229; eDepartment of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229; fDivision of Developmental Biology, Perinatal Institute, Cincinnati Children’s Research Foundation, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229; gDepartment of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229; hDepartment of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; iDepartment of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; jIntegrative Program for Biological and Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; kThe University of North Carolina McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; lDepartment of Pediatrics, University of Minnesota, Minneapolis, MN 55455; mLillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455; nNational Xenopus Resource, Marine Biological Laboratory, Woods Hole, MA 02543; oEugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543; pDepartment of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637; qGladstone Institute of Cardiovascular Disease, San Francisco, CA 94158; rRoddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158; and sAmsterdam Cardiovascular Sciences, Department of Medical Biology, Amsterdam University Medical Center, University of Amsterdam, 1105AZ Amsterdam, The Netherlands Edited by Roeland Nusse, Stanford University School of Medicine, Stanford, CA, and approved September 27, 2018 (received for review July 9, 2018) Codevelopment of the lungs and heart underlies key evolutionary these structures comprise common forms of human congenital innovations in the transition to terrestrial life. Cardiac specializations heart disease. that support pulmonary circulation, including the atrial septum, are Recent work has highlighted the common developmental or- generated by second heart field (SHF) cardiopulmonary progenitors igin of multiple mesodermal derivatives in both the heart and the (CPPs). It has been presumed that transcription factors required in lung (5, 6). This lateral plate mesodermal population has been the SHF for cardiac septation, e.g., Tbx5,directlydriveacardiacmor- termed the “second heart field” (SHF) or “cardiopulmonary phogenesis gene-regulatory network. Here, we report instead that progenitors” (CPPs). This population originates dorsal to the TBX5 directly drives Wnt ligands to initiate a bidirectional signaling cardiac inflow tract and ventral to the anterior foregut and loop between cardiopulmonary mesoderm and the foregut endo- generates multiple structures in the heart, e.g., the atrial septum, derm for endodermal pulmonary specification and, subsequently, and in the lungs, e.g., smooth muscle and vascular endothelium atrial septation. We show that Tbx5 is required for pulmonary spec- (5, 6). This essential CPP region is labeled by expression of the ification in mice and amphibians but not for swim bladder develop- canonical Wnt signaling ligand Wnt2, the Hedgehog (Hh) ment in zebrafish. TBX5 is non–cell-autonomously required for Wnt2 pulmonary endoderm specification by directly driving and Significance Wnt2b expression in cardiopulmonary mesoderm. TBX5 ChIP- sequencing identified cis-regulatory elements at Wnt2 sufficient for In the 20 years since the discovery of the genetic link between endogenous Wnt2 expression domains in vivo and required for Wnt2 the transcription factor TBX5 and congenital heart defects, few expression in precardiac mesoderm in vitro. Tbx5 cooperated with direct targets of TBX5 in cardiac morphogenesis have been ShhsignalingtodriveWnt2b expression for lung morphogenesis. identified. In this work, we demonstrate that TBX5 directly Tbx5 haploinsufficiency in mice, a model of Holt–Oram syndrome, regulates canonical Wnt ligands required for initiation of lung caused a quantitative decrement of mesodermal-to-endodermal Wnt development. Lung endoderm forms a Hedgehog signaling signaling and subsequent endodermal-to-mesodermal Shh signaling source required for morphogenesis of both the lungs and the required for cardiac morphogenesis. Thus, Tbx5 initiates a mesoderm– cardiac inflow septum. Our work expands the role of TBX5 to endoderm–mesoderm signaling loop in lunged vertebrates that include a non–cell-autonomous component for atrial septation. provides a molecular basis for the coevolution of pulmonary and We find the mesoderm–endoderm–mesoderm signaling loop cardiac structures required for terrestrial life. initiated by TBX5 is evolutionarily conserved from amphibians to mammals. This work suggests that the evolutionary origin lung development | heart development | TBX5 | Wnt signaling | of lungs may have involved the recruitment of cardiac TBX5. Hedgehog signaling Author contributions: J.D.S., S.A.R., C.E.S., E.A.T.B.A., M.E.H., R.K.H., B.J., A.M.Z., F.L.C., tilization of atmospheric oxygen revolutionized the ability of and I.P.M. designed research; J.D.S., S.A.R., C.E.S., J.B., A.B.R., J.K., K.I., R.D.N., M.R., Uvertebrates to adapt to terrestrial life (1). At the center of A.D.H., S.L., W.T., E.A.T.B.A., L.L.-Z., and B.J. performed research; S.S.-K.C. and M.K. con- tributed new reagents/analytic tools; J.D.S., S.A.R., C.E.S., and X.H.Y. analyzed data; and this revolution are the lungs, a foregut-derived gas-exchange J.D.S., S.A.R., A.M.Z., F.L.C., and I.P.M. wrote the paper. BIOLOGY structure (1, 2). The derived cardiovascular system, utilizing The authors declare no conflict of interest. pulmonary oxygen, must manage blood from both the body and DEVELOPMENTAL This article is a PNAS Direct Submission. the lungs simultaneously (2). While most lungfish, amphibians, Published under the PNAS license. and reptiles exhibit a three-chambered heart with an atrial Data deposition: RNA-sequencing, ChIP-sequencing, and ATAC-sequencing data have septum separating pulmonary and systemic circulation enter- been deposited in the Gene Expression Omnibus databank (accession nos. GSE75077 ing the heart (3), the two-sided, four-chambered crocodilian, and GSE119885). avian, and mammalian hearts have independently evolved to 1To whom correspondence should be addressed. Email: [email protected]. completely separate pulmonary from systemic circulation (4). This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. The proper development and placement of the cardiac septa 1073/pnas.1811624115/-/DCSupplemental. are critical for the efficient handling of blood, and defects in Published online October 23, 2018. www.pnas.org/cgi/doi/10.1073/pnas.1811624115 PNAS | vol. 115 | no. 45 | E10615–E10624 Downloaded by guest on September 28, 2021 signaling-responsive transcription factor Gli1, and the T-box A Transcriptional Profiling of Microdissected CPP-containing Region family transcription factor Tbx5 (5, 7–9). Mate Tbx5+/- Mice Mutations in TBX5 have been implicated as the primary genetic Tbx5 cause of Holt–Oram syndrome (HOS), a human syndrome that Dissect E9.5 Embryos includes cardiac septal defects (10–14). Previous work has demon- Heart strated that Tbx5 is required in the posterior SHF (pSHF) for atrial Pool Tissue from 4 Embryos septation (7, 9, 15). In addition, Sonic hedgehog (Shh) signaling has each Tbx5+/+ and Tbx5-/- been implicated in cardiac septation (7–9, 16). Shh, expressed in the pulmonary endoderm (PE), activates GLI-mediated transcription in CPP RNA-Sequencing BCE9.5 CPP Tbx5+/+ and Tbx5-/- RNA-seq the CPPs (7, 8). Shh and Tbx5 genetically interact for cardiac sep- Spearman’s ρ tation, and constitutive activation of Hh signaling in CPPs rescues 50 atrial septal defects caused by reduced Tbx5 dose (7, 9). Further- 0.95 1.00 Tbx5+/+ Tbx5-/- Nkx2-1 more, TBX5 and GLI transcription factors directly cooperate at Shh 0.95 0.95 0.95 0.97 0.96 0.99 1 Wnt2 enhancers for genes required for cardiac septation (7, 9). This has 0.97 0.96 0.96 0.98 0.98 1 0.99 NANCI generated a model in which TBX5 and GLI transcription fac- 5 Tbx4 0.99 0.99 0.98 0.99 1 0.98 0.96 Wnt2b tors directly activate gene expression in the CPPs of the pSHF for Fzd10 −log10 FDR Gli1 cardiac morphogenesis. 0.99 0.99 0.98 1 0.99 0.98 0.97 Wnt7b 1 CPPs are an important source of signals that induce the pul- 0.99 0.99 1 0.98 0.98 0.96 0.95 monary lineage in the ventral foregut endoderm and contribute – 0.99 1 0.99 0.99 0.99 0.96 0.95 directly to the atrial septum and cardiopulmonary vasculature (17 −5 −2 −1 0 1 2 5 19). An evolutionarily conserved paracrine signaling cascade in- 1 0.99 0.99 0.99 0.99 0.97 0.95 log2 fold change volving retinoic acid, Hh, Wnt signaling, and bone morphogenic D qRT-PCR Validation of Early Pulmonary Development Genes 1.5 protein (BMP) regulates the induction of pulmonary progenitors * p<0.05 NS from amphibians to mammals (5, 17–21).
Recommended publications
  • Genetic Variants in WNT2B and BTRC Predict Melanoma Survival
    ACCEPTED MANUSCRIPT Genetic Variants in WNT2B and BTRC Predict Melanoma Survival Qiong Shi1, 2, 3, 9, Hongliang Liu2, 3, 9, Peng Han2, 3, 4, 9, Chunying Li1, Yanru Wang2, 3, Wenting Wu5, Dakai Zhu6, Christopher I. Amos6, Shenying Fang7, Jeffrey E. Lee7, Jiali Han5, 8* and Qingyi Wei2, 3* 1Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710032, China; 2Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA, 3Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA, 4Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, China; 5Department of Epidemiology, Fairbanks School of Public Health, Indiana University Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis,MANUSCRIPT IN 46202, USA 6Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA; 7Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA. 8Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA 9These authors contributed equally to this work. ACCEPTED *Correspondence: Qingyi Wei, M.D., Ph.D., Duke Cancer Institute, Duke University Medical Center and Department of Medicine, Duke School of Medicine, 905 S LaSalle Street, Durham, NC 27710, USA, Tel.: (919) 660-0562, E-mail: [email protected] and Jiali Han, M.D., Ph.D., 1 _________________________________________________________________________________ This is the author's manuscript of the article published in final edited form as: Shi, Q., Liu, H., Han, P., Li, C., Wang, Y., Wu, W., … Wei, Q.
    [Show full text]
  • Wnt Signaling in Vertebrate Neural Development and Function
    J Neuroimmune Pharmacol (2012) 7:774–787 DOI 10.1007/s11481-012-9404-x INVITED REVIEW Wnt Signaling in Vertebrate Neural Development and Function Kimberly A. Mulligan & Benjamin N. R. Cheyette Received: 18 June 2012 /Accepted: 10 September 2012 /Published online: 27 September 2012 # Springer Science+Business Media, LLC 2012 Abstract Members of the Wnt family of secreted signaling immature neurons migrate to populate different cortical proteins influence many aspects of neural development and layers, nuclei, and ganglia in the forebrain, midbrain, hind- function. Wnts are required from neural induction and axis brain, and spinal cord. Each mature neuron elaborates an formation to axon guidance and synapse development, and axon and numerous dendrites while forming hundreds to even help modulate synapse activity. Wnt proteins activate a thousands of synapses with other neurons, enabling electro- variety of downstream signaling pathways and can induce a chemical communication throughout the nervous system. similar variety of cellular responses, including gene tran- All these developmental processes must be coordinated to scription changes and cytoskeletal rearrangements. This re- ensure proper construction and function of the CNS, and view provides an introduction to Wnt signaling pathways this requires the coordinated developmental activity of a and discusses current research on their roles in vertebrate vast array of genes. neural development and function. Members of the Wnt family of secreted signaling proteins are implicated in every step of neural development men- Keywords Wnt signaling . Neural development . tioned above. Wnt proteins provide positional information CNS patterning . Axon guidance . Dendrite growth . within the embryo for anterior-posterior axis specification of Synapse formation .
    [Show full text]
  • The Wnt Signaling Pathway in Tumorigenesis, Pharmacological
    Wang et al. Biomarker Research (2021) 9:68 https://doi.org/10.1186/s40364-021-00323-7 REVIEW Open Access The Wnt signaling pathway in tumorigenesis, pharmacological targets, and drug development for cancer therapy Zhuo Wang1,2†, Tingting Zhao1,2†, Shihui Zhang3, Junkai Wang1, Yunyun Chen1,2, Hongzhou Zhao1,2, Yaxin Yang4, Songlin Shi2, Qiang Chen5 and Kuancan Liu1,2* Abstract Wnt signaling was initially recognized to be vital for tissue development and homeostasis maintenance. Further studies revealed that this pathway is also important for tumorigenesis and progression. Abnormal expression of signaling components through gene mutation or epigenetic regulation is closely associated with tumor progression and poor prognosis in several tissues. Additionally, Wnt signaling also influences the tumor microenvironment and immune response. Some strategies and drugs have been proposed to target this pathway, such as blocking receptors/ligands, targeting intracellular molecules, beta-catenin/TCF4 complex and its downstream target genes, or tumor microenvironment and immune response. Here we discuss the roles of these components in Wnt signaling pathway in tumorigenesis and cancer progression, the underlying mechanisms that is responsible for the activation of Wnt signaling, and a series of drugs targeting the Wnt pathway provide multiple therapeutic values. Although some of these drugs exhibit exciting anti-cancer effect, clinical trials and systematic evaluation should be strictly performed along with multiple-omics technology. Keywords: Wnt signaling, beta-catenin, Epigenetic modification, Tumor microenvironment, Drug development Background polyposis coli (APC), glycogen synthase kinase-3β (GSK- The Wnt signaling cascade is critical for tissue morpho- 3β), Axin, casein kinase 1(CK1). Degradation of beta- genesis, homeostasis, and regeneration.
    [Show full text]
  • Contribution of Enhancer-Driven and Master-Regulator Genes to Autoimmune Disease Revealed Using Functionally Informed SNP-To-Gene Linking Strategies Kushal K
    bioRxiv preprint doi: https://doi.org/10.1101/2020.09.02.279059; this version posted March 31, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Contribution of enhancer-driven and master-regulator genes to autoimmune disease revealed using functionally informed SNP-to-gene linking strategies Kushal K. Dey1, Steven Gazal1, Bryce van de Geijn 1,3, Samuel Sungil Kim 1,4, Joseph Nasser5, Jesse M. Engreitz5, Alkes L. Price 1,2,5 1 Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA 2 Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 3 Genentech, South San Francisco, CA 4 Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 5 Broad Institute of MIT and Harvard, Cambridge, MA, USA Abstract Gene regulation is known to play a fundamental role in human disease, but mechanisms of regulation vary greatly across genes. Here, we explore the con- tributions to disease of two types of genes: genes whose regulation is driven by enhancer regions as opposed to promoter regions (Enhancer-driven) and genes that regulate many other genes in trans (Master-regulator). We link these genes to SNPs using a comprehensive set of SNP-to-gene (S2G) strategies and apply stratified LD score regression to the resulting SNP annotations to draw three main conclusions about 11 autoimmune diseases and blood cell traits (average Ncase=13K across 6 autoimmune diseases, average N=443K across 5 blood cell traits).
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • Accompanies CD8 T Cell Effector Function Global DNA Methylation
    Global DNA Methylation Remodeling Accompanies CD8 T Cell Effector Function Christopher D. Scharer, Benjamin G. Barwick, Benjamin A. Youngblood, Rafi Ahmed and Jeremy M. Boss This information is current as of October 1, 2021. J Immunol 2013; 191:3419-3429; Prepublished online 16 August 2013; doi: 10.4049/jimmunol.1301395 http://www.jimmunol.org/content/191/6/3419 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2013/08/20/jimmunol.130139 Material 5.DC1 References This article cites 81 articles, 25 of which you can access for free at: http://www.jimmunol.org/content/191/6/3419.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists by guest on October 1, 2021 • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2013 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Global DNA Methylation Remodeling Accompanies CD8 T Cell Effector Function Christopher D. Scharer,* Benjamin G. Barwick,* Benjamin A. Youngblood,*,† Rafi Ahmed,*,† and Jeremy M.
    [Show full text]
  • Negative Regulators of Colonic Peripheral Regulatory T Cell Development Teresa Lei Ai Washington University in St
    Washington University in St. Louis Washington University Open Scholarship Arts & Sciences Electronic Theses and Dissertations Arts & Sciences Spring 5-15-2018 Negative Regulators of Colonic Peripheral Regulatory T Cell Development Teresa Lei Ai Washington University in St. Louis Follow this and additional works at: https://openscholarship.wustl.edu/art_sci_etds Part of the Allergy and Immunology Commons, Immunology and Infectious Disease Commons, and the Medical Immunology Commons Recommended Citation Ai, Teresa Lei, "Negative Regulators of Colonic Peripheral Regulatory T Cell Development" (2018). Arts & Sciences Electronic Theses and Dissertations. 1509. https://openscholarship.wustl.edu/art_sci_etds/1509 This Dissertation is brought to you for free and open access by the Arts & Sciences at Washington University Open Scholarship. It has been accepted for inclusion in Arts & Sciences Electronic Theses and Dissertations by an authorized administrator of Washington University Open Scholarship. For more information, please contact [email protected]. WASHINGTON UNIVERSITY IN ST. LOUIS Division of Biology and Biomedical Sciences Immunology Dissertation Examination Committee: Chyi-Song Hsieh, Chair Paul M. Allen Kenneth M. Murphy Gwendalyn J. Randolph Wayne M. Yokoyama Negative Regulators of Colonic Peripheral Regulatory T Cell Development By Teresa Lei Ai A dissertation presented to The Graduate School of Washington University in partial fulfillment of the requirements for the degree of Doctor of Philosophy May 2018 St. Louis, Missouri
    [Show full text]
  • The Role of Tbx5 in Sinoatrial Node Differentiation in Mouse Embryonic Stem Cell Derived Cardiomyocytes Yunkai Dai Clemson University, [email protected]
    Clemson University TigerPrints All Dissertations Dissertations May 2019 The Role of Tbx5 in Sinoatrial Node Differentiation in Mouse Embryonic Stem Cell Derived Cardiomyocytes Yunkai Dai Clemson University, [email protected] Follow this and additional works at: https://tigerprints.clemson.edu/all_dissertations Recommended Citation Dai, Yunkai, "The Role of Tbx5 in Sinoatrial Node Differentiation in Mouse Embryonic Stem Cell Derived Cardiomyocytes" (2019). All Dissertations. 2377. https://tigerprints.clemson.edu/all_dissertations/2377 This Dissertation is brought to you for free and open access by the Dissertations at TigerPrints. It has been accepted for inclusion in All Dissertations by an authorized administrator of TigerPrints. For more information, please contact [email protected]. THE ROLE OF TBX5 IN SINOATRIAL NODE DIFFERENTIATION IN MOUSE EMBRYONIC STEM CELL DERIVED CARDIOMYOCYTES A Dissertation Presented to the Graduate School of Clemson University In Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy Bioengineering by Yunkai Dai May 2019 Accepted by: Dr Ann Foley, Committee Chair Dr Agneta Simionescu Dr Robin C. Muise-Helmericks Dr Ying Mei ABSTRACT The sinoatrial node of the mouse embryo arises from the wall of the right atrium near the border of the sinus venosus. Early in development this region expresses the transcription factor Tbx5. Because of this, Tbx5 is thought to sit at the apex of a transcriptional cascade leading to sinoatrial node (SAN) differentiation. To test this we produced a mouse embryonic stem cell line B1 (pTripZ-mTbx5; αMHC::GFP) that conditionally overexpresses Tbx5, to determine if this would lead to enhanced SAN differentiation. We found that ES cells overexpressing Tbx5 showed enhanced overall cardiac differentiation and that cardiac cells showed increased beat rates as compared control embryos.
    [Show full text]
  • REVIEW ARTICLE the Genetics of Autism
    REVIEW ARTICLE The Genetics of Autism Rebecca Muhle, BA*; Stephanie V. Trentacoste, BA*; and Isabelle Rapin, MD‡ ABSTRACT. Autism is a complex, behaviorally de- tribution of a few well characterized X-linked disorders, fined, static disorder of the immature brain that is of male-to-male transmission in a number of families rules great concern to the practicing pediatrician because of an out X-linkage as the prevailing mode of inheritance. The astonishing 556% reported increase in pediatric preva- recurrence rate in siblings of affected children is ϳ2% to lence between 1991 and 1997, to a prevalence higher than 8%, much higher than the prevalence rate in the general that of spina bifida, cancer, or Down syndrome. This population but much lower than in single-gene diseases. jump is probably attributable to heightened awareness Twin studies reported 60% concordance for classic au- and changing diagnostic criteria rather than to new en- tism in monozygotic (MZ) twins versus 0 in dizygotic vironmental influences. Autism is not a disease but a (DZ) twins, the higher MZ concordance attesting to ge- syndrome with multiple nongenetic and genetic causes. netic inheritance as the predominant causative agent. By autism (the autistic spectrum disorders [ASDs]), we Reevaluation for a broader autistic phenotype that in- mean the wide spectrum of developmental disorders cluded communication and social disorders increased characterized by impairments in 3 behavioral domains: 1) concordance remarkably from 60% to 92% in MZ twins social interaction; 2) language, communication, and and from 0% to 10% in DZ pairs. This suggests that imaginative play; and 3) range of interests and activities.
    [Show full text]
  • Tsukushi Functions As a Wnt Signaling Inhibitor by Competing with Wnt2b for Binding to Transmembrane Protein Frizzled4
    Tsukushi functions as a Wnt signaling inhibitor by competing with Wnt2b for binding to transmembrane protein Frizzled4 Kunimasa Ohtaa,b,1,2, Ayako Itoa,c,1, Sei Kuriyamaa,d,3, Giuseppe Lupoe,f, Mitsuko Kosakag,4, Shin-ichi Ohnumah, Shinichi Nakagawai, and Hideaki Tanakaa,c,d aDepartment of Developmental Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan; bPrecursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan; cGlobal Center of Excellence, Kumamoto University, Kumamoto 860-8556, Japan; d21st Century Center of Excellence, Kumamoto University, Kumamoto 860-8556, Japan; eDepartment of Biology and Biotechnology “C. Darwin,” University of Rome “La Sapienza,” 00185 Rome, Italy; fIstituto Pasteur–Fondazione Cenci Bolognetti, 00185, Rome, Italy; gRIKEN Center for Developmental Biology, Kobe 650-0047, Japan; hInstitute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom; and IRIKEN Advanced Science Institute, Nakagawa RNA Biology Laboratory, Saitama 351-0198, Japan Edited* by Lynn T. Landmesser, Case Western Reserve University, Cleveland, OH, and approved July 8, 2011 (received for review January 11, 2011) The Wnt signaling pathway is essential for the development of We previously described the isolation of Tsukushi (TSK) protein diverse tissues during embryogenesis. Signal transduction is acti- isoforms (13), soluble molecules belonging to the small leucine- vated by the binding of Wnt proteins to the type I receptor low- rich proteoglycan (SLRP) family (14), and showed that they work density lipoprotein receptor–related protein 5/6 and the seven-pass as extracellular modulators of pivotal signaling cascades during transmembrane protein Frizzled (Fzd), which contains a Wnt- early embryonic development in chicks and frogs (13, 15–17).
    [Show full text]
  • Deregulated Wnt/Β-Catenin Program in High-Risk Neuroblastomas Without
    Oncogene (2008) 27, 1478–1488 & 2008 Nature Publishing Group All rights reserved 0950-9232/08 $30.00 www.nature.com/onc ONCOGENOMICS Deregulated Wnt/b-catenin program in high-risk neuroblastomas without MYCN amplification X Liu1, P Mazanek1, V Dam1, Q Wang1, H Zhao2, R Guo2, J Jagannathan1, A Cnaan2, JM Maris1,3 and MD Hogarty1,3 1Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA; 2Department of Biostatistics and Epidemiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA and 3Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA Neuroblastoma (NB) is a frequently lethal tumor of Introduction childhood. MYCN amplification accounts for the aggres- sive phenotype in a subset while the majority have no Neuroblastoma (NB) is a childhood embryonal malig- consistently identified molecular aberration but frequently nancy arising in the peripheral sympathetic nervous express MYC at high levels. We hypothesized that acti- system. Half of all children with NB present with features vated Wnt/b-catenin (CTNNB1) signaling might account that define their tumorsashigh riskwith poor overall for this as MYC is a b-catenin transcriptional target and survival despite intensive therapy (Matthay et al., 1999). multiple embryonal and neural crest malignancies have A subset of these tumors are characterized by high-level oncogenic alterations in this pathway. NB cell lines without genomic amplification of the MYCN proto-oncogene MYCN amplification express higher levels of MYC and (Matthay et al., 1999) but the remainder have no b-catenin (with aberrant nuclear localization) than MYCN- consistently identified aberration to account for their amplified cell lines.
    [Show full text]
  • In Situ Detection of Tbx5 Expression in Developing Chick Embryonic Heart
    San Jose State University SJSU ScholarWorks Master's Theses Master's Theses and Graduate Research Fall 2010 In Situ Detection Of Tbx5 Expression In Developing Chick Embryonic Heart Vaishali Agarwal San Jose State University Follow this and additional works at: https://scholarworks.sjsu.edu/etd_theses Recommended Citation Agarwal, Vaishali, "In Situ Detection Of Tbx5 Expression In Developing Chick Embryonic Heart" (2010). Master's Theses. 3901. DOI: https://doi.org/10.31979/etd.84mn-c4vy https://scholarworks.sjsu.edu/etd_theses/3901 This Thesis is brought to you for free and open access by the Master's Theses and Graduate Research at SJSU ScholarWorks. It has been accepted for inclusion in Master's Theses by an authorized administrator of SJSU ScholarWorks. For more information, please contact [email protected]. IN SITU DETECTION OF Tbx 5 EXPRESSION IN DEVELOPING EMBRYONIC CHICK HEART A Thesis Presented to The Faculty of the Department of Biological Sciences San Jose State University In Partial Fulfillment of the Requirements for the Degree Master of Science by Vaishali Agarwal December 2010 © 2010 Vaishali Agarwal ALL RIGHTS RESERVED The Designated Thesis Committee Approves the Thesis Titled IN SITU DETECTION OF Tbx 5 EXPRESSION IN DEVELOPING EMBRYONIC CHICK HEART by Vaishali Agarwal APPROVED FOR THE DEPARTMENT OF BIOLOGICAL SCIENCES SAN JOSE STATE UNIVERSITY December 2010 Dr. Steven White Department of Biological Sciences Dr. Michael Sneary Department of Biological Sciences Dr. Bob Fowler Department of Biological Sciences ABSTRACT IN SITU DETECTION OF Tbx 5 EXPRESSION IN DEVELOPING EMBRYONIC CHICK HEART by Vaishali Agarwal The Tbx 5 gene codes for a highly conserved transcription factor containing a DNA-binding motif called the T box (or T-domain).
    [Show full text]