1
|
Preusse K, Cochran K, Dai Q, Kopan R. Notch dimerization provides robustness against environmental insults and is required for vascular integrity. PLoS One 2025; 20:e0311353. [PMID: 39854367 DOI: 10.1371/journal.pone.0311353] [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: 07/09/2024] [Accepted: 09/18/2024] [Indexed: 01/26/2025] Open
Abstract
The Notch intracellular domain (NICD) regulates gene expression during development and homeostasis in a transcription factor complex that binds DNA either as monomer, or cooperatively as dimers. Mice expressing Notch dimerization-deficient (NDD) alleles of Notch1 and Notch2 have defects in multiple tissues that are sensitized to environmental insults. Here, we report that cardiac phenotypes and DSS (Dextran Sodium Sulfate) sensitivity in NDD mice can be ameliorated by housing mice under hypo-allergenic conditions (food/bedding). However, compound heterozygote NDD mice (N1RA/-; N2RA/-) in hypo-allergenic conditions subsequently develop severe hydrocephalus and hemorrhages. Further analysis revealed multiple vascular phenotypes in NDD mice including leakage, malformations of brain vasculature, and vasodilation in kidneys, leading to demise around P21. This mouse model is thus a hypomorphic allele useful to analyze vascular phenotypes and gene-environment interactions. The possibility of a non-canonical Notch signal regulating barrier formation in the gut, skin, and blood systems is discussed.
Collapse
Affiliation(s)
- Kristina Preusse
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Kim Cochran
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Quanhui Dai
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Raphael Kopan
- Division of Developmental Biology, Department of Pediatrics, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| |
Collapse
|
2
|
Favarolo MB, López SL. Notch signaling in the division of germ layers in bilaterian embryos. Mech Dev 2018; 154:122-144. [PMID: 29940277 DOI: 10.1016/j.mod.2018.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/08/2018] [Accepted: 06/18/2018] [Indexed: 01/09/2023]
Abstract
Bilaterian embryos are triploblastic organisms which develop three complete germ layers (ectoderm, mesoderm, and endoderm). While the ectoderm develops mainly from the animal hemisphere, there is diversity in the location from where the endoderm and the mesoderm arise in relation to the animal-vegetal axis, ranging from endoderm being specified between the ectoderm and mesoderm in echinoderms, and the mesoderm being specified between the ectoderm and the endoderm in vertebrates. A common feature is that part of the mesoderm segregates from an ancient bipotential endomesodermal domain. The process of segregation is noisy during the initial steps but it is gradually refined. In this review, we discuss the role of the Notch pathway in the establishment and refinement of boundaries between germ layers in bilaterians, with special focus on its interaction with the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- María Belén Favarolo
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN), Facultad de Medicina, Laboratorio de Embriología Molecular "Prof. Dr. Andrés E. Carrasco", Buenos Aires, Argentina
| | - Silvia L López
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN), Facultad de Medicina, Laboratorio de Embriología Molecular "Prof. Dr. Andrés E. Carrasco", Buenos Aires, Argentina.
| |
Collapse
|
3
|
Herman AM, Rhyner AM, Devine WP, Marrelli SP, Bruneau BG, Wythe JD. A novel reporter allele for monitoring Dll4 expression within the embryonic and adult mouse. Biol Open 2018; 7:bio026799. [PMID: 29437553 PMCID: PMC5898260 DOI: 10.1242/bio.026799] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 01/29/2018] [Indexed: 12/23/2022] Open
Abstract
Canonical Notch signaling requires the presence of a membrane bound ligand and a corresponding transmembrane Notch receptor. Receptor engagement induces multiple proteolytic cleavage events culminating in the nuclear accumulation of the Notch intracellular domain and its binding to a transcriptional co-factor to mediate gene expression. Notch signaling networks are essential regulators of vascular patterning and angiogenesis, as well as myriad other biological processes. Delta-like 4 (Dll4) encodes the earliest Notch ligand detected in arterial cells, and is enriched in sprouting endothelial tip cells. Dll4 expression has often been inferred by proxy using a lacZ knockin reporter allele. This is problematic, as a single copy of Dll4 is haploinsufficient. Additionally, Notch activity regulates Dll4 transcription, making it unclear whether these reporter lines accurately reflect Dll4 expression. Accordingly, precisely defining Dll4 expression is essential for determining its role in development and disease. To address these limitations, we generated a novel BAC transgenic allele with a nuclear-localized β-galactosidase reporter (Dll4-BAC-nlacZ). Through a comparative analysis, we show the BAC line overcomes previous issues of haploinsufficiency, it recapitulates Dll4 expression in vivo, and allows superior visualization and imaging. As such, this novel Dll4 reporter is an important addition to the growing Notch toolkit.
Collapse
Affiliation(s)
- Alexander M Herman
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander M Rhyner
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - W Patrick Devine
- Department of Pathology, University of California San Francisco, San Francisco, CA 94113, USA
- Gladstone Institute of Cardiovascular Disease, University of California San Francisco, San Francisco, CA 94110, USA
| | - Sean P Marrelli
- Department of Neurology, McGovern Medical School at UT Health, Houston, TX 77005, USA
| | - Benoit G Bruneau
- Gladstone Institute of Cardiovascular Disease, University of California San Francisco, San Francisco, CA 94110, USA
| | - Joshua D Wythe
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
4
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
5
|
Li Y, Tzatzalos E, Kwan KY, Grumet M, Cai L. Transcriptional Regulation of Notch1 Expression by Nkx6.1 in Neural Stem/Progenitor Cells during Ventral Spinal Cord Development. Sci Rep 2016; 6:38665. [PMID: 27924849 PMCID: PMC5141430 DOI: 10.1038/srep38665] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 11/14/2016] [Indexed: 12/22/2022] Open
Abstract
Notch1 signaling plays a critical role in maintaining and determining neural stem/progenitor cell (NSPC) fate, yet the transcriptional mechanism controlling Notch1 specific expression in NSPCs remains incomplete. Here, we show transcription factor Nkx6.1 interacts with a cis-element (CR2, an evolutionarily conserved non-coding fragment in the second intron of Notch1 locus) and regulates the expression of Notch1 in ventral NSPCs of the developing spinal cord. We show that the Notch1 expression is modulated by the interaction of Nkx6.1 with a 139 bp enhancer sequence within CR2. Knockdown or overexpression of Nkx6.1 leads to down- or up-regulated Notch1 expression, respectively. In CR2-GFP transgenic mouse, GFP expression was found prominent in the ventricular zone and neural progenitor cells from embryonic day 9.5 to postnatal day 7. GFP+ cells were mainly neural progenitors for interneurons and not for motoneurons or glial cells. Moreover, GFP expression persisted in a subset of ependymal cells in the adult spinal cord, suggesting that CR2 is active in both embryonic and adult NSPCs. Together our data reveal a novel mechanism of Notch1 transcriptional regulation in the ventral spinal cord by Nkx6.1 via its binding with Notch1 enhancer CR2 during embryonic development.
Collapse
Affiliation(s)
- Ying Li
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Evangeline Tzatzalos
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Kelvin Y Kwan
- W.M. Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Martin Grumet
- W.M. Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Li Cai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| |
Collapse
|
6
|
Martin-Lannerée S, Halliez S, Hirsch TZ, Hernandez-Rapp J, Passet B, Tomkiewicz C, Villa-Diaz A, Torres JM, Launay JM, Béringue V, Vilotte JL, Mouillet-Richard S. The Cellular Prion Protein Controls Notch Signaling in Neural Stem/Progenitor Cells. Stem Cells 2016; 35:754-765. [PMID: 27641601 DOI: 10.1002/stem.2501] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 08/29/2016] [Accepted: 08/31/2016] [Indexed: 12/26/2022]
Abstract
The prion protein is infamous for its involvement in a group of neurodegenerative diseases known as Transmissible Spongiform Encephalopathies. In the longstanding quest to decipher the physiological function of its cellular isoform, PrPC , the discovery of its participation to the self-renewal of hematopoietic and neural stem cells has cast a new spotlight on its potential role in stem cell biology. However, still little is known on the cellular and molecular mechanisms at play. Here, by combining in vitro and in vivo murine models of PrPC depletion, we establish that PrPC deficiency severely affects the Notch pathway, which plays a major role in neural stem cell maintenance. We document that the absence of PrPC in a neuroepithelial cell line or in primary neurospheres is associated with drastically reduced expression of Notch ligands and receptors, resulting in decreased levels of Notch target genes. Similar alterations of the Notch pathway are recovered in the neuroepithelium of Prnp-/- embryos during a developmental window encompassing neural tube closure. In addition, in line with Notch defects, our data show that the absence of PrPC results in altered expression of Nestin and Olig2 as well as N-cadherin distribution. We further provide evidence that PrPC controls the expression of the epidermal growth factor receptor (EGFR) downstream from Notch. Finally, we unveil a negative feedback action of EGFR on both Notch and PrPC . As a whole, our study delineates a molecular scenario through which PrPC takes part to the self-renewal of neural stem and progenitor cells. Stem Cells 2017;35:754-765.
Collapse
Affiliation(s)
- Séverine Martin-Lannerée
- INSERM UMR 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Sophie Halliez
- VIM, UR 892, INRA, Université Paris-Saclay, Jouy-en-Josas, France
| | - Théo Z Hirsch
- INSERM UMR 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Julia Hernandez-Rapp
- INSERM UMR 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Bruno Passet
- Department of Pharma Research, INRA UMR 1313, Génétique animale et biologie intégrative, Jouy-en-Josas, France
| | - Céline Tomkiewicz
- INSERM UMR 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| | - Ana Villa-Diaz
- Centro de Investigación en Sanidad Animal-INIA, U 942 Madrid, Spain
| | | | - Jean-Marie Launay
- AP-HP Service de Biochimie, Fondation FondaMental, INSERM U942 Hôpital Lariboisière, Paris, France.,Pharma Research Department, F. Hoffmann-La-Roche Ltd, Basel, Switzerland
| | - Vincent Béringue
- VIM, UR 892, INRA, Université Paris-Saclay, Jouy-en-Josas, France
| | - Jean-Luc Vilotte
- Department of Pharma Research, INRA UMR 1313, Génétique animale et biologie intégrative, Jouy-en-Josas, France
| | - Sophie Mouillet-Richard
- INSERM UMR 1124, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, UMR 1124, Paris, France
| |
Collapse
|
7
|
Ota T, Takekoshi S, Takagi T, Kitatani K, Toriumi K, Kojima T, Kato M, Ikoma N, Mabuchi T, Ozawa A. Notch signaling may be involved in the abnormal differentiation of epidermal keratinocytes in psoriasis. Acta Histochem Cytochem 2014; 47:175-83. [PMID: 25392571 PMCID: PMC4164705 DOI: 10.1267/ahc.14027] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 06/24/2014] [Indexed: 12/20/2022] Open
Abstract
Localization of each keratin isoform differs among epidermal layers. Proliferating basal cells synthesize keratin 14 (K14) and suprabasal cells express keratin 10 (K10) in normal skin. Notch signaling is essential for keratinocyte differentiation. Notch1 is expressed in all epidermal layers, Notch2 in the basal cell layer and Notch3 in basal cell and spinous cell layers in normal epidermis. It has been poorly elucidated how localization and expression levels of Notch molecules are related to epidermal molecular markers K10 and K14 in psoriatic skin with abnormal differentiation of epidermal tissue. This study aimed to investigate the relationship between abnormal differentiation of epidermal cells in psoriatic skin and expression of Notch molecules. We investigated keratins (K14 and K10) and Notches (1, 2, 3 and 4) using immunohistochemistry in psoriatic skin (n=30) and normal skin (n=10). In normal skin, K14 and K10 were discretely observed in the basal cell layer and suprabasal layer, respectively. In psoriatic skin, K14 was expressed in the pan epidermal layer while it and K10 were co-expressed in some middle suprabasal layer cells. Notch1, 2, 3, and 4 localized in all epidermal layers in normal skin. In psoriatic skin, Notch1, 2, and 4 mainly localized in suprabasilar layers and Notch3 is lacalized in pan epidermal, suprabasilar, and basilar layers. Protein and mRNA of Notch1, 2, and 3 isoforms decreased in psoriatic epidermis compared with normal epidermis. These data suggest that decrements in these Notch molecules might cause aberrant expression of K10 and K14 leading to anomalous differentiation of the epidermis in psoriatic lesions.
Collapse
Affiliation(s)
- Tami Ota
- Department of Dermatology, Tokai University School of Medicine
| | - Susumu Takekoshi
- Department of Cell Biology, Division of Host Defense Mechanism, Tokai University School of Medicine
| | - Tatsuya Takagi
- Department of Cell Biology, Division of Host Defense Mechanism, Tokai University School of Medicine
| | - Kanae Kitatani
- Department of Cell Biology, Division of Host Defense Mechanism, Tokai University School of Medicine
| | - Kentaro Toriumi
- Department of Cell Biology, Division of Host Defense Mechanism, Tokai University School of Medicine
| | - Tomoko Kojima
- Department of Dermatology, Tokai University School of Medicine
| | - Masayuki Kato
- Department of Dermatology, Tokai University School of Medicine
| | - Norihiro Ikoma
- Department of Dermatology, Tokai University School of Medicine
| | | | - Akira Ozawa
- Department of Dermatology, Tokai University School of Medicine
| |
Collapse
|
8
|
James AC, Szot JO, Iyer K, Major JA, Pursglove SE, Chapman G, Dunwoodie SL. Notch4 reveals a novel mechanism regulating Notch signal transduction. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1272-84. [PMID: 24667410 DOI: 10.1016/j.bbamcr.2014.03.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/13/2014] [Accepted: 03/17/2014] [Indexed: 01/08/2023]
Abstract
Notch4 is a divergent member of the Notch family of receptors that is primarily expressed in the vasculature. Its expression implies an important role for Notch4 in the vasculature; however, mice homozygous for the Notch4(d1) knockout allele are viable. Since little is known about the role of Notch4 in the vasculature and how it functions, we further investigated Notch4 in mice and in cultured cells. We found that the Notch4(d1) allele is not null as it expresses a truncated transcript encoding most of the NOTCH4 extracellular domain. In cultured cells, NOTCH4 did not signal in response to ligand. Moreover, NOTCH4 inhibited signalling from the NOTCH1 receptor. This is the first report of cis-inhibition of signalling by another Notch receptor. The NOTCH4 extracellular domain also inhibits NOTCH1 signalling when expressed in cis, raising the possibility that reported Notch4 phenotypes may not be due to loss of NOTCH4 function. To better address the role of NOTCH4 in vivo, we generated a Notch4 null mouse in which the entire coding region was deleted. Notch4 null mice exhibited slightly delayed vessel growth in the retina, consistent with our novel finding that NOTCH4 protein is expressed in the newly formed vasculature. These findings indicate a role of NOTCH4 in fine-tuning the forming vascular plexus.
Collapse
Affiliation(s)
- A C James
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia.
| | - J O Szot
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, UNSW, Sydney, Australia.
| | - K Iyer
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia.
| | - J A Major
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia.
| | - S E Pursglove
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia.
| | - G Chapman
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, Australia.
| | - S L Dunwoodie
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, UNSW, Sydney, Australia.
| |
Collapse
|
9
|
Kotasová H, Procházková J, Pacherník J. Interaction of Notch and gp130 signaling in the maintenance of neural stem and progenitor cells. Cell Mol Neurobiol 2014; 34:1-15. [PMID: 24132391 PMCID: PMC11488917 DOI: 10.1007/s10571-013-9996-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/30/2013] [Indexed: 01/10/2023]
Abstract
Notch and gp130 signaling are involved in the regulation of multiple cellular processes across various tissues during animal ontogenesis. In the developing nervous system, both signaling pathways intervene at many stages to determine cell fate-from the first neural lineage commitment and generation of neuronal precursors, to the terminal specification of cells as neurons and glia. In most cases, the effects of Notch and gp130 signaling in these processes are similar. The aim of the current review was to summarize the knowledge regarding the roles of Notch and gp130 signaling in the maintenance of neural stem and progenitor cells during animal ontogenesis, from early embryo to adult. Recent data show a direct crosstalk between these signaling pathways that seems to be specific for a particular type of neural progenitors.
Collapse
Affiliation(s)
- Hana Kotasová
- Faculty of Science, Institute of Experimental Biology, Masaryk University, Brno, Czech Republic
| | - Jiřina Procházková
- Faculty of Science, Institute of Experimental Biology, Masaryk University, Brno, Czech Republic
| | - Jiří Pacherník
- Faculty of Science, Institute of Experimental Biology, Masaryk University, Brno, Czech Republic
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
10
|
Smolarkiewicz M, Skrzypczak T, Wojtaszek P. The very many faces of presenilins and the γ-secretase complex. PROTOPLASMA 2013; 250:997-1011. [PMID: 23504135 PMCID: PMC3788181 DOI: 10.1007/s00709-013-0494-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 03/01/2013] [Indexed: 05/02/2023]
Abstract
Presenilin is a central, catalytic component of the γ-secretase complex which conducts intramembrane cleavage of various protein substrates. Although identified and mainly studied through its role in the development of amyloid plaques in Alzheimer disease, γ-secretase has many other important functions. The complex seems to be evolutionary conserved throughout the Metazoa, but recent findings in plants and Dictyostelium discoideum as well as in archeons suggest that its evolution and functions might be much more diversified than previously expected. In this review, a selective survey of the multitude of functions of presenilins and the γ-secretase complex is presented. Following a brief overview of γ-secretase structure, assembly and maturation, three functional aspects are analyzed: (1) the role of γ-secretase in autophagy and phagocytosis; (2) involvement of the complex in signaling related to endocytosis; and (3) control of calcium fluxes by presenilins.
Collapse
Affiliation(s)
- Michalina Smolarkiewicz
- Department of Molecular and Cellular Biology, Faculty of Biology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznań, Poland
| | - Tomasz Skrzypczak
- Department of Molecular and Cellular Biology, Faculty of Biology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznań, Poland
| | - Przemysław Wojtaszek
- Department of Molecular and Cellular Biology, Faculty of Biology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznań, Poland
| |
Collapse
|
11
|
Samsa LA, Yang B, Liu J. Embryonic cardiac chamber maturation: Trabeculation, conduction, and cardiomyocyte proliferation. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2013; 163C:157-68. [PMID: 23720419 PMCID: PMC3723796 DOI: 10.1002/ajmg.c.31366] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Congenital heart diseases are some of the most common human birth defects. Though some congenital heart defects can be surgically corrected, treatment options for other congenital heart diseases are very limited. In many congenital heart diseases, genetic defects lead to impaired embryonic heart development or growth. One of the key development processes in cardiac development is chamber maturation, and alterations in this maturation process can manifest as a variety of congenital defects including non-compaction, systolic dysfunction, diastolic dysfunction, and arrhythmia. During development, to meet the increasing metabolic demands of the developing embryo, the myocardial wall undergoes extensive remodeling characterized by the formation of muscular luminal protrusions called cardiac trabeculae, increased cardiomyocyte mass, and development of the ventricular conduction system. Though the basic morphological and cytological changes involved in early heart development are clear, much remains unknown about the complex biomolecular mechanisms governing chamber maturation. In this review, we highlight evidence suggesting that a wide variety of basic signaling pathways and biomechanical forces are involved in cardiac wall maturation.
Collapse
Affiliation(s)
- Leigh Ann Samsa
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Betsy Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Liu N, Zhang J, Ji C. The emerging roles of Notch signaling in leukemia and stem cells. Biomark Res 2013; 1:23. [PMID: 24252593 PMCID: PMC4177577 DOI: 10.1186/2050-7771-1-23] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 07/15/2013] [Indexed: 12/16/2022] Open
Abstract
The Notch signaling pathway plays a critical role in maintaining the balance between cell proliferation, differentiation and apoptosis, and is a highly conserved signaling pathway that regulates normal development in a context- and dose-dependent manner. Dysregulation of Notch signaling has been suggested to be key events in a variety of hematological malignancies. Notch1 signaling appears to be the central oncogenic trigger in T cell acute lymphoblastic leukemia (T-ALL), in which the majority of human malignancies have acquired mutations that lead to constitutive activation of Notch1 signaling. However, emerging evidence unexpectedly demonstrates that Notch signaling can function as a potent tumor suppressor in other forms of leukemia. This minireview will summarize recent advances related to the roles of activated Notch signaling in human lymphocytic leukemia, myeloid leukemia, stem cells and stromal microenvironment, and we will discuss the perspectives of Notch signaling as a potential therapeutic target as well.
Collapse
Affiliation(s)
- Na Liu
- Department of Hematology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Jinan, Shandong 250012, P, R, China.
| | | | | |
Collapse
|
13
|
Tang Y, Bai H, Urs S, Wang Z, Liaw L. Notch1 activation in embryonic VE-cadherin populations selectively blocks hematopoietic stem cell generation and fetal liver hematopoiesis. Transgenic Res 2012; 22:403-10. [PMID: 22851140 DOI: 10.1007/s11248-012-9637-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 07/19/2012] [Indexed: 10/28/2022]
Abstract
Hematopoietic stem cells (HSC) are found in several independent sites embryonically. Loss-of-function studies indicated that Notch1, but not Notch2 signaling was required for HSC emergence from the aortic-gonado-mesonephros (AGM) region. We previously showed that constitutive Notch1 activation impaired primitive erythroid differentiation, but its effects on HSC emergence from the AGM region were not studied. To further define specific roles of Notch receptors, we characterized HSC in mouse embryos expressing either Notch1 intracellular domain (ICD) or Notch4ICD in VE-cadherin or SM22α expressing populations. Although embryonic Notch1 activation in VE-cadherin populations led to lethality after E13.5, earlier defects in the fetal liver were observed. Embryos were analyzed at E12.5 to assess hematopoiesis and the phenotype of developing cells in the AGM region. We found that activation of Notch1 in the endothelial compartment in VE-cadherin expressing cells resulted in the absence of intra-aortic clusters and defects in fetal liver hematopoiesis. In contrast, although Notch4 expression is regulated during fetal hematopoiesis, activation of Notch4 in VE-cadherin expressing populations did not affect HSC phenotype, although later vascular remodeling was impaired. Likewise, activation of Notch1 in SM22α positive populations had no significant effect on hematopoiesis. Our results indicate a cell type-dependent activity and distinct features of Notch1 versus Notch4 signaling and their impact on HSC generation.
Collapse
Affiliation(s)
- Yuefeng Tang
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA
| | | | | | | | | |
Collapse
|
14
|
Granados-Riveron JT, Brook JD. The impact of mechanical forces in heart morphogenesis. ACTA ACUST UNITED AC 2012; 5:132-42. [PMID: 22337926 DOI: 10.1161/circgenetics.111.961086] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Javier T Granados-Riveron
- Institute of Genetics, School of Biology, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom.
| | | |
Collapse
|
15
|
Abstract
Notch is a well-conserved signaling pathway and its function in cell fate determination is crucial in embryonic development and in the maintenance of tissue homeostasis during adult life. Notch activation depends on cell-cell interactions that are essential for the generation of cell diversity from initially equivalent cell populations. In the adult hematopoiesis, Notch is undoubtedly a very efficient promoter of T-cell differentiation, and this has masked for a long time the effects of Notch on other blood lineages, which are gradually being identified. However, the adult hematopoietic stem cell (HSC) remains mostly refractory to Notch intervention in experimental systems. In contrast, Notch is essential for the generation of the HSCs, which takes place during embryonic development. This review summarizes the knowledge accumulated in recent years regarding the role of the Notch pathway in the different stages of HSC ontology from embryonic life to fetal and adult bone marrow stem cells. In addition, we briefly examine other systems where Notch regulates specific stem cell capacities, in an attempt to understand how Notch functions in stem cell biology.
Collapse
|
16
|
Shah DK, Zúñiga-Pflücker JC. Notch receptor-ligand interactions during T cell development, a ligand endocytosis-driven mechanism. Curr Top Microbiol Immunol 2012; 360:19-46. [PMID: 22581027 DOI: 10.1007/82_2012_225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Notch signaling plays an important role during the development of different cell types and tissues. The role of Notch signaling in lymphocyte development, in particular in the development and commitment to the T cell lineage, has been the focus of research for many years. Notch signaling is absolutely required during the commitment and early stages of T cell development. Activation of the Notch signaling pathway is initiated by ligand-receptor interactions and appears to require active endocytosis of Notch ligands. Studies addressing the mechanism underlying endocytosis of Notch ligands have helped to identify the main players important and necessary for this process. Here, we review the Notch ligands, and the proposed models of Notch activation by Notch ligand endocytosis, highlighting key molecules involved. In particular, we discuss recent studies on Notch ligands involved in T cell development, current studies aimed at elucidating the relevance of Notch ligand endocytosis during T cell development and the identification of key players necessary for ligand endocytosis in the thymus and during T cell development.
Collapse
Affiliation(s)
- Divya K Shah
- Department of Immunology, Sunnybrook Research Institute, University of Toronto, 2075 Bayview Avenue, Toronto, ON, M4 N 3M5, Canada.
| | | |
Collapse
|
17
|
Aubin-Houzelstein G. Notch signaling and the developing hair follicle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 727:142-60. [PMID: 22399345 DOI: 10.1007/978-1-4614-0899-4_11] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Notch function in the hair follicle has been mainly studied by use of transgenic mice carrying either loss or gain of function mutations in various members of the pathway. These studies revealed that whereas embryonic development of the hair follicle can be achieved without Notch, its postnatal development requires an intact Notch signaling in the hair bulb and the outer root sheath. Among the many roles played by Notch in the hair follicle, two can be highlighted: in the bulge, Notch controls a cell fate switch in hair follicle stem cells or their progenitors, preventing them from adopting an epidermal fate. In the hair bulb, Notch controls cell differentiation, ensuring the proper development of every layer of the hair shaft and inner root sheath. Notch function in the hair follicle is both cell autonomous and cell non autonomous and involves intercellular communication between adjacent layers.
Collapse
|
18
|
Beres BJ, George R, Lougher EJ, Barton M, Verrelli BC, McGlade CJ, Rawls JA, Wilson-Rawls J. Numb regulates Notch1, but not Notch3, during myogenesis. Mech Dev 2011; 128:247-57. [DOI: 10.1016/j.mod.2011.02.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 02/18/2011] [Accepted: 02/22/2011] [Indexed: 12/18/2022]
|
19
|
Li X, von Boehmer H. Notch Signaling in T-Cell Development and T-ALL. ISRN HEMATOLOGY 2011; 2011:921706. [PMID: 22111016 PMCID: PMC3200084 DOI: 10.5402/2011/921706] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 12/15/2010] [Indexed: 11/23/2022]
Abstract
The Notch signaling pathway is an evolutionarily conserved cell signaling system present in most multicellular organisms, as it controls cell fate specification by regulating cell proliferation, differentiation, apoptosis, and survival. Regulation of the Notch signaling pathway can be achieved at multiple levels. Notch proteins are involved in lineage fate decisions in a variety of tissues in various species. Notch is essential for T lineage cell differentiation including T versus B and αβ versus γδ lineage specification. In this paper, we discuss Notch signaling in normal T-cell maturation and differentiation as well as in T-cell acute lymphoblastic lymphoma/leukemia.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | - Harald von Boehmer
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| |
Collapse
|
20
|
Le Bras A, Samson C, Trentini M, Caetano B, Lelievre E, Mattot V, Beermann F, Soncin F. VE-statin/egfl7 expression in endothelial cells is regulated by a distal enhancer and a proximal promoter under the direct control of Erg and GATA-2. PLoS One 2010; 5:e12156. [PMID: 20808444 PMCID: PMC2922337 DOI: 10.1371/journal.pone.0012156] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 07/20/2010] [Indexed: 11/24/2022] Open
Abstract
Angiogenesis is the process by which new blood vessels arise from existing ones by the budding out of endothelial cell capillaries from the luminal side of blood vessels. Blood vessel formation is essential for organ development during embryogenesis and is associated with several physiological and pathological processes, such as wound healing and tumor development. The VE-statin/egfl7 gene is specifically expressed in endothelial cells during embryonic development and in the adult. We studied here the regulatory mechanisms that control this tissue-specific expression. RT-qPCR analyses showed that the specificity of expression of VE-statin/egfl7 in endothelial cells is not shared with its closest neighbor genes notch1 and agpat2 on the mouse chromosome 2. Chromatin-immunoprecipitation analysis of histone modifications at the VE-statin/egfl7 locus showed that the chromatin is specifically opened in endothelial cells, but not in fibroblasts at the transcription start sites. A 13 kb genomic fragment of promoter was cloned and analyzed by gene reporter assays which showed that two conserved regions are important for the specific expression of VE-statin/egfl7 in endothelial cells; a −8409/−7563 enhancer and the −252/+38 region encompassing the exon-1b transcription start site. The latter contains essential GATA and ETS-binding sites, as assessed by linker-scanning analysis and site-directed mutagenesis. An analysis of expression of the ETS and GATA transcription factors showed that Erg, Fli-1 and GATA-2 are the most highly expressed factors in endothelial cells. Erg and GATA-2 directly control the expression of the endogenous VE-statin/egfl7 while Fli-1 probably exerts an indirect control, as assessed by RNA interference and chromatin immunoprecipitation. This first detailed analysis of the mechanisms that govern the expression of the VE-statin/egfl7 gene in endothelial cells pinpoints the specific importance of ETS and GATA factors in the specific regulation of genes in this cell lineage.
Collapse
Affiliation(s)
- Alexandra Le Bras
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Chantal Samson
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Matteo Trentini
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Bertrand Caetano
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Etienne Lelievre
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Virginie Mattot
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
| | - Friedrich Beermann
- Swiss Institute for Experimental Cancer Research (ISREC), Centre de Phénotypage Génomique (CPG), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Fabrice Soncin
- CNRS, Institut de Biologie de Lille, UMR 8161, Equipe labellisée La Ligue, Lille, France
- Université Lille-Nord de France, Lille, France
- Institut Pasteur de Lille, F-59019 Lille, France
- * E-mail:
| |
Collapse
|
21
|
Liu H, Zhang W, Kennard S, Caldwell RB, Lilly B. Notch3 is critical for proper angiogenesis and mural cell investment. Circ Res 2010; 107:860-70. [PMID: 20689064 DOI: 10.1161/circresaha.110.218271] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE The heterotypic interactions of endothelial cells and mural cells (smooth muscle cells or pericytes) are crucial for assembly, maturation, and subsequent function of blood vessels. Yet, the molecular mechanisms underlying their association have not been fully defined. OBJECTIVE Our previous in vitro studies indicated that Notch3, which is expressed in mural cells, mediates these cell-cell interactions. To assess the significance of Notch3 on blood vessel formation in vivo, we investigated its role in retinal angiogenesis. METHODS AND RESULTS We show that Notch3-deficient mice exhibit reduced retinal vascularization, with diminished sprouting and vascular branching. Moreover, Notch3 deletion impairs mural cell investment, resulting in progressive loss of vessel coverage. In an oxygen-induced retinopathy model, we demonstrate that Notch3 is induced in hypoxia and interestingly, pathological neovascularization is decreased in retinas of Notch3-null mice. Analysis of oxygen-induced retinopathy mediators revealed that angiopoietin-2 expression is significantly reduced in the absence of Notch3. Furthermore, in vitro experiments showed that Notch3 is sufficient for angiopoietin-2 induction, and this expression is additionally enhanced in the presence of hypoxia-inducible factor 1α. CONCLUSIONS These results provide compelling evidence that Notch3 is important for the investment of mural cells and is a critical regulator of developmental and pathological blood vessel formation.
Collapse
Affiliation(s)
- Hua Liu
- National Children's Research Institute, The Ohio State University, Columbus, OH 43205, USA
| | | | | | | | | |
Collapse
|
22
|
Hedgehog regulates distinct vascular patterning events through VEGF-dependent and -independent mechanisms. Blood 2010; 116:653-60. [PMID: 20339091 DOI: 10.1182/blood-2009-12-256644] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Despite the clear importance of Hedgehog (Hh) signaling in blood vascular development as shown by genetic analysis, its mechanism of action is still uncertain. To better understand the role of Hh in vascular development, we further characterized its roles in vascular development in mouse embryos and examined its interaction with vascular endothelial growth factor (VEGF), a well-known signaling pathway essential to blood vascular development. We found that VEGF expression in the mouse embryo depended on Hh signaling, and by using genetic rescue approaches, we demonstrated that the role of Hh both in endothelial tube formation and Notch-dependent arterial identity was solely dependent on its regulation of VEGF. In contrast, overactivation of the Hh pathway through deletion of Patched1 (Ptch1), a negative regulator of Hh signaling, resulted in reduced vascular density and increased Delta-like ligand 4 expression. The Ptch1 phenotype was independent of VEGF pathway dysregulation and was not rescued when Delta-like ligand 4 levels were restored to normal. These findings establish that Hh uses both VEGF- and Notch-dependent and -independent mechanisms to pattern specific events in early blood vascular development.
Collapse
|
23
|
Masumura T, Yamamoto K, Shimizu N, Obi S, Ando J. Shear stress increases expression of the arterial endothelial marker ephrinB2 in murine ES cells via the VEGF-Notch signaling pathways. Arterioscler Thromb Vasc Biol 2009; 29:2125-31. [PMID: 19797707 DOI: 10.1161/atvbaha.109.193185] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Arterial-venous specification in the embryo has been assumed to depend on the influence of fluid mechanical forces, but its cellular and molecular mechanisms are still poorly understood. Our previous in vitro study revealed that fluid shear stress induces endothelial cell (EC) differentiation by murine embryonic stem (ES) cells. In the present study we investigated whether shear stress regulates the arterial-venous specification of ES-cell-derived ECs. METHODS AND RESULTS When murine ES cell-derived VEGFR2(+) cells were exposed to shear stress, expression of the arterial EC marker protein ephrinB2 increased dose-dependently. The ephrinB2 mRNA levels also increased in response to shear stress, whereas the mRNA levels of the venous EC marker EphB4 decreased. Notch cleavage and translocation of the Notch intracellular domain (NICD) into the nucleus occurred as early as 30 minutes after the start of shear stress and increased with time. Gamma-Secretase inhibitors (DAPT and L685 458) and the recombinant extracellular domain of the Notch ligand DLL4 abolished the shear stress-induced NICD translocation, and that, in turn, blocked the shear stress-induced upregulation of ephrinB2 expression. In addition, the VEGF receptor kinase inhibitor SU1498 was found to suppress both the shear-stress-induced Notch cleavage and up-regulation of ephrinB2 expression. CONCLUSIONS Exposure to shear stress induces an increase in expression of ephrinB2 in murine ES cells via VEGF-Notch signaling pathways.
Collapse
Affiliation(s)
- Tomomi Masumura
- Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Japan
| | | | | | | | | |
Collapse
|
24
|
Miazga CM, McLaughlin KA. Coordinating the timing of cardiac precursor development during gastrulation: A new role for Notch signaling. Dev Biol 2009; 333:285-96. [DOI: 10.1016/j.ydbio.2009.06.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 06/16/2009] [Accepted: 06/27/2009] [Indexed: 10/20/2022]
|
25
|
Abstract
The Notch pathway is an ancient, highly conserved signaling mechanism that participates in essential cell-cell communication events between adjacent cells. Mutations in Notch-signaling elements cause cardiac abnormalities in mice and humans, demonstrating an essential role for Notch in heart development. Studies with targeted mutant mice indicate that Notch signaling promotes the epithelial-to-mesenchyme transition that gives rise to the cardiac valve primordium, which later is sculpted into mature valves. During ventricular chamber development, the myocardium differentiates into two layers: an outer compact zone and an inner trabecular zone. Trabeculae provide a pumping function during early phases of ventricular development and contribute to the cardiac conduction system in the mature heart. Notch regulates the endocardium-to-myocardium signals that balance proliferation and differentiation of trabecular myocytes. Recent evidence demonstrates that defective NOTCH signaling leads to aortic valve degeneration in humans. Future research will be informative about the involvement of altered NOTCH signaling in chamber abnormalities and other cardiac disorders.
Collapse
|
26
|
Abstract
The major arteries and veins of the vertebrate circulatory system are formed early in embryonic development, before the onset of circulation, following de novo aggregation of "angioblast" progenitors in a process called vasculogenesis. Initial embryonic determination of artery or vein identity is regulated by variety of genetic factors that work in concert to specify endothelial cell fate, giving rise to 2 distinct components of the circulatory loop possessing unique structural characteristics. Work in multiple in vivo animal model systems has led to a detailed examination of the interacting partners that determine arterial and venous specification. We discuss the hierarchical arrangement of many signaling molecules, including Hedgehog (Hh), vascular endothelial growth factor (VEGF), Notch, and chicken ovalbumin upstream-transcription factor II (COUP-TFII) that promote or inhibit divergent pathways of endothelial cell fate. Elucidation of the functional role of these genetic determinants of blood vessel specification together with the epigenetic factors involved in subsequent modification of arterial-venous identity will allow for potential new therapeutic targets for vascular disorders.
Collapse
Affiliation(s)
- Matthew R Swift
- Laboratory of Molecular Genetics, NICHD, NIH, Bethesda, MD 20892, USA.
| | | |
Collapse
|
27
|
Formation and Differentiation of Avian Somite Derivatives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 638:1-41. [DOI: 10.1007/978-0-387-09606-3_1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
28
|
Venkatesh DA, Park KS, Harrington A, Miceli-Libby L, Yoon JK, Liaw L. Cardiovascular and hematopoietic defects associated with Notch1 activation in embryonic Tie2-expressing populations. Circ Res 2008; 103:423-31. [PMID: 18617694 PMCID: PMC2654335 DOI: 10.1161/circresaha.108.177808] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Notch signaling is critical for the development and maintenance of the cardiovasculature, with loss-of-function studies defining roles of Notch1 in the endothelial/hematopoietic lineages. No in vivo studies have addressed complementary gain-of-function strategies within these tissues to define consequences of Notch activation. We developed a transgenic model of Cre recombinase-mediated activation of a constitutively active mouse Notch1 allele (N1ICD(+)) and studied transgene activation in Tie2-expressing lineages. The in vivo phenotype was compared to effects of Notch1 activation on endothelial tubulogenesis, paracrine regulation of smooth muscle cell proliferation, and hematopoiesis. N1ICD(+) embryos showed midgestation lethality with defects in angiogenic remodeling of embryonic and yolk sac vasculature, cardiac development, smooth muscle cell investment of vessels, and hematopoietic differentiation. Angiogenic defects corresponded with impaired endothelial tubulogenesis in vitro following Notch1 activation and paracrine inhibition of smooth muscle cells when grown with Notch1-activated endothelial cells. Flow cytometric analysis of hematopoietic and endothelial precursor populations demonstrated a significant loss of CD71(+)/Ter119(+) populations with an active N1ICD(+) allele and a corresponding increase in c-Kit(+)/CD71 and Flk1(+) populations, suggesting a developmental block during the transition between c-Kit- and Ter119-expressing erythroblasts. Cardiovascular lineages are sensitive to an imbalance in Notch signaling, with aberrant activation reflecting a vascular phenotype comparable to a loss-of-function Notch1 mutation.
Collapse
MESH Headings
- Animals
- Cardiovascular System/embryology
- Cardiovascular System/metabolism
- Cells, Cultured
- Embryo, Mammalian/blood supply
- Embryo, Mammalian/metabolism
- Embryonic Development/genetics
- Endothelium, Vascular/cytology
- Endothelium, Vascular/embryology
- Endothelium, Vascular/metabolism
- Gene Expression Regulation, Developmental
- Hematopoietic System/embryology
- Hematopoietic System/metabolism
- Mice
- Mice, Transgenic
- Models, Animal
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/embryology
- Muscle, Smooth, Vascular/metabolism
- Mutation
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Receptor, TIE-2/genetics
- Receptor, TIE-2/metabolism
- Signal Transduction/physiology
- Yolk Sac/blood supply
- Yolk Sac/metabolism
Collapse
Affiliation(s)
- Deepak A Venkatesh
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME 04074, USA
| | | | | | | | | | | |
Collapse
|
29
|
Chae J, Yu K, Cho S, Kim J, Koo D, Lee K, Han Y. Aberrant expression of developmentally important signaling molecules in cloned porcine extraembryonic tissues. Proteomics 2008; 8:2724-34. [DOI: 10.1002/pmic.200701134] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
30
|
Gordon-Thomson C, Botto SA, Cam GR, Moore GPM. Notch pathway gene expression and wool follicle cell fates. ACTA ACUST UNITED AC 2008. [DOI: 10.1071/ea07315] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The Notch family of genes has been implicated in specifying cell fates during hair follicle morphogenesis. We examined Notch gene expression during wool follicle formation, as an understanding of genes that influence cell distributions in the developing follicle is a prerequisite for devising molecular strategies to manipulate fibre characters and follicle density. We identified transcripts for the Notch1 receptor and one of its ligands, Jagged1, in fetal sheep skin by reverse transcriptase polymerase chain reaction. The sheep-specific cDNA sequences were used as templates to produce probes to investigate the expression patterns of Notch1 and Jagged1 in developing ovine fetal skin by in situ hybridisation. Notch1 and Jagged1 were detected in the epidermis and in a subpopulation of mesenchymal cells before follicle initiation. At day 70 during follicle initiation, transcripts were also detected in cells at the tip of the epidermal plug and in dermal condensates. By day 86, Notch1 and Jagged1 were detected in the distal cells of the epidermal downgrowths and epidermis and Notch1 was no longer detected in the mesenchyme and dermal condensates. After day 96, transcripts were absent from the epidermis, but localised to differentiating outer root sheath (ORS) cells. The distributions of transcripts implicate a Notch1–Jagged1 signal pathway in the fates of prospective ORS cells. The transient appearance of Notch1 in cells at the epidermal–mesenchymal junction during early follicle morphogenesis suggests that the receptor may be responsible for the specification of a cell subpopulation committed to a prepapilla fate at initiation.
Collapse
|
31
|
Del Monte G, Grego-Bessa J, González-Rajal A, Bolós V, De La Pompa JL. Monitoring Notch1 activity in development: evidence for a feedback regulatory loop. Dev Dyn 2007; 236:2594-614. [PMID: 17685488 DOI: 10.1002/dvdy.21246] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Signaling through Notch receptors, which regulate cell fate decisions and embryonic patterning, requires ligand-induced receptor cleavage to generate the signaling active Notch intracellular domain (NICD). Here, we show an analysis at specific developmental stages of the distribution of active mouse Notch1. We use an antibody that recognizes N1ICD, and a highly sensitive staining technique. The earliest N1ICD expression was observed in the mesoderm and developing heart, where we detected expression in nascent endocardium, presumptive cardiac valves, and ventricular and atrial endocardium. During segmentation, N1ICD was restricted to the presomitic mesoderm. N1ICD expression was also evident in arterial endothelium, and in kidney and endodermal derivatives such as pancreas and thymus. Ectodermal N1ICD expression was found in central nervous system and sensory placodes. We found that Notch1 transcription and activity was severely reduced in zebrafish and mouse Notch pathway mutants, suggesting that vertebrate Notch1 expression is regulated by a positive feedback loop.
Collapse
Affiliation(s)
- Gonzalo Del Monte
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología/CSIC, Campus Cantoblanco, Madrid, Spain
| | | | | | | | | |
Collapse
|
32
|
Compartmentalised expression of Delta-like 1 in epithelial somites is required for the formation of intervertebral joints. BMC DEVELOPMENTAL BIOLOGY 2007; 7:68. [PMID: 17572911 PMCID: PMC1924847 DOI: 10.1186/1471-213x-7-68] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Accepted: 06/17/2007] [Indexed: 01/23/2023]
Abstract
Background Expression of the mouse Delta-like 1 (Dll1) gene in the presomitic mesoderm and in the caudal halves of somites of the developing embryo is required for the formation of epithelial somites and for the maintenance of caudal somite identity, respectively. The rostro-caudal polarity of somites is initiated early on within the presomitic mesoderm in nascent somites. Here we have investigated the requirement of restricted Dll1 expression in caudal somite compartments for the maintenance of rostro-caudal somite polarity and the morphogenesis of the axial skeleton. We did this by overexpressing a functional copy of the Dll1 gene throughout the paraxial mesoderm, in particular in anterior somite compartments, during somitogenesis in transgenic mice. Results Epithelial somites were generated normally and appeared histologically normal in embryos of two independent Dll1 over-expressing transgenic lines. Gene expression analyses of rostro-caudal marker genes suggested that over-expression of Dll1 without restriction to caudal compartments was not sufficient to confer caudal identity to rostral somite halves in transgenic embryos. Nevertheless, Dll1 over-expression caused dysmorphologies of the axial skeleton, in particular, in morphological structures that derive from the articular joint forming compartment of vertebrae. Accordingly, transgenic animals exhibited missing or reduced intervertebral discs, rostral and caudal articular processes as well as costal heads of ribs. In addition, the midline of the vertebral column did not develop normally. Transgenic mice had open neural arches and split vertebral bodies with ectopic pseudo-growth plates. Endochondral bone formation and ossification in the developing vertebrae were delayed. Conclusion The mice overexpressing Dll1 exhibit skeletal dysmorphologies that are also evident in several mutant mice with defects in somite compartmentalisation. The Dll1 transgenic mice demonstrate that vertebral dysmorphologies such as bony fusions of vertebrae and midline vertebral defects can occur without apparent changes in somitic rostro-caudal marker gene expression. Also, we demonstrate that the over-expression of the Dll1 gene in rostral epithelial somites is not sufficient to confer caudal identity to rostral compartments. Our data suggest that the restricted Dll1 expression in caudal epithelial somites may be particularly required for the proper development of the intervertebral joint forming compartment.
Collapse
|
33
|
Abstract
Notch is an evolutionarily conserved local cell signaling mechanism that participates in a variety of cellular processes: cell fate specification, differentiation, proliferation, apoptosis, adhesion, epithelial-mesenchymal transition, migration, and angiogenesis. These processes can be subverted in Notch-mediated pathological situations. In the first part of this review, we will discuss the role of Notch in vertebrate central nervous system development, somitogenesis, cardiovascular and endocrine development, with attention to the mechanisms by which Notch regulates cell fate specification and patterning in these tissues. In the second part, we will review the molecular aspects of Notch-mediated neoplasias, where Notch can act as an oncogene or as a tumor suppressor. From all these studies, it becomes evident that the outcome of Notch signaling is strictly context-dependent and differences in the strength, timing, cell type, and context of the signal may affect the final outcome. It is essential to understand how Notch integrates inputs from other signaling pathways and how specificity is achieved, because this knowledge may be relevant for future therapeutic applications.
Collapse
Affiliation(s)
- Victoria Bolós
- Departmento de Inmunología y Oncología, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Científicas, Darwin 3, Campus de Cantoblanco, E-28049 Madrid, Spain
| | | | | |
Collapse
|
34
|
Grego-Bessa J, Luna-Zurita L, Monte GD, Bolós V, Melgar P, Arandilla A, Garratt AN, Zang H, Mukouyama YS, Chen H, Shou W, Ballestar E, Esteller M, Rojas A, Pérez-Pomares JM, de la Pompa JL. Notch signaling is essential for ventricular chamber development. Dev Cell 2007; 12:415-29. [PMID: 17336907 PMCID: PMC2746361 DOI: 10.1016/j.devcel.2006.12.011] [Citation(s) in RCA: 393] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2006] [Revised: 11/26/2006] [Accepted: 12/24/2006] [Indexed: 12/19/2022]
Abstract
Ventricular chamber morphogenesis, first manifested by trabeculae formation, is crucial for cardiac function and embryonic viability and depends on cellular interactions between the endocardium and myocardium. We show that ventricular Notch1 activity is highest at presumptive trabecular endocardium. RBPJk and Notch1 mutants show impaired trabeculation and marker expression, attenuated EphrinB2, NRG1, and BMP10 expression and signaling, and decreased myocardial proliferation. Functional and molecular analyses show that Notch inhibition prevents EphrinB2 expression, and that EphrinB2 is a direct Notch target acting upstream of NRG1 in the ventricles. However, BMP10 levels are found to be independent of both EphrinB2 and NRG1 during trabeculation. Accordingly, exogenous BMP10 rescues the myocardial proliferative defect of in vitro-cultured RBPJk mutants, while exogenous NRG1 rescues differentiation in parallel. We suggest that during trabeculation Notch independently regulates cardiomyocyte proliferation and differentiation, two exquisitely balanced processes whose perturbation may result in congenital heart disease.
Collapse
Affiliation(s)
- Joaquín Grego-Bessa
- Dpto. de Inmunología y Oncología, Centro Nacional de Biotecnología/CSIC, Darwin 3, Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Luis Luna-Zurita
- Dpto. de Inmunología y Oncología, Centro Nacional de Biotecnología/CSIC, Darwin 3, Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Gonzalo del Monte
- Dpto. de Inmunología y Oncología, Centro Nacional de Biotecnología/CSIC, Darwin 3, Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Victoria Bolós
- Dpto. de Inmunología y Oncología, Centro Nacional de Biotecnología/CSIC, Darwin 3, Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Pedro Melgar
- Dpto. de Inmunología y Oncología, Centro Nacional de Biotecnología/CSIC, Darwin 3, Campus de Cantoblanco, E-28049 Madrid, Spain
| | - Alejandro Arandilla
- Dpto. de Inmunología y Oncología, Centro Nacional de Biotecnología/CSIC, Darwin 3, Campus de Cantoblanco, E-28049 Madrid, Spain
| | | | - Heesuk Zang
- Laboratory of Developmental Biology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, USA
| | - Yoh-suke Mukouyama
- Laboratory of Developmental Biology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, USA
| | - Hanying Chen
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, USA
| | - Weinian Shou
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, USA
| | - Esteban Ballestar
- Programa de Patología Molecular, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Manel Esteller
- Programa de Patología Molecular, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Ana Rojas
- Structural Bioinformatics Group, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | | | - José Luis de la Pompa
- Dpto. de Inmunología y Oncología, Centro Nacional de Biotecnología/CSIC, Darwin 3, Campus de Cantoblanco, E-28049 Madrid, Spain
| |
Collapse
|
35
|
Tanaka M, Kokubo M, Marunouchi T. Asymmetric localization of Notch2 on the microvillous surface in choroid plexus epithelial cells. Histochem Cell Biol 2007; 127:449-56. [PMID: 17219215 DOI: 10.1007/s00418-006-0260-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2006] [Indexed: 10/23/2022]
Abstract
Notch family molecules are transmembrane receptors that play various roles in contact-dependent cell-cell interactions in a wide range of organs. In the brain, Notch2, but not the other members of Notch, is expressed in the choroid plexus at an exceptionally high level. We immunohistochemically examined the cellular and subcellular localization of Notch2 protein in the choroid plexus using confocal and electron microscopy. Unexpectedly, Notch2 was asymmetrically localized on the microvillous surface of epithelial cells in the choroid plexus of both postnatal and adult rats. This localization pattern of Notch2 suggests its novel and unknown role independent of contact with adjacent cells in the choroid plexus. In organotypic cultures of the choroid plexus, the addition of anti-Notch2 antibody resulted in deformation of microvilli in epithelial cells, which suggests a role of Notch2 in the maintenance of the microvillous structure in choroid plexus epithelial cells.
Collapse
Affiliation(s)
- Masahiko Tanaka
- Division of Cell Biology, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi, 470-1192, Japan.
| | | | | |
Collapse
|
36
|
Abstract
Somites are segments of paraxial mesoderm that give rise to a multitude of tissues in the vertebrate embryo. Many decades of intensive research have provided a wealth of data on the complex molecular interactions leading to the formation of various somitic derivatives. In this review, we focus on the crucial role of the somites in building the body wall and limbs of amniote embryos. We give an overview on the current knowledge on the specification and differentiation of somitic cell lineages leading to the development of the vertebral column, skeletal muscle, connective tissue, meninges, and vessel endothelium, and highlight the importance of the somites in establishing the metameric pattern of the vertebrate body.
Collapse
Affiliation(s)
- Bodo Christ
- Institute of Anatomy und Cell Biology, Department of Molecular Embryology, University of Freiburg, Albertstr. 17, 79104 Freiburg, Germany.
| | | | | |
Collapse
|
37
|
Chau MDL, Tuft R, Fogarty K, Bao ZZ. Notch signaling plays a key role in cardiac cell differentiation. Mech Dev 2006; 123:626-40. [PMID: 16843648 PMCID: PMC1567976 DOI: 10.1016/j.mod.2006.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Revised: 05/22/2006] [Accepted: 06/07/2006] [Indexed: 11/17/2022]
Abstract
Results from lineage tracing studies indicate that precursor cells in the ventricles give rise to both cardiac muscle and conduction cells. Cardiac conduction cells are specialized cells responsible for orchestrating the rhythmic contractions of the heart. Here, we show that Notch signaling plays an important role in the differentiation of cardiac muscle and conduction cell lineages in the ventricles. Notch1 expression coincides with a conduction marker, HNK-1, at early stages. Misexpression of constitutively active Notch1 (NIC) in early heart tubes in chick exhibited multiple effects on cardiac cell differentiation. Cells expressing NIC had a significant decrease in expression of cardiac muscle markers, but an increase in expression of conduction cell markers, HNK-1, and SNAP-25. However, the expression of the conduction marker connexin 40 was inhibited. Loss-of-function study, using a dominant-negative form of Suppressor-of-Hairless, further supports that Notch1 signaling is important for the differentiation of these cardiac cell types. Functional studies show that the expression of constitutively active Notch1 resulted in abnormalities in ventricular conduction pathway patterns.
Collapse
Affiliation(s)
- Mary D L Chau
- Department of Medicine and Cell Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
38
|
Weller M, Krautler N, Mantei N, Suter U, Taylor V. Jagged1 ablation results in cerebellar granule cell migration defects and depletion of Bergmann glia. Dev Neurosci 2006; 28:70-80. [PMID: 16508305 DOI: 10.1159/000090754] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2005] [Accepted: 06/21/2005] [Indexed: 11/19/2022] Open
Abstract
Jagged1 is a ligand for members of the Notch family of receptors. Mutations in the human JAG1 gene are the major cause of Alagille syndrome, an autosomal dominant disorder affecting the liver, heart, eye, skeleton, kidneys, and craniofacial structures. Although expressed throughout mammalian embryonic development and in the adult, the function of Jagged1 in the central nervous system is not clear. Jagged1 is broadly expressed in the cerebellum suggesting an important role in Notch signaling. In order to address the function of Jagged1 in the mouse central nervous system, we have inactivated the Jag1 gene in the cerebellar primordium at mid-embryogenesis. Loss of Jagged1 results in aberrant granule cell migration and ectopic differentiation in the external germinal layer and molecular layer of the early postnatal cerebellum. We show that Bergmann glia in the cerebellum lose contact to the pial surface and have stunted processes. In vitro analysis revealed a depletion of Bergmann glia in the Jagged1 mutant mice. Our findings suggest that Jagged1 plays a role in cell fate specification and survival in the cerebellum.
Collapse
Affiliation(s)
- Mathias Weller
- Department of Molecular Embryology, Max Planck Institute of Immunobiology, Freiburg, Germany
| | | | | | | | | |
Collapse
|
39
|
Beaster-Jones L, Horton AC, Gibson-Brown JJ, Holland ND, Holland LZ. The amphioxus T-box gene, AmphiTbx15/18/22, illuminates the origins of chordate segmentation. Evol Dev 2006; 8:119-29. [PMID: 16509891 DOI: 10.1111/j.1525-142x.2006.00083.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Amphioxus and vertebrates are the only deuterostomes to exhibit unequivocal somitic segmentation. The relative simplicity of the amphioxus genome makes it a favorable organism for elucidating the basic genetic network required for chordate somite development. Here we describe the developmental expression of the somite marker, AmphiTbx15/18/22, which is first expressed at the mid-gastrula stage in dorsolateral mesendoderm. At the early neurula stage, expression is detected in the first three pairs of developing somites. By the mid-neurula stage, expression is downregulated in anterior somites, and only detected in the penultimate somite primordia. In early larvae, the gene is expressed in nascent somites before they pinch off from the posterior archenteron (tail bud). Integrating functional, phylogenetic and expression data from a variety of triploblast organisms, we have reconstructed the evolutionary history of the Tbx15/18/22 subfamily. This analysis suggests that the Tbx15/18/22 gene may have played a role in patterning somites in the last common ancestor of all chordates, a role that was later conserved by its descendents following gene duplications within the vertebrate lineage. Furthermore, the comparison of expression domains within this gene subfamily reveals similarities in the genetic bases of trunk and cranial mesoderm segmentation. This lends support to the hypothesis that the vertebrate head evolved from an ancestor possessing segmented cranial mesoderm.
Collapse
Affiliation(s)
- Laura Beaster-Jones
- Marine Biology Research Division, Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA 92093-0202, USA.
| | | | | | | | | |
Collapse
|
40
|
Williams CK, Li JL, Murga M, Harris AL, Tosato G. Up-regulation of the Notch ligand Delta-like 4 inhibits VEGF-induced endothelial cell function. Blood 2005; 107:931-9. [PMID: 16219802 PMCID: PMC1895896 DOI: 10.1182/blood-2005-03-1000] [Citation(s) in RCA: 294] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Delta-like 4 (Dll4), a membrane-bound ligand for Notch1 and Notch4, is selectively expressed in the developing endothelium and in some tumor endothelium, and it is induced by vascular endothelial growth factor (VEGF)-A and hypoxia. Gene targeting studies have shown that Dll4 is required for normal embryonic vascular remodeling, but the mechanisms underlying Dll4 regulatory functions are currently not defined. In this study, we generated primary human endothelial cells that overexpress Dll4 protein to study Dll4 function and mechanism of action. Human umbilical vein endothelial cells retrovirally transduced with Dll4 displayed reduced proliferative and migratory responses selectively to VEGF-A. Expression of VEGF receptor-2, the principal signaling receptor for VEGF-A in endothelial cells, and coreceptor neuropilin-1 was significantly decreased in Dll4-transduced endothelial cells. Consistent with Dll4 signaling through Notch, expression of HEY2, one of the transcription factors that mediates Notch function, was significantly induced in Dll4-overexpressing endothelial cells. The gamma-secretase inhibitor L-685458 significantly reconstituted endothelial cell proliferation inhibited by immobilized extracellular Dll4 and reconstituted VEGFR2 expression in Dll4-overexpressing endothelial cells. These results identify the Notch ligand Dll4 as a selective inhibitor of VEGF-A biologic activities down-regulating 2 VEGF receptors expressed on endothelial cells and raise the possibility that Dll4 may be exploited therapeutically to modulate angiogenesis.
Collapse
Affiliation(s)
- Cassin Kimmel Williams
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
41
|
Limbourg FP, Takeshita K, Radtke F, Bronson RT, Chin MT, Liao JK. Essential role of endothelial Notch1 in angiogenesis. Circulation 2005; 111:1826-32. [PMID: 15809373 PMCID: PMC2633594 DOI: 10.1161/01.cir.0000160870.93058.dd] [Citation(s) in RCA: 215] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Notch signaling influences binary cell fate decisions in a variety of tissues. The Notch1 receptor is widely expressed during embryogenesis and is essential for embryonic development. Loss of global Notch1 function results in early embryonic lethality, but the cell type responsible for this defect is not known. Here, we identify the endothelium as the primary target tissue affected by Notch1 signaling. METHODS AND RESULTS We generated an endothelium-specific deletion of Notch1 using Tie2Cre and conditional Notch1(flox/flox) mice. Mutant embryos lacking endothelial Notch1 died at approximately embryonic day 10.5 with profound vascular defects in placenta, yolk sac, and embryo proper, whereas heterozygous deletion had no effect. In yolk sacs of mutant embryos, endothelial cells formed a primary vascular plexus indicative of intact vasculogenesis but failed to induce the secondary vascular remodeling required to form a mature network of well-organized large and small blood vessels, which demonstrates a defect in angiogenesis. These vascular defects were also evident in the placenta, where blood vessels failed to invade the placental labyrinth, and in the embryo proper, where defective blood vessel maturation led to pericardial and intersomitic hemorrhage. Enhanced activation of caspase-3 was detected in endothelial and neural cells of mutant mice, which resulted in enhanced apoptotic degeneration of somites and the neural tube. CONCLUSIONS These findings recapitulate the vascular phenotype of global Notch1-/- mutants and indicate an essential cell-autonomous role of Notch1 signaling in the endothelium during vascular development. These results may have important clinical implications with regard to Notch1 signaling in adult angiogenesis.
Collapse
MESH Headings
- Animals
- Apoptosis
- Caspase 3
- Caspases/metabolism
- Embryo Loss
- Embryo, Mammalian
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/embryology
- Endothelium, Vascular/physiology
- Genotype
- Hemorrhage/etiology
- Mice
- Mice, Mutant Strains
- Neovascularization, Physiologic
- Neural Tube Defects/etiology
- Neural Tube Defects/pathology
- Placenta/blood supply
- Receptor, Notch1
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Signal Transduction
- Somites/pathology
- Transcription Factors/deficiency
- Transcription Factors/genetics
- Transcription Factors/physiology
Collapse
Affiliation(s)
- Florian P Limbourg
- Vascular Medicine Research Unit, Brigham & Women's Hospital, and Harvard Medical School, Boston, Mass, USA
| | | | | | | | | | | |
Collapse
|
42
|
Ishibashi M, Saitsu H, Komada M, Shiota K. Signaling cascade coordinating growth of dorsal and ventral tissues of the vertebrate brain, with special reference to the involvement of Sonic hedgehog signaling. Anat Sci Int 2005; 80:30-6. [PMID: 15794128 DOI: 10.1111/j.1447-073x.2005.00096.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The vertebrate brain is a complex and highly organized structure with numerous neurons and glial cells. During development, undifferentiated progenitor cells proliferate from neural stem/precursor cells and gradually restrict their fates according to their environment. Differentiated cells are arranged precisely to accomplish their function and to maintain integrity as a whole brain. In this respect, cells must receive signals to know where and when they determine their fates. Secreted and membrane molecules convey the information between cells. The secreted glycoprotein Sonic hedgehog (Shh) is one of such signaling molecules. Sonic hedgehog is widely known to specify ventral neuronal types according to the concentration of Shh, whereas differentiation of dorsal neurons is largely independent of Shh. However, in the diencephalon and midbrain, dorsal parts are also affected in Shh-mutant embryos. Detailed analysis demonstrated that Shh signaling indirectly regulates the growth of the dorsal tissue in these regions. One of the fibroblast growth factor (FGF) members, namely FGF15, has been reported to be downstream to Shh signaling in the mouse embryonic brain. Luciferase assays and transgenic analysis revealed that the Fgf15 gene is a direct target of Shh. Downregulation of Tcf4 and upregulation of Bmp4 in Shh mutants suggest that Wnt and BMP signals from the dorsal midline are also involved in the dorsal brain phenotype. These data suggest the coordinating role of the Shh-FGF15-Wnt/BMP signaling cascade between the ventral and dorsal parts of the brain.
Collapse
Affiliation(s)
- Makoto Ishibashi
- Department of Anatomy and Developmental Biology and Kyoto University, Kyoto, Japan.
| | | | | | | |
Collapse
|
43
|
Abstract
The embryonic vasculature develops in a conserved manner in all vertebrates. Endothelial progenitor cells differentiate from mesodermal cells, then migrate and assemble into the dorsal aorta and the cardinal vein. This primitive circulatory loop undergoes sprouting and branching via a two-step navigation mechanism to form the trunk vascular network. Various studies using several model systems have uncovered a number of signaling mechanisms that regulate these complex processes. A genetic approach in zebrafish has led to identification of mutations and molecules that are responsible for specification of endothelial progenitor cells, differentiation of arterial and venous cells, and patterning of the dorsal aorta and intersegmental vessels. These studies highlight the unique utilities and benefits of the zebrafish system for studying development of embryonic blood vessels.
Collapse
Affiliation(s)
- Tao P Zhong
- Departments of Medicine and Cell and Developmental Biology Vanderbilt University School of Medicine Nashville, Tennessee 37232, USA
| |
Collapse
|
44
|
Hadland BK, Huppert SS, Kanungo J, Xue Y, Jiang R, Gridley T, Conlon RA, Cheng AM, Kopan R, Longmore GD. A requirement for Notch1 distinguishes 2 phases of definitive hematopoiesis during development. Blood 2004; 104:3097-105. [PMID: 15251982 PMCID: PMC5998659 DOI: 10.1182/blood-2004-03-1224] [Citation(s) in RCA: 185] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Notch1 is known to play a critical role in regulating fates in numerous cell types, including those of the hematopoietic lineage. Multiple defects exhibited by Notch1-deficient embryos confound the determination of Notch1 function in early hematopoietic development in vivo. To overcome this limitation, we examined the developmental potential of Notch1(-/-) embryonic stem (ES) cells by in vitro differentiation and by in vivo chimera analysis. Notch1 was found to affect primitive erythropoiesis differentially during ES cell differentiation and in vivo, and this result reflected an important difference in the regulation of Notch1 expression during ES cell differentiation relative to the developing mouse embryo. Notch1 was dispensable for the onset of definitive hematopoiesis both in vitro and in vivo in that Notch1(-/-) definitive progenitors could be detected in differentiating ES cells as well as in the yolk sac and early fetal liver of chimeric mice. Despite the fact that Notch1(-/-) cells can give rise to multiple types of definitive progenitors in early development, Notch1(-/-) cells failed to contribute to long-term definitive hematopoiesis past the early fetal liver stage in the context of a wild-type environment in chimeric mice. Thus, Notch1 is required, in a cell-autonomous manner, for the establishment of long-term, definitive hematopoietic stem cells (HSCs).
Collapse
Affiliation(s)
- Brandon K Hadland
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis MO 63110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kuan CYK, Tannahill D, Cook GMW, Keynes RJ. Somite polarity and segmental patterning of the peripheral nervous system. Mech Dev 2004; 121:1055-68. [PMID: 15296971 DOI: 10.1016/j.mod.2004.05.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2004] [Revised: 04/29/2004] [Accepted: 05/03/2004] [Indexed: 10/26/2022]
Abstract
The analysis of the outgrowth pattern of spinal axons in the chick embryo has shown that somites are polarized into anterior and posterior halves. This polarity dictates the segmental development of the peripheral nervous system: migrating neural crest cells and outgrowing spinal axons traverse exclusively the anterior halves of the somite-derived sclerotomes, ensuring a proper register between spinal axons, their ganglia and the segmented vertebral column. Much progress has been made recently in understanding the molecular basis for somite polarization, and its linkage with Notch/Delta, Wnt and Fgf signalling. Contact-repulsive molecules expressed by posterior half-sclerotome cells provide critical guidance cues for axons and neural crest cells along the anterior-posterior axis. Diffusible repellents from surrounding tissues, particularly the dermomyotome and notochord, orient outgrowing spinal axons in the dorso-ventral axis ('surround repulsion'). Repulsive forces therefore guide axons in three dimensions. Although several molecular systems have been identified that may guide neural crest cells and axons in the sclerotome, it remains unclear whether these operate together with considerable overall redundancy, or whether any one system predominates in vivo.
Collapse
Affiliation(s)
- C-Y Kelly Kuan
- Department of Anatomy, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | | | | | | |
Collapse
|
46
|
Krebs LT, Xue Y, Norton CR, Sundberg JP, Beatus P, Lendahl U, Joutel A, Gridley T. Characterization of Notch3-deficient mice: normal embryonic development and absence of genetic interactions with a Notch1 mutation. Genesis 2004; 37:139-43. [PMID: 14595837 DOI: 10.1002/gene.10241] [Citation(s) in RCA: 200] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The Notch signaling pathway is an evolutionarily conserved signaling mechanism and mutations in its components disrupt cell fate specification and embryonic development in many organisms. To analyze the in vivo role of the Notch3 gene in mice, we created a deletion allele by gene targeting. Embryos homozygous for this mutation developed normally and homozygous mutant adults were viable and fertile. We also examined whether we could detect genetic interactions during early embryogenesis between the Notch3 mutation and a targeted mutation of the Notch1 gene. Double homozygous mutant embryos exhibited defects normally observed in Notch1-deficient embryos, but we detected no obvious synergistic effects in the double mutants. These data demonstrate that the Notch3 gene is not essential for embryonic development or fertility in mice, and does not have a redundant function with the Notch1 gene during early embryogenesis.
Collapse
Affiliation(s)
- Luke T Krebs
- The Jackson Laboratory, Bar Harbor, Maine 04609, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Two distinct mechanisms, vasculogenesis and angiogenesis implement the formation of the vascular network in the embryo. Vasculogenesis gives rise to the heart and the first primitive vascular plexus inside the embryo and in its surrounding membranes, as the yolk sac circulation. Angiogenesis is responsible for the remodeling and expansion of this network. While vasculogenesis refers to in situ differentiation and growth of blood vessels from mesodermal derived hemangioblasts, angiogenesis comprises two different mechanisms: endothelial sprouting and intussusceptive microvascular growth (IMG). The sprouting process is based on endothelial cell migration, proliferation and tube formation. IMG divides existing vessel lumens by formation and insertion of tissue folds and columns of interstitial tissue into the vessel lumen. The latter are termed interstitial or intervascular tissue structures (ITSs) and tissue pillars or posts. Intussusception also includes the establishment of new vessels by in situ loop formation in the wall of large veins. The molecular regulation of these distinct mechanisms is discussed in respect to the most important positive regulators, VEGF and its receptors flk-1 (KDR) and flt-1, the Angiopoietin/tie system and the ephrin-B/EpH-B system. The cellular mechanisms and the molecular regulation of angiogenesis in the pathological state are summarized and the differences of physiological and pathological angiogenesis elaborated.
Collapse
Affiliation(s)
- Sybill Patan
- Division of Cardiology, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York 10461, USA
| |
Collapse
|
48
|
Affiliation(s)
- Lisa D Urness
- Division of Cardiology, University of Utah School of Medicine, Salt Lake City, Utah 84112, USA
| | | |
Collapse
|
49
|
Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM, Díez J, Aranda S, Palomo S, McCormick F, Izpisúa-Belmonte JC, de la Pompa JL. Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev 2003; 18:99-115. [PMID: 14701881 PMCID: PMC314285 DOI: 10.1101/gad.276304] [Citation(s) in RCA: 730] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) is fundamental to both embryogenesis and tumor metastasis. The Notch intercellular signaling pathway regulates cell fate determination throughout metazoan evolution, and overexpression of activating alleles is oncogenic in mammals. Here we demonstrate that Notch activity promotes EMT during both cardiac development and oncogenic transformation via transcriptional induction of the Snail repressor, a potent and evolutionarily conserved mediator of EMT in many tissues and tumor types. In the embryonic heart, Notch functions via lateral induction to promote a selective transforming growth factor-beta (TGFbeta)-mediated EMT that leads to cellularization of developing cardiac valvular primordia. Embryos that lack Notch signaling elements exhibit severely attenuated cardiac snail expression, abnormal maintenance of intercellular endocardial adhesion complexes, and abortive endocardial EMT in vivo and in vitro. Accordingly, transient ectopic expression of activated Notch1 (N1IC) in zebrafish embryos leads to hypercellular cardiac valves, whereas Notch inhibition prevents valve development. Overexpression of N1IC in immortalized endothelial cells in vitro induces EMT accompanied by oncogenic transformation, with corresponding induction of snail and repression of VE-cadherin expression. Notch is expressed in embryonic regions where EMT occurs, suggesting an intimate and fundamental role for Notch, which may be reactivated during tumor metastasis.
Collapse
Affiliation(s)
- Luika A Timmerman
- University of California Comprehensive Cancer Center, San Francisco, California 94115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tanaka M, Marunouchi T. Immunohistochemical localization of Notch receptors and their ligands in the postnatally developing rat cerebellum. Neurosci Lett 2003; 353:87-90. [PMID: 14664907 DOI: 10.1016/j.neulet.2003.08.080] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although mRNAs of Notch receptors and their ligands are known to be expressed in the postnatally developing rodent cerebellum, their protein localization has been poorly investigated. In the present study, we immunohistochemically examined localization of Notch receptors and their ligands during postnatal cerebellar development in rats. During the first two postnatal weeks, intense signals of Notch1-3 were localized in Bergmann fibers (radial fibers of Bergmann glia), as confirmed by double fluorescent immunohistochemistry. After that, the signals gradually declined into adulthood. Among Notch ligands, Jagged1 and 2 were also localized in Bergmann fibers. These results suggest that cell-cell interactions through Jagged-Notch signaling can occur between Bergmann glia during postnatal cerebellar development.
Collapse
Affiliation(s)
- Masahiko Tanaka
- Division of Cell Biology, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | | |
Collapse
|