1
|
Papalamprou A, Yu V, Jiang W, Sheyn J, Stefanovic T, Chen A, Castaneda C, Chavez M, Sheyn D. Single Cell Transcriptomics-Informed Induced Pluripotent Stem Cells Differentiation to Tenogenic Lineage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.10.536240. [PMID: 37090543 PMCID: PMC10120682 DOI: 10.1101/2023.04.10.536240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
During vertebrate embryogenesis, axial tendons develop from the paraxial mesoderm and differentiate through specific developmental stages to reach the syndetome stage. While the main roles of signaling pathways in the earlier stages of the differentiation have been well established, pathway nuances in syndetome specification from the sclerotome stage have yet to be explored. Here, we show stepwise differentiation of human iPSCs to the syndetome stage using chemically defined media and small molecules that were modified based on single cell RNA-sequencing and pathway analysis. We identified a significant population of branching off-target cells differentiating towards a neural phenotype overexpressing Wnt. Further transcriptomics post-addition of a WNT inhibitor at the somite stage and onwards revealed not only total removal of the neural off-target cells, but also increased syndetome induction efficiency. Fine-tuning tendon differentiation in vitro is essential to address the current challenges in developing a successful cell-based tendon therapy.
Collapse
Affiliation(s)
- Angela Papalamprou
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Victoria Yu
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Wensen Jiang
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Julia Sheyn
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Tina Stefanovic
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Angel Chen
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Chloe Castaneda
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Melissa Chavez
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Dmitriy Sheyn
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
2
|
Ayad NM, Lakins JN, Ghagre A, Ehrlicher AJ, Weaver VM. Tissue tension permits β-catenin phosphorylation to drive mesoderm specification in human embryonic stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.14.549074. [PMID: 37503095 PMCID: PMC10370032 DOI: 10.1101/2023.07.14.549074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The role of morphogenetic forces in cell fate specification is an area of intense interest. Our prior studies suggested that the development of high cell-cell tension in human embryonic stem cells (hESC) colonies permits the Src-mediated phosphorylation of junctional β-catenin that accelerates its release to potentiate Wnt-dependent signaling critical for initiating mesoderm specification. Using an ectopically expressed nonphosphorylatable mutant of β-catenin (Y654F), we now provide direct evidence that impeding tension-dependent Src-mediated β-catenin phosphorylation impedes the expression of Brachyury (T) and the epithelial-to-mesenchymal transition (EMT) necessary for mesoderm specification. Addition of exogenous Wnt3a or inhibiting GSK3β activity rescued mesoderm expression, emphasizing the importance of force dependent Wnt signaling in regulating mechanomorphogenesis. Our work provides a framework for understanding tension-dependent β-catenin/Wnt signaling in the self-organization of tissues during developmental processes including gastrulation.
Collapse
Affiliation(s)
- Nadia M.E. Ayad
- Graduate Program in Bioengineering, University of California, San Francisco and University of California Berkeley, San Francisco, CA 94143, USA; Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Johnathon N. Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ajinkya Ghagre
- Department of Bioengineering, McGill University, Montreal, QC H3A 0E9, Canada
| | - Allen J. Ehrlicher
- Department of Bioengineering, Department of Anatomy and Cell Biology, Department of Biomedical Engineering, Department of Mechanical Engineering, Centre for Structural Biology, Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; UCSF Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
3
|
Nakayama J, Makinoshima H, Gong Z. Gastrulation Screening to Identify Anti-metastasis Drugs in Zebrafish Embryos. Bio Protoc 2022; 12:e4525. [PMID: 36313195 PMCID: PMC9548519 DOI: 10.21769/bioprotoc.4525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/04/2022] [Accepted: 08/25/2022] [Indexed: 12/29/2022] Open
Abstract
Few models exist that allow for rapid and effective screening of anti-metastasis drugs. Here, we present a drug screening protocol utilizing gastrulation of zebrafish embryos for identification of anti-metastasis drugs. Based on the evidence that metastasis proceeds through utilizing the molecular mechanisms of gastrulation, we hypothesized that chemicals interrupting zebrafish gastrulation might suppress the metastasis of cancer cells. Thus, we developed a phenotype-based chemical screen that uses epiboly, the first morphogenetic movement in gastrulation, as a marker. The screen only needs zebrafish embryos and enables hundreds of chemicals to be tested in five hours by observing the epiboly progression of chemical-treated embryos. In the screen, embryos at the two-cell stage are firstly corrected and then developed to the sphere stage. The embryos are treated with a test chemical and incubated in the presence of the chemical until vehicle-treated embryos develop to the 90% epiboly stage. Finally, positive 'hit' chemicals that interrupt epiboly progression are selected by comparing epiboly progression of the chemical-treated and vehicle-treated embryos under a stereoscopic microscope. A previous study subjected 1,280 FDA-approved drugs to the screen and identified adrenosterone and pizotifen as epiboly-interrupting drugs. These were validated to suppress metastasis of breast cancer cells in mice models of metastasis. Furthermore, 11β-hydroxysteroid dehydrogenase 1 (HSD11β1) and serotonin receptor 2C (HTR2C), the primary targets of adrenosterone and pizotifen, respectively, promoted metastasis through induction of epithelial-mesenchymal transition (EMT). Therefore, this screen could be converted into a chemical genetic screening platform for identification of metastasis-promoting genes. Graphical abstract.
Collapse
Affiliation(s)
- Joji Nakayama
- Tsuruoka Metabolomics Laboratory, National Cancer Center, Tsuruoka, Japan
,
Shonai Regional Industry Promotion Center, Tsuruoka, Japan
,
Department of Biological Science, National University of Singapore, Singapore
,
*For correspondence:
| | - Hideki Makinoshima
- Tsuruoka Metabolomics Laboratory, National Cancer Center, Tsuruoka, Japan
,
Division of Translational Research, Exploratory Oncology Research, and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Zhiyuan Gong
- Department of Biological Science, National University of Singapore, Singapore
| |
Collapse
|
4
|
Abstract
Neural crest cells (NCCs) are a dynamic, multipotent, vertebrate-specific population of embryonic stem cells. These ectodermally-derived cells contribute to diverse tissue types in developing embryos including craniofacial bone and cartilage, the peripheral and enteric nervous systems and pigment cells, among a host of other cell types. Due to their contribution to a significant number of adult tissue types, the mechanisms that drive their formation, migration and differentiation are highly studied. NCCs have a unique ability to transition from tightly adherent epithelial cells to mesenchymal and migratory cells by altering their polarity, expression of cell-cell adhesion molecules and gaining invasive abilities. In this Review, we discuss classical and emerging factors driving NCC epithelial-to-mesenchymal transition and migration, highlighting the role of signaling and transcription factors, as well as novel modifying factors including chromatin remodelers, small RNAs and post-translational regulators, which control the availability and longevity of major NCC players.
Collapse
Affiliation(s)
| | - Crystal D. Rogers
- Department of Anatomy, Physiology, and Cell Biology, UC Davis School of Veterinary Medicine, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA
| |
Collapse
|
5
|
Godden AM, Antonaci M, Ward NJ, van der Lee M, Abu-Daya A, Guille M, Wheeler GN. An efficient miRNA knockout approach using CRISPR-Cas9 in Xenopus. Dev Biol 2022; 483:66-75. [PMID: 34968443 PMCID: PMC8865746 DOI: 10.1016/j.ydbio.2021.12.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Abstract
In recent years CRISPR-Cas9 knockouts (KO) have become increasingly ultilised to study gene function. MicroRNAs (miRNAs) are short non-coding RNAs, 20-22 nucleotides long, which affect gene expression through post-transcriptional repression. We previously identified miRNAs-196a and -219 as implicated in the development of Xenopus neural crest (NC). The NC is a multipotent stem-cell population, specified during early neurulation. Following EMT, NC cells migrate to various points in the developing embryo where they give rise to a number of tissues including parts of the peripheral nervous system, pigment cells and craniofacial skeleton. Dysregulation of NC development results in many diseases grouped under the term neurocristopathies. As miRNAs are so small, it is difficult to design CRISPR sgRNAs that reproducibly lead to a KO. We have therefore designed a novel approach using two guide RNAs to effectively 'drop out' a miRNA. We have knocked out miR-196a and miR-219 and compared the results to morpholino knockdowns (KD) of the same miRNAs. Validation of efficient CRISPR miRNA KO and phenotype analysis included use of whole-mount in situ hybridization of key NC and neural plate border markers such as Pax3, Xhe2, Sox10 and Snail2, q-RT-PCR and Sanger sequencing. To show specificity we have also rescued the knockout phenotype using miRNA mimics. MiRNA-219 and miR-196a KO's both show loss of NC, altered neural plate and hatching gland phenotypes. Tadpoles show gross craniofacial and pigment phenotypes.
Collapse
Affiliation(s)
- Alice M Godden
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Marco Antonaci
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Nicole J Ward
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Michael van der Lee
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Anita Abu-Daya
- King Henry Building, King Henry I St, Portsmouth, PO1 2DY, United Kingdom
| | - Matthew Guille
- King Henry Building, King Henry I St, Portsmouth, PO1 2DY, United Kingdom
| | - Grant N Wheeler
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom.
| |
Collapse
|
6
|
Wu Y, Devotta A, José-Edwards DS, Kugler JE, Negrón-Piñeiro LJ, Braslavskaya K, Addy J, Saint-Jeannet JP, Di Gregorio A. Xbp1 and Brachyury establish an evolutionarily conserved subcircuit of the notochord gene regulatory network. eLife 2022; 11:e73992. [PMID: 35049502 PMCID: PMC8803312 DOI: 10.7554/elife.73992] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Gene regulatory networks coordinate the formation of organs and structures that compose the evolving body plans of different organisms. We are using a simple chordate model, the Ciona embryo, to investigate the essential gene regulatory network that orchestrates morphogenesis of the notochord, a structure necessary for the proper development of all chordate embryos. Although numerous transcription factors expressed in the notochord have been identified in different chordates, several of them remain to be positioned within a regulatory framework. Here, we focus on Xbp1, a transcription factor expressed during notochord formation in Ciona and other chordates. Through the identification of Xbp1-downstream notochord genes in Ciona, we found evidence of the early co-option of genes involved in the unfolded protein response to the notochord developmental program. We report the regulatory interplay between Xbp1 and Brachyury, and by extending these results to Xenopus, we show that Brachyury and Xbp1 form a cross-regulatory subcircuit of the notochord gene regulatory network that has been consolidated during chordate evolution.
Collapse
Affiliation(s)
- Yushi Wu
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
| | - Arun Devotta
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
| | - Diana S José-Edwards
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
| | - Jamie E Kugler
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
| | - Lenny J Negrón-Piñeiro
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
| | - Karina Braslavskaya
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
| | - Jermyn Addy
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
| | | | - Anna Di Gregorio
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
| |
Collapse
|
7
|
Monroy BY, Adamson CJ, Camacho-Avila A, Guerzon CN, Echeverria CV, Rogers CD. Expression atlas of avian neural crest proteins: Neurulation to migration. Dev Biol 2022; 483:39-57. [PMID: 34990731 DOI: 10.1016/j.ydbio.2021.12.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/11/2021] [Accepted: 12/30/2021] [Indexed: 11/20/2022]
Abstract
Neural crest (NC) cells are a dynamic population of embryonic stem cells that create various adult tissues in vertebrate species including craniofacial bone and cartilage and the peripheral and enteric nervous systems. NC development is thought to be a conserved and complex process that is controlled by a tightly-regulated gene regulatory network (GRN) of morphogens, transcription factors, and cell adhesion proteins. While multiple studies have characterized the expression of several GRN factors in single species, a comprehensive protein analysis that directly compares expression across development is lacking. To address this lack in information, we used three closely related avian models, Gallus gallus (chicken), Coturnix japonica (Japanese quail), and Pavo cristatus (Indian peafowl), to compare the localization and timing of four GRN transcription factors, PAX7, SNAI2, SOX9, and SOX10, from the onset of neurulation to migration. While the spatial expression of these factors is largely conserved, we find that quail NC cells express SNAI2, SOX9, and SOX10 proteins at the equivalent of earlier developmental stages than chick and peafowl. In addition, quail NC cells migrate farther and more rapidly than the larger organisms. These data suggest that despite a conservation of NC GRN players, differences in the timing of NC development between species remain a significant frontier to be explored with functional studies.
Collapse
Affiliation(s)
- Brigette Y Monroy
- Anatomy, Physiology, and Cell Biology Department, University of California Davis, Davis, CA, 95616, USA
| | - Carly J Adamson
- Anatomy, Physiology, and Cell Biology Department, University of California Davis, Davis, CA, 95616, USA
| | - Alexis Camacho-Avila
- Department of Biology, California State University Northridge, Northridge, CA, 91330, USA
| | - Christian N Guerzon
- Anatomy, Physiology, and Cell Biology Department, University of California Davis, Davis, CA, 95616, USA
| | - Camilo V Echeverria
- Anatomy, Physiology, and Cell Biology Department, University of California Davis, Davis, CA, 95616, USA
| | - Crystal D Rogers
- Anatomy, Physiology, and Cell Biology Department, University of California Davis, Davis, CA, 95616, USA.
| |
Collapse
|
8
|
Nakayama J, Tan L, Li Y, Goh BC, Wang S, Makinoshima H, Gong Z. A zebrafish embryo screen utilizing gastrulation identifies the HTR2C inhibitor pizotifen as a suppressor of EMT-mediated metastasis. eLife 2021; 10:e70151. [PMID: 34919051 PMCID: PMC8824480 DOI: 10.7554/elife.70151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
Metastasis is responsible for approximately 90% of cancer-associated mortality but few models exist that allow for rapid and effective screening of anti-metastasis drugs. Current mouse models of metastasis are too expensive and time consuming to use for rapid and high-throughput screening. Therefore, we created a unique screening concept utilizing conserved mechanisms between zebrafish gastrulation and cancer metastasis for identification of potential anti-metastatic drugs. We hypothesized that small chemicals that interrupt zebrafish gastrulation might also suppress metastatic progression of cancer cells and developed a phenotype-based chemical screen to test the hypothesis. The screen used epiboly, the first morphogenetic movement in gastrulation, as a marker and enabled 100 chemicals to be tested in 5 hr. The screen tested 1280 FDA-approved drugs and identified pizotifen, an antagonist for serotonin receptor 2C (HTR2C) as an epiboly-interrupting drug. Pharmacological and genetic inhibition of HTR2C suppressed metastatic progression in a mouse model. Blocking HTR2C with pizotifen restored epithelial properties to metastatic cells through inhibition of Wnt signaling. In contrast, HTR2C induced epithelial-to-mesenchymal transition through activation of Wnt signaling and promoted metastatic dissemination of human cancer cells in a zebrafish xenotransplantation model. Taken together, our concept offers a novel platform for discovery of anti-metastasis drugs.
Collapse
Affiliation(s)
- Joji Nakayama
- Department of Biological Science, National University of SingaporeSingaporeSingapore
- Cancer Science Institute of Singapore, National University of SingaporeSingaporeSingapore
- Tsuruoka Metabolomics Laboratory, National Cancer CenterTsuruokaJapan
- Shonai Regional Industry Promotion CenterTsuruokaJapan
| | - Lora Tan
- Department of Biological Science, National University of SingaporeSingaporeSingapore
| | - Yan Li
- Department of Biological Science, National University of SingaporeSingaporeSingapore
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of SingaporeSingaporeSingapore
| | - Shu Wang
- Department of Biological Science, National University of SingaporeSingaporeSingapore
- Institute of Bioengineering and NanotechnologySingaporeSingapore
| | - Hideki Makinoshima
- Tsuruoka Metabolomics Laboratory, National Cancer CenterTsuruokaJapan
- Division of Translational Research, Exploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Zhiyuan Gong
- Department of Biological Science, National University of SingaporeSingaporeSingapore
| |
Collapse
|
9
|
Shellard A, Mayor R. Collective durotaxis along a self-generated stiffness gradient in vivo. Nature 2021; 600:690-694. [PMID: 34880503 DOI: 10.1038/s41586-021-04210-x] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/02/2021] [Indexed: 02/07/2023]
Abstract
Collective cell migration underlies morphogenesis, wound healing and cancer invasion1,2. Most directed migration in vivo has been attributed to chemotaxis, whereby cells follow a chemical gradient3-5. Cells can also follow a stiffness gradient in vitro, a process called durotaxis3,4,6-8, but evidence for durotaxis in vivo is lacking6. Here we show that in Xenopus laevis the neural crest-an embryonic cell population-self-generates a stiffness gradient in the adjacent placodal tissue, and follows this gradient by durotaxis. The gradient moves with the neural crest, which is continually pursuing a retreating region of high substrate stiffness. Mechanistically, the neural crest induces the gradient due to N-cadherin interactions with the placodes and senses the gradient through cell-matrix adhesions, resulting in polarized Rac activity and actomyosin contractility, which coordinates durotaxis. Durotaxis synergizes with chemotaxis, cooperatively polarizing actomyosin machinery of the cell group to prompt efficient directional collective cell migration in vivo. These results show that durotaxis and dynamic stiffness gradients exist in vivo, and gradients of chemical and mechanical signals cooperate to achieve efficient directional cell migration.
Collapse
Affiliation(s)
- Adam Shellard
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London, UK.
| |
Collapse
|
10
|
Piacentino ML, Hutchins EJ, Bronner ME. Essential function and targets of BMP signaling during midbrain neural crest delamination. Dev Biol 2021; 477:251-261. [PMID: 34102166 PMCID: PMC8277753 DOI: 10.1016/j.ydbio.2021.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/30/2022]
Abstract
BMP signaling plays iterative roles during vertebrate neural crest development from induction through craniofacial morphogenesis. However, far less is known about the role of BMP activity in cranial neural crest epithelial-to-mesenchymal transition and delamination. By measuring canonical BMP signaling activity as a function of time from specification through early migration of avian midbrain neural crest cells, we found elevated BMP signaling during delamination stages. Moreover, inhibition of canonical BMP activity via a dominant negative mutant Type I BMP receptor showed that BMP signaling is required for neural crest migration from the midbrain, independent from an effect on EMT and delamination. Transcriptome profiling on control compared to BMP-inhibited cranial neural crest cells identified novel BMP targets during neural crest delamination and early migration including targets of the Notch pathway that are upregulated following BMP inhibition. These results suggest potential crosstalk between the BMP and Notch pathways in early migrating cranial neural crest and provide novel insight into mechanisms regulated by BMP signaling during early craniofacial development.
Collapse
Affiliation(s)
- Michael L Piacentino
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Erica J Hutchins
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
11
|
George RM, Maldonado-Velez G, Firulli AB. The heart of the neural crest: cardiac neural crest cells in development and regeneration. Development 2020; 147:147/20/dev188706. [PMID: 33060096 DOI: 10.1242/dev.188706] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiac neural crest cells (cNCCs) are a migratory cell population that stem from the cranial portion of the neural tube. They undergo epithelial-to-mesenchymal transition and migrate through the developing embryo to give rise to portions of the outflow tract, the valves and the arteries of the heart. Recent lineage-tracing experiments in chick and zebrafish embryos have shown that cNCCs can also give rise to mature cardiomyocytes. These cNCC-derived cardiomyocytes appear to be required for the successful repair and regeneration of injured zebrafish hearts. In addition, recent work examining the response to cardiac injury in the mammalian heart has suggested that cNCC-derived cardiomyocytes are involved in the repair/regeneration mechanism. However, the molecular signature of the adult cardiomyocytes involved in this repair is unclear. In this Review, we examine the origin, migration and fates of cNCCs. We also review the contribution of cNCCs to mature cardiomyocytes in fish, chick and mice, as well as their role in the regeneration of the adult heart.
Collapse
Affiliation(s)
- Rajani M George
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | - Gabriel Maldonado-Velez
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | - Anthony B Firulli
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| |
Collapse
|
12
|
Li J, Perfetto M, Materna C, Li R, Thi Tran H, Vleminckx K, Duncan MK, Wei S. A new transgenic reporter line reveals Wnt-dependent Snai2 re-expression and cranial neural crest differentiation in Xenopus. Sci Rep 2019; 9:11191. [PMID: 31371771 PMCID: PMC6672020 DOI: 10.1038/s41598-019-47665-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
During vertebrate embryogenesis, the cranial neural crest (CNC) forms at the neural plate border and subsequently migrates and differentiates into many types of cells. The transcription factor Snai2, which is induced by canonical Wnt signaling to be expressed in the early CNC, is pivotal for CNC induction and migration in Xenopus. However, snai2 expression is silenced during CNC migration, and its roles at later developmental stages remain unclear. We generated a transgenic X. tropicalis line that expresses enhanced green fluorescent protein (eGFP) driven by the snai2 promoter/enhancer, and observed eGFP expression not only in the pre-migratory and migrating CNC, but also the differentiating CNC. This transgenic line can be used directly to detect deficiencies in CNC development at various stages, including subtle perturbation of CNC differentiation. In situ hybridization and immunohistochemistry confirm that Snai2 is re-expressed in the differentiating CNC. Using a separate transgenic Wnt reporter line, we show that canonical Wnt signaling is also active in the differentiating CNC. Blocking Wnt signaling shortly after CNC migration causes reduced snai2 expression and impaired differentiation of CNC-derived head cartilage structures. These results suggest that Wnt signaling is required for snai2 re-expression and CNC differentiation.
Collapse
Affiliation(s)
- Jiejing Li
- Department of Biology, West Virginia University, Morgantown, WV, 26506, USA.,Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming, 650032, China
| | - Mark Perfetto
- Department of Biology, West Virginia University, Morgantown, WV, 26506, USA.,Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Christopher Materna
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Rebecca Li
- Brown University, Providence, RI, 02912, USA
| | - Hong Thi Tran
- Department for Molecular Biomedical Research and Center for Medical Genetics, Ghent University, B-9052, Ghent, Belgium
| | - Kris Vleminckx
- Department for Molecular Biomedical Research and Center for Medical Genetics, Ghent University, B-9052, Ghent, Belgium
| | - Melinda K Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
13
|
Physiological effects of KDM5C on neural crest migration and eye formation during vertebrate development. Epigenetics Chromatin 2018; 11:72. [PMID: 30522514 PMCID: PMC6282277 DOI: 10.1186/s13072-018-0241-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/22/2018] [Indexed: 02/07/2023] Open
Abstract
Background Lysine-specific histone demethylase 5C (KDM5C) belongs to the jumonji family of demethylases and is specific for the di- and tri-demethylation of lysine 4 residues on histone 3 (H3K4 me2/3). KDM5C is expressed in the brain and skeletal muscles of humans and is associated with various biologically significant processes. KDM5C is known to be associated with X-linked mental retardation and is also involved in the development of cancer. However, the developmental significance of KDM5C has not been explored yet. In the present study, we investigated the physiological roles of KDM5C during Xenopus laevis embryonic development. Results Loss-of-function analysis using kdm5c antisense morpholino oligonucleotides indicated that kdm5c knockdown led to small-sized heads, reduced cartilage size, and malformed eyes (i.e., small-sized and deformed eyes). Molecular analyses of KDM5C functional roles using whole-mount in situ hybridization, β-galactosidase staining, and reverse transcription-polymerase chain reaction revealed that loss of kdm5c resulted in reduced expression levels of neural crest specifiers and genes involved in eye development. Furthermore, transcriptome analysis indicated the significance of KDM5C in morphogenesis and organogenesis. Conclusion Our findings indicated that KDM5C is associated with embryonic development and provided additional information regarding the complex and dynamic gene network that regulates neural crest formation and eye development. This study emphasizes the functional significance of KDM5C in Xenopus embryogenesis; however, further analysis is needed to explore the interactions of KDM5C with specific developmental genes. Electronic supplementary material The online version of this article (10.1186/s13072-018-0241-x) contains supplementary material, which is available to authorized users.
Collapse
|
14
|
Rogers CD, Nie S. Specifying neural crest cells: From chromatin to morphogens and factors in between. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2018; 7:e322. [PMID: 29722151 PMCID: PMC6215528 DOI: 10.1002/wdev.322] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 12/16/2022]
Abstract
Neural crest (NC) cells are a stem-like multipotent population of progenitor cells that are present in vertebrate embryos, traveling to various regions in the developing organism. Known as the "fourth germ layer," these cells originate in the ectoderm between the neural plate (NP), which will become the brain and spinal cord, and nonneural tissues that will become the skin and the sensory organs. NC cells can differentiate into more than 30 different derivatives in response to the appropriate signals including, but not limited to, craniofacial bone and cartilage, sensory nerves and ganglia, pigment cells, and connective tissue. The molecular and cellular mechanisms that control the induction and specification of NC cells include epigenetic control, multiple interactive and redundant transcriptional pathways, secreted signaling molecules, and adhesion molecules. NC cells are important not only because they transform into a wide variety of tissue types, but also because their ability to detach from their epithelial neighbors and migrate throughout developing embryos utilizes mechanisms similar to those used by metastatic cancer cells. In this review, we discuss the mechanisms required for the induction and specification of NC cells in various vertebrate species, focusing on the roles of early morphogenesis, cell adhesion, signaling from adjacent tissues, and the massive transcriptional network that controls the formation of these amazing cells. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Signaling Pathways > Cell Fate Signaling.
Collapse
Affiliation(s)
- Crystal D. Rogers
- Department of Biology, College of Science and Mathematics, California State University Northridge, Northridge, California
| | - Shuyi Nie
- School of Biological Sciences and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
15
|
Rashid D, Puettmann P, Roy E, Bradley RS. Neural crest development in Xenopus requires Protocadherin 7 at the lateral neural crest border. Mech Dev 2018; 149:41-52. [PMID: 29366801 PMCID: PMC5820198 DOI: 10.1016/j.mod.2018.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/04/2017] [Accepted: 01/15/2018] [Indexed: 12/12/2022]
Abstract
In vertebrates, the neural crest is a unique population of pluripotent cells whose development is dependent on signaling from neighboring tissues. Cadherin family members, including protocadherins, are emerging as major players in neural crest development, largely through their roles in cell adhesion and sorting in embryonic tissues. Here, we show that Protocadherin 7 (Pcdh7), previously shown to function in sensorial layer integrity and neural tube closure in Xenopus, is also involved in neural crest specification and survival. Pcdh7 expression partly overlaps the neural crest domain at the lateral neural crest border. Pcdh7 knockdown in embryos does not alter neural crest induction; however, neural crest specification markers, including Snail2 and Sox9, are lost, due to apoptosis of the neural crest starting after stage 13. Pcdh7 knockdown also results in downregulation of Wnt11b; both of which are co-expressed in the sensorial layer lateral to the neural crest, suggestive of a role for Wnt11b in the neural crest apoptosis. Confirming this role, apoptosis, Snail2 expression and the developmental fate of the neural crest can be partially rescued by ectopic expression of Wnt11b. These results indicate that Pcdh7 plays an important role in maintaining the sensorial layer at the lateral neural crest border, which is necessary for the secretion of survival factors, including Wnt11b.
Collapse
Affiliation(s)
- Dana Rashid
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Paul Puettmann
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Ethan Roy
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Roger S. Bradley
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| |
Collapse
|
16
|
Schille C, Schambony A. Signaling pathways and tissue interactions in neural plate border formation. NEUROGENESIS 2017; 4:e1292783. [PMID: 28352644 DOI: 10.1080/23262133.2017.1292783] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 02/04/2023]
Abstract
The neural crest is a transient cell population that gives rise to various cell types of multiple tissues and organs in the vertebrate embryo. Neural crest cells arise from the neural plate border, a region localized at the lateral borders of the prospective neural plate. Temporally and spatially coordinated interaction with the adjacent tissues, the non-neural ectoderm, the neural plate and the prospective dorsolateral mesoderm, is required for neural plate border specification. Signaling molecules, namely BMP, Wnt and FGF ligands and corresponding antagonists are derived from these tissues and interact to induce the expression of neural plate border specific genes. The present mini-review focuses on the current understanding of how the NPB territory is formed and accentuates the need for coordinated interaction of BMP and Wnt signaling pathways and precise tissue communication that are required for the definition of the prospective NC in the competent ectoderm.
Collapse
Affiliation(s)
- Carolin Schille
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nuremberg , Erlangen, Germany
| | - Alexandra Schambony
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nuremberg , Erlangen, Germany
| |
Collapse
|
17
|
Liu Z, Cheng TTK, Shi Z, Liu Z, Lei Y, Wang C, Shi W, Chen X, Qi X, Cai D, Feng B, Deng Y, Chen Y, Zhao H. Efficient genome editing of genes involved in neural crest development using the CRISPR/Cas9 system in Xenopus embryos. Cell Biosci 2016; 6:22. [PMID: 27042291 PMCID: PMC4818404 DOI: 10.1186/s13578-016-0088-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/15/2016] [Indexed: 03/28/2023] Open
Abstract
Background The RNA guided CRISPR/Cas9 nucleases have been proven to be effective for gene disruption in various animal models including Xenopus tropicalis. The neural crest (NC) is a transient cell population during embryonic development and contributes to a large variety of tissues. Currently, loss-of-function studies on NC development in X. tropicalis are largely based on morpholino antisense oligonucleotide. It is worthwhile establishing targeted gene knockout X. tropicails line using CRISPR/Cas9 system to study NC development. Methods We utilized CRISPR/Cas9 to disrupt genes that are involved in NC formation in X. tropicalis embryos. A single sgRNA and Cas9 mRNA synthesized in vitro, were co-injected into X. tropicalis embryos at one-cell stage to induce single gene disruption. We also induced duplex mutations, large segmental deletions and inversions in X. tropicalis by injecting Cas9 and a pair of sgRNAs. The specificity of CRISPR/Cas9 was assessed in X. tropicalis embryos and the Cas9 nickase was used to reduce the off-target cleavages. Finally, we crossed the G0 mosaic frogs with targeted mutations to wild type frogs and obtained the germline transmission. Results Total 16 target sites in 15 genes were targeted by CRISPR/Cas9 and resulted in successful indel mutations at 14 loci with disruption efficiencies in a range from 9.3 to 57.8 %. Furthermore, we demonstrated the feasibility of generation of duplex mutations, large segmental deletions and inversions by using Cas9 and a pair of sgRNAs. We observed that CRISPR/Cas9 displays obvious off-target effects at some loci in X. tropicalis embryos. Such off-target cleavages was reduced by using the D10A Cas9 nickase. Finally, the Cas9 induced indel mutations were efficiently passed to G1 offspring. Conclusion Our study proved that CRISPR/Cas9 could mediate targeted gene mutation in X. tropicalis with high efficiency. This study expands the application of CRISPR/Cas9 platform in X. tropicalis and set a basis for studying NC development using genetic approach. Electronic supplementary material The online version of this article (doi:10.1186/s13578-016-0088-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhongzhen Liu
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China ; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Hong Kong, SAR, China
| | - Tina Tsz Kwan Cheng
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Zhaoying Shi
- Shenzhen Key Laboratory of Cell Microenvironment, Department of Biology, South University of Science and Technology of China, Shenzhen Guangdong, 518055 China
| | - Ziran Liu
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Yong Lei
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Chengdong Wang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Weili Shi
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Xiongfeng Chen
- Advanced Biomedical Computing Center, National Cancer Institute, National Institutes of Health, Frederick, MD 21702 USA
| | - Xufeng Qi
- Key Laboratory for Regenerative Medicine of Ministry of Education, Ji Nan University, Guangzhou, 510632 China ; Department of Developmental and Regenerative Biology, College of Life Science and Technology, Ji Nan University, Guangzhou, 510632 China
| | - Dongqing Cai
- Key Laboratory for Regenerative Medicine of Ministry of Education, Ji Nan University, Guangzhou, 510632 China ; Department of Developmental and Regenerative Biology, College of Life Science and Technology, Ji Nan University, Guangzhou, 510632 China
| | - Bo Feng
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Yi Deng
- Shenzhen Key Laboratory of Cell Microenvironment, Department of Biology, South University of Science and Technology of China, Shenzhen Guangdong, 518055 China
| | - Yonglong Chen
- Shenzhen Key Laboratory of Cell Microenvironment, Department of Biology, South University of Science and Technology of China, Shenzhen Guangdong, 518055 China
| | - Hui Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China ; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Hong Kong, SAR, China
| |
Collapse
|
18
|
Zhao Y, Shi J, Winey M, Klymkowsky MW. Identifying domains of EFHC1 involved in ciliary localization, ciliogenesis, and the regulation of Wnt signaling. Dev Biol 2016; 411:257-265. [PMID: 26783883 DOI: 10.1016/j.ydbio.2016.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/16/2015] [Accepted: 01/07/2016] [Indexed: 12/24/2022]
Abstract
EFHC1 encodes a ciliary protein that has been linked to Juvenile Myoclonic Epilepsy. In ectodermal explants, derived from Xenopus laevis embryos, the morpholino-mediated down-regulation of EFHC1b inhibited multiciliated cell formation. In those ciliated cells that did form, axoneme but not basal body formation was inhibited. EFHC1b morphant embryos displayed defects in central nervous system (CNS) and neural crest patterning that were rescued by a EFHC1b-GFP chimera. EFHC1b-GFP localized to ciliary axonemes in epidermal, gastrocoele roof plate, and neural tube cells. In X. laevis there is a link between Wnt signaling and multiciliated cell formation. While down-regulation of EFHC1b led to a ~2-fold increase in the activity of the β-catenin/Wnt-responsive TOPFLASH reporter, EFHC1b-GFP did not inhibit β-catenin activation of TOPFLASH. Wnt8a RNA levels were increased in EFHC1b morphant ectodermal explants and intact embryos, analyzed prior to the on-set of ciliogenesis. Rescue of the EFHC1b MO's ciliary axonemal phenotypes required the entire protein; in contrast, the EFHC1b morpholino's Wnt8a, CNS, and neural crest phenotypes were rescued by a truncated form of EFHC1b. The EFHC1b morpholino's Wnt8a phenotype was also rescued by the injection of RNAs encoding secreted Wnt inhibitors, suggesting that these phenotypes are due to effects on Wnt signaling, rather than the loss of cilia, an observation of potential relevance to understanding EFHC1's role in human neural development.
Collapse
Affiliation(s)
- Ying Zhao
- Molecular, Cellular & Developmental Biology, University of Colorado Boulder Boulder, Colorado 80309-0347, USA
| | - Jianli Shi
- Molecular, Cellular & Developmental Biology, University of Colorado Boulder Boulder, Colorado 80309-0347, USA
| | - Mark Winey
- Molecular, Cellular & Developmental Biology, University of Colorado Boulder Boulder, Colorado 80309-0347, USA
| | - Michael W Klymkowsky
- Molecular, Cellular & Developmental Biology, University of Colorado Boulder Boulder, Colorado 80309-0347, USA.
| |
Collapse
|
19
|
Lobikin M, Lobo D, Blackiston DJ, Martyniuk CJ, Tkachenko E, Levin M. Serotonergic regulation of melanocyte conversion: A bioelectrically regulated network for stochastic all-or-none hyperpigmentation. Sci Signal 2015; 8:ra99. [PMID: 26443706 DOI: 10.1126/scisignal.aac6609] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Experimentally induced depolarization of resting membrane potential in "instructor cells" in Xenopus laevis embryos causes hyperpigmentation in an all-or-none fashion in some tadpoles due to excess proliferation and migration of melanocytes. We showed that this stochastic process involved serotonin signaling, adenosine 3',5'-monophosphate (cAMP), and the transcription factors cAMP response element-binding protein (CREB), Sox10, and Slug. Transcriptional microarray analysis of embryos taken at stage 15 (early neurula) and stage 45 (free-swimming tadpole) revealed changes in the abundance of 45 and 517 transcripts, respectively, between control embryos and embryos exposed to the instructor cell-depolarizing agent ivermectin. Bioinformatic analysis revealed that the human homologs of some of the differentially regulated genes were associated with cancer, consistent with the induced arborization and invasive behavior of converted melanocytes. We identified a physiological circuit that uses serotonergic signaling between instructor cells, melanotrope cells of the pituitary, and melanocytes to control the proliferation, cell shape, and migration properties of the pigment cell pool. To understand the stochasticity and properties of this multiscale signaling system, we applied a computational machine-learning method that iteratively explored network models to reverse-engineer a stochastic dynamic model that recapitulated the frequency of the all-or-none hyperpigmentation phenotype produced in response to various pharmacological and molecular genetic manipulations. This computational approach may provide insight into stochastic cellular decision-making that occurs during normal development and pathological conditions, such as cancer.
Collapse
Affiliation(s)
- Maria Lobikin
- Biology Department and Center for Regenerative and Developmental Biology, Tufts University, Medford, MA 02155, USA
| | - Daniel Lobo
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Douglas J Blackiston
- Biology Department and Center for Regenerative and Developmental Biology, Tufts University, Medford, MA 02155, USA
| | - Christopher J Martyniuk
- Center for Environmental and Human Toxicology and Department of Physiological Sciences, UF Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Elizabeth Tkachenko
- Biology Department and Center for Regenerative and Developmental Biology, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Biology Department and Center for Regenerative and Developmental Biology, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
20
|
Barriga EH, Trainor PA, Bronner M, Mayor R. Animal models for studying neural crest development: is the mouse different? Development 2015; 142:1555-60. [PMID: 25922521 DOI: 10.1242/dev.121590] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The neural crest is a uniquely vertebrate cell type and has been well studied in a number of model systems. Zebrafish, Xenopus and chick embryos largely show consistent requirements for specific genes in early steps of neural crest development. By contrast, knockouts of homologous genes in the mouse often do not exhibit comparable early neural crest phenotypes. In this Spotlight article, we discuss these species-specific differences, suggest possible explanations for the divergent phenotypes in mouse and urge the community to consider these issues and the need for further research in complementary systems.
Collapse
Affiliation(s)
- Elias H Barriga
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA Department of Anatomy and Cell Biology, University of Kansas Medical Centre, Kansas City, KS 66160, USA
| | - Marianne Bronner
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
21
|
Shi J, Zhao Y, Vonderfecht T, Winey M, Klymkowsky MW. Centrin-2 (Cetn2) mediated regulation of FGF/FGFR gene expression in Xenopus. Sci Rep 2015; 5:10283. [PMID: 26014913 PMCID: PMC4650658 DOI: 10.1038/srep10283] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 03/27/2015] [Indexed: 01/06/2023] Open
Abstract
Centrins (Cetns) are highly conserved, widely expressed, and multifunctional Ca2+-binding eukaryotic signature proteins best known for their roles in ciliogenesis and as critical components of the global genome nucleotide excision repair system. Two distinct Cetn subtypes, Cetn2-like and Cetn3-like, have been recognized and implicated in a range of cellular processes. In the course of morpholino-based loss of function studies in Xenopus laevis, we have identified a previously unreported Cetn2-specific function, namely in fibroblast growth factor (FGF) mediated signaling, specifically through the regulation of FGF and FGF receptor RNA levels. Cetn2 was found associated with the RNA polymerase II binding sites of the Cetn2-regulated FGF8 and FGFR1a genes, but not at the promoter of a gene (BMP4) whose expression was altered indirectly in Cent2 morphant embryos. These observations point to a previously unexpected role of Cetn2 in the regulation of gene expression and embryonic development.
Collapse
Affiliation(s)
- Jianli Shi
- Molecular, Cellular &Developmental Biology University of Colorado Boulder, Boulder, Colorado 80309
| | - Ying Zhao
- Molecular, Cellular &Developmental Biology University of Colorado Boulder, Boulder, Colorado 80309
| | - Tyson Vonderfecht
- Molecular, Cellular &Developmental Biology University of Colorado Boulder, Boulder, Colorado 80309
| | - Mark Winey
- Molecular, Cellular &Developmental Biology University of Colorado Boulder, Boulder, Colorado 80309
| | - Michael W Klymkowsky
- Molecular, Cellular &Developmental Biology University of Colorado Boulder, Boulder, Colorado 80309
| |
Collapse
|
22
|
LI YOUQIANG, WU YANYUAN, ABBATIELLO THOMASC, WU WARRENL, KIM JURI, SARKISSYAN MARIANNA, SARKISSYAN SUREN, CHUNG SEYUNGS, ELSHIMALI YAHYA, VADGAMA JAYDUTTV. Slug contributes to cancer progression by direct regulation of ERα signaling pathway. Int J Oncol 2015; 46:1461-72. [PMID: 25652255 PMCID: PMC4356499 DOI: 10.3892/ijo.2015.2878] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 11/24/2014] [Indexed: 02/07/2023] Open
Abstract
Hormone therapy targeting estrogen receptor α (ERα) is the most effective treatment for breast cancer. However, this treatment eventually fails as the tumor develops resistance. Although reduced expression of ER-α is a known contributing factor to endocrine resistance, the mechanism of ER-α downregulation in endocrine resistance is still not fully understood. The present study shows that Slug has an inverse relationship with ERα in breast and prostate cancer patient samples. Also the inhibition of Slug blocks mammary stem cell activity in primary mammary epithelial cells. We hypothesize that Slug may be a key transcription factor in the regulation of ERα expression. To understand the Slug-ERα signaling pathway, we employed resistant cell line MCF-TAMR (ERα relatively negative) derived from its parental MCF-7 (ERα positive) cell line and assessed changes in cell phenotype, activity and response to therapy. Conversely, we performed knockdown of Slug in the high-Slug expressing cell line MDA-MB-231 and assessed reversal of the mesenchymal phenotype. Microarray analysis showed that Slug is overexpressed in high grade breast and prostate cancer tissues. Additionally, Slug overexpression leads to drug resistance. Furthermore, we demonstrated that Slug binds directly to ERα promoter E-boxes and represses ERα expression. This resulted in decrease in epithelial-to-mesenchymal transition in cancer cells. These findings demonstrate that Slug, by regulation of ERα expression, contributes to tumor progression and could serve as an important target for cancer therapy.
Collapse
Affiliation(s)
- YOUQIANG LI
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- University of California at Los Angeles David Geffen School of Medicine, and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90024, USA
| | - YANYUAN WU
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- University of California at Los Angeles David Geffen School of Medicine, and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90024, USA
| | - THOMAS C. ABBATIELLO
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - WARREN L. WU
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - JU RI KIM
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - MARIANNA SARKISSYAN
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - SUREN SARKISSYAN
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - SEYUNG S. CHUNG
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - YAHYA ELSHIMALI
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - JAYDUTT V. VADGAMA
- Division of Cancer Research and Training, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- University of California at Los Angeles David Geffen School of Medicine, and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90024, USA
| |
Collapse
|
23
|
Savagner P. Epithelial-mesenchymal transitions: from cell plasticity to concept elasticity. Curr Top Dev Biol 2015; 112:273-300. [PMID: 25733143 DOI: 10.1016/bs.ctdb.2014.11.021] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a developmental cellular process occurring during early embryo development, including gastrulation and neural crest cell migration. It can be broken down in distinct functional steps: (1) loss of baso-apical polarization characterized by cytoskeleton, tight junctions, and hemidesmosomes remodeling; (2) individualization of cells, including a decrease in cell-cell adhesion forces, (3) emergence of motility, and (4) invasive properties, including passing through the subepithelial basement membrane. These phases occur in an uninterrupted process, without requiring mitosis, in an order and with a degree of completion dictated by the microenvironment. The whole process reflects the activation of specific transcription factor families, called EMT transcription factors. Several mechanisms can combine to induce EMT. Some are reversible, involving growth factors and cytokines and/or environmental signals including extracellular matrix and local physical conditions. Others are irreversible, such as genomic alterations during carcinoma progression, along a selective and irreversible clonal drift. In carcinomas, these signals can converge to initiate a metastable phenotype. In this state, similarly to activated keratinocytes during re-epithelialization, cells can initiate a cohort migration and engage into a transient and reversible EMT controlled by the local environment prior to efficient intravasation and metastasis. EMT transcription factors also participate in cancer progression by inducing apoptosis resistance and maintaining stem-like properties exposed in tumor recurrences. These properties, very important on a clinical point of view, are not intrinsically linked to EMT, but can share common pathways.
Collapse
Affiliation(s)
- Pierre Savagner
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U896, Institut régional du cancer Université Montpellier1, Montpellier, France.
| |
Collapse
|
24
|
Muñoz WA, Trainor PA. Neural crest cell evolution: how and when did a neural crest cell become a neural crest cell. Curr Top Dev Biol 2015; 111:3-26. [PMID: 25662256 DOI: 10.1016/bs.ctdb.2014.11.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
As vertebrates evolved from protochordates, they shifted to a more predatory lifestyle, and radiated and adapted to most niches of the planet. This process was largely facilitated by the generation of novel vertebrate head structures, which were derived from neural crest cells (NCC). The neural crest is a unique vertebrate cell population that is frequently termed the "fourth germ layer" because it forms in conjunction with the other germ layers and contributes to a diverse array of cell types and tissues including the craniofacial skeleton, the peripheral nervous system, and pigment cells among many other tissues and cell types. NCC are defined by their origin at the neural plate border, via an epithelial-to-mesenchymal transition (EMT), together with multipotency and polarized patterns of migration. These defining characteristics, which evolved independently in the germ layers of invertebrates, were subsequently co-opted through their gene regulatory networks to form NCC in vertebrates. Moreover, recent data suggest that the ability to undergo an EMT was one of the latter features co-opted by NCC. In this review, we discuss the potential origins of NCC and how they evolved to contribute to nearly all tissues and organs throughout the body, based on paleontological evidence together with an evaluation of the evolution of molecules involved in NCC development and their migratory cell paths.
Collapse
Affiliation(s)
- William A Muñoz
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, USA.
| |
Collapse
|
25
|
Bisson N, Wedlich D, Moss T. The p21-activated kinase Pak1 regulates induction and migration of the neural crest in Xenopus. Cell Cycle 2014; 11:1316-24. [DOI: 10.4161/cc.19685] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
26
|
Noisa P, Raivio T. Neural crest cells: From developmental biology to clinical interventions. ACTA ACUST UNITED AC 2014; 102:263-74. [DOI: 10.1002/bdrc.21074] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 08/22/2014] [Indexed: 12/14/2022]
Affiliation(s)
- Parinya Noisa
- Institute of Biomedicine/Physiology; University of Helsinki; Finland
- School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology; Nakhon Ratchasima Thailand
| | - Taneli Raivio
- Institute of Biomedicine/Physiology; University of Helsinki; Finland
- Children's Hospital, Helsinki University Central Hospital; Finland
| |
Collapse
|
27
|
Chibby functions in Xenopus ciliary assembly, embryonic development, and the regulation of gene expression. Dev Biol 2014; 395:287-98. [PMID: 25220153 DOI: 10.1016/j.ydbio.2014.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/03/2014] [Accepted: 09/05/2014] [Indexed: 12/21/2022]
Abstract
Wnt signaling and ciliogenesis are core features of embryonic development in a range of metazoans. Chibby (Cby), a basal-body associated protein, regulates β-catenin-mediated Wnt signaling in the mouse but not Drosophila. Here we present an analysis of Cby's embryonic expression and morphant phenotypes in Xenopus laevis. Cby RNA is supplied maternally, negatively regulated by Snail2 but not Twist1, preferentially expressed in the neuroectoderm, and regulates β-catenin-mediated gene expression. Reducing Cby levels reduced the density of multiciliated cells, the number of basal bodies per multiciliated cell, and the numbers of neural tube primary cilia; it also led to abnormal development of the neural crest, central nervous system, and pronephros, all defects that were rescued by a Cby-GFP chimera. Reduction of Cby led to an increase in Wnt8a and decreases in Gli2, Gli3, and Shh RNA levels. Many, but not all, morphant phenotypes were significantly reversed by the Wnt inhibitor SFRP2. These observations extend our understanding of Cby's role in mediating the network of interactions between ciliogenesis, signaling systems and tissue patterning.
Collapse
|
28
|
Smoczer C, Hooker L, Sachani SS, Crawford MJ. Microinjection manipulations in the elucidation of Xenopus brain development. Methods Mol Biol 2014; 1082:143-54. [PMID: 24048932 DOI: 10.1007/978-1-62703-655-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Microinjection has a long and distinguished history in Xenopus and has been used to introduce a surprisingly diverse array of agents into embryos by both intra- and intercellular means. In addition to nuclei, investigators have variously injected peptides, antibodies, biologically active chemicals, lineage markers, mRNA, DNA, morpholinos, and enzymes. While enumerating many of the different microinjection approaches that can be taken, we will focus upon the mechanical operations and options available to introduce mRNA, DNA, and morpholinos intracellularly into early stage embryos for the study of neurogenesis.
Collapse
|
29
|
Morosan-Puopolo G, Balakrishnan-Renuka A, Yusuf F, Chen J, Dai F, Zoidl G, Lüdtke THW, Kispert A, Theiss C, Abdelsabour-Khalaf M, Brand-Saberi B. Wnt11 is required for oriented migration of dermogenic progenitor cells from the dorsomedial lip of the avian dermomyotome. PLoS One 2014; 9:e92679. [PMID: 24671096 PMCID: PMC3966816 DOI: 10.1371/journal.pone.0092679] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 02/24/2014] [Indexed: 12/23/2022] Open
Abstract
The embryonic origin of the dermis in vertebrates can be traced back to the dermomyotome of the somites, the lateral plate mesoderm and the neural crest. The dermal precursors directly overlying the neural tube display a unique dense arrangement and are the first to induce skin appendage formation in vertebrate embryos. These dermal precursor cells have been shown to derive from the dorsomedial lip of the dermomyotome (DML). Based on its expression pattern in the DML, Wnt11 is a candidate regulator of dorsal dermis formation. Using EGFP-based cell labelling and time-lapse imaging, we show that the Wnt11 expressing DML is the source of the dense dorsal dermis. Loss-of-function studies in chicken embryos show that Wnt11 is indeed essential for the formation of dense dermis competent to support cutaneous appendage formation. Our findings show that dermogenic progenitors cannot leave the DML to form dense dorsal dermis following Wnt11 silencing. No alterations were noticeable in the patterning or in the epithelial state of the dermomyotome including the DML. Furthermore, we show that Wnt11 expression is regulated in a manner similar to the previously described early dermal marker cDermo-1. The analysis of Wnt11 mutant mice exhibits an underdeveloped dorsal dermis and strongly supports our gene silencing data in chicken embryos. We conclude that Wnt11 is required for dense dermis and subsequent cutaneous appendage formation, by influencing the cell fate decision of the cells in the DML.
Collapse
Affiliation(s)
- Gabriela Morosan-Puopolo
- Department of Anatomy and Molecular Embryology, Ruhr-University of Bochum, Bochum, Germany
- Department of Molecular Embryology, Freiburg University, Freiburg, Germany
- Faculty of Biology, Freiburg University, Freiburg, Germany
| | - Ajeesh Balakrishnan-Renuka
- Department of Anatomy and Molecular Embryology, Ruhr-University of Bochum, Bochum, Germany
- Department of Molecular Embryology, Freiburg University, Freiburg, Germany
- Faculty of Biology, Freiburg University, Freiburg, Germany
| | - Faisal Yusuf
- Department of Anatomy and Molecular Embryology, Ruhr-University of Bochum, Bochum, Germany
| | - Jingchen Chen
- Department of Anatomy and Molecular Embryology, Ruhr-University of Bochum, Bochum, Germany
| | - Fangping Dai
- Department of Molecular Embryology, Freiburg University, Freiburg, Germany
| | - Georg Zoidl
- Department of Anatomy and Molecular Embryology, Ruhr-University of Bochum, Bochum, Germany
| | - Timo H.-W. Lüdtke
- Institute for Molecular Biology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Andreas Kispert
- Institute for Molecular Biology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Carsten Theiss
- Department of Anatomy and Molecular Embryology, Ruhr-University of Bochum, Bochum, Germany
| | - Mohammed Abdelsabour-Khalaf
- Department of Anatomy and Molecular Embryology, Ruhr-University of Bochum, Bochum, Germany
- Faculty of Biology, Freiburg University, Freiburg, Germany
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr-University of Bochum, Bochum, Germany
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, Albert-Ludwigs University, Freiburg, Germany
| |
Collapse
|
30
|
Nassour M, Idoux-Gillet Y, Selmi A, Côme C, Faraldo MLM, Deugnier MA, Savagner P. Slug controls stem/progenitor cell growth dynamics during mammary gland morphogenesis. PLoS One 2012; 7:e53498. [PMID: 23300933 PMCID: PMC3531397 DOI: 10.1371/journal.pone.0053498] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 11/29/2012] [Indexed: 12/22/2022] Open
Abstract
Background Morphogenesis results from the coordination of distinct cell signaling pathways controlling migration, differentiation, apoptosis, and proliferation, along stem/progenitor cell dynamics. To decipher this puzzle, we focused on epithelial-mesenchymal transition (EMT) “master genes”. EMT has emerged as a unifying concept, involving cell-cell adhesion, migration and apoptotic pathways. EMT also appears to mingle with stemness. However, very little is known on the physiological role and relevance of EMT master-genes. We addressed this question during mammary morphogenesis. Recently, a link between Slug/Snai2 and stemness has been described in mammary epithelial cells, but EMT master genes actual localization, role and targets during mammary gland morphogenesis are not known and we focused on this basic question. Methodology/Principal Findings Using a Slug–lacZ transgenic model and immunolocalization, we located Slug in a distinct subpopulation covering about 10–20% basal cap and duct cells, mostly cycling cells, coexpressed with basal markers P-cadherin, CK5 and CD49f. During puberty, Slug-deficient mammary epithelium exhibited a delayed development after transplantation, contained less cycling cells, and overexpressed CK8/18, ER, GATA3 and BMI1 genes, linked to luminal lineage. Other EMT master genes were overexpressed, suggesting compensation mechanisms. Gain/loss-of-function in vitro experiments confirmed Slug control of mammary epithelial cell luminal differentiation and proliferation. In addition, they showed that Slug enhances specifically clonal mammosphere emergence and growth, cell motility, and represses apoptosis. Strikingly, Slug-deprived mammary epithelial cells lost their potential to generate secondary clonal mammospheres. Conclusions/Significance We conclude that Slug pathway controls the growth dynamics of a subpopulation of cycling progenitor basal cells during mammary morphogenesis. Overall, our data better define a key mechanism coordinating cell lineage dynamics and morphogenesis, and provide physiological relevance to broadening EMT pathways.
Collapse
Affiliation(s)
- Mayssa Nassour
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherche Médicale U896, Université Montpellier, Centre Régional de Lutte contre le Cancer Val d’Aurelle-Paul Lamarque, Montpellier, France
| | - Ysia Idoux-Gillet
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherche Médicale U896, Université Montpellier, Centre Régional de Lutte contre le Cancer Val d’Aurelle-Paul Lamarque, Montpellier, France
| | - Abdelkader Selmi
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherche Médicale U896, Université Montpellier, Centre Régional de Lutte contre le Cancer Val d’Aurelle-Paul Lamarque, Montpellier, France
| | - Christophe Côme
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherche Médicale U896, Université Montpellier, Centre Régional de Lutte contre le Cancer Val d’Aurelle-Paul Lamarque, Montpellier, France
| | | | - Marie-Ange Deugnier
- Institut Curie UMR144, Centre National de la Recherche Scientifique, Paris, France
| | - Pierre Savagner
- Institut de Recherche en Cancérologie de Montpellier, Institut National de la Santé et de la Recherche Médicale U896, Université Montpellier, Centre Régional de Lutte contre le Cancer Val d’Aurelle-Paul Lamarque, Montpellier, France
- * E-mail:
| |
Collapse
|
31
|
Medeiros DM. The evolution of the neural crest: new perspectives from lamprey and invertebrate neural crest-like cells. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:1-15. [PMID: 23799627 DOI: 10.1002/wdev.85] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The neural crest is an embryonic cell population that gives rise to an array of tissues and structures in adult vertebrates including most of the head skeleton. Because neural crest cells (NCCs), and many of their derivatives, are unique to vertebrates, the evolution of the neural crest is thought to have potentiated vertebrate origins and diversification. However, the lack of clear NCC homologs in invertebrate chordates has made it difficult to reconstruct the evolutionary history of modern NCCs. In this review, the development of NCCs in the basal jawless vertebrate, lamprey, is compared with the development of neural crest-like cells in a range of invertebrates to deduce features of the first NCCs and their evolutionary precursors. These comparisons demonstrate that most of the defining attributes of NCCs are widespread features of invertebrate embryonic ectoderm. In addition, they suggest ancient origins for the neural border domain and chondroid skeletal tissue in the first bilaterian, and show that NCCs must have evolved in a chordate with an unduplicated invertebrate-type genome. On the basis of these observations, a stepwise model for the evolution of NCCs involving heterotopic and heterochronic activation of ancient ectodermal gene programs and new responsiveness to preexisting inducing signals is proposed. In light of the phylogenetic distribution of neural crest-like cells, the deep homology of developmental gene networks, and the central role of evolutionary loss in deuterostome evolution, this article concludes with suggestions for future studies in a broad range of bilaterians to test key aspects of this model. WIREs Dev Biol 2013, 2:1-15. doi: 10.1002/wdev.85 For further resources related to this article, please visit the WIREs website.
Collapse
|
32
|
Stuhlmiller TJ, García-Castro MI. Current perspectives of the signaling pathways directing neural crest induction. Cell Mol Life Sci 2012; 69:3715-37. [PMID: 22547091 PMCID: PMC3478512 DOI: 10.1007/s00018-012-0991-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 03/12/2012] [Accepted: 04/02/2012] [Indexed: 01/05/2023]
Abstract
The neural crest is a migratory population of embryonic cells with a tremendous potential to differentiate and contribute to nearly every organ system in the adult body. Over the past two decades, an incredible amount of research has given us a reasonable understanding of how these cells are generated. Neural crest induction involves the combinatorial input of multiple signaling pathways and transcription factors, and is thought to occur in two phases from gastrulation to neurulation. In the first phase, FGF and Wnt signaling induce NC progenitors at the border of the neural plate, activating the expression of members of the Msx, Pax, and Zic families, among others. In the second phase, BMP, Wnt, and Notch signaling maintain these progenitors and bring about the expression of definitive NC markers including Snail2, FoxD3, and Sox9/10. In recent years, additional signaling molecules and modulators of these pathways have been uncovered, creating an increasingly complex regulatory network. In this work, we provide a comprehensive review of the major signaling pathways that participate in neural crest induction, with a focus on recent developments and current perspectives. We provide a simplified model of early neural crest development and stress similarities and differences between four major model organisms: Xenopus, chick, zebrafish, and mouse.
Collapse
Affiliation(s)
- Timothy J Stuhlmiller
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520-8103, USA
| | | |
Collapse
|
33
|
Shi J, Zhang H, Dowell RD, Klymkowsky MW. sizzled function and secreted factor network dynamics. Biol Open 2012; 1:286-94. [PMID: 23213419 PMCID: PMC3507283 DOI: 10.1242/bio.2012019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Studies on the role of the E-box binding transcription factor Snail2 (Slug) in the induction of neural crest by mesoderm (Shi et al., 2011) revealed an unexpected increase in the level of sizzled RNA in the dorsolateral mesodermal zone (DMLZ) of morphant Xenopus embryos. sizzled encodes a secreted protein with both Wnt and BMP inhibitor activities. Morpholino-mediated down-regulation of sizzled expression in one cell of two cell embryos or the C2/C3 blastomeres of 32-cell embryos, which give rise to the DLMZ, revealed decreased expression of the mesodermal marker brachyury and subsequent defects in neural crest induction, pronephros formation, and muscle patterning. Loss of sizzled expression led to decreases in RNAs encoding the secreted Wnt inhibitor SFRP2 and the secreted BMP inhibitor Noggin; the sizzled morphant phenotype could be rescued by co-injection of RNAs encoding Noggin and either SFRP2 or Dickkopf (a mechanistically distinct Wnt inhibitor). Together, these observations reveal that sizzled, in addition to its established role in dorsal-ventral patterning, is also part of a dynamic BMP and Wnt signaling network involved in both mesodermal patterning and neural crest induction.
Collapse
Affiliation(s)
- Jianli Shi
- Molecular, Cellular and Developmental Biology, University of Colorado , Boulder, CO 80309-0347 , USA
| | | | | | | |
Collapse
|