Ectoderm: Difference between revisions

From Embryology
mNo edit summary
mNo edit summary
Line 21: Line 21:
|-bgcolor="F5FAFF"  
|-bgcolor="F5FAFF"  
|
|
* '''Notch signaling in the division of germ layers in bilaterian embryos'''{{#pmid:29940277|PMID29940277}}
* '''Notch signaling in the division of germ layers in bilaterian embryos'''{{#pmid:29940277|PMID29940277}} "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."
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.


* '''A molecular atlas of the developing ectoderm defines neural, neural crest, placode, and nonneural progenitor identity in vertebrates'''{{#pmid:29049289|PMID29049289}} "During vertebrate neurulation, the embryonic ectoderm is patterned into lineage progenitors for neural plate, neural crest, placodes and epidermis. Here, we use Xenopus laevis embryos to analyze the spatial and temporal transcriptome of distinct ectodermal domains in the course of neurulation, during the establishment of cell lineages. In order to define the transcriptome of small groups of cells from a single germ layer and to retain spatial information, dorsal and ventral ectoderm was subdivided along the anterior-posterior and medial-lateral axes by microdissections. Principal component analysis on the transcriptomes of these ectoderm fragments primarily identifies embryonic axes and temporal dynamics. This provides a genetic code to define positional information of any ectoderm sample along the anterior-posterior and dorsal-ventral axes directly from its transcriptome. In parallel, we use nonnegative matrix factorization to predict enhanced gene expression maps onto early and mid-neurula embryos, and specific signatures for each ectoderm area. The clustering of spatial and temporal datasets allowed detection of multiple biologically relevant groups (e.g., Wnt signaling, neural crest development, sensory placode specification, ciliogenesis, germ layer specification). We provide an interactive network interface, EctoMap, for exploring synexpression relationships among genes expressed in the neurula, and suggest several strategies to use this comprehensive dataset to address questions in developmental biology as well as stem cell or cancer research."
* '''A molecular atlas of the developing ectoderm defines neural, neural crest, placode, and nonneural progenitor identity in vertebrates'''{{#pmid:29049289|PMID29049289}} "During vertebrate neurulation, the embryonic ectoderm is patterned into lineage progenitors for neural plate, neural crest, placodes and epidermis. Here, we use Xenopus laevis embryos to analyze the spatial and temporal transcriptome of distinct ectodermal domains in the course of neurulation, during the establishment of cell lineages. In order to define the transcriptome of small groups of cells from a single germ layer and to retain spatial information, dorsal and ventral ectoderm was subdivided along the anterior-posterior and medial-lateral axes by microdissections. Principal component analysis on the transcriptomes of these ectoderm fragments primarily identifies embryonic axes and temporal dynamics. This provides a genetic code to define positional information of any ectoderm sample along the anterior-posterior and dorsal-ventral axes directly from its transcriptome. In parallel, we use nonnegative matrix factorization to predict enhanced gene expression maps onto early and mid-neurula embryos, and specific signatures for each ectoderm area. The clustering of spatial and temporal datasets allowed detection of multiple biologically relevant groups (e.g., Wnt signaling, neural crest development, sensory placode specification, ciliogenesis, germ layer specification). We provide an interactive network interface, EctoMap, for exploring synexpression relationships among genes expressed in the neurula, and suggest several strategies to use this comprehensive dataset to address questions in developmental biology as well as stem cell or cancer research."

Revision as of 19:59, 11 May 2019

Embryology - 28 Mar 2024    Facebook link Pinterest link Twitter link  Expand to Translate  
Google Translate - select your language from the list shown below (this will open a new external page)

العربية | català | 中文 | 中國傳統的 | français | Deutsche | עִברִית | हिंदी | bahasa Indonesia | italiano | 日本語 | 한국어 | မြန်မာ | Pilipino | Polskie | português | ਪੰਜਾਬੀ ਦੇ | Română | русский | Español | Swahili | Svensk | ไทย | Türkçe | اردو | ייִדיש | Tiếng Việt    These external translations are automated and may not be accurate. (More? About Translations)

Introduction

The trilaminar embryo

The top layer of the early trilaminar embryo germ layers (ectoderm, mesoderm and endoderm) formed by gastrulation. The ectoderm can be though of as having 4 early regions: neural plate, neural crest, surface ectoderm and placodes. Note that there are other pages describing neural (central nervous system; brain and spinal cord) and neural crest (peripheral nervous system; sensory and sympathetic ganglia). Epidermis (integumentary, skin contribution) development will be briefly mentioned due to its ectoderm origin.


The ectoderm contributes to the human embryo:

  1. nervous system, both central (neural plate) and peripheral (neural crest).
  2. epidermis of the skin (surface ectoderm) and pigmented cells (neural crest).
  3. head regions that contribution sensory and endocrine structures (placodes).
  4. adrenal gland medullary cells (neural crest).


Ectoderm Links: gastrulation | endoderm | mesoderm | ectoderm | Lecture - Ectoderm Development | Lecture - Neural Development | Lecture - Integumentary Development | neural | neural crest | integumentary | placode | Category:Ectoderm

Some Recent Findings

Zebrafish ectodermal patterning model[1]
  • Notch signaling in the division of germ layers in bilaterian embryos[2] "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."
  • A molecular atlas of the developing ectoderm defines neural, neural crest, placode, and nonneural progenitor identity in vertebrates[3] "During vertebrate neurulation, the embryonic ectoderm is patterned into lineage progenitors for neural plate, neural crest, placodes and epidermis. Here, we use Xenopus laevis embryos to analyze the spatial and temporal transcriptome of distinct ectodermal domains in the course of neurulation, during the establishment of cell lineages. In order to define the transcriptome of small groups of cells from a single germ layer and to retain spatial information, dorsal and ventral ectoderm was subdivided along the anterior-posterior and medial-lateral axes by microdissections. Principal component analysis on the transcriptomes of these ectoderm fragments primarily identifies embryonic axes and temporal dynamics. This provides a genetic code to define positional information of any ectoderm sample along the anterior-posterior and dorsal-ventral axes directly from its transcriptome. In parallel, we use nonnegative matrix factorization to predict enhanced gene expression maps onto early and mid-neurula embryos, and specific signatures for each ectoderm area. The clustering of spatial and temporal datasets allowed detection of multiple biologically relevant groups (e.g., Wnt signaling, neural crest development, sensory placode specification, ciliogenesis, germ layer specification). We provide an interactive network interface, EctoMap, for exploring synexpression relationships among genes expressed in the neurula, and suggest several strategies to use this comprehensive dataset to address questions in developmental biology as well as stem cell or cancer research."
  • Meis transcription factor maintains the neurogenic ectoderm and regulates the anterior-posterior patterning in embryos of a sea urchin, Hemicentrotus pulcherrimus[4] "Precise body axis formation is an essential step in the development of multicellular organisms, for most of which the molecular gradient and/or specifically biased localization of cell-fate determinants in eggs play important roles. In sea urchins, however, any biased proteins and mRNAs have not yet been identified in the egg except for vegetal cortex molecules, suggesting that sea urchin development is mostly regulated by uniformly distributed maternal molecules with contributions to axis formation that are not well characterized. Here, we describe that the maternal Meis transcription factor regulates anterior-posterior axis formation through maintenance of the most anterior territory in embryos of a sea urchin, Hemicentrotus pulcherrimus. Loss-of-function experiments revealed that Meis is intrinsically required for maintenance of the anterior neuroectoderm specifier foxQ2 after hatching and, consequently, the morphant lost anterior neuroectoderm characteristics. In addition, the expression patterns of univin and VEGF, the lateral ectoderm markers, and the mesenchyme-cell pattern shifted toward the anterior side in Meis morphants more than they did in control embryos, indicating that Meis contributes to the precise anteroposterior patterning by regulating the anterior neuroectodermal fate.
More recent papers  
Mark Hill.jpg
PubMed logo.gif

This table allows an automated computer search of the external PubMed database using the listed "Search term" text link.

  • This search now requires a manual link as the original PubMed extension has been disabled.
  • The displayed list of references do not reflect any editorial selection of material based on content or relevance.
  • References also appear on this list based upon the date of the actual page viewing.


References listed on the rest of the content page and the associated discussion page (listed under the publication year sub-headings) do include some editorial selection based upon both relevance and availability.

More? References | Discussion Page | Journal Searches | 2019 References | 2020 References

Search term: Ectoderm Development | Images | Ectoderm

Older papers  
These papers originally appeared in the Some Recent Findings table, but as that list grew in length have now been shuffled down to this collapsible table.

See also the Discussion Page for other references listed by year and References on this current page.

  • A BMP regulatory network controls ectodermal cell fate decisions at the neural plate border.[1] "During ectodermal patterning the neural crest and preplacodal ectoderm are specified in adjacent domains at the neural plate border. BMP signalling is required for specification of both tissues, but how it is spatially and temporally regulated to achieve this is not understood. Here, using a transgenic zebrafish BMP reporter line in conjunction with double-fluorescent in situ hybridisation, we show that, at the beginning of neurulation, the ventral-to-dorsal gradient of BMP activity evolves into two distinct domains at the neural plate border: one coinciding with the neural crest and the other abutting the epidermis. In between is a region devoid of BMP activity, which is specified as the preplacodal ectoderm. We identify the ligands required for these domains of BMP activity. We show that the BMP-interacting protein Crossveinless 2 is expressed in the BMP activity domains and is under the control of BMP signalling. We establish that Crossveinless 2 functions at this time in a positive-feedback loop to locally enhance BMP activity, and show that it is required for neural crest fate. We further demonstrate that the Distal-less transcription factors Dlx3b and Dlx4b, which are expressed in the preplacodal ectoderm, are required for the expression of a cell-autonomous BMP inhibitor, Bambi-b, which can explain the specific absence of BMP activity in the preplacodal ectoderm. Taken together, our data define a BMP regulatory network that controls cell fate decisions at the neural plate border."
<html5media width='316' height='500' image="http://php.med.unsw.edu.au/embryology/images/a/a6/Neuralplate_001_icon.jpg">File:Neuralplate_001.mp4</html5media> This animation shows the embryonic disc from the amniotic cavity side ectoderm (human week 3) onward.
  • Neural plate (blue) in central region of the ectoderm.
  • Primitive streak extending from the bottom of the neural plate.
  • Epidermis primordia (white) region surrounding the neural plate. Integumentary (skin) development will be briefly covered here.
  • Buccopharnygeal and Cloacal membranes (circular region above and below the neural plate).


Stage10 neural sm.jpg Stage10 SEM1.jpg

Objectives

  • Understanding of events during the third and fourth week of development
  • Understanding the process of notochord formation
  • Understanding the process of early neural development
  • Brief understanding of neural crest formation
  • Brief understanding of epidermis formation
  • Understanding of the adult components derived from ectoderm
  • Brief understanding of early neural abnormalities

Textbook References

  • Human Embryology (3rd ed.) Chapter 5 p107-125
  • The Developing Human: Clinically Oriented Embryology (6th ed.)
  • Moore and Persaud Chapter 18 p451-489
  • Essentials of Human Embryology Larson Chapter 5 p69-79
  • Before We Are Born (5th ed.) Moore and Persaud Chapter 19 p423-458

Overview

ectoderm
The nested tables below show an overview of the different ectoderm-derived tissues (links go to topic pages).
Template Only - content to be added. Mesoderm has been completed.
embryonic
neural plate  
neural tube
central nervous system - brain, spinal cord
neural crest 
neural crest
placode
otic - inner ear cochlea, vestibular optic - lens nasal anterior pituitary
surface ectoderm  
integumentary - epithelium, glands
extra-embryonic
placental membrane
amnion
Overview: Ectoderm | Mesoderm | Endoderm Layers: ectoderm | mesoderm | endoderm


Notochord

  • forms initially as the Axial Process, a hollow tube which extends from the primitive pit , cranially to the oral membrane
  • the axial process then allow transient communication between the amnion and the yolk sac through the neuroenteric canal.
  • the axial process then merges with the Endodermal layer to form the Notochordal Plate.
  • the notochordal plate then rises back into the Mesodermal layer as a solid column of cells which is the Notochord.

Ectoderm

  • 2 parts
  • midline neural plate
    • columnar
  • lateral surface ectoderm
    • cuboidal
    • sensory placodes
    • epidermis of skin, hair, glands, anterior pituitary, teeth enamel

Neural Plate

Neuralplate cartoon.png
Stage 10 neural groove to tube
<html5media height="500" width="316">File:Neuralplate_001.mp4</html5media>

Click Here to play on mobile device

Development of the neural plate region at the embryonic disc stage.

Dorsal view of the embryonic disc from the amniotic cavity side showing the ectoderm with the central region developing into the neural plate.

The neural plate extends from buccopharyngeal membrane to primitive node and forms above the notochord and paraxial mesoderm.The neuroectodermal cells form a broad brain plate and narrower spinal cord region.

Three specific regions medial to lateral can also be identified: midline region floor plate, neural plate, edge of neural plate neural crest

  • extends from buccopharyngeal membrane to primitive node
  • forms above notochord and paraxial mesoderm
  • neuroectodermal cells
    • broad brain plate
    • narrower spinal cord
  • 3 components form: floor plate, neural plate, neural crest

Neural Determination- neuronal populations are specified before plate folds

  • signals from notochord and mesoderm - secrete noggin, chordin,follistatin
    • all factors bind BMP-4 an inhibitor of neuralation
    • bone morphogenic protein acts through membrane receptor
  • lateral inhibition generates at spinal cord level 3 strips of cells
  • expression of delta inhibits nearby cells, which express notch receptor, from becoming neurons
  • Delta-Notch inetraction- generates Neural strips

Neural Groove

<html5media height="480" width="480">File:Neuraltube_001.mp4</html5media>

Click Here to play on mobile device

This animation of early neural development from week 3 onward shows the neural groove fusing to form the neural tube.

View - Dorsolateral of the whole early embryo and yolk sac. Cranial (head) to top and caudal (tail) to bottom. Yolk sac is shown to the left.

Beginning with the neural groove initially fusing at the level of the 4th somite to form the neural tube and closing in both directions to leave 2 openings or neuropores: a cranial neuropore (anterior neuropore) and a caudal neuropore (posterior neuropore).

The animation also shows as the embryo grows and folds it increases in size relative to the initial yolk sac. Note also the increasing number of somites over time.

  • forms in the midline of the neural plate (day 18-19)
  • either side of which are the neural folds which continues to deepen until about week 4
  • neural folds begins to fuse, beginning at 4th somite level

Neural Tube

Stage 12 caudal neuropore
  • the neural tube forms the brain and spinal cord
  • fusion of neural groove extends rostrally and caudally
  • begins at the level of 4th somite
  • closes neural groove "zips up" in some species.
    • humans appear to close at multiple points along the tube.
  • leaves 2 openings at either end - Neuropores
    • cranial neuropore closes before caudal

Failure for the neural tube to close correctly or completely results in a neural tube defect.

Secondary Neuralation

<html5media height="300" width="320">File:Secondary_neurulation_01.mp4</html5media>

Click Here to play on mobile device

This animation shows the early developmental process often described as secondary neurulation.

Red - site of secondary neurulation | Blue - neural tube

  • caudal end of neural tube formed by secondary neuralation
  • develops from primitive streak region
  • solid cord canalized by extension of neural canal
  • mesodermal caudal eminence
Links: MP4 version | Neural System Development


Neural Crest

  • a population of cells at the edge of the neural plate that lie dorsally when the neural tube fuses
    • dorsal to the neural tube, as a pair of streaks
    • pluripotential, forms many different types of cells
    • cells migrate throughout the embryo
    • studied by quail-chick chimeras
    • transplanted quail cells have obvious nucleoli compared with chicken

Neural Crest Derivitives

  • dorsal root ganglia
  • autonomic ganglia
  • adrenal medulla
  • drg sheath cells, glia
  • pia-arachnoid sheath
  • skin melanocytes
  • connective tissue of cardiac outflow
  • thyroid parafollicular cells
  • craniofacial skeleton
  • teeth odontoblasts
Links: Neural Crest Development

Ectodermal Placodes

  • Specialized ectodermal "patches" in the head region
  • Contribute sensory structures - otic placode (otocyst), nasal placode, lens placode
  • Contribute teeth

Human Neuralation - Early Stages

The stages below refer to specific Carneigie stages of development.

  • Carnegie stage 8 (about 18 postovulatory days) neural groove and folds are first seen
  • Carnegie stage 9 the three main divisions of the brain, which are not cerebral vesicles, can be distinguished while the neural groove is still completely open.
  • Carnegie stage 10 (two days later) neural folds begin to fuse near the junction between brain and spinal cord, when neural crest cells are arising mainly from the neural ectoderm
  • Carnegie stage 11 (about 24 days) the rostral (or cephalic) neuropore closes within a few hours; closure is bidirectional, it takes place from the dorsal and terminal lips and may occur in several areas simultaneously. The two lips, however, behave differently.
  • Carnegie stage 12 (about 26 days) The caudal neuropore takes a day to close.
    • the level of final closure is approximately at future somitic pair 31
    • corresponds to the level of sacral vertebra 2
  • Carnegie stage 13 (4 weeks) the neural tube is normally completely closed.

Secondary neurulation begins at stage 12 - is the differentiation of the caudal part of the neural tube from the caudal eminence (or end-bud) without the intermediate phase of a neural plate.

Above text modified from[5]

Movies

Neural Development
Neuralplate 001 icon.jpg
 ‎‎Neural Plate
Page | Play
Neuraltube 001 icon.jpg
 ‎‎Neural Tube
Page | Play
Secondary neurulation 01 icon.jpg
 ‎‎Secondary Neurulation
Page | Play
Stage13-CNS-icon.jpg
 ‎‎Stage 13 Neural
Page | Play
Stage13 MRI 3D02 icon.jpg
 ‎‎Embryo CNS
Page | Play
Mouse neural tube 01 movie icon.jpg
 ‎‎Neural Tube Close
Page | Play
Stage16 MRI 3D02 icon.jpg
 ‎‎Embryo CNS
Page | Play
Stage16 MRI S01 icon.jpg
 ‎‎Embryo Stage 16
Page | Play
Human embryo tomography Carnegie stage 17.jpg
 ‎‎Stage 17 Embryo
Page | Play
Stage22-CNS-icon.jpg
 ‎‎Stage 22 Neural
Page | Play
Stage23 MRI 3D02 icon.jpg
 ‎‎Embryo CNS
Page | Play
Stage23 MRI S01 icon.jpg
 ‎‎Sagittal Head
Page | Play
Abnormalities Ultrasound
Brain fissure development 03.jpg
 ‎‎Sylvian Fissure
Page | Play
Adult human brain tomography.jpg
 ‎‎Adult Brain
Page | Play
US Dandy-Walker 01.jpg
 ‎‎Dandy-Walker
Page | Play
US Spina bifida GA19week.jpg
 ‎‎Spina Bifida
Page | Play
Fetal-Brain-icon.jpg
 ‎‎Neural
Page | Play


References

  1. 1.0 1.1 Reichert S, Randall RA & Hill CS. (2013). A BMP regulatory network controls ectodermal cell fate decisions at the neural plate border. Development , 140, 4435-44. PMID: 24089471 DOI.
  2. Favarolo MB & López SL. (2018). Notch signaling in the division of germ layers in bilaterian embryos. Mech. Dev. , 154, 122-144. PMID: 29940277 DOI.
  3. Plouhinec JL, Medina-Ruiz S, Borday C, Bernard E, Vert JP, Eisen MB, Harland RM & Monsoro-Burq AH. (2017). A molecular atlas of the developing ectoderm defines neural, neural crest, placode, and nonneural progenitor identity in vertebrates. PLoS Biol. , 15, e2004045. PMID: 29049289 DOI.
  4. Yaguchi J, Yamazaki A & Yaguchi S. (2018). Meis transcription factor maintains the neurogenic ectoderm and regulates the anterior-posterior patterning in embryos of a sea urchin, Hemicentrotus pulcherrimus. Dev. Biol. , 444, 1-8. PMID: 30266259 DOI.
  5. <pubmed>8005032</pubmed>

Textbooks

Online Textbooks

Search

Search NLM Online Textbooks: "Ectoderm" : Developmental Biology | The Cell- A molecular Approach | Molecular Biology of the Cell

Reviews

Kutejova E, Briscoe J & Kicheva A. (2009). Temporal dynamics of patterning by morphogen gradients. Curr. Opin. Genet. Dev. , 19, 315-22. PMID: 19596567 DOI.

Charron F & Tessier-Lavigne M. (2007). The Hedgehog, TGF-beta/BMP and Wnt families of morphogens in axon guidance. Adv. Exp. Med. Biol. , 621, 116-33. PMID: 18269215 DOI.

Charron F & Tessier-Lavigne M. (2005). Novel brain wiring functions for classical morphogens: a role as graded positional cues in axon guidance. Development , 132, 2251-62. PMID: 15857918 DOI.


External Links

External Links Notice - The dynamic nature of the internet may mean that some of these listed links may no longer function. If the link no longer works search the web with the link text or name. Links to any external commercial sites are provided for information purposes only and should never be considered an endorsement. UNSW Embryology is provided as an educational resource with no clinical information or commercial affiliation.


Take the Quiz

1 Ectoderm refers only to the neural plate region of the trilaminar embryo

true
false

2 The central nervous system forms in the sequence:

norochord to neural plate to neural tube
neural tube to neural plate to neural groove
neural plate to neural groove to neural tube
neural plate to neural crest to neural zone

3 The neural plate is narrower at the caudal (tail) end and therefore closes earlier than the broad cranial (head) end.

true
false

4 The correct sequence from cranial to caudal of the secondary brain vesicles is:

prosencephalon, mesencephalon, metencephalon, myelencephalon, rhombencephalon
telencephalon, diencephalon, metencephalon, mesencephalon, myelencephalon
telencephalon, diencephalon, mesencephalon, metencephalon, myelencephalon
prosencephalon, diencephalon, mesencephalon, myelencephalon, metencephalon

Glossary Links

Glossary: A | B | C | D | E | F | G | H | I | J | K | L | M | N | O | P | Q | R | S | T | U | V | W | X | Y | Z | Numbers | Symbols | Term Link

Cite this page: Hill, M.A. (2024, March 28) Embryology Ectoderm. Retrieved from https://embryology.med.unsw.edu.au/embryology/index.php/Ectoderm

What Links Here?
© Dr Mark Hill 2024, UNSW Embryology ISBN: 978 0 7334 2609 4 - UNSW CRICOS Provider Code No. 00098G