• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/255

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

255 Cards in this Set

  • Front
  • Back
. You dissect the animal and vegetal portions of a Xenopus blastula stage embryo and recombine them in an explant assay. After incubating for several days, describe the cell identity portions derived from the animal and vegetal tissues.
The animal cap will be converted to mesoderm. The endoderm will give rise to endodermal derivatives only.
You experimentally, deplete embryos for b-catenin, and repeat the assay from #1 (don't worry about possible secondary effects from losing cadherin-based cell adhesion). What will be the cell identity of the animal cap and vegetal portions? Why?
A loss of Beta-catenin in the vegetal endoderm means there will be no dorsalizing signal secreted from the endoderm to the mesoderm. This means that the vegetal region of the embryo will give rise only to ventral endodermal tissue types. There is a caveat / trick to this question. The beta-catenin would have to be inhibited before the end of the first cell division and would more realistically have to be done in the oocyte (looking at the lecture slides, compare phenotype of beta-catenin depleted embryos from oocytes verses 1 hour after fertilization).
List two of the general mesodermal inducing factors?
Any of the following: Vg1 and VegT, TGF-beta/nodal related genes (Xnr, activin). Remember that nodal signaling is a subset of the TGF-beta superfamily.
What is the primary purpose of the Nieuwkoop center?
To induce the organizer cells (remember that organizer cells are mesodermal, but are induced by the vegetal-specific Nieuwkoop center. The combination of beta-catenin and Vg1/nodal also directly induces gene expression of some of the BMP antagonists expressed by the organizer cells. Remember, B-catenin induces dorsal tissue identity in both the mesoderm and ectoderm (becomes neural) but does not directly induce mesoderm.
Describe the role of Dishevelled in the Wnt pathway. Can it be used to determine the difference between canonical and non-canonical signaling?
Wnt ligand binding to frizzled activates disheveled (Dsh). In canonical signaling, Dsh causes the stabilization of Beta catenin and the activation of Wnt signaling. Dsh is also involved in non-canonical Wnt signaling, which is indendent of B-catenin stabilization. So Dsh cannot be used to distinguish different types of Wnt signaling. In contrast, a dominant negative GSK3B construct would result in activation of beta-catenin. So comparing experiments in which each pathway was disrupted allowed Wallingford et al to conclude that the planar cell polarity (PCP) pathway was required for convergent extension during frog gastrulation.
List the four morphogenetic processes involved in Xenopus gastrulation. For each, write a sentence that summarizes the specific area of gastrulation that each is involved with.
A) Invagination
B) Involution
C) Convergent extension
D) Epiboly
Invagination, through the bottle cells, causes the emergence of the blastopore lip
As the marginal cells reach the dorsal lip, they involute over the inner layer of the animal cap cells.
The mesoderm undergoes convergent extension, which acts as a driving force to push the mesoderm further into the embryo (remember not all mesoderm undergoes this process (NIMZ = head mesoderm and also pharyngeal endoderm)..
The animal cap (and some of the marginal zone that does not involute) then cover the entire outer layer of the embryo by epiboly; this covers the vegetal cells (think of pulling a cap down from your head to completely cover your face).
What is one major molecules that is secreted by the dorsal lip (organizer region) of Xenopus embryos. What is its biochemical function? What happens if it is also expressed in the ventral marginal zone?
Chordin, Noggin, Follistatin are all BMP inhibitors. By blocking BMP signals, the ectoderm is induced to become neural tissue and also causes mesoderm to become dorsal. By expressing these molecules at the ventral marginal zone (instead of their normal dorsal region), a second axis is formed, and you get a twin embryo.
You experimentally generate a Keller explant. What is this? If you culture this in isolation, what structures do you predict will be specified? Will all the tissues undergo convergent extension?
This is an explant that includes the mesoderm that will migrate into the dorsal side of the embryo during gastrulation. By removing it from the embryo, we can study some of the morhpological processes. It will make a dorsal mesodermal structures (only dorsal since you take explant from dorsal lip side at inception of gastrulation). Essentially, it makes tissues that undergo convergent extension (mainly notohord); at the most anterior end it has the NIMZ that makes the head mesoderm. Thus you ultimately see a stick like structure (notochord) with a knob at the most rostral end (very anterior head mesoderm; remember Cereberus and Lim from lecture).
What is the archenteron?
The embryonic gut; it is formed during gastrulation
In the following image, increasing amounts of a certain protein are added to a neural plate-like tissue that are all stained for the presence of Nkx2.2. The top has no protein, the middle has a low amount of protein, while the bottom image has a higher amount. What protein do you think this is? Why do you need a lot of it?
Sonic hedgehog (Shh). You need a lot because Nkx2.2 is one of the most ventral targets of Shh (V3 interneurons). Remember that Shh activates targets in different classes of the ventral neurons in a dose-dependent fashion. V3 interneurons lie right above the floorplate and require the second highest doses of Shh protein.
What is an inhibitory signal in the somite that helps guide trunk neural cell migration? Where is it expressed?
EphrinB1 which is expressed in the posterior somite.
You want to study neural tube closture in mouse embryos. Design an experimental setup that would allow you to monitor this process occurring in real time development.
You could use the strategy outline in class lecture, which was to generate a transgenic mouse that has myristoylated YFP (yellow fluorescent protein that is specifically expressed in the cell membrane). You could then culture mouse embryos and take movies of the process in real time under a fluorescent microscope.
What happens when you electroporate chick embryos with Sox9 in the neural tube?
You get the expression of neural crest markers on the electroporated side of the neural tube, but these cells are unable to migrate out of the neurepithelium.
Where do neural crest cells form in relation to the neural plate of the epidermis?
The form at the boundary of the neural plate and epidermis.
What are the human birth defects corresponding to neural tube closure defects in the a) anterior neural tube and b) in the posterior neural tube?
a) anencephaly b) spinal bifida
Describe the 3 primary vesicles of the brain. What will each of them become?
prosencephalon = forebrain
mescencephalon = midbrain
rhombencephalon = hindbrain
In classic experiments, embryologists took naive neural plate and cultured it with notochord tissue to form ventral neuronal fates (including floorplate and motor neurons). What do you predict would happen if you performed these same experiments, substituting notochord tissue from a) Shh -/- embryos? b) Ptch -/- embryos? What if you incubated wt notochord with neural plate tissue from Smo -/- embryos?
The active component from the notochord is secreted Shh. In a) there is no Shh so the neural explants will not become ventralized. b) Ptch1 is a gain-of-function mutation in the Shh pathway, but the notochord will still make Shh so you will get ventral tissue. c) The notochord will still make Shh, but the neural explant tissue can't receive it (Smo is required for signal transduction of the hedgehog pathway). Therefore, you will not get ventralized neuronal tissue (it will be dorsal).
You want to misexpress Dbx2 throughout the right side (and only the right side) of a chick neural tube. How do you do this? What type of gene is Dbx2? Would you still form motor neurons? Why or why not?
You need to electroporate a plasmid that is expressing Dbx2 into the neural tube (electroporation hits only one side of the neural tube). Dbx2 is a type 1 Shh target that represses the type 2 Shh target Nkx6.1. Misexpressing Dbx2 will cause a loss of Nkx6.1 expression on the right side of the neural tube. Loss of Nikx6.1 will result in a failure to generate motor neurons on that side of the neural tube.
You want to further characterize the switch that tells trunk neural crest cells to start becoming melanocytes. a) Which protein, essential for making all neural crest cells, needs to be downregulated for this to happen?
FoxD3 needs to be downregulated.
You remain unimpressed by the supporting data from lecture (which is all about cell culture) and decide to test the model by performing an experiment where you forcibly maintain expression of this protein in neural crest cells. How do you do this? How could you determine if this supported the model?
The key is that you need to maintain FoxD3 expression in neural crest cells. If the model is true, all trunk neural crest cells should become sensory neurons/glia or at the very least should not express Mitf1. One strategy would be to electroporate the neural tube at an early stage before the neural crest cells migrate with a vector driving FoxD3 expression as well as some other marker (such as GFP). This marker is important because it is unlikely that all cells will get the electroporated construct. This expression would be maintainted well after neural crest migrationYou would to monitor for a general NCC antibody that is not specific to either lineage (eg HNK) as well as for your fluorescent maker (GFP). If the GFP+ cells all migrate through the ventral somites, this suggests they are forming glial cells (could confirm that they do not express Mitf1 with a second immunostaining). If most cells are electroporated, you would predict that you would also have a strong depletion of melanocyte cells.
Another way is to misexpress FoxD3 throughout most NCCs. You could give the embryo a virus or you could make the embryo express FoxD3 ubiquitously and see it with GFP and transplant pre-migratory NCCs to a recipient embryo where controls would be NCC transplants from donor embryos only expressing GFP.
You wish to generate a deletion of Pax6 in mice
A) Draw a schematic showing the targeting vector below the relevant region of the HoxD cluster.
-Should have a homology arm of roughly >=2Kb before the Pax6 coding region and a second homology arm (roughly >=2Kb) immediately after HoxD12. The region in between should contain a positive selection marker such as Neomycin resistance and may contain additional add-ons (like a reporter gene).
You wish to generate a deletion of Pax6 in mice. B) Once you have generated ES cells containing the targeting vector, describe the steps you would take to obtain homozygous null embryos for experimental analysis.
The ES cells are injected into a blastula stage mouse embryo, and this is then reimplanted into the uterus of a host mother. If the ES cells contribute to the forming embryo, mother will give birth to chimeric embryos, which can be seen with a different coat color (usually ES cells make a brown color while the blastocysts give rise to embryos with a black coat color. So chimeric embryos are black and brown - the more brown the better). Cross the chimera with wild-type mice to generate F1 heterozygous animals. Intercross F1 hets to make F2s. Mendelian genetics predicts that 25% of these should be homozygous for the null allele.
FYI, Pax6 not only has roles in neural tube development (e.g. the antagonistic role between Pax6 and Nkx2.2 that establishes one of the progenitor boundaries in the neural tube) but is also important for eye development (recall the second lecture of class).
You knock out a novel gene and are disappointed to discover that it has no embryonic phenotype. What are some reasons why there is no phenotype?
Perhaps you did not create a null allele. Could check this by Western blot with an antibody against a region of the protein not deleted; could also check by RT-PCR. Perhaps the gene is not required during embryogenesis. Is it expressed in the embryo (hopefully you did this before making the knockout!), perhaps there is redundancy with other genes. Are their closely related genes?
Velocardial facial syndrome (also called DiGeorge Syndrome) is a genetic disorder with a complex range of symptoms. Patients typically have cardiac problems, facial defects, and cleft palates, and may also have immune system problems. The syndrome is caused by a deletion of 1.5 - 3 million base pairs on chromosome 22. This large deletion removes many genes, and it has been challenging to determine which ones are causing the syndrome. Using your knowledge of developmental biology, speculate on what might be the embryonic origin of this phenotype (i.e. cell type). Using this speculation, devise an set of experiments using a model organism that will allow you to identify candidate gene(s) in the deleted region that might be responsible for the disorder.
Pharyngeal arch-derived neural crest cells generate the vast majority of the face and the aortic arches, but the thymus, which is responsible for T-cell maturation in the immune system, is derived from the 3rd and 4th pharyngeal arch (not mentioned in class). Cranial neural crest defects, as mentioned in class, also cause cleft palate. Defects in pharyngeal-arch derived crest cells could explain this complex set of symptoms. One experiment to explore this would be to identify all the candidate genes in the deleted region, perform in situ hybridization on a vertebrate model system (probably mouse; fish, frog and chick would work as well), and look for genes that are expressed in pharyngeal arch neural crest cells.
What would you predict would be the phenotype of a netrin mutant mouse?
Netrin acts as an intermediate-range chemoattractant that causes commissural axons (upon blocking Robo1 signaling via Robo3) to migrate towards the floorplate of the neural tube. In mouse embryos lacking functional netrin gene expression, commissural axons should still migrate ventrally down the neural tube but would not attracted to the floorplate. As a result, netrin deficient commissural axonal projections would fail to cross over to the opposite side of the neural tube.
You are so impressed with the Brainbow mouse that you decide to use it to study cranial neural crest migration. A) Describe an experimental strategy that would allow you to adapt the Brainbow approach to studying cranial neural crest (hint: we described a method for general genetic fate mapping of neural crest cells in the Feb 28th lecture). B) Would such an approach be useful for studying cranial neural crest - why or why not?
First, you need to convert the Brainbow construct from a neural specific promoter (Thy1) to one that will be active in neural crest. A couple of ways to do this. The easiest is to just use the ubiquitous Rosa26 promoter used in the genetic fate mapping. Using standard molecular techniques, insert the Brainbow cassette downstream of this promoter, where LaZ was previously. It is now in an OFF state, since the LoxP-STOP-LoxP cassette is in place. It could then be crossed to the Wnt1Cre construct used previously (acceptable answer). Alternatively, you could generate a Wnt1CreER mouse that allowed for transient activation of Cre in the neural crest cells of the embryos at a specific timepoint only via Tamoxifen injection (also acceptable answer).
B)The advantage to the brainbow approach is that it allows specific axonal projections and connections to be cleanly mapped back to the source. The cranial neural crest cells are not interconnected with anything; they migrate as single cells, and so aside from generating a pretty picture, it would not be particularly useful.
Describe the features of the Notch pathway that allow it to behave like a clock or oscillator. On a theoretical level, how could you modify the Notch proteins (any protein or proteins in the pathway) to speed up or slow down the oscillator?
The Delta (or Delta-like) ligand activates the Notch receptor. This in turn activates 1) the transcription of a transcriptional repressor (Hairy aka Hey aka Hes); in amniotes (chick/mouse) this core oscillatory pathway also has the transcription of Lunatic fringe (Lfng) a negative regulator of Notch signaling. Hairy represses its own transcription and also represses the transcription of Lfng.

The keys are the time-lag between the induction of Notch signaling and the production of Hairy and Lfng proteins (which have to be transcribed then translated). During this lag, Notch signaling is active. The presence of Lfng protein causes an inhibition of Notch signaling. At the same time, the presence of Hes proteins represses the transcription of both itself and Lfng. Notch inhibition will occur until Lfng and Hes mRNA/protein decays. Once they decay, the pathway can be activated again, setting up another oscillation.

The timing components involve the half-life of Lfng and/or Hes. If you were to generate more stable forms of these proteins (or mRNAs), this would increase the amount of time between oscillations. Conversely, if you were to destabilize the mRNA/protein, the oscillator would move faster.
LMC motor neurons are only present at specific axial levels of the spinal cord - why? In contrast, MMC motor neurons are rpesent throughout the entire spinal cord - why? Where are motor neurons found within the neural tube?
They only innervate the limbs, and are only found at the levels of the spinal column that project to the limbs. The MMC motor neurons innervate the body wall muscles and so are needed at all axial levels. Motor neurons are found in the ventral neural tube - just dorsal to the V3 interneurons.
What do you think would happen if you forced expression of ephrinA2 throughout the entire limb bud?
-This experiment hasn’t been done. But one prediction would be the complete absence of dorsally projecting motor axons, which are repelled by ephrins.
Based on material covered in lecture, what do you think would happen to neuronal migration in a Robo1 mutant embryo? Why?
Robo1 is the receptor for the midline chemorepellant ligand, Slit. In its absence, the migrating commisural axons would not avoid the midline. At the same time, Robo1 normally inhibits the Netrin receptor, DCC (except in the more ventral neural tube where Robo3 is expressed). So the axons would still be attracted ventrally - but would likely migrate straight down. Also, they might cross over and then cross back across the ventral midline.
What is the predicted phenotype of Robo3 null embryos? Why?
Without Robo3, commissural axons will not cross over the midline and will be stuck at the ventral lateral side of the neural tube. This is because Robo1/2 –mediated repulsion prevents the axons from going toward the midline. In addition, Robo1/2 prevents DCC from sensing a chemotactic netrin gradient.
Describe a way of generating genetically fate mapping the fate of the posterior sceleretome of the somites using -galactosidase activity. Mention the name of genes and how this system works. What are two tissues you might expect to see labeled? What are your controls?
Generate a transgenic mouse line that expresses Cre in the posterior scleretome. A great marker would be to use Ephrin B1::Cre (could generate a transgenic with EphrinB1 promoter enhancer as long as you showed / knew ahead of time that the construct contained enhancer elements active in the posterior scleretome. Alternatively, you could also generate a targeted mouse where you insert Cre and Neomycin in place of the EprhinB1 coding region (so mouse is an Ephrin B1 heterezygote – with the non-active gene copy driving Cre activity in the native Ephrin B1 domain. Typically hemizygous (Tg::EprhinB1 +/-)
2. Obtain a RosaLacZ reporter line. Rosa26 drives expression of LacZ, but only after Cre-mediated excision of a ‘STOP’ cassette. Typically homozygous (RosaLacZ c/c)
3. Cross the mice from steps 1 and 2 together. Stain for -galactosidase activity and visualize expression. Should be present in many tissues: vertebrate, ribs, blood vesses. Genotype embryos. Genotype all embryos. The only ones with staining should be Tg::EphrinB1 +/-; RosaLacZ c/+. The controls are Tg::EphrinB1 -/-; RosaLacZ c/+ (no staining should be present). Important note: this strategy will not label the migratory motor neurons or neural crest cells in this region; these are not somite-derived.
How is the gray crescent formed?
Cortical rotation
What are the domains of the blastula?
Animal cap, marginal zone, and vegetal base
What is the animal cap made of?
Ectoderm
What is the marginal zone made of?
Mesoderm
What is the vegetal base made of?
Endoderm
What happens when the sperm enters the frog egg?
There's a cortical rotation of 30* which makes the gray crescent.
What do microtubules do following sperm entry in a frog egg?
They're negative at the centriole and grow outward with positive ends. These positive ends move up along the sides.
What does beta-catenin do?
It's the dorsalizing signal.
What happens to embryos when they're severely depleted of beta catenin?
They become severely ventralized = belly pieces.
What's the experiment you use to find out if beta-catenin plays a role in DV patterning?
You dissect a frog embryo into dorsal and ventral sides and perform qPCR. There is higher Wnt 11 dorsally which makes sense because it's part of the canonical Wnt pathway.
What happens if you inject Wnt into ooctyes before fertilization?
They become super dorsalized.
Why should you inject Wnt before fertilization to see dorsalization?
Wnt is a maternal factor.
What does Wnt do when the sperm gets inside the egg?
It rotates 30* and activates the canoncial Wnt pathway.
Where does the wnt come from in the DV axis formation?
From the vegetal cells.
How many cells in the blastula?
20K
Name 3 mesodermal markers?
Xbra
chordin
FGF8
What happens when you stick a wt cap on a wt base of a blastula?
You get an increase of FGF8 and a decrease of chordin.
Mesoderm induction requires what two signals?
Vg1 and VegT
What happens in Vg1 and VegT mutants?
You get belly pieces.
Where is Vg1 and VegT located in the blastula?
In the bottom vegetal portion.
Another name for Vg1
Nodal
What does Vg1 do?
It activates Smad 2,3 trxn factors.
What happens if you stick a wt animal cap on a vg1- base?
You get a severe loss of mesoderm markers. No chordin and reduced Xbra and fgf8.
What does chordin do?
It's a BMP antagonist
What's the nieuwkoop center?
The dorsal vegetal region of the blastula
What factors are high in the Nieuwkoop center?
beta-catenin and VegT/Vg1. There's also a higher level of Xnr and a gradient of BMP4.
What specific structure is in the Nieuwkoop center?
The organizer.
What does beta-catenin in the Nieuwkoop center lead to? Show the pathway?
beta-catenin w/ VegT/Vg1 --> high nodal --> organizer
What's the pathway for VegT/Vg1 alone?
VegT/Vg1 --> low Nodal --> Ventral mesoderm
What happens when you transfer a doral blastopore lip onto a blastula?
You make a two-headed monster
What does the blastopore lip do?
It is the organizer and induces host tissue to form the body axis.
4 properties of the organizer:
1. initiates the movements of gastrulation
2. differentiates into dorsal mesoderm
3. induces ventral mesoderm to become paraxial mesoderm (somites)
4. induces dorsal ectoderm and neural tube formation
What does Noggin antagonize?
BMP
What does BMP do?
It's involved in ventral patterning
Where is noggin expressed?
In the organizer and notochord
What happens if you inject BMP4 into a blastula?
You get a belly piece.
What is the default of the organizer?
Neural ectoderm
What is a great neural marker?
Sox2
What's the initiating step of gastrulation?
The marginal zone cells become bottle shaped.
How does the blastopore form?
Via invagination at the marginal zone.
What is the archenteron?
The beginning of the gut of the embryo.
Name the example of epiboly in gastrulation.
The ectoderm engulfs the endoderm via epiboly.
What is the convergent extension?
It's the stretching and organization of the organizer.
Explain what a Keller explant is:
You take off the organizer and you put it under a microscrope and you will see that it will undergo convergent extension.
What molecules are involved in convergent extension?
Non-canonical Wnt signaling is used to induce cell polarity by activating Dsh (Disheveled).
What can convergent extension live without?
Canonical Wnt signaling
Where does the head mesoderm come from?
The mesoderm that is pushed up from the blastopore.
How can you make multiple heads from head mesoderm?
You inject Cerberus which inhibits Wnt8, BMPs, Nodal and causes more head mesoderm.
Where is Cerberus expressed?
In the anterior endomesoderm.
What happens if you mess with the anterior endomesoderm?
You get headless embryos.
What happens to Lim1 -/- mutants?
They don't have heads.
How common are neural tube defects?
1/1000 live births
What happens in primary neurulation?
Neural plate folds to generate hollow neural tube.
What happens in secondary neurulation?
A solid cylinder of cells cavitates to form hollow neural tube.
What are the 3 structures of the neural plate?
1. The neural plate
2. Neural crest
3. Epidermis
What is the neural plate made out of?
Ectoderm
What drives the folding of the neural tube?
Microtubes and actin.
How does the cephalic region of the neural tube close?
It zippers toward the anterior neuropore.
What happens if your neural tube fails to close?
You get spinal bifida or anencephaly.
How does the neural tube close at the midbrain?
A process called buttoning created by cellular bridges.
What factor is needed for neural tube closure?
B9 (folic acid).
Where is the folate receptor expressed?
In the dorsal neural tube.
How can you decrease neural tube defects?
By taking folic acid at the beginning of the pregnancy. It decreases it by as much as 70%.
what is Mthfd11?
An enzyme in the pathway that makes folic acid available to the cell.
What happens in Mthfd11 mutants?
They have an open hindbrain. Their neural tube doesn't close.
What is the result from the neural tube closing?
Occlusion of the neural tube allows pressure to swell in the lumen of the developing brain.
In zebrafish, what causes the ventricles to swell?
The Na/K ATPase which causes an osmotic gradient.
Name the 3 developmental structures of the brain:
1. prosencephalon
2. mesenchephalon
3. rhombencephalon
forebrain comes from:
prosencephalon
midbrain comes from:
mesencephalon
hindbrain comes from:
rhombencephalon
What happens to Otx2 mutants?
They are missing their forebrain and midbrain.
What happens to Gbx2 mutants?
They're missing their metencephalon (rostral hindbrain).
What does Gbx2 do?
It restricts Otx2 expression
What does the Otx/Gbx border do?
It sets up the mid/hindbrain boundary.
What does Gbx2 establish?
The metencephalon
What does Otx2 establish?
The mesencephalon
What is the isthumus?
The major molecular organizing center in brain development.
Another name for rostral?
Anterior
Another name for caudal?
Posterior
What is Hoxb8 a marker for?
caudal (posterior) neural identity
Where is RA expressed?
In the caudal paraxial mesoderm
What does RA induce?
RA from the paraxial mesoderm induces caudal neurons.
What is Shh a morphogen for?
It's a ventral morphogen
Where is the highest level of Shh?
In the notochord.
What is the Shh pathway?
Shh binds --> Patched --I Smo --> Upregulation trxn
How can you visualize the Hh gradient?
You make a mouse with Shh::GFP
What happens to Ptch1-/- mutants?
They way overexpress Shh.
What happens to Shh mutant embryos?
They lack ventral neurons
Name 2 factors induced by high Shh.
Nkx6.1 and Nkx2.2
Name two factors induced by low Shh.
Pax7 and Dbx1
What factor is repressed in high Shh?
Pax6
How do you electroporate something?
You shock the cells and DNA will move to one side.
What does ectopic Nkx6.1 do?
It causes the loss of Dbx2
What does ectopic Dbx2 do?
Leads to loss of Nkx6.1
What is the fourth germ layer?
Neural crest
Where are neural crest cells born?
In the dorsal neural tube.
What is EMT?
The change that neural crest cells undergo called the epithelial to mesenchymal transition and then they migrate to the periphery.
What is RhoB?
It is the factor expressed during neural crest initiating migration
When do crest cells express RhoB?
When they undergo EMT.
What do neural crest cells express just prior to migration?
Sox9.
What do Sox9 and RhoB do?
They induce Neural Crest differentiation and EMT.
What do neural crest cells end up generating?
Sensory and sympathetic neurons.
What is the sclerotome?
They'll make sensory and sympathetic neurons.
Trunk crest cells only migrate through...
that anterior half of somites.
What is expressed in the posterior half-somite?
Ephrin B1
What do crest cells express?
EphB3.
What repells crest cells from going into the posterior portion of the somite?
Ephrin B1.
What is the stripe assay?
The way to show that neural crest cells are repelled by ephrin B1.
What do path 2 crest cells generate?
They make melanocytes.
Where do path 2 crest cells migrate?
Between the dermamyotome and the surface ectoderm.
What does FoxD3 do?
It's essential for generating NCC and says on Pathway 1.
What does FoxD3 repress?
A gene called Mitf, which is the master regulator of melanocyte fate.
What does Mitf do?
It's the regulator of melanocyte fate.
Mitf1 is essential for...
melanocyte stem cells.
Why do we turn gray?
the cells expressing mitf1 are lost.
Where do the pharyngeal arches come from?
The migration of specific streams of cranial neural crest cells.
Pharyngeal arches are populated by what kind of cell?
Neural crest cells.
What happens to pdgfra mutants?
They're defective for neural crest migration
What is Pdgf?
It's a cranial neural crest chemoattractant.
What is the anatomy of an arch?
Muscle in the middle, wrapped in crest cells with endoderm on top and ectoderm on the bottom.
What does the first pharyngeal arch contribute to?
Malleus
Incus
Mechel's cartilage
What does the second pharyngeal arch contribute to?
The stapes and styloid process
What does the 3rd pharyngeal arch make?
Hyoid bone
What do pharyngeal arch 4 and 5 make?
Thyroid cartilage
When genetically fate mapping a mouse, where do the facial structures come from?
Neural crest cells and mesodermally derived cells.
Where do facial prominences come from?
The first pharyngeal arch.
3 facial structures from the 1st pharyngeal arch.
1. midline facial skeleton
2. upper jaw and palatal skeleton
3. lower jaw
What occurs at the border of Shh and Fgf8?
It's the distal tip of the upper break
What happens if you make extra Fgf8-Shh boundaries?
You get multiple ectopic beaks in a bird.
What makes up an axon growth cone?
Filopodia and lamellipodia.
What are filopodia?
The microspike fingers of the growth cone
What are lamellipodia?
The webbing of the growth cone.
What does the axon growth cone do?
It senses the environment and connects to the appropriate targets.
What is pathway selection?
Axon chooses route through the environment to its target.
What is target selection?
Axon recognizes target and initiates synaptogenesis.
Addess selection:
Synapses are strengthed and lost
Name the two structures of the neural tube that contribute to motor axon projections?
Lateral motor column
Medial motor column
Where does the medial motor column project to?
The dermamyotome
What are the subcategories of the lateral motor column?
Lateral subdivision --> dorsal
Medial subdivision --> ventral
Where do LMCl project to?
Dorsal musculature
Where do LMCm project to?
Ventral musculature
Ephrins are concentrated in what part of the limb bud?
The ventral part
EphA4 expressing axons project...
dorsally
EphA4 is sufficient to drive axons...
dorsally.
What does Lim1 do?
It activates Eph4A.
What does Isl1 do?
It represses Eph4A.
What is the pathway for LMCl axons?
Lim1 --> Eph4A --> dorsal projection
What must axons to at the ventral midline?
They must project to the ventral midline and only cross once.
What are Robo and Slit?
Slit is a ligand in the midline of drosophila.
Robo is a receptor on axons.
What happens to slit mutants?
Axons never leave the midline.
How do Slit and Robo interact?
Robo1 is a receptor and when Slit binds to it, it inhibits DCC.
Slit binding to Robo1 results in what?
Chemorepulsion
What happens when Robo3 is expressed?
It inhibits Robo1 so now DCC can work and the axon can cross the midline.
What does Netrin bind to?
DCC.
What happens after the axon crosses the midline?
Robo1 turns back on and blocks DCC chemoattraction
What does the brainbow help you do?
You can see the neuronal connections based on the random turning on of GFPs.
How does brainbow work?
It's a Cre-base recombination resulting in expression of 3 different fluorescent proteins.
What is the axon chemoattractant?
Netrin binding to DCC receptor.
What is the axon chemorepellant?
Slit binding to Robo1.
Where does somitic formation occur?
In the presomitic mesoderm
What is the somite clock?
The time period of somite formation.
What is the Notch pathway?
Delta (membrane-bound ligand) --> Notch (receptor) --> Lfng --I Hairy --I trxn of Hes and Lfng
What is the Clock and Wavefront model?
There is an oscillator (clock) that regulates the periodic generation of somites and a wavefront that defines the region competent to respond to the clock and become a somite.
What does the clock part of the Clock and Wavefront model do?
It regulates the periodic generation of somites.
What does the Wavefront part of the model do?
It defines the region that will become a somite.
What is the central oscillator controlled by?
Notch/Delta signaling
Another name for Hairy...
Hes
What is the oscillating expression of Hairy?
Hairy1 expression moves from posterior plate to anterior. The posterior expression boundary matches the posterior of what will be the next somite. Oscillation happens every 90 minutes in a chick.
How does Notch affect the Wavefront model?
It provides signaling for the border information. Notch signaling is sufficient to cause segmentation.
What happens to Delta-like 3 mutants?
They have defects in somite formation, leading to rib fusions.
What signaling is necessary for somite formation?
Notch signaling
What happens to MESP2 mutants?
They have defects in ribs and vertebrae.
What happens to Lfng mutant mice?
They lose Notch cycling
How does Lnfg aid in oscillating expression?
Lnfg is a negative regulator of Notch, but is very unstable. So it leads to an oscillating signal.
How does Wnt signaling affect somite segmentation?
It helps position determination front.
What does Axin2 do?
Inhibits Wnt activity and is short-lived.
If Notch can't oscillate, how does Wnt react?
They still show axin2 expression oscillation so Wnt pathways target genes still oscillate.
What happens if beta-catenin can't be degraded? This is relating to somite formation.
There's no Wnt activity. There are no visible somites.
What happens to somite formation when there's no beta-catenin?
Oscillation still occurs, but it's abnormal. Wnts do not appear to be a pacemaker for oscillations.
What regulates the size of somites?
Fgf
What happens if you stick an FGF-soaked bead near the undetermined zone?
It makes a smaller somite.
How is FGF related to the wavefront?
It regulates segmentation size by changing the "size" of the wavefront.
What happens at somite IV?
Somite segmentation is determined. The receding Fgf8 marks a boundary.
What is the determination front in a molecular definition?
It is the posterior boundary of Mesp2 gene expression.
What happens when the wavefront breaks?
It's the region at which presomitic mesoderm first acquires segmental identity.
What happens to cells anterior to the determination front?
They are more epithelialized due to a downregulation of fgf8. They undero a mesenchyal - epithelial transformation.
How is somitogenesis in fgf8 nulls?
It's normal.
Why do fgf8 nulls still have normal somitogenesis?
fgf8 is redundant for fgf4.
How can you knock out fgf8/4?
You have do conditionally KO.
What happens when you conditionally KO fgf4/8?
They have major somite defects. They make a bunch of somites.
What happens to the the presomitic mesoderm in fgf4/8 cKOs?
It all undergoes somitogenesis.
What does the wavefront prevent?
Early differentiation.
How is the determination front in fgf4/8 cKOs?
It's expanded in the PSM.
In the Clock and Wavefront model, what is keeping the somites from differentiating prematurely?
the fgf/Wnt gradient.
How are snakes somitogenesis different from others?
They have slower wavefront
A slower wavefront leads to...
lots of somites.
What pinches off somites?
Eph/ephrin interactions.
What does Eph4A do in somite formation?
It precedes somite formation and is restricted to the anterior half of the somite.
Where are ephrins expressed during somitogenesis?
In the posterior somite.
How does Eph signaling affect somitogenesis?
It positions somite boundaries.
How does epithelialization of a somite work?
Integrins and N-cadherin polarize outer cells into an epithelium. It occurs immediately after somite fission.
Which process provies the major force to extend the anterior-posterior axis of frog embryos during gastrulation?
Convergent extension
Name a general mesodermal inducing factor.
Vg1
Name 4 neural crest derivatives:
1. The stapes
2. The peripheral neuvous system
3. The hyoid bone
4. The mandible
What branchial arch gives rise to most of the face?
First
Which molecular interaction is most important for helping scult the motor neuron in the neural tube?
Nkx2.2 -- Pax6
Name the pathway that mediates convergent extension.
Non-canonical Wnt signaling.
You take the dorsal life of a gastrulating frog embryo and transplant it to the ventral side of the host embryo. What happens?
The embryo forms another body axis.
You electroporate a chick neural tube with Sox9. What happens to the electroporated cells?
They become neural crest cells.
3 things you need to make a targeting vector that can modify the genome of mouse ES cells.
1. left homology arm
2. right homology arm
3. Positive selection cassette eg NeoR
You express Eph4A in all motor neurons. What happens?
All the projections will go in the dorsal part of the limb bud.
What general route do non-melanocyte trunk neural cells take when they migrate? How could you block or inhibit this route?
They migrate through the anterior sclerotome. You could reroute them by forcible expression of mitf (They'll now become a melanocyte). Or you could block the route by misexpressing Ephrin B1 throughout the whole somite.
Embryologist took naive neural plate and cultured it with notochord tissue to form ventral neuronal fates. What do you predict would happen if you performed these same experiments substituting notochord tissue from Smo-/- embryos? B: Shh-/-? What if you incubated with wt notochord with neural plate tissue from Shh-/- embryos?
A. Will not express ventral neural tube markers
B. Will not express ventral neural tube markers
C. Will express ventral neural tube markers.
How could you use genetic fate mapping to determine the fate of somites?
Somite-specific Cre crossed with Cre reporter construct (a construct with a ubiquitous promoter then a loxP-stop-loxP followed by reporter gene that will only be trxned when Cre removes the stop cassette).
You treat recently fertilized frog eggs with Nocodazole. What will this do? What will the embryo look like as a result?
This will block microtubule polymerization, preventing cortical rotation. The embryo will be a belly piece.
You have just generated a genetically modified ES cell line in which one copy of a gene is deleted, creating a null allele. Briefly list the steps you need to take to generate and analyze homozygous null embryos.
1. Inject ES cells into host mouse embryo and implant injected embryos into surrogate mom.
2. Mom will give birth to chimeras. You can tell chimeras by coat color contribution.
3. Breed chimeras to wt mice to test for germline transmission. These progeny are heterozygous embryos.
4. Cross heterozygous animals and collect embryos for genotyping and analysis.
Why are pregnant women encouraged to take folic acid?
Because folic acid supplements reduce the prevalence of neural tube defects.
What do you think would happen to somite development if you incubated embryos in an inhibitor that reduces but does not eliminate fgf signaling.
You would have a reduced gradient of the wavefront. This would likely give rise to fewer and bigger somites.
In class, we discussed the general features of an oscillator. The Hh pathway has some of these same features although it does not appear to oscillate. Describe the feature of the Hh pathway that could make it act as an oscillator? How you could modify this in a way that might make it into an oscillator?
The Hh lignad can activate the trxn of Ptch1 mRNA would make Ptch1 protein. Ptch1 inhibits Hh signaling (unless boud by more ligand) and therefore acts as a negative regulator of the Hh pathway. You could modify the Ptch protein to make it more unstable. This would allow the pathway to oscillate between off and on in the presence of the Hh ligand.
LMC motor neurons are only present at specific axial levels of the spinal cord - why? In contrast, MMC motor neurons are present throughout the entire spinal cord - why? Where are motor neurons found within the neural tube?
A. LMC neurons only project to the forelimb and hindlimb and are only found at these levels.
B. MMC neurons project to axial musculature of the body at all levels.
C. Motor neurons are found in the ventral neural tube.
You take vegetal tissue from a frog embryo and culture it together with an animal cap tissue. What happens to both tissues?
The vegetal tissue will form endoderm. The animal cap tissue will form mesoderm.
You have just discovered a new gene that, when depleted in frog embryos with siRNA antisense oligonucleotides, gives
rise to an embryo that lacks all neuroepithelium.

A) Based on what we learned in class, propose a molecular mechanism for how this gene might work? Describe one
additional piece of experimental data that would be helpful for you to refine your model?
B) What do you think would happen if you over-expressed this molecule? Devise an experiment to test your
hypothesis. Be as specific as you can be and include appropriate controls.
Secreted BMP antagonists from the organizer inhibit the BMP pathway, resulting in the formation of neural tissue. So the BMP pathway is activated in all ectodermal cells when the gene is lost. It could be the loss of a major BMP antagonist like noggin, chordin, or follistatin. There's a lot of redundancy though. It could also be a problem in the BMP pathway to make it constantly expressed. B. Overexpression could result in neural expression in all neural tissue. You could do insitu for neural marker like Sox2. Or overexpressing some aspect of the BMP pathway could turn off BMP.