Desmosomes and adherence junctions: What do they have in common, what are the differences?
In common: Both cell-cell adhesion structures, mediated by Cadherins
Differences: Desmosomes-TM-adhesion protein-Non classical cadherins (Desmoglein, desmocollin), Intracellular Cytoskeletal attachment- Intermediate filaments
Adherens junction - TM-adhesion protein- Classical cadherins, ntracellular Cytoskeletal attachment- Actin filaments
Desmosomes and hemidesmosome: What do they have in common, what are the differences?
In Common: Intracellular Cytoskeletal attachment: Intermediate filaments, both cell adhesion structures
Differences: Desmosomes- Mediate Cell-Cell adhesion, TM-adhesion proteins -Cadherine (Desmoglein, desmocollin),
Hemidesmosomes - Mediate Cell-ECM adhesion, TM-adhesion proteins-Integrin, type XVII collagen
Also different Intracellular adaptor proteins
1. Adherens junctions
a) Please draw or describe the structure of adherens junctions and name the core components.
b) In which type of tissue do you find adherens junctions?
c) To dissociate cells in culture, these are often treated with solutions containing EDTA. Please explain the effect of EDTA in this process.
d) One of the components of adherens junctions has a second function in a signaling pathway that is important during embryonic development and tumorigenesis. Please name this component and briefly explain its function in signaling.
a) Clasical cadherin (E-Cad, N-Cad) Neighbouring cells are linked through homophilic interactions between the extracellular domains of the cadherins, p120 Catenin, ß-Catenin linked to Cadherins on Cytosolic side, Anchorprotein (Vinculin) link Complex to Actinfilaments
b) In epithelia
c) Cadherin cell adhesion is Ca2+ dependent. EDTA forms a chelate complex with Ca2+ and therefore destabilizes the adhesion. Cadherins contain Ca2+ -binding sites in their extracellular domain and Ca2+ binding is required for maintaining of this domain in a conformation that allows homophilic interaction. Removal of Ca2+ induces a conformational change that prevents homophilic interaction
d) ß-Catenin is a component of the canonical Wnt pathway. Upon activation of the pathway cytoplasmatic ß-Catenin is stabilized, translocates to the nucleus and associates with Lef/Tcf Proteins to form a transcriptional activator
Epidermolysis bullosa (EB) ( is a congenital skin disease caused by mutations in genes encoding components of the epidermo-dermal junction and is characterized by severe skin blistering already in response to minor traumata. So far mutations in 15 genes have been identified. Different disease types are known that differ in the level of the skin, where the blistering occurs. In junctional EB (JEB) the tear forms between epidermis and basal lamina, in dystrophic EB (DEB) there is a separation of epidermis and basal lamina from the underlying dermis. Which genes would you screen for mutations, and why, in a patient with
a) JEB
b) DEB
a) Integrin a3, Integrin a6ß4, collagen XVII, Laminin-332 make up connection of epithelia to Basal lamina in Hemidesmosomes
b) Kollagen VII mediates connection Dermis to Basallamina
Name at least two core components of the basal lamina and explain their function.
Laminin: Primary organizer of basal lamina structures, joins to each other and binds to Surface Receptors (Integrins) to form a network -> Stabilization
Collagen IV: ropelike superhelix provides tensile strenght to basal lamina
Integrins: Connect Epithelial cells to ECM
Additional Components: Nidogen-Glycoprotein, Perlecan-proteoglycan -> Bring protein stability
Schematically draw the structure of a hemidesmosome and label its components.
What is the function of a hemidesmosome?
Cell-Matrix adhesion structures that anchor epithelial cells to underlying basement membrane and provide structural stability promoting tissue integrity
Cell-junctions
a) name 4 junctions in the epidermal layer (apical to basal)
b) name for each the transmembrane protein for cell contact
c) which one of the 4 have contact to cytoskeleton and name the filaments
a) Tight junctions-seals gap between epithelial cells
Adherens junction- connects actin filament bundle in one cell with the adjastend cell
Desmosomes- connect IF in one cell to those in adjastend cell
Gap junctions- allows passage of small water soluble molecules from cell to cell
b) Tight junction: Occludin & Claudins
Adherens junction: Cadherins (E-Cadherin)
Desmosome: Desmogleins & Desmocollins (Nonclassical cadherins)
Gap junctions: Integrins
c) Desmosomes have contact with Cytoskeleton & connect to Intermediate Filaments (Keratins)
What are the functions in apoptosis of
A) pro survival BCL-2
B) pro apopt. BCL-2
C) mitochondria
D) caspase
E) IAP
A) Function is to inhibit Apoptose, Prevent activation of pro-apoptotic members of BCl-2 family -> inhibiting Cytochrome c release in Mitochondria
B) promote apoptose by facilitating the release of pro-apoptotic factors (Cytochrome c) - Promote Activation of downstream caspases and apoptosis initiation, Acts opposite to pro-survival BCL-2
C) Important role in Intrinsic apoptotic pathway - Mitochondria release Cytochrome c in Cytoplasm -> triggers formation of the apoptosome -> Caspase activation & Caspase cascade
D) Protease Enzymes Key executioners of Apoptose, Activated in response to apoptotic signals. Initiator Caspase (9) cleave & activate effector caspases (3,7) -> proteolytic cleavage of cellular substrates
E) Apoptose Inhibitors that inhibit caspases, regulate both intrinsic & extrinsic pathway
At what levels do changes in the human genome occur?
· Genome [numerical alteration of the complete genome or numerical alteration of a single chromosome]
· Chromosome [insertion, deletion, duplication, inversion, translocation (defects every 3 – 5 Mb)]
· Gene [point mutation, mutations of several kilobases, mutations in regulatory sequences. 1-300bp. point mutations account for up to 70% of all types of variation]
What type of DNA variations can be found in the human genome? Which type of DNA variation is the most common one?
· SNP`s [= ”single nucleotide polymorphism”: occur ~ 1 in 300 nucleotides]
· CNV`s [= “Copy number variations”: are responsible for by far the greatest number of nucleotides that differ between two genomes,]
· Microsatellites [= “short tandem repeats” (used for DNA profiling)]
-> SNPs most common type in humans
How are DNA variations generated?
· SNP`s and small DNA changes are caused by DNA damage, that can occur via
- External causes: [ionizing radiation, ultraviolet radiation, environmental chemicals]
- Internal causes: [depurination, deamidation, reactive oxygen species (ROS), nonenzymatic methylation, normal DNA metabolism]
· CNV`s (and microsatellites and SNP`s) are generated by errors in recombination and errors in replication
· Microsattelites: Errors in replication (backward spillage)
· All DNA Variations are caused by failure in repairing DNA
What type of DNA variation is a “nonsense mutation”? What is a possible consequence for the gene/protein?
· Derived from a SNP
· Exchange of the encoding amino acid leads to premature stop codon ( ->termination signal)
· Possible consequence truncated protein, protein degradation, NMD (nonsense-mediated mRNA decay)
What is a “CNV”? What are possible consequences?
· Copy number variation = sections of the genome are repeated
· Surprisingly frequent in human genome (~5%)
· Possible consequence: parts of a chromosome are deleted or duplicated
a) What is the pattern of inheritance for this disease?
b) What is the risk for children of the patient to be affected?
c) What could be the pathology of this pattern of inheritance? Do you know an example of a disease matching the criteria?
a) Autosomal dominant
b) 50%
c) Dominant negative effect (Osteogenesis imperfecta)
Haploinsufficiency (Aniridia)
a) What is the pattern of inheritance? Explain why?
b) Do you know an example?
a) X-linked recessive. Only male individuals are affected females are only carriers
b) Red-green color blindness, Haemophilia A & B, Duchenne muscular dystrophy, Fra (X) syndrome
What is the pattern of inheritance? Explain why?
Autosomal recessive - both parents are carriers but only homzygous mutation leads to phenotype
What does a homozygous mutation mean?
Both alleles carry the same mutation - individual homozygous for that mutation
What does compound heterozygous mean?
Presence of 2 different mutant alleles at the same locus
What is a Loss of function mutation? Do you know any disease?
· A loss-of-function mutation is a genetic mutation that leads to a reduction or complete loss of the normal function of a gene -> no protein, non-functional protein, disrupted functional domain
· Examples: Cystic fibrosis (CF), Hemophilia, Tay-Sachs disease
usually (autosomal) recessive pattern of inheritance
What are the most important models of inheritance for monogenetic diseases?
· Autosomal recessive - One affected allele is tolerated, typically unaffected carrier parents, affected individuals homozygous or compound heterozygous E.g. Cystic fibrosis
· Autosomal dominant - One affected allele is not tolerated, affected individuals usually 1 affected parent, 50% chance of passing mutant allele to offspring, E.g. Huntington disease
· X-Chromosomal - Mutations on X chromosome either Dominant or Recessive (in Males single copy sufficient, females homozygous, E.g. Hemophilia)
(Y-chromosomal - Only males affected from father to all sons)
What is the reference genome?
· A reference genome (also known as a reference assembly) is a digital nucleic acid sequence database, assembled by scientists as a representative example of a species’ set of genes. They are often assembled from the sequencing of DNA from a number of donors.
Which information is missed by using a reference genome?
· Reference genomes do not accurately represent every individual among a population.
Which type of DNA variants can you detect using Sanger sequencing?
· SNPs (inducing small insertions, deletion of a few base pairs)
Describe the main principle of Next Generation Sequencing
the main principle of NGS involves parallel sequencing of millions of DNA fragments, generating massive amounts of sequence data simultaneously.
Outline the general principle (i.e. steps) of next generation sequencing technologies.
· Fragmentation of DNA & adaptor ligation (library preparation)
· Immobilization on a glass slide surface (flow cell) & amplification of DNA fragments (clonal amplification by bridge PCR, cluster generation)
· Cyclic sequencing of clusters with fluorescently labelled nucleotides
· Imaging of flow cell after each cycle (emission from each cluster is recorded, emission wavelength are used to identify the base)
· Cycling is repeated ‘n’ times to create read length of ‘n’ bases
· Millions of short reads are aligned to a reference sequence with bioinformatics software
What kind of mutations in regulatory sequences are there?
in Promotor (e.g. TATA Box) - affects expression levels
in splice site (Enhancer, Silencer…) - affects splicing, exons skipping, intron retention
in 5` UTR (cap) - affects mRNA stability
in 3’ UTR (Poly-A) - affects stability & export mRNA
Name 3 mutations without change of reading frame, what are the results?
Silent Mutation - same amino acid (rarely pathogenic), different tRNA
Missense Mutation - different amino acid by single nucleotide substitution, conservative change (AA similar properties), non conservative change ( AA different properties -> Alters structure, function of protein) E.g. Sickle Cell Disease
Nonsense Mutation - nucleotide substitution leads to premature stop-codon (termination) -> truncated protein, protein degradation, NMD-nonsense mediated mRNA decay
Readthrough Mutation - Nucleotide substitution -> Stop codon is lost -> Extended Protein
What are Mutations with change of reading frame ?
Frameshift mutations:
Insertion (addition of nucleotides)
Deletion ( Loss of nucleotides)
-> can lead to stop codon, degradation by NMD
What is the key event initiating D-V and A-P asymmetry in the Drosophila oocyte?
(Pattern signals Pyrow.)
A-P
· mRNAs provided by the nurse cells use the microtubule scaffold to differentially localize to the two poles of the oocyte. Location of the mRNAs define the anterior (bicoid) and posterior (nanos trapped by Oscar) pole
Anterior: Bicoid transported by Dynein to minus pole of microtubule
Posterior: Oscar transported by Kinesin to plus pole of microtubule where it traps nanos
· Oocyte nucleus associates with the microtubuli network and is transported to the minus end. The location of the nucleus defines the dorsal pole. Nucleus travels to anterior dorsal side -> Gruken recieved by Torpedo in dorsal follicle cells -> Inhibition of pipe dorsally
Ventral secretion of pipe - cleavage of spätzle binds toll & activates dorsl
Please describe the genetic hierarchy in A-P patterning in Drosophila
Maternal gradients (Bicoid,nanos…)
Gap genes: regulated by maternal gradients, expression controlled by concentration of bicoid & hunchback at different points along A-P axis, Regulation of Gap genes by themselves ( Krüppel, Hunchback…)
Pair rule genes: formation of striped pattern that’s a precursor for segmentation. Each stripe requires a specific gap protein activity. e.g. evenskipped: hunchback and bicoid would activate evenskipped in a broad domain but only where giant and krüppel activity is lowest the stripe is formed.
Segment Polarity genes: equip the parasegment with polarity, forming an anterior/posterior pattern in each segment. Expression is initiated by gap and pair rule genes and maintained by autoregulatory circuits. produce signalling molecules / transducers. e.g. wingless & engrailed: neither evenskipped nor ftz is present wingless is expressed, wherever one of those pair rule genes is present engrailed is expressed.
Homeotic Genes (Hox): specify the segment identity, appear in gene clusters, regulated by gap and pair rule genes and cross-regulation. gene order on chromosome corresponds to spatial and temporal order of gene expression along the A-P axis = colinarity. homeobox = DNA binding domain, transcription factors. Mutated HOX genes -> defect body structures (e.g. 4 wing pairs [ultrabithorax HOX gene] or legs for antenna [antennapedia])
Please explain different mechanisms for generation of positional information.
(Pattern formation Pyrow.)
Patterning of cellular fields:
Morphogen gradients: signalling molecules, that form concentration gradients across developing tissue. Concentration at particular location determines fate of cells in that region. E.g. Maternal gradients of bicoid (anterior) & caudal (posterior)
Lateral inhibition: Cells with a certain fate inhibit neighboring cells from adopting the same fate. E.g. Delta-Notch inhibition
Induction/Induction cascades: Group of cells influences the fate of adjacent cells. Induction cascades lead to sequential determination of cell fates. E.g. Lens formation
Oscillating systems: generation of periodic patterns of gene expression or signaling. periodicity contributes to the establishment of positional information. E.g. Segmentation clock in somitogenesis
Reaction diffusion systems: Interaction between activators and inhibitors that diffuse through tissues -> results in the formation of spatial patterns. E.g. WNT-DKK specifies hair distribution, Zebrafish stripes
Organization by cell sorting:
Cell adhesion: Cells with similar adhesion properties tend to sort out and form distinct tissues mediated by Adhesion molecules.E.g Cadherin mediated cell adhesion and sorting leads to tissue organization
Making cells different:
Localized determinants: positional info by establishing e.g anterior and posterior axis through asymmetric distribution. Mechanisms: Diffusion and local anchoring (Nanos), Localized protection (hsp83), Active transport along cytoskeleton (Oskar and Bicoid mRNA on microtubules)
Asymmetric cell divisions: Daughter cells with different fates – unequal distribution of cellular components or fate determinants. E.g. Asymmetric division in Neuroepithelium
Please explain how cells may sort on their own into distinct tissues in development!
Through Cell Adhesion. E.g. Cadherin mediated: Different cadherin types in different cell. Some have stronger adhesion and form stronger aggregates. Strong aggregates form the center, low adhesion in the peripherie. N-, P-, and E caherins(Cells with high adhesion maximize adhesion to each other and minimize contact to cells with low adhesion) Bsp. Neural plate epidermis
Please explain August Weismann's theory of localized determinants.
Each cell inherits a specific determinants which determines the cells development. Only the germline cells are consistent. That’s why mutation in the germ cell are inherited by the offspring and mutations in the somatic cells are not.
Please explain different mechanisms how localized determinants may be generated.
· Diffusion and local anchoring: The nanos mRNA diffuses in the cell, posteriorly is an anchor (oskar) that fixates nanos to the posterior site -> local translation of nanos
· Localized protection: hsp83 diffuses in the cell and gets protected by Protector protein complex otherwise it will get degraded – only proted mRna will get translated
· Active transport along cytoskeleton: Oskar and Bicoid mRNA use microtubules as transport mediated by Dynein and Kinesin, Oskar moves posterior via kinesin, bicoid moves anterior via Dynein
How may asymmetry be generated to initiate asymmetric cell divisions?
Symmetry Breaking (Polarity Determinants/Proteins are distributed asymmetrically within cell E.g. PAR-Proteins) -> Polarity establishment -> Determination segregation -> Spindle positioning -> Distinct daughter cells
Cell fate determining proteins in one of daughter cells
What is induction?
Please explain a serial induction (= induction cascade, relay mechanisms).
Induction:
Group of cells influences the fate of adjacent cells.
formation of a different tissue, induced by a signal from tissue B, the signal reaches up to a certain threshold into tissue A and leads to the formation of tissue C
one signal induces only one new cell type with one concentration threshold.
serial induction (= induction cascade, relay mechanisms):
From the newly formed tissue C another signal induces the formation of tissues D and E, the signal acts on tissues A and B up to a certain threshold
e.g. formation of the lens
Please define a "morphogen".
signaling molecule that plays a key role in embryonic development by forming concentration gradients within developing tissues.
concentration gradients provide positional information to cells, influencing their fate and behavior based on the concentration of the morphogen they are exposed to.
essential for the process of pattern formation, establish the spatial organization of different cell types and structures in an organism.
Examples: Sonic hedgehog, Bone Morphogenetic Proteins (BMPs), and Wnt proteins
How can a stable gradient of a morphogen be established?
stable source of signal and a limited stability of the signal leads to a balance between synthesis, diffusion and degradation to establish a stable gradient after a startup time
How can inhibitors generate a morphogen field?
An inducer is extending across a field of cells and uniformly distributed, an inhibitor is then distributed from a source resulting in a gradient of inducer activity
What does it mean morphogen mechanisms are scalable? How does this work?
A morphogen gradient can expand proportionally with tissue growth, it can pattern small and large scale tissue fields, an adjustment of stability, diffusion and transport rate may be needed
Please explain how lateral inhibition works!
Field of cells that all have the same potential to differentiate- some cells inhibit adjacent cells from developing in a similar way. Individual cells within a field of cells get defined. E.g Delta-Notch forms a contact dependent signalling system between adjacent cells. Competition of Notch and Delta receptors of each cell. Delta binds to Notch on adjacent cells. Activation of Notch leads to inhibition of Delta. One cell wins.
How can repeat body segments be made and the number of segments evolve rapidly?
Oscillating systems generating repeat units through cycles of gene expression, the mechanism works on 3 tiers: the bottom tier is single cell oscillators where the translation of a gene expresses a protein that inhibits the gene (negative feedback) the middle tier is local synchronisation of neighbouring cells and the upper tier is global control of slowing and arrest which means the signal gradient gets slower and at some point arrested which forms a new segment. During each cycle one new segment is determined. (clock and wavefront model)
Please explain the fundamental principles of a "reaction - diffusion" system!
Circuit of diffusion activator and inhibitor where the activator activates itself and the inhibitor, the inhibitor then inhibits the self-activation of the activator. The Reaction diffusion system has a self-organizing property and generates patterns independent of size of the cellular field, whenever the concentration of the activator exceeds a certain level there
What regulates the density of hair growth in the skin of a mouse?
Example for Reaction-Diffusion System hat influences the patterning and density of hair follicles
Wnt ligands are expressed in localized regions, promoting hair follicle development, while DKK proteins act as inhibitors, restricting Wnt signaling in specific areas. spatial distribution of Wnt and DKK establishes patterns of high and low Wnt activity,
Why has the striping pattern of fish similar size in small, young and big old fish (and not small fish small stripes and big fish big stripes)?
Once developed stripes do not change anymore, but over time new stripes are added.
Example for an Reaction-Diffusion System Interaction between activators & Inhibitors leads to pattern formation -> Diffusible signalling molecules establish a stable pattern
Is bicoid phylogenetically conserved? Why (not)?
(Pattern-signals Pyro.)
Bicoid plays crucial role in embryonic development in certain organisms (Drosophila) -> well-conserved between close related species of Drosophila, may not be conserved across all phyla
Mechanisms of embryonic development can differ among diff. organisms
What principles govern the transcriptional regulation of zygotic A-P genes?
· Cross regulation (e.g. gap genes, pair rule genes and segment polarity genes repressing or activating each other)
· Regulation by maternal gradients (bicoid, hunchback, caudal regulating gap genes)
· Chromatin Modification and Epigenetic Regulation
Drosophila embryos lacking Dorsal protein are heavily “dorsalized” (i.e. form exclusively dorsal cell types and structures) Which of the statements below is correct:
The dorsalized embryo is homozygous for a dorsal mutant allele
The mother of the dorsalized embryos is homozygous for the mutant dorsal allele
The father of the dorsalized embryo is homozygous for the mutant dorsal allele
Embryos lacking Dorsal protein are homozygous for a mutant dorsal allele -> Therefore the first statement is correct
Since dorsal is a maternal gradient the second statement is also correct - If the mother lacks functional Dorsal protein due to being homozygous for a mutant allele, the embryo she produces will be dorsalized
What is the contribution of the soma and the germline in the establishment of the D-V axis of Drosophila?
??? please refine ???
Somatic cells: maternal gene products mRNAs & Proteins E.g Dorsal, Toll
Dorsal: Contributes the genetic information for genes involved in D-V patterning
The somatic cells contribute to the formation of the D-V axis through the expression and localization of signaling molecules -> Toll receptor activated by spätzle (produced by ventral follicle cells) leading to nuclear localization of dorsal Protein on ventral side -> TF for ventral developement
The germline cells also contribute to the establishment of the D-V axis by providing maternally deposited factors in the egg -> e.g. gurken which determines Anterior - Posterior but also indirectly D-V
Please describe the BMP signal transduction pathway.
BMP binds to type 1 and 2 serine threonine receptor -> phosphrylation cascade
Phosphorylation cascade: Type 2 -> type 1 -> smads (TF, regulates severall genes)
Smad builds complex with co-smads and translocates into the nucleus
High bmp activity dorsal, ventral low activity -> gradient
Sog (DPP antagonist) gradient, graded inhibition of dpp(BMP) generates a reverse gradient of dpp signaling activity
Sog is involved in the transport from dpp to dorsal side, by binding to dpp
Please name possibilities to generate morphogen gradient. Can you give some examples from the early Drosophila development?
Bicoid gradient – source and sink gradient (diffusion) e.g. anterior posterior dev.
BMP gradient – time resolved inhibitor e.g. dorsal ventral dpp sog
Dorsal gradient – nuclear translocation signal via active transport e.g. dorsal ventral dev.
The dorsal half of an early Drosophila embryo is patterned by a BMP/Dpp signaling gradient. How does the gradient form and what is the contribution of Dorsal in the process?
Threshold dependent activation of different genes through Dorsal gradient inhibits dpp at the ventral side
Dorsal inhibits dpp
Sog is distributed in a gradient and binds to dpp preventing dpp to bind to BMP receptor
Graded inhibition of dpp via sog generates a reverse gradient of dpp signalling activity
Sog pushes dpp from the flanks to the dorsal side (amnioserosa AS)
How can you assign morphogen activity to a protein
Loss of function: Mutations, deletions in the gene -> no gradient -> no morphogenic activty ergo no morphogen
Gain of function: Overexpression
Liveimaging
Dose response analysis
Morphogen gradients regulate transcription of target genes in a concentration dependent manner. How can you explain gradient-dependent expression of GenA, B and C in the schematic bellow?
Gene A expressed at high gradient level -> inhibition of gene C
Gene C expressed at gradient threshold too low for gene A activation
Gene B active when gradient is low because the gradient acts as inhibitor for B
What is a “fate map” (Anlagenplan), and how can it be investigated?
(Early Vertebrate Development, Driever)
A fate map is a diagram or representation that illustrates the developmental destiny or fate of cells in an embryo. It shows how different regions or groups of cells will differentiate and contribute to specific tissues, organs, or structures in the fully formed organism.
Investigation by: Tracing techiques, cell ablation (removal), transplantational studies, genetic markers and imaging/microscopy
Is it possible to assign all cells of the embryo to one of the three germ layers?
No, the germ cells cant be assigned to the three germ layers, other cells can be assigned to the endo-, meso- and ectoderm.
Which 3 major cell movements constitute gastrulation?
Epiboly: process during embryonic development in which one or more cell layers spread over and cover another layer of cells. In Xenopus gastrulation, the ectodermal cells from the animal hemisphere spread over the vegetal hemisphere, covering the entire embryo. Radial intercalation of cells of the blastocoel roof drives epiboly.
Emboly: cells undergo movements such as invagination, involution, and ingression, leading to the formation of germ layers. Group of cells on the surface of an embryo rolls or folds inward to form an underlying layer. key process in the formation of the mesoderm & Endoderm. Invagination- sea urchin, Involution- frog, Ingression- fish
Convergent Extension: cellular movement during which cells in a tissue layer intercalate, causing the tissue to both narrow in one dimension and extend in another. Directed migration of ventral & lateral cells toward the dorsal midline results in anterio-posterior extension of the body axis
Please explain how gastrulation is initiated at the dorsal blastopore lip in amphibian embryos.
Cells that will become endoderm & mesoderm in the marginal zone move inside the gastrula through the blastopore by rolling under the lip as a coherent cell sheet -> Involution
How do cells move at the dorsal blastopore lip (at surface and deeper)?
Surface: involution of meso- and endoderm through blastopore by rolling under the lip as coherent cell sheet
Deeper:Tissues converge towards midline & extend along antero-posterior axis (Convergent Extension)
Ectoderm spreads downward to cover the whole embryo ( Epiboly)
Inward movement of Endoderm & Mesoderm begins dorsally & spreads laterally & ventrally to form a complete circle of involuting cells around blastopore
How do non-polar cell manage to become polarized and initiate directional movement?
Reorganization of the Cytoskeleton to establish polarity: Microtubule & Actin dynamic
Polarized Membrane domains: E.g PCP pathway components like Receptors apical & planar distribution
Cell Adhesion & ECM interactions – Integrins & Cell Adhesion molecules
Please explain the mechanism of planar cell polarity signalling!
Planar Cell Polarity (PCP) pathway maintains the coordinated polarization of cells within a tissue.
After Cell Polarity is established Frizzled receptors are activated by signaling molecules, which leads to the recruitment and activation of Disheveled (Dsh) in the cytoplasm. Activated Dsh initiates a cascade of intracellular signaling events that result in the asymmetrical distribution of signaling components within the cell. The asymmetrical signaling results in the recruitment and interaction of proteins like Vang/Stbm and Flamingo, contributing to the establishment of planar cell polarity.
PCP pathway is regulated by Canonical WNT signalling:
Wnt ligands can activate the PCP pathway
Please explain mechanisms for initial generation of the dorsoventral asymmetry in the amphibian embryo!
After fertilization cortical layer rotates towards the side of the sperm entry -> Cortical rotation establishes initial asymmetry. Corticale Rotation causes the activation of ß-Catenin on the dorsal side of the blastula and Overlap with TGF-ß/Nodal induce the formation of the Nieuwkoop Center
Wnt/β-catenin pathway, is involved in specifying dorsal cell fates.
How is the mesoderm induced?
(Early Vertebrate Development)
Check
The mesoderm is induced from the animal cap tissue. Nodal ( TGF-ß Signal) acts as a mesoderm inducer. Presence of ß-Catenin on the dorsal side stimulates nodal transcription -> Dorsal-Ventral gradient in Nodal related proteins -> Mesoderm is induced dorsally forms the Spemann Organizer. Maternal VegT, Vg1 activate expression of Nodal related proteins.
What is the "Community Effect"?
A single animal cap cell or a small number of animal cap cells in contact with vegetal tissue are not induced to become mesodermal cells and do not begin to express mesodermal markers. A minimum of 200 animal cap cells must be present for induction of Muscle differentiation.
Please explain the "Organizer-Experiment"
The experiment involved transplantation of a specific region from one embryo to another, demonstrating the ability of this tissue to induce the formation of a secondary body axis. tissue from the donor's dorsal blastopore lip was transplanted into the ventral side of a recipient blastula. transplanted organizer tissue induced the formation of a secondary embryonic axis in the recipient embryo. This secondary axis included structures such as the notochord and neural tube.
What is the "Organizer" (anatomically? molecularly? functionally?)
Anatomically: Specialized region or group of cells that plays a crucial role in pattern formation and the establishment of body axes. E.g Amphibians -Spemann Mangold localized at dorsal blastopore lip
Molecularly: Releases signalling molecules E.g: Amphibians Chordin, Noggin, Follistatin antagonize BMP signalling
Functionally: primary function is to induce the formation of specific tissues and body axes. plays a role in establishing the dorso-ventral and anterior-posterior axes.
By what types of experimental approaches have molecules been identified that mediate organizer activities?
Classic Organizer Transplantation: Transplanting a small piece of the dorsal blastopore lip or organizer tissue from one embryo to another can demonstrate the ability of this tissue to induce secondary axes, revealing the presence of organizer activity.
Tissue Explants: Analyzing the behavior of tissue explants derived from different regions of the embryo to understand the intrinsic properties of organizer tissues.
Loss-of Function Studies: Inhibiting the function of specific genes suspected to be involved in organizer activity to observe the resulting developmental effects.
or Gain of fuction studies by Overexpression
What are the main "signalling activities" of the organizer?
Wnt antagonists enable the formation of dorsal & ventral structures:
Chordin, Noggin, Follistatin
Frzb also Wnt antagonist resembles Wnt binding domain of Wnt receptor Frizzled
Please explain the principle mechanism of WNT signalling!
WNT Ligand Binding: WNT ligands bind to surface receptors E.g Frizzled
Activation of Frizzled Receptor: WNT binding induces conformational change leading to its activation
Disheveled Activation: Activated Frizzled receptors recruit & activate Disheveled
Inhibition of Destruction Complex: Without WNT signal – destruction complex active (including GSK3, APC & CK1) phosphorylates ß-Catenin -> marking it for degradation. With WNT Signal Dishevelled inhibits GSK3 – preventing phosphorylation & degradation of ß-Catenin
Accumulation ß-Catenin: unphosphorylated ß-Catenin acuumulates & migrates to the nucleus binds to TF and promotes gene expression of WNT target genes.
How does the BMP signalling pathway work?
Ligand Binding: Binding of BMP ligands (BMP2, BMP4…) to BMP receptors
Receptor Complex Formation: upon ligand binding Receptor Type I & II form a complex and Type II receptors phosphorylate & activate Type I recepors
Activated Type I activates Smads: Phosphorylation and activation of Smads, dimerize with Co-Smads & regulate gene expression
How is the BMP activity gradient generated?
Chordin, Noggin & Follistatin are BMP antagonists, synthesized & localized on dorsal side establish the gradient of BMP activity via Inhibition
BMP antagonists accumulate on dorsal side inhibit BMP activity -> High BMP ventral and No BMP Dorsal
How can you visualize BMP signalling activity directly in situ in the embryo?
By using phosphor-Smad specific antibodies -> Shows Pathway activity
Reporter Gene constructs with fluorescent proteins like GFP etc.
Can single cells in the embryo simultaneously receive two separate TGF beta signals and respond to each correctly? How?
Yes! TGF-β ligands typically bind to specific cell surface receptors. Different TGF-β ligands can activate distinct receptor complexes, allowing cells to discriminate between signals. Receptor Specificity. Different Intracellular Signal Transduction leads to activation of different Smads and therefore regulate different gene expression
Please explain how dorsoventral and anteroposterior patterning are linked during gastrulation!
Formation & Signals of the Nieuwkoop Center induce the formation of the dorsoventral axis (Cortical rotation)
Vegetal cells induce the mesoderm and the DV Nieuwkoop center induces dorsal mesoderm and the gastrula organizer
The gastrula Organizer (Spemann) responsible for pattern formation along the DV and AP axis. Gastrulation movements, including involution and convergent extension, contribute to the establishment of the anteroposterior axis.
Please explain the mechanics of neurulation!
Neurulation is the process during embryonic development in which the neural plate folds and transforms into the neural tube.
Notochord induces formation of Central nervous system by signaling the Ectoderm to form thick & flat neural plate.
Formation of neural folds and the elevation of such. Converging movement pushes neural folds upwards (n-Cadherin) -> Closure of neural tubes -> disconnection from epidermis
Does the neural anlage formed during neural induction have anterior or posterior CNS characteristics?
the neural anlage, which refers to the early neural tissue or precursor to the central nervous system (CNS), initially does not have distinct anterior or posterior characteristics. Neural induction sets the stage for the development of the neural plate, and the subsequent processes of neurulation and regionalization further define the anterior-posterior (A-P) axis
WNT signaling (and frzb as WNT inhibition) affect A-P patterning
Please explain the concept of planar and vertical signals in neural patterning!
Anteroposterior patterning in the neural plate:
—> (Spemann) Organizer - planar signals (patterning along a single layer or plane, often determining the anterior-posterior axis e.g. WNT pathway)
—> axial mesoderm - vertical signals (patterning along the vertical axis, specifying dorsal-ventral identity in tissues like the neural tube e.g. Shh specifying ventral fate)
Which mechanisms control dorso-ventral patterning in the spinal cord?
Sonic Hedgehog (SHH) Signaling: SHH is secreted by the notochord and floor plate cells, which are located at the ventral midline of the developing neural tube. SHH forms a concentration gradient, with higher levels ventrally and lower levels dorsally. Shh gradient determines neuronal populations along D-V axis of the spinal cord. Pathway actively repressed when no signal. Signal available binding to Patched & Smoothened -> Ci Protein made activator -> Transcription of Hedgehog genes.
Bone Morphogenetic Protein (BMP) Signalling: BMPs are secreted by the roof plate and dorsal ectoderm, creating a gradient with higher concentrations dorsally and lower concentrations ventrally.
Shh represses class I hox genes (pax6, pax7 ...) and activates class II hox genes (nkx2.2, nkx6.1). These hox genes interpret the shh gradient and control the differentiation of ventral neural cell fate (V0, V1, V2, V3, MN). Shh gradient determines neuronal population along d-v axis
Please describe an experiment that identified a ventral signaling center in the spinal cord.
Transplantation assay of Notochord. Notochord induces floor plate & ventral motor neurons. Notochord removed -> no floorplate & motor neurons. Notochord transplanted second floorplate & motor neurons.
Shh
Which mechanisms "posteriorize" the neural plate?
Global organization: wnt signaling —> high WNT activity on posterior, ANti-WNTs (frzb) on anterior side (forebrain)
retinoic acid gradient along A-P axis or FGF signalling promoting posterior cell fate
Interaction with organizer
Regional organization: Rhombencephalon segmentation
Which organizing centers have been identified in the early CNS?
anterior neural border
zona limitans intrathalamica
midbrain hindbrain boundary
What is neural induction? Which mechanisms mediate neural induction?
(Neural Development, Driever)
the process by which embryonic cells in the ectoderm acquire a neural fate (to form the neural plate) rather than differentiate as epidermis or mesoderm
Inhibition of BMP Signaling: Chordin, Noggin, and Follistatin: Secreted proteins from the organizer inhibit Bone Morphogenetic Protein (BMP) signaling. Blocking BMP signaling is crucial for neural induction, as BMPs are potent inhibitors of neural differentiation. (“Default model “ Permissive induction, cells without BMP inhibition differentiate into Epidermis)
FGF Signaling: Induces neural fate by inhibiting Smad phosphorylation -> Represses BMP transcription and induce BMP antagonist expression
ß-Catenin & VegT: Nuclear distribution of ß-Catenin & vegetallocalisation of VegT determines the distribution of Chd, Nog, BMP, Fgf & Xnrs (Nodal related factors)
The mesoderm also contributes cells to all organs derived from the primitiv gut tube. From which part of the mesoderm are these cells derived? What cell types do these mesodermal cells form (e.g. in the lung, the pancreas and the gut)?
(Organogenesis Neubüser)
developement of mesoderm around endoderm: lateral plate and Splanchnic mesoderm (Visceral mesoderm)
(airway) smooth mucle cells (ASMC), connective tissue
The development of which organs involves the process referred to as branching morphogenesis? What principle mechanisms can you imagine to induce branching?
-> Lungs and kidneys
Initiation of Branching -> Bud formation, outgrowth from existing structure. Kidney: GDNF binds to Ret receptors on nephric duct. Binding initiates branching -> Uretric duct. Lung: Primary Lung buds formation via ventral Nkx2.1 expression
Proliferation -> at the tip of the bud - extension into surrounding tissue, branched structure. Lung: Fgf10 expression at next branching event, negative-feedback loop Fgf10 -> BMP4, Shh. Kidney: GDNF & Ret Receptor binding
Proliferation -> at the tip of the bud - extension into surrounding tissue, branched structure
Interaction between different cell types
ECM composition & organization influence migration & differentiation
Apical-Basal polarity
Remodeling & Maturation -> cell differentiate into specific cell types, branched network differntiates into functional organ
Feed-back loops -> control branching process, Lung:
How can you explain, that at early stages of endodermal organ development in the foregut region the loss or addition of single (or a small number of related) transcription factors in the precursor population of an organ is sometimes sufficient to reprogram these cells towards the developmental program of a neighboring organ?
precursor cells in the foregut region may have common developmental origins
a specific combination of TFs form a specific organ, they overlap at some points that is why an alteration in these TF can reprogram cells to become a different organ
Signaling pathways from the surrounding microenvironment also influence cell fate decisions. Altering the activity of specific transcription factors may impact how cells respond to signaling cues, leading to a shift in developmental fate.
-> Siganls from Mesosderm affect TF in Endoderm several genes expressed at specific location leads to time dependent regionalization
The initial maintenance of an undifferentiated, proliferative progenitor/precursor/stem cell population during organ development is an important general aspect in organogenesis. What is a typical outcome if there is a premature differentiation of these cells in respect to organ size?
Premature differentiation leads to small organ size due to the depletion of the undifferentiated progenitor pool -> reduced number of cells available for further growth and development
Impaired tissue architecture & infomplete Organ formation - rely on proliferation & differentiation of precursor cells
Loss of regenerative Capacity -> reduction of progenitor pool limits self repair ability
How are lung precursors specified?
(Lung Development, Neubüser)
Nkx2.1 induces seperation from the gut tube -> lung precursor formation
Nkx2.1 accumulates ventrally by upregulation from BMP, FGF and Wnt
Wnt signalling is upregulated by RA
Sox2 inhibited by BMP -> accumulates dorsally
Noggin at dorsal side inhibits BMP -> BMP only ventrally
Which signaling pathways are involved in regulating branching morphogenesis of the lung primordium? What regulatory interactions between them do you know?
-> repetitive process with self-limiting feedback loops
Pathways involved: FGF pathway, Wnt pathway, Shh Pathway, BMP pathway
Fgf10 from mesoderm (Receptor: Fgfr2) at tips of branches induces expression of BMP4 & Shh, Fgf10 is downregulated by Bmp4 & Shh -> limits outgrowth of individual buds, important for the development of adjacent side branches
How is patterning and cell differentiation in the developing lung coupled to branching morphogenesis?
Early Patterning:
the ends of the branches grow further (directed by the mesenchymal cells that send FGF signaling) the cells outside of the FGF / BMP/Shh / WNT threshold (more proximal cells) already start to differentiate
Distal parts of each branch have proliferative progenitor populations that induce side branching. They are exposed to high levels of Fgf10 which induces Bmp4 expression
When the side branch grows cells escape the influence of Fgf10 & BMP4 which initiates differentiation to bronchial progenitor cells
They express Sox2 and initiate the differentiation programm into Basal, Ciliated, Clb & Goblet cells
ASMC (airway smooth muscle cells) differentiate from mesenchyme
Late Patterning:
Alveolar Development - Sox9 expressing distal progenitor cells can differentiate into ATI & ATII cells
What is the mechanism to initiate repair after injury of the distal airways?
Upon injury -> surviving cells expand to cover gaps -> change of cell shape induces WNT7b expression & induces Fgf10 expression in underlying mesoderm (Stem cell niche)-> cells get reprogrammed to proliferate and regenerate injured cellls
In stem cells: fgf10 produced by mesenchyme (stem cell niche) binds fgfr2b receptor & controls Wnt7b expression -> Fgf10 -> feed forward loop
Explain the most important steps of pancreas development and their molecular regulation.
(Pancreas Development, Neubüser)
Pancreas develops from Pdx1 positive endoderm in the posterior foregut region through fusion of a dorsal & ventral bud that are induced by independent mechanisms.
- Signals from the cardiogenic mesoderm induce ventral foregut-> Low dosage of Fgf signaling needed (compared to lung)
- Signals from the notochord allow dorsal pancreas development by suppressing endodermal Shh expression. Shh is downregulated by Fgf & Activin. -> Pdx1 expression
Proliferation: Feed-forward loop involving Fgf10 from pancreatic mesenchyme. Sox9 initiates expression of Fgf receptor and makes cells responsive for Fgf10 signals
Development of branched pancreas ductal system by joining of Microlumina & following Plexus remodeling
Specification:
- Early progenitor give rise to trunk & tip progenitor cells (Binary cell decision controlled by Notch pathway activity, Notch active -> Trunk cell, inactive -> Tip cells).
- Trunk cells give rise to duct cells & endocrine precursors ( bipotent, High Notch -> Duct cells, Low Notch -> Endocrine precursors (Sox9 -> Ngn3)
- Tip cells give rise to acinar cell of exocrine pancreas
-Endocrine precursors delaminate from the ducts migrate into mesenchyme cluster & form islets of Langenhans -> vascularize
Proliferation and maintenance of the pancreas progenitor cells requires FGF10 from the surrounding mesoderm
Cell types and architecture of the adult pancreas:
1. Exocrine pancreas: acinar tissue (digestive enzymes) and ducts
2. Endocrine pancreas: islets of langerhans with alpha-cells: Glukagon, beta-cells: Insulin; delta-cells: Somatostatin, PP-cells und Epsilon-cells: Ghrelin
Pancreas morphogenesis: Development of the branched pancreas ductal system
Cell type specification in the pancreas (delta - notch)
Specification of Pancreatic Progenitors:
Early in embryonic development, cells in the endodermal layer of the gut tube undergo specification to become pancreatic progenitor cells. This process is influenced by signaling molecules, including fibroblast growth factors (FGFs(10) —> upregulate to inhibit Shh), bone morphogenetic proteins (BMPs), and sonic hedgehog (Shh —> inhibited for pancreas).
Formation of the Dorsal and Ventral Pancreatic Buds:
Pancreatic progenitors give rise to two buds, the dorsal and ventral pancreatic buds, which will fuse to form the mature pancreas. The transcription factor Pdx1 (Pancreatic and duodenal homeobox 1) plays a crucial role in the specification and maintenance of pancreatic progenitors.
Signals from the aorta and vitellin veins further support pancreas development
Fusion of Pancreatic Buds:
The dorsal and ventral pancreatic buds fuse to form a single organ. This process is mediated by signaling centers and involves the coordinated expression of various genes, including Ptf1a (Pancreas transcription factor 1a).
Endocrine Cell Differentiation:
Within the developing pancreas, some cells differentiate into endocrine cells, which will later form the islets of Langerhans. Neurogenin 3 (Ngn3) is a key transcription factor involved in the specification of endocrine progenitors. These cells then differentiate into specific endocrine cell types, such as insulin-producing beta cells and glucagon-producing alpha cells.
Exocrine Cell Development:
Other cells in the pancreas differentiate into exocrine cells, primarily acinar cells responsible for producing digestive enzymes. Transcription factors such as Ptf1a and Rbpjl (Recombination signal-binding protein for immunoglobulin kappa J region-like) play essential roles in exocrine cell development.
Ductal Cell Formation:
Ductal cells, which form the pancreatic ducts, are also essential for proper pancreas function. Transcription factors such as Sox9 (SRY-Box Transcription Factor 9) are involved in ductal cell specification.
Mature Pancreas Formation:
The coordinated development and differentiation of endocrine, exocrine, and ductal cells lead to the formation of the mature pancreas. This process involves intricate signaling pathways, including Notch signaling, which plays a role in cell fate decisions.
Islet Maturation and Functional Integration:
Following the initial differentiation of endocrine cells, further maturation occurs as these cells integrate into functional islets. The maturation process involves the expression of additional transcription factors, such as Nkx6.1 and MafA, which are crucial for beta cell function.
How can you prove that signals from the notochord are necessary for induction of the dorsal pancreas bud using the chick as a model organism?
Notochord Ablation or Notochord tissue transplantation to different localization:
Surgical removal/ablation or relocalization of the notochord (tissue) in chick embryos
compare pancreas developement between control group and manipulated chick
analyze development of pancreatic buds in both groups -> Examine pancreatic buds, especially dorsal pancreas bud -> Immunostaining for expression markers like Shh & Pdx1
If removal of the notochord results in a significant alteration in dorsal pancreas development compared to the control group -> evidence that signals from the notochord are necessary for the induction of the dorsal pancreas bud in the chick embryo
Inhibition /Overexpression of Notochordal Signaling
pathway inhibitors or blocking antibodies specific to notochord-derived signaling molecules
How can you show that the Pdx1 positive endoderm of a day 8.5 mouse requires interactions with surrounding mesodermal structures for the differentiation into pancreas specific cell types?
Tissue Recombination Experiments:
isolate the Pdx1 positive endoderm and combine it with different mesodermal tissues
Removal or transplantation of the surrounding mesodermal structures and compare the effects on the mouse
genetic or chemical blocking of Signaling Pathways from the mesoderm
Live imaging techniques
FGF10 is an important signal from the mesoderm to the endoderm in both the developing lung and pancreas, but in both organs different genes are regulated by FGF10 signaling. How can you explain this?
Context-Dependent Signaling:
Target genes of Fgf signaling can be cell type specific – therefore distinct transcriptional response
Expressions of Receptors, Inhibitors and Activators of FGF10 signalling varies and there might be Crosstalk with other pathway that influence downstream gene expression
What phenotype would you expect in mouse embryos with a pancreas specific knockout of Sox9?
Sox9 initiates the expression of the Fgf receptor and makes cells responsive for fgf10 signals – feed forward loop -> (disruption leads to respicification of pancreas progenitors into liver progenitors)
Could result in overall decrease in Pancreatic size since Sox9 drives proliferation & survival of pancreatic progenitor cells -> Since Sox9 is involved in Cell type specification it could lead to altered tissue architecture, irregular cell organization, and disrupted cellular arrangements
What pancreas phenotype would you expect in mice with a Neurogenin3 knockout?
Neurogenin 3 (Ngn3) is a key transcription factor involved in the specification of endocrine progenitors. These cells then differentiate into specific endocrine cell types, such as insulin-producing beta cells and glucagon-producing alpha cells.
knock out phenotypes would have:
Loss of Endocrine Cell Differentiation
Decreased Beta Cell Mass (insulin producers —> type 1 diabetes) and alpha cell mass (glucagon)
Islets of Langerhans altered, which contain clusters of endocrine cells, may exhibit altered morphology
Since Ngn3 is particularly important for the specification of endocrine precursor cells -> Could result in absence of endocrine cells -> deficiency in Hormone producing cells
No hormone production like Insulin could lead to Diabetes Phenotype
Isles of Langerhans cells may not develop since they are made of endocrine cells
On the other hand cells would preferably differentiate into ductal cells, increased cell production of these cells
How can you find out what the Neurogenin 3-positive cells of the early pancreas primordium will give rise to at later stages, using the mouse as a model organism?
Lineage Tracing:
Utilize lineage tracing techniques to mark and track the descendants of Ngn3-positive cells over time —> genetic reporter system under control of Ngn3 promotor
Generate transgenic mouse lines that express reporter genes (e.g., green fluorescent protein, LacZ) under the control of the Ngn3 promoter
Specific marker such as immunohistochemistry or immunofluorescence on the Ngn3 pos cells
knock out mice —> inhibit or delete Ngn3 and see which cell do not deveope later
Explain the most important steps of mammalian kidney development and their molecular regulation.
(Kidney Development, Neubüser)
Development of 3 different transient kidneys that develop from intermediate mesoderm: Pronephros ( no function), Mesonephros (embryonic kidney), Metanephros (permanent kidney)
Formation of the nephric duct which elongates through proliferation -> Chemotaxis towards FGF8 produced in tail bud region
Metanephros development:
GDNF binds to Ret receptors on nephric duct. Binding initiates branching -> Uretric duct formation
Negative Regulators in adjacent cells of uretric bud inhibit GDNF (FOXC1) & Ret receptor expression (SPRY1)
GDNF also controls further branching morphogenesis
Wnt signals essential for nephron formation & branching morphogenesis of collecting ducts ( WNT11 tip positive feedback loop GDNF & RET, WNT9b acts on underlying mesenchyme -> mesenchym condensation -> WNT9b (stalk) -> ß-Catenin-> FGF8 -> WNT4. 3 Progenitor pools exposure to different concentrations of WNT9b in the stalk -> differentiation -> fusion with duct -> Nephron patterning with Glomerulus
What happens if you inactivate the GDNF Gene at different times of development? How could you achieve this experimentally?
GDNF functions at different times of development:
Prior to Ureteric Bud Formation: Initiation of Bud fails and no collecting duct system is formed
During Ureteric Bud Growth: Branching of uretric bud -> Inactivation would cause reduced collecting duct complexity with limited numbers of branches
After Bud Branching: Further Growth and Maturation compromised
Experiments: Fusion to gene switch (adding of substrate -> gene switched off), adding GDNF inhibitors during different stages, Knockout of GDNF gene during different stages of development
Wnt signaling is important for kidney development. What signal transduction pathways downstream of Wnt do you know? How could you investigate whether the canonical Wnt pathway is involved in kidney development? Discuss the advantages and drawbacks of different experimental strategies.
Wnt -> frz -> dishevelled -> destruction complex inactive -> beta catenin into nucleus (transcription)
Overexpression/Inhibition of Wnt ligands Adv: distinct phenotype Disadv: wnt signalling is everywhere -> off target effect limited specificity -> can effect multiple tissues & pathways
Fusionprotein with gene switch -> makes Wnts visible + modulating amount (kp ob das geht)
How could you visualize, in which cells of the kidney primordium Wnt signaling is activated? Discuss different possible approaches.
β-Catenin Staining
fluorescence in situ hybridization
immunohistochemistry: antibody staining
reportergene with responsive molecule e.g. beta-galactosidase
How could you visualize, in which cells of the nephric ducts GDNF signaling is active?
GDNF Staining
GDNF Reporter mice/construct
Oncogenes and tumour suppressor genes represent a major concept in tumour biology. Please define these terms and describe their different mode of action and how they are altered in cancer.
(Cancer, Brummer)
Oncogenes: Tumourigenesis due to dysregulated gene expression and/or gene product activity ->gain of function mutation, often in a dominant way (E.g. Ras,Src)
Tumorsupressorgenes: Tumorigenesis by loss of gene expression and/or function – loss of function mutations usually recessive (E.g. RB, p53)
Please give examples for oncogenes and tumour suppressor genes.
Oncogenes: Ras (H-Ras, K-Ras), Raf, Src (v-Src), Bcr-Abl, Myc
Tumorsuppressor genes: BRCA1/2, Rb, p53, p16, p21
Which genetic alterations are found in oncogenes and tumour suppressor genes?
gain of function (oncogenes hyperactive) —> point mutations, insertions, translocations
loss of function (inactivation of tumor supressor genes) —> premature stop codons, epigenetic silencing, loss of genes/chromosomes
Is just a mutation in RAS sufficient to cause cancer? What do you think?
No needs mutation in Oncogene & Tumor suppressor gene -> Otherwise tumor suppressor buffers over proliferative cell -> Example Leberflecken
Can cancer be inherited?
Predisposition for Cancer can be inherited e.g the recessive character of tumor suppressorgenes can be inherited E.g. BRCA1 Mutations
The Cancer itself, the tumors can not be inherited ( you also need 2 mutated copies to sustain cancer for recessive genes)
Why is retinoblastoma always occuring in the eye?
Retinoblastoma is a type of cancer that occurs in the retina, Mutations in the rb gene responsible for this type of cancer specifically affect retinal cells (only in the eye) leading to the development of tumors in the retina
Tumor viruses: Are tumor cells lytic? What is the the difference between slowly and acute transforming retroviruses?
tumor cells are not lytic
Acute transforming retroviruses – Produce a dominant acting oncoprotein (v-ONC) that is derived from a cellular proto-oncogene (C-onc), more rapid mechanism
Slowly transforming retroviruses – transformation by insertional mutagenesis in the promotor of proto-oncogenes, induce cell transformation gradually
The discovery of Bcr-Abl led to one of the first successfully applied “targeted cancer therapies”? What is Bcr-Abl, how does the chemotherapy work and what is the conceptual difference to chemotherapy.
Bcr-Abl is a fusion oncogene that generates a constitutively active kinase with new properties
ATP competitive Inhibitors like Imatinib block active centre of Bcr-Abl and inhibit its tyrosine activity therefore disrupts signaling pathways crucial for cancer cell survival
Difference to other chemotherapies is the specificity of targeting cancer cell growth with reduced impact on normal cells
You discover that a rare tumour type often overexpresses a receptor tyrosine kinase and this protein represents an oncogenic driver. What therapeutic approach(es) can be designed?
targeted therapeutic approaches can be designed to specifically inhibit the aberrantly activated signaling pathway. RTK Inhibitors that bind the kinase region of the RTK thus inhibits oncogenic signalling pathway.
Antibodies that specifically target the receptor
The occurrence of distant organ metastases is the main cause of cancer-related death. Which are the steps that formally describe the formation of metastases of solid tumours?
(Cancer, Hecht)
Local invasion: Tumor cells break through basal lamina & enter stroma
Intravasation: Cells invade the blood vessels
Dissemination: Cancer cells travel through the blood stream
Arrest in Microvessels: Cells adhere to blood vessel wall & arrest
Extravasation: Cells exit the blood stream & enter the tissue
Micrometastases: Cancer Cells form small clusters
Colonization: Formation of Macro metastases by proliferation -> Tumor is formed
What are the key characteristics of epithelial cell assemblies?
Apical-basal polarity
cell-cell-adhesion (tight junctions, adherens junctions, desmosome, gap junctions)
cell-matrix-adhesion (anchorage to the basal-lamina)
Basal lamina ( Laminin, Collagen IV, Integrin, Perlecan, Nidogen)
What is E-cadherin? What is its biological function in normal tissues? What is its role in tumour invasion and metastasis?
E-cadherin is an adherens junction component in epithelial cell-cell adhesion. The transmembrane component forms homophilic interactions with each other and links to the actin cytoskeleton via catenins. Ca2+ dependent.
Downregulation of E-Cadherin in Cancer leads to the disruption of Cell-cell adhesion contributing to the disintegration of adherens junctions and the breakdown of tissue structure. Downregulation associated with Epithelial- Mesenchymal Transition (EMT) -> Tumor cells can disseminate more easily and metastasize. (Or via Proteases MMPs)
What are integrins? What are their biological functions in normal epithelial tissues
transmembrane proteins that form intracellular connections to cytokeratins in hemidesmosomes. Form the extracellular link to basement membrane components (Intermediate filaments). Are Ca2+ dependent. Mediate cell-ECM interactions – aberrant integrin signaling contributes to tumor progression & metastasis
What is a basement membrane (basal lamina)? What are its main building blocks?
Specialized extracellular matrix that underlies epithelial tissue. Main components are Collagen Type IV, Laminins, Nidogens, Perlecans & Integrins
How can the E-Cadherin gene and/or protein be inactivated?
(Cancer,Hecht)
Transcriptional downregulation by transcription factors like SNAIL
Mutations
Proteolysis (Degradation by metallo proteases )
downregulation of gene activity (epithelial-mesenchymal transition [EMT])
What are the possible roles of proteases in tumor cell invasion?
Protease secretion like metalloproteases allow ECM removal & degradation therefore loss of adhesion -> Enable Tumor to break through the basal lamina, disconnect Cell-cell adhesion & create channel for further invasion (ECM degradation) -> Metastasis formation
Substrates for MMPs: Integrins, Collagen IV, Laminin, pro-MMP,
The epithelial-mesenchymal transition is a cellular program that is thought to facilitate metastases formation in the course of tumour progression. What are the molecular and cellular changes that are induced by this program and how do these changes promote metastasis?
Change in gene Expression leads to:
Loss of epithelial features -> weakend cell-cell junctions & disruption of epithelial tissue
Gain of motility -> ability to move through ECM
Gain of invasiveness -> Penetration through Basal lamina
Protease secretion (MMP-2, MMP-9) -> allow ECM components (E-cadherin) removal therefore loss of adhesion
How can sequencing of tumour genomes inform therapy decisions?
Precision oncology: Inventory of mutations that are present -> Which potential targets are present and therefore tailor-made & personalized therapy
Development of male & female gonads
Bipotential gonad with Müllerian & Wolffian duct
Gonad primordium can form ovaries & Testes depending on chromosomes - Wolffian duct maintained in males, Müllerian duct in females. Develops adjacent to Mesonephros
XY -> Sox9 induction in Gonad primordium -> Sertoli & Leydig cells -> Testosterone from Leydig cells acts on Wolffian duct, AMH leads to regression of Müllerian duct -> Male genitalia
XX -> WNT signaling activity leads to Estrogen production-> Müllerian duct forms female genitalia
pseudokinase, which is a correctly folded protein that, however, does not contain a functional kinase domain because of such a mutation. Discuss how it is possible that a pseudokinase gene still can be necessary for an organism.
Baumeister
Even though the Pseudokinase does not exert any kinase activities it could still retain its Protein-Protein Interactions and act as scaffolds, or adapters to form certain protein complexes needed for other signalling processes -> Therefore still plays a role in regulating signalling cascades and pathways.
Could act as allosteric regulators like inhibitor and activator of other kinases
targeting proteins to other compartments.
act on gene expression as transcriptional regulators or influence the activity of other TFs.
What is the difference between Atg8/LC3-I and Atg8/LC3-II? Why is this protein particularly interesting in cell biological research? How can you distinguish LC3-I and LC3-II and why is this distinction important?
Atg8/LC3-I – cytosolic form -> Conversion to LC3-II to initiate formation of autophagosome
Atg8/LC3-II – membrane bound form
Interesting for Research because they play a key role in the regulation of autophagy – stress mechanism that allows cells to adapt to nutrient, glucose deprivation -> Since autophagy dysregulation is involved in the progression of various diseases they are potential drug targets
Distinction: LC3-II has lipid extensions -> higher molecular weight (Gel electrophorese) Ratio of LC3-II to LC3-I shows progression of autophagy
Many eukaryotic transcriptional repressor proteins differ from prokaryotic repressors in the way they block transcription. Please describe the mode-of-action of a typical bacterial repressor and at least three ways, how eukaryotic repressors can work.
??
Prokaryotes: Binding of Operator region on gene (Consensus sequence) -> prevents RNA Polymerase binding -> No Transcription initiation
Eukaryotes: Binding of Promotor region of a gene in case of repressors -> binding of Silencer site
Protein-Protein Interactions with TF that act as co-repressors
Interaction with chromatin structure, Chromatin remodeling
You want to study whether a single gene contributes to long lifespan in a single tissue of an
organism.
For this purpose, design an experiment that allows you the expression of e.g. a gene like daf-
16 (encoding a FOXO transcription factor) in a single tissue of C. elegans, the nervous system.
How do you accomplish single cell expression of daf-16?
Tissue specific promotor for nervous system
TetR system: Tet repressor binds DNA in the absence of tetracycline-> Precisely, the goal is to use TetR to activate a GFP reporter gene in neurons in the absence of tetracycline, whereas addition of tetracycline will induce a conformational change in TetR that abolishes its DNA binding.
Briefly describe the general design of the two components of this system (repressor/activator gene and GFP reporter gene)
eukaryotic promotor which is neuron specific
eukaryotic 3’ UTR for Stop codon
docking site for TetR -> tetO
Fusion of TetR with Activator domain
GFP reporter gene
-> TetR binds as activator to docking sequence in promotor -> Transcription of GFP reporter gene in neurons
-> Mutation of TetR (not losing DNA binding ability after Tc binding) -> addition of Tc in cell -> DNA binding -> activating transcription
Your lab provides you with a DNA copy of a gene encoding APP, the Amyloid Precursor Protein, fused to GFP. This gene is expressed in many tissues in the mouse. You know methods to introduce this gene into mice, so that you can monitor expression and GFP localization (to the membrane). Now, in a next step you only want to express APP::GFP only in neurons. How do you accomplish this?
Tissue specific promotor
1) Think about how you could experimentally induce ER stress in the nervous systems.
2) How do you generate a UPRER reporter gene? Which gene do you choose (see lecture slides), how would you generate such a reporter gene? Which components does it need to contain to be expressed and, in particular, to be responsive to the ER stress response pathway?
1) Need C.elegans neuron specific promotor
either: mutation in APP encoding gene + ER secretion signal -> misfolded Protein -> induces UPRER
or: spliced xbp1 -> induces stress response directly (spliced because IRE1 not active, no stress signal)
2) GFP fusion to Promotor activated by hsp4 (hsp4 expression only induced by ER stress) -> Reporter gene expressed in all cells + only when ER stress (Neurons -> Intestine)
We have seen that mechanistic target-of-rapamycin complex I (mTORC1) is a negative regulator of autophagy.
a) What is the main substrate of mTORC1 in the control of autophagy? Describe in few sentences how this regulation works.
b) Another phosphorylation target of mTORC1 is the protein 4E-BP. This affects gene expression in a rather general way. Describe in few words how phosphorylated 4EBP interferes with gene expression (interactor, role of the interactor, consequences: which step(s) in gene expression is/are affected?)
a)
Main target of mTORC1 is ULK1. If there are plenty of nutrients mTORC1 is active and inactivates ULK1 by phosphorylation. Phosphorylated ULK1 cannot activate Autophagy Initiation.
Low nutrient levels -> mTORC1 activity inhibited by AMPK – prevents ULK1 phosphorylation. Release of the Ulk1 complex -> Activation of ULK1 via AMPK phosphorylation-> activates multiple downstream proteins initiating Autophagy and formation of Autophagosome
b) Dephosphorylated - binds and inhibits Translation Initiation Factor eIF4E -> no binding of eIF4E to 5’ cap -> no translation
Phosphorlation of 4EBP by mTORC1 - cannot inhibit eIF4E -> Translation active
Human geneticists have identified a polymorphism in the human gene Ras and found that this polymorphism correlates with an increased likelihood to develop cancer. Ras is represented by homologous genes in all known multicellular model organisms, its C. elegans name, e.g., is let-60. Your bioinformatician provides you with the Ras cDNA and protein sequences and information about the nature of the polymorphism. You want to study the function/dysfunction of this gene (or its homolog) in a reverse genetics approach using a model organism, either Drosophila or C. elegans.
a) What is a gene polymorphism? Why does a gene polymorphism not always interfere with the function of the affected protein?
b) Describe four principal ways to manipulate, using reverse genetics, the activity of this gene, its isoform, or its homolog in your model organism and one advantage or one disadvantage of each method.
c) Which information do you need in addition, if you want to find out whether the particular mutation also affects the function of the homologous gene in Drosophila or C. elegans?
a) Multiple variations (alleles) within a population ( at least 1 %) at one specific locus. E.g SNPs (Single nucleotide Polymorphisms), Insertion/Deletion Polymorphism, Tandem Repeats (Microsattelites), Copy number variations (CNV)
doesn’t always affect function: - variations in non-coding regions, regions that do not effect the proteins function, Silent-mutations.
b) Gene Knockout - Advantage: distinct Phenotype, Disadvantage: Gene Knockout might be lethal
Gene Silencing- Advantage: Reversible, Disadvanatage: Challenging across tissues
Different protein signatures are required to target a protein to (a) nucleus, (b) endoplasmic reticulum, (c) mitochondria, (d) extracellular space. Describe the nature of the respective targeting signal and how this signal is interpreted by the cell (what are the proteins recognizing the signal?).
d???
a) Nucleus: Nuclear localization signal (NLS, typically positively charged Aa residues like Lysine & arginine) is recognized by nuclear import receptors (Importins) and binds the NLS containing cargo protein in the cytosol and facilitates translocation through nuclear pore complex
b) ER signal sequence (rich in hydrophobic AA, L,V,I,F…) is recognized by the signal recognition particle (SRP) and halts protein synthesis. SRP guides ribosome complex to ER where the complex binds SRP receptors. SRP receptor is released and complex changes onto a protein translocator. The synthesis resumes and peptide is transferred across membrane
c) Mitochondria: Targeting signal with mitochondria targeting sequence (MTS). Signal sequences are amphiphilic a-helices with positive charge on one side and nonpolar on the other. Signal sequence recognized by outer membrane receptor (TOM). Associated protein translocator transports signal sequence across outer Mito. Membrane to IM space- signal sequence recognized by 2nd Translocator in inner membrane (TIM) which transports protein across both membranes
d) Exocytose ??
The sensor for the mitochondrial stress response, the transcription factor ATFS-1 (from C. elegans, functionally similar to human ATF4/5 and CHOP), contains both a mitochondrial and a nuclear localization signal.
a) Where in the amino acid sequence of the protein would you expect the mitochondrial localization signal to be localized? Describe principle features of the mitochondrial and nuclear localization signals.
b) Describe what will happen to ATFS-1 variants that have been engineered to remove one of these targeting signals?
c) By default, ATFS-1 mitochondrial targeting signal dominates over the nuclear targeting signal. Discuss ways how the cell could mask a nuclear targeting signal. Do you know other transcription factors in which nuclear access can be temporarily masked? Describe the underlying principles.
d) If ATFS-1 gets transported into the mitochondria, it becomes target of the mitochondrial matrix protease LONP-1. Describe how the mitochondrial import machinery contributes to allowing physical contacts between LONP- and ATFS-1.
e) Lowering the efficacy of the electron transport chain will typically prevent mitochondrial import of proteins. Discuss what will happen to ATFS-1 in such a case.
a) Mitochondrial localization signal in ATFS-1 is probably located at the N-terminus -rich in positively charged amino acids, creating an amphipathic helix -> interaction with negatively charged mitochondrial outer membrane.
Nuclear Localization Signal (NLS): rich in basic AA (Lysine, Arginine)
b) predominant entering of either Mitochondria or Nucleus
when ATFS-1 is imported into Mito. -> it gets degraded and if it enters Nucleus the TF activates UPRmt by transcription activation
c) Post-Translational modifications or Protein-interactions that mask nuclear signal -> Mitochondria direction
d) Import machinery via Translocase on outer membrane (TOM) & inner membrane (TIM) transports ATFS-1 acrosss mitochondrial membranes -> allowing Mitochondrial matrix protease (LONP-1) to interact with ATFS-1 -> Degradation
e) Accumulation in the Cytoplasm leads to translocation in the nucleus & triggering of nuclear stress response genes
1.A genetic cross is being performed between C. elegans mutants daf-2 and daf-16. The phenotypic characteristics of each mutant are displayed below. Daf-2 = daf-2 / daf-2. daf-2 is localized on chromosome III, daf-16 is localized on chromosome I. The F1 generation of this cross is phenotypically inconspicuous (no phenotypic differences from wild type can be observed).
a)How do you interprete this result with respect to the genetic characteristics of the mutants involved?
b)Describe the genotypes you expect to get in the F1 generation progeny of this cross.
c)The F2 generation phenotypes are a mixture of: - wild type animals - dauer-constitutive animals - dauer-defective animals You will isolate 100 progeny of the F2 generation. Which segregation (representation by percentage) of phenotypes do you expect to get among the F2 animals?
d) In an alternative cross of two mutants, unc-2 and daf-16, you receive, in addition to Unc and Daf-d mutants, also Unc Daf-d double mutants. Describe why you do not receive F2 generation progeny with a Daf-c and Daf-d double mutant phenotype in the daf-2 x daf-16 cross. Please provide a genetic interpretation of this effect.
a) F1 generation is not homozygous mutated for daf-2 or daf-16 therefore you do not get one of the 2 phenotypes. Functional copy of either one of the alleles compensates loss of function in the other
b) The F1 generation will inherit one allele from each parent. Possible F1 Genotypes: Daf-2/daf-16, daf-2/+ (WT allele of daf-16), +/daf-16 (WT allele of daf-2), +/+ (WT)
Inheritance of daf-2 allele, daf-16 allele or respective WT alleles -> 4 possible genotypes
c) Possible genotypes:
WT phenotype- 2/+, +/2, +/+, 16/+, +/16
Daf-2( dauer constitutive) phenotype: 2/2
Daf-16 (Dauer-defective) phenotype: 16/16
d) Compared to Daf-2 & daf-16 mutants that generate opposing phenotypes you get unrelated phenotypes for Unc-2 & daf-16 crossing and therefore get a combination off unc-2 & daf-16 phenotype. The underlying genetic mechanism for the daf-1, daf-16 cross is called Epistasis where mutations of 1 gene supresses the effect of mutations in the other gene therefore one phenotype prevails over the other.
The protein Cas9 from the bacterium Thermus thermophilus belongs to type III Cas systems and cleaves single-stranded RNA (instead of double-stranded DNA), but otherwise functions very similar to DNA-cleaving Cas9. You want to express this Cas9 from genomic DNA of Thermus thermophilus to express it in the nematode C. elegans to allow cleavage (and, thus, destruction) of the mRNA of daf-2, exclusively in the intestine.
a) Describe a standard method to allow expression of Tth Cas9 in C. elegans intestinal cells.
b) What, in addition to the expression of Cas9, do you also need to do in order to get this CRISPR/Cas9 system working in the worm?
c) How do you monitor efficacy of this Cas9 function in the worm?
????
a) Construct Cas9 expression plasmid with a tissue specific promotor that drives expression in C.elegans – Include marker (Fluorescent or drug) to identify & selecting transgenic worms expressing Cas9 – microinjection of plasmid
Zuletzt geändertvor 13 Tagen