Midterm REVIEW cards Flashcards
What is epigenetic regulation?
Transcription in eukaryotes takes place on DNA that is wrapped in chromatin. Chromatin
needs to open for a gene to be activated and transcription to proceed. Chromatin-
mediated regulation is an entirely new mechanism as compared to prokaryotes. It is often
called EPIGENETIC REGULATION of gene expression
heterochromatin
regions of chromosomes that are intensely stained
- DNA is more densely packed
- rich in repetitive DNA (Transposons, centromeres
and telomeres)
- not accessible to transcriptional machinery
- Inactive genes are found in heterochromatin
Euchromatin
- lightly stained chromosome regions
- Active genes are found in euchromatin
- accessible to transcriptional machinery
What kind of RNA does Polymerase I transcribe?
Pre-rRNA (28S, 18S,
5.8S rRNAs)
(These are Ribosome components, used for protein
synthesis)
What kind of RNA does RNA pol II transcribe
mRNA - Encodes protein
snRNAs - RNA splicing
siRNAs - Chromatin-mediated repression,
translation control
miRNAs - Translation contro
What kind of RNA does RNA pol III transcribe?
tRNAs - Protein synthesis
5S rRNA - Ribosome component, protein
synthesis
snRNA U6 - RNA splicing
7S RNA - Signal recognition particle for
insertion of polypeptides into the
endoplasmic reticulum
Other small stable
RNAs with Various functions, unknown for
many
How many polypeptides does RNA pol II consist of?
12
The 5 main components of RNA polymerase - and their functions?
- Clamp domain - opens to accomodate DNA and closes/swings shut during transcription to anchor polymerase to DNA
- Bridge - closes clamp domain
- Catalytic centre - where the synthesis of RNA takes place (with the participation of Mg++)
- Channel - where newly synthezed RNA exits and then is immediately capped by 7MGuanosine
- Carboxyterminal domain - domain involved in many regulatory interactions and plays a key role in initiation, release and elongation of synthesized mRNAs
Carboxyterminal Domain
part of RPB1 subunit
This is a specialized domain not found in any other polymerase, prokaryotic or eukaryotic.
* The CTD is involved in multiple regulatory interactions and plays a key role in the initiation,
release, elongation and processing of the synthesized mRNAs
* The CTD in yeast contains of 26 repeats of Tyr-Ser-Pro-Thr-Ser-Pro-Ser
– in mammals it contains 52 repeats
* The Ser residues in the CTD are phosphorylated upon transition from initiation to
elongation
* The CTD is not structured. It is not shown on the previous two or three slides because it
can not be analysed by crystallography!!
Core promoter sequences
TATA box – a tight consensus sequence
o prevalent in highly transcribed genes
- -31 to -26
- Initiator – less conserved element
o some genes contain Initiator but no TATA - -2 to +4
- BRE (TFIIB Recognition Element) and/or
o influence the activity of the promoter - -38 to -32
- DPE (Downstream Promoter Element)
o influence the activity of the promoter
+28 to +32
What techniques do we use to analyse these processes?
- DNA sequencing
- PCR
- RT-PCR
- RNA sequencing
Techniques with antibodies: - immunofluorescence, immunoprecipitation, ChIP
PCR
Polymerase Chain Reaction
PCR: amplifying DNA by repetitive cycles of denaturing and renaturing of
DNA in the presence of thermostable DNA polymerase (Taq polymerase).
* You need two primers that anneal to the ends of the amplified DNA
fragment at 50 - 60°C.
* Taq polymerase synthesizes new strands of DNA starting at the 3’-ends of
the annealed primers (72°C).
* Newly synthesized DNA is denatured at high temperature (95°C).
* The temperature is lowered (50 - 60°C) and more primers anneal to the new
strands.
* The cycle is repeated 30 times. This means that if you start with one
molecule of DNA you will end up with 230 molecules
RNA sequencing and qRT-PCR
QUANTITATIVE
techniques for the analysis of gene expression
9
* RNA is isolated from a sample
* RNA is converted to DNA by the use of specific primers directed to
a specific gene and RNA-Dependent-DNA Polymerase
RT-PCR (Reverse Transcription-PCR)
* Use the produced DNA in a PCR reaction with Taq polymerase and
the specific primers directed to a specific gene
* By quantifying the DNA produced by PCR you indirectly quantify
the abundance of the corresponding RNA in the sample
RNA sequencing
RNA is isolated from a sample and converted to DNA by the use of random primers and
RNA-Dependent-DNA Polymerase
* Break the produced DNA in small (200 bp) pieces.
* Sequence the DNA by Massive Parallel DNA sequencing
* The produced sequences are analyzed by a software and aligned to the sequence of the
genome (see the animation on the next slides)
* The number of sequences of that align to each locus in the genome are quantified and
then plotted.
* Resolution in the plot is very high, sometimes within a base.
* The plot is giving a quantitative presentation of the levels of transcription at each
position of the genome.
- In lecture 4 (Fig. 9-17) this technique is used for the analysis of uni-directional and bi-
directional transcription from a eukaryotic promoter
What are antibodies?
Antibodies are natural immunoglobulins produced by animals to combat
invading exogenous proteins of any kind
* A specialised class of B-lymphocytes rearrange the Ig genes
- Each B-lymphocyte produces ONE UNIQUE ANTIBODY against an exogenous
protein (THE ANTIGEN). - Upon invasion by an antigen the B-lymphocytes recognise this antigen,
proliferate and produce large amounts of the antibody to destroy it
monoclonal antibody
The antibody produced by one B-lymphocyte (one clone)
polyclonal antibodies
In the blood of the animal with multiple B-lymphocytes produce multiple
antibodies.
Structure of an antibody
ntibodies consist of two heavy and two light chains, joined by disulphide bonds.
Each light and heavy chains consist of a constant domains and a hyper-
variable domain. In the complete antibody the hyper-variable domains of the heavy
and lights chains come together to form the antigen binding site.
Epitopes
the part of the antigen that the antibody recognizes and binds to via the antigen binding site/paritope!
epitopes can be:
- Linear, made up of continuous sequence of amino acid
* Conformational made up of animo acids close together in the folded structure of
the protein
Production of antibodies against a desired antigen
We can “trick” an animal (mouse, rabbit, sheep, donkey) by injecting it
with a protein (an antigen) of choice. The animal will respond by
producing multiple antibodies against the antigen.
* We can take out some blood from these animals, purify the
immunoglobulins and prepare POLYCLONAL ANTIBODIES AGANST THE
ANTIGEN
* We can isolate single clones of B-lymphocytes, maintain them in culture
to produce MONOCLONAL ANTBODIES
* Now we have highly specific antibodies against a protein of interest.
We can use these antibodies for various techniques of identification and
quantifying
Immuno-fluorescence
We can use specific antibodies coupled to
a fluorescent dye and localize the antigen in the cell
1) prepare sample and place on microscope slide
2) incubate with primary antibody
3) incubate with fluorochrome-conjugated secondary antibody: wash away unbound antibody
4) mount specimen and observe in fluorescence microscope
Immuno-precipitation
We can hook the antibodies to large
beads and mix them with extract, then wash away the extract.
The antigen (and its associated proteins) remain associated with
the beads via the antibody
1) primary antibody is added to mixed protein solution
2) agarose beads are added which form a complex with ab-antigen
3) centrifuge and wash to separate antigen complex from mix
4) elute antigen (protein of interest) and detect via western blot
ChIP
Chromatin Immuno-Precipitation
- multi-step technique that detects the binding of a specific protein to a specific DNA element (or multiple DNA elements) in vivo
ChIP steps
1) treat living cells with a membrane-permeating cross-linker such as formaldehyde
2) sonicate to shear cellular chromatin to short fragments and add antibody to pol II
3) immunoprecipitate to isolate pol II cross-linked to dna
4) revers cross-linking, isolate DNA, and subject to massively parallel DNA sequencing
Formation of the PIC steps:
1) association of TFIID to the promoter
2) followed by TFIIB and TFIIA
3) RNA polymerase is recruited and The non-phosphorylated CTD
(Carboxy Terminal Domain) of
RNA pol II establishes contact
with several GTFs
4) TFIIE and TFIIH are recruited
5) TFIIH helicase activity facilitates
the opening of the double helix of
DNA: Open PIC
6) RNA Polymerase II initiates
transcription
7) TFIIH kinase activity
phosphorylates the CTD to release RNA polymerase from
the promoter
Transition from initiation to elongation steps / pausing steps
After the initial phosphorylation of CTD by TFIIH kinase,
1) two negative
elongation factors, NELF and DSIF, associate with RNA polymerase II
2) they pause it downstream of the initiation site.
3) * At this point another kinase, CDK9/CycT (also called PTEFb (Positive
Transcription Elongation Factor b) phosphorylates the CTD and NELF.
4) * RNA polymerase II with fully phosphorylated CTD is released
5) * Positive elongation factors PAF and Spt16 join the polymerase.
6) * DSIF becomes a positive elongation factor
What happens in HIV/AIDs with Tar and Tat
The 5’ end of the short RNA is synthesized before the polymerase pauses
* The short RNA forms a secondary structure called TAR. TAR holds the polymerase and has an
inhibitory effect on Cdk9/CycT.
* HIV1 (human Immuno-deficiency virus) encodes the protein called Tat. Tat binds to TAR.
* Upon cell stress other factors release the polymerase and some Tat is made.
* When bound to TAR, Tat ACTIVATES CDK9/CycT kinase and releases the paused RNA
polymerase II
* Now more Tat is made and viral genes are fully expressed
what is the key to HIV latency
HIV is dormant but activates upon stress when the organism is
weak, slowly killing T-cells and abolishing immunity
* The patients die from lost immunity and any opportunistic infections
The Tat protein of HIV holds RNA polymerase II until conditions for the spreading of the virus are favorable
TATA-containing promoters vs. CpG islands
At most TATA-containing and TATA-less promoters transcription initiates
and proceeds in one direction
* !!!Your textbook does not have a good illustration on this type of uni-
directional transcription initiation. The figure shown on the next slide
is from the 8th edition of the textbook.
- At other promoters (called CpG islands) transcription initiates in both directions, but the polymerase stalls and falls off DNA in one of the directions and then proceeds in the other direction only
WHat are CpG islands
CpG islands occupy the promoters of about 70%
percent of the genes in vertebrates. These genes
are transcribed at a low rate.
* CpG rich DNA contains less nucleosomes and are easier to transcribe.
* At CpG islands transcription initiates at any position (not defined) within the island.
* Transcription from CpG island initiates in both directions, but proceeds in the direction of the
Open Reading Frame (ORF) only.
How do we find gene regulatory sequences?
Linker scanning-reporter gene technique for identifying gene regulatory elements on DNA
What is linker scanning mutagenesis?
You buy a plasmid that contains a reporter gene, usually a gene that encodes an enzyme (tk in figure 8-17)
The tk ORF (Open Reading Frame) on the plasmid has no promoter.
Upstream of the tk ORF you clone a large piece of DNA that you know is controlling YOUR gene of interest (not tk )
You systematically replace small pieces (10 bp) of the cloned regulatory DNA by a “linker”.
You produce multiple plasmids with linker substitutions (9 in figure 8-17)
You transform each individual plasmid in a cell culture and measure the activity of the reporter (a simple enzyme reaction of cell extract)
Loss of reporter activity means that you have hit a regulatory sequence
Reporter plasmids contain:
- an ORF for an enzyme (Luciferase (luc) in this case)
- NO promoter-proximal regulatory sequences
- A multiple cloning site (MCS) for the cloning a regulatory sequence
- Translation initiation elements upstream of the reporter gene
- Poly A+ signal downstream of the reporter gene
linker scanning mutation steps
- Eukaryotic promoter region DNA that supports high-level expression of a reporter gene (light purple) is cloned in a plasmid vector.
- Overlapping linker scanning (LS) mutations (insertions of scrambled sequence) are introduced from one end of the region to the other.
- Mutant plasmids are transfected separately into cultured cells.
The expression/activity of the reporter gene product (thymidine kinase gene) is assayed.
Enhancers
distal regulatory elements,
could be upstream or downstream of the initiation site
position independent
orientation independent
promoter proximal regions
Function within a short distance (several hundred bps from the transcription initiation site
Immediately upstream or downstream from the initiation site
Position dependent: usually non-functional if moved
How do enhancers communicate with promotors
via bending DNA
Pioneer transcription factor
Binds to a specific regulatory sequence within the condensed chromatin
Interacts with chromatin-remodeling enzymes and histone acetylases that decondense the chromatin, making it accessible to RNA polymerase II and general transcription factors.
Activator proteins
Bind to specific transcription-control elements in both promoter-proximal sites and distant enhancers
Interact with one another and with the multisubunit Mediator complex to assemble general transcription factors and RNA polymerase II (Pol II) on promoters
Transcriptional activation
Pol II initiates transcription.
Pol II pauses after transcribing fewer than 100 nucleotides because of action of the elongation inhibitor NELF (negative elongation factor) associated with DSIF (DRB sensitivity-inducing factor).
Activators in transcription
Promote association of the Pol II-NELF-DSIF complex with elongation factor P-TEFb (cyclin T-CDK9 [kinase]), which releases NELF
NELF release allows resumption of RNA transcription.
How can transcriptional activators and repressors be identified?
by deletion analysis of the protein
What are the two main domains of transcriptional activators and repressors?
Transcriptional activators or repressors often have independent DNA-binding domains and effector (activation or repression) domains
Details of deletion expereiment for transcriptional activators and repressors
Activators are composed of distinct functional domains.
(a) Reporter-gene construct containing a lacZ reporter gene (encoding β-galactosidase) and a TATA box ligated to UASGAL, a regulatory element that contains several Gal4-binding sites.
The reporter-gene construct and DNAs encoding wild-type or mutant (deleted) Gal4 introduced simultaneously into mutant (gal4) yeast cells.
Activity of β-galactosidase expressed from lacZ was assayed.
High activity if introduced GAL4 DNA encodes a functional protein.
(b) Wild-type and mutant Gal4 — results:
N-terminal end 50 amino acid deletion:
Eliminates UASGAL binding.
Contains DNA-binding site.
C-terminal end deletions:
β-galactosidase expression activation eliminated only when between 126 and 189 or more amino acids deleted from the C-terminal end.
End contains activation domain.
Internal deletions — Gal4 central region is not crucial for its function in this assay.
Motif
Motif is a sequence in a protein that, based on similarity with other known functional domains, is likely to produce a distinct 3D structure and a distinct domain.
- WITHIN DOMAINS
ex. zinc-finger, leu zipper
These motifs are easy to recognize by bioinformatic tools
What is a domain
DOMAIN is a portion of a polypeptide with a specified function: Transactivation Domain, dimerization domain, dna binding domain, hormone binding domain.
The domains are identified by deletion analysis of actual peptides
Zn finger motif
regularly spaced C and H amino acids
have a zn2+ in middle
in DNA binding domains
leu zipper motif
is found in protein dimerization domains: regualrily spaced L amino acid residues:
HLH motif
helix turn helix motif
found in many dimerization domains
Dimers
This means that if two proteins (green and yellow) can combine, they will have three binding sites (gg, yy, gy), three proteins (green, yellow, blue) can have six binding sites (gy, gb, yb, gg, yy, bb)
These six combinations can recognize six DNA binding sites
Adding an inhibitor for one of the three proteins increase the possible outcomes for activation (sort of a ‘fine tuning’)
Enhanceosome
Combinatorial binding to DNA-binding proteins from different classes
Eukaryotes have very complex promoters and enhancers.
In these, various transcription activators and repressors can bind
The transcriptional activators or repressors often interact with each other and cooperate to establish a strong activator or repressor site to tightly control the expression of the target genes
What are silencers?
There are positions in the genomes where large heterochromatin blocks are established and maintained
We will call these positions “SILENCERS”
Genes that are close to these SILENCERS are strongly repressed
The repression of the genes depends on their position (close to, or within heterochromatin) relative to the silencer and does not depend on their promoters.
What are the two mating type loci in yeast?
HML and HMR
they are constantly repressed thanks to Rap1 and Sir proteins
How does acetylation of histone tails affect the activity of genes?
Acetylation of the histone tails reduces their positive charge, allows for the opening of chromatin (euchromatin) and for transcription
Deacetylation of histone tails reconstitutes positive charge, strengthens DNA-histone interactions, compacts chromatin(heterochromatin) and suppresses transcription
How does Rap1 and Sir proteins regulate the repression of HML and HMR?
Two mating type loci , HML and HMR, are constantly repressed
Genetic studies identified proteins that regulate this repression
Rap1 binds next to the repressed loci and recruits Sir (Silent Information Region) proteins
Sir proteins spread away and cover the chromatin of nearby genes
Sir2 is a HISTONE DEACETYLASE. It deacetylates the histones within HML and HMR
The deacetylated histones bind tighter to DNA and form heterochromatin
If the genes in HML and HMR are moved away from HML and HMR loci, they become active
Conclusion: Histone hypo-acetylation is necessary for positional gene repression
Lysine to arginine substitution in histone tails result
PERMANENT GENE REPRESSION
Arginine retains a positive charge only, can not be acetylated.
The DNA-histone interaction is strong, chromatin is compact, gene repression can not be reversed by acetylating the histones
These mutants maintain permanent gene repression at telomeres.
Lysine to glutamine substitutions in histone tails affect
CANNOT BE DEACETYLATED - CANNOT BE REPRESSED
Glutamine has a neutral charge, resembles acetylated Lysine, can not be deacetylated to gain a positive charge
Chromatin is decondensed, gene can not be repressed.
These mutations maintain permanent gene activation at telomeres
How are genes silenced at the telomeres of S. cerevisiae?
Rap1 binds directly to the telomeric repeats
SIR proteins are recruited by Rap1
Sir proteins spread in the sub-telomeric regions and cover them with condensed hypoacetylated nucleosomes
More detailed:
Many telomeres cluster at the periphery of the nucleus and form large 3D heterochromatin block
RAP1 –
binds HML and HMR silencer sequences that are repeated multiple times at each yeast-chromosome telomere
nucleates formation of heterochromatin
SIR2 – histone deacetylase that removes acetyl groups on histone tail lysines
RAP1 and SIR2, 3, and 4 proteins – bind to each other
SIR3 and SIR4 – bind to histone H3 and H4 N-terminal tails that are maintained in a largely nonacetylated state by SIR2 deacetylase
What does that telomere and sir protein light up diagram represent?
(b,c) Yeast cells labeled with:
(b) a telomere-specific hybridization probe and
(c) fluorescent-labeled specific anti-SIR3 antibody
SIR3 – localized in the repressed telomeric heterochromatin. (RAP1, SIR2, and SIR4 also colocalize with the repressed telomeric heterochromatin.)
What is rap 1
the REPRESSOR
binds to the silencer region of the dna and recruits the corepressors
What are sir 3 and sir 4
co-repressors - they bind to H3 and H4 N-terminal tails (deacetylated)
What is sir 2?
a histone deacetylase - removes acetyl groups on histone tail lysines
What type of repression is RAP1 and Sir
Position-dependent repression of genes
The repression of the genes depends on their position (close to, or within heterochromatin) relative to the silencer and does not depend on their promoters
these positions are called silencers
If the genes in HML and HMR are moved away from HML and HMR loci, they become active
Conclusion: Histone hypo-acetylation is necessary for positional gene repression
Promoter dependents repression and activation
promotor-dependent and position-independent regulation also works via histone acetylation/deacetylation, however different enzymes and different co-repressors and co-activators are employed
EX. Ume6 and Rpd3
The repressors and activators bind to promoters and enhancers and recruit the co-repressors or co-activators
The co-repressors and co-activators do not spread, they stay localized at the regulatory elements
In these situations the position of the genes is not a critical factor; the promotor is.
What kind of enzyme complex will Gcn4 recruit? Which specific domain of Gcn4 will interact with this enzyme complex? What would be the consequence of its activity?
1) the SAGA complex - a histone hyperacetylase
2) the activation domain
3) ACTIVATION of the genes with the UAS promoter
What kind of enzyme complex will Ume6 recruit? Which specific domain of Ume6 will interact with this enzyme complex? What would be the consequence of its activity?
1) Rpd3L - a histone deacetylase
2) the repression domain (RD)
3) Repression of the genes with URS1 promoter
Primary sex traits
changes in the body part of reproduction (menstration)
What kind of enzymes do transcriptional activators of SAGA and Gcn5 complex recruit?
Histone acetyl transferases
and a class of co-activators called chromatin remodeling complexes
What do chromatin remodeling complexes do?
Chromatin-Remodeling Complexes then use ATP to push nucleosomes along DNA to “open” promoters
ex. SWI and SNF
What are SWI and SNF?
Chromatin remodeling factors
SWI/SNF (switching/sucrose non-fermenting) ATPases
Move nucleosomes along the DNA by sliding or transferring them
Shift nucleosomes away from the promoter/enhancer sites and gives transcription factors access to the DNA
Chromatin-Remodeling Factors use ATP to move nucleosomes
What is the mediator
an example of a co-activator
(a major “bridging” factor between transcriptional activators, GTFs, histone modifiers and chromatin remodelers)
The Mediator is a huge complex containing variety of proteins including enzymes, proteins that recognize activators, proteins that recognize GTFs and other coactivators
Different mediator complexes contain proteins required for specific genes
The Mediator(s) directly interacts with transcriptional activators
One mediator complex can interact with multiple transcriptional activators
The Mediator(s) directly interacts with GTFs
Transmission of memory by epigenetic means
Cell differentiate and select the genes they express.
Then the cells divide multiple times but continue to express the same genes after each cell division.
DNA replication is a major disruptor of chromatin and bound transcription factors.
The “memory” of gene expression is achieved mainly through reconstitution of the same chromatin structure after the passage of the replication fork
We call this epigenetic memory of transcription
epigenetic marks
Epigenetic control of transcription is maintained by DNA methylation and methylation and acetylation of histones.
We call these epigenetic marks
Epigenetic marks are faithfully rebuilt during and/or soon after the passage of the replication fork
Epigenetic marks on histones
(Histone post-translational modifications (PTMs): Methylation, Acetylation, Phosphorylation
which is the only methylated histone site that leads to repression
H3 (K9, K27) Me2/3
DNA methylation
Methylation of CpG islands leads to condensation of chromatin
Methylated DNA recruits special class of Me-DNA-biding proteins (MeBPs).
MeBPs recruit factors that deacetylate histones and condense chromatin
IMPORTANT: the methylation of DNA is rebuilt immediately after the passage of the replication forks
At what levels do noncoding RNAs regulate gene expression?
At the levels of transcription, RNA processing and translation
miRNA
Short interfering RNAs, 22 bp long, that base-pair extensively, but not completely with mRNAs, especially over bases 2 to 7 at the 5’ end of the miRNA (the seed sequence)
this pairing inhibits translation of the target mRNA and targets it for degradation
miRNAs
microRNAs, 22 bases long, base pair extensively, but not completely (imperfect match) with mRNA, especially over bases 2 to 7 at the 5’ end of the miRNA
THis pairing inhibits translation of the target mRNA
What modifications happen to premRNA to produce a mature mRNA
Pre-mRNA (the primary transcript) is capped, then spliced, then polyadenylated before being exported to the cytoplasm
splicing
means linking two ropes without a knot
A large ribonucleoprotein spliceosome complex catalyzes the joining of two exons
Alternative splicing
and alternative cleavage at different poly(A) sites yield a significant variety of different mRNAs from the same gene in different cell types or at different developmental stages.
Some of the proteins generated by alternative splicing have drastically different activities
Two examples of alternative splicing in in
1) Alternative splicing in neurons
Perception of sound
Neuron connectivity
2) Sex determination in Drosophila
Dscam isoform expression in Drosophila neurons
Most extreme example of regulated alternative RNA processing yet discovered
Helps to specify tens of millions of different specific synaptic connections between neurons in the Drosophila brain
(bottom) Dscam neuron cell-surface protein:
10 different immunoglobulin (Ig) domains (ovals)
6 different fibronectin type III domains (rectangles)
One transmembrane domain (yellow)
C-terminal cytoplasmic domain
Fully processed mRNA contains:
One of 12 Ig2 exons
One of 48 Ig3 exons
One of 33 Ig7 exons
One of 2 transmembrane exons
Alternative splicing – can generate 12 x 48 x 33 x 2 = 38,016 possible isoforms
Dendrite self avoidance wildtype vs. dscam mutants
Dendrites are branches that extend out of the neural cell bodies
The branches of the same neuron repel each other while overlapping with branches from other neurons
This is controlled by Dscam
Dscam mutants show evidence of reduced dendrite self-avoidance
autism vs. normal spicing
In one-third of patients with autism spectrum disorder (ASD), more skipping of cassette exons and microexons are observed than in normals
Abnormalities in the frequency of splicing of alternative 5′ and 3′ splice sites are observed only rarely in ASD.
What is sxl
Sxl is an RNA binding protein, acts as a suppressor of splicing
- present in females
it suppresses the splicing of sxl gene btw exons 2 and 3
and it suppresses the splicing of tra protein btw exons 1 and 2
Tra
Tra is an RNA binding protein, acts as an activator of splicing
activates splizing of dsx
Dsx
is a transcription activator/repressor
activates transcription of genes in females encoding female parts
represses transcription the transcription of those genes in males
What other protein regulates splicing of Dsx
Rpb1 (along with tra) form a complex that activates splicing of the Dsx gene
Tra-Rbp1-Tra2 complexes interact with six exonic splicing enhancers in exon 4.
Activates exon 3–4 splicing
Lin-4, Let-7
The development of C. elegans is regulated by multiple lineage (LIN) determining and lethality (LET) genes
Interestingly, the genes lin-4 and let-7 genes are NOT encoding for any protein
They produce small RNAs that are complementary to other mRNAs
The imperfect binding of the lin-4 or let-7 micro RNAs (miRNAs) to the 3’-UTR (untranslated region) of the mRNAs dramatically reduce the translation of the mRNAs
Difference between miRNAs and siRNAs
miRNA would IMPERFECTLY (there are mismatches) bind to the target mRNA
The binding is usually at the 3’-UTR
The imperfect binding leads to repression of translation
siRNA are produced from cleavage of double stranded RNA
They bind PERFECTLY (no mismatches) to the target RNA and cause its rapid degradation by a mechanism called RNA interference
miRNA function in limb development
Dicer gene knockout – eliminates generation of miRNAs in mammals – embryonic lethal
Experiment – Dicer knockout only in limb tissue (13-day mouse embryos): (immunostained for Gd5 protein, a marker of joint formation)
(left) Dicer-expressing control – normal limb development
(right) Dicer-knockout in limbs (Conditional expression of Cre induces dicer gene deletion only in limb cells.)
Result – fundamental pattern maintained but abnormal limb development
Conclusion – Dicer (miRNA) is required for morphogenesis but not patterning of the vertebrate limb
miRNA processing in nucleus
miRNA transcription and processing:
RNA polymerase II transcribes primary miRNA transcripts (pri-miRNA) – folds to form double strand region
Nuclear double-strand RNA–specific endoribonuclease Drosha and double-strand RNA–binding protein DGCR8 (Pasha in Drosophila) bind pri-miRNA double strand regions
Drosha cleaves the pri-miRNA – generates a ~70-nucleotide pre-miRNA
WHat transports miRNA to the cytoplasm
Exportin 5
– nuclear transporter transports processed pri-miRNA to the cytoplasm
RNA interference steps once in cytoplasm
Step 1: dsRNA is cleaved to small (21-28 bp) RNA fragments by a dsRNA-endonuclease called DICER
Step 2: Small dsRNAs bind to RISC (RNA Induced Silencing Complex) and form an inactive RISC complex.
Step 3: Argonaut in RISC is a RNA helicase. It unwinds dsRNA. Active RISC* is formed.
Step 4: RISC*, guided by single-stranded siRNA, cleaves target mRNA (“SLICER” activity by Argonaut)
Argonaut
Argonaut (RISC):
An RNA helicase
Removes one of the strands of the dsRNA
Leads the trimmed siRNA to the target mRNA
What is PAX6
PAX6 regulates several processes during the development of mammals
PAX6 is a developmentally regulated gene
it encodes a transcriptional activator
It contains three promoters and three transcription initiation sites
It has more than three enhancers
It is expressed at different times and in different organs during embryonal development
It is expressed in certain parts of the brain in adult organisms
The translation of PAX6 mRNAs is regulated by several miRNAs
How many promotors does pax6 have?
3 alternative promoters
10.5 days after fertilization - the embryo
the gene is inactive in all cells of the body: heterochromatin
Close to 10.5 days after fertilization:
pioneer factors open heterochromatin at the PAX6 locus in the pancreas (P) and the lens plate (LP)
In all other organs, PAX6 remains in heterochromatin
10.5 days after fertilization:
the pancreatic and lens/cornea enhancers activate PAX6 by bending DNA and come in proximity of the promoter
The pancreatic and the lens/cornea enhancers will be occupied by multiple, but not identical transcriptional activators (enhanceosome)
Transcriptional activators communicate with the GTFs, Mediator, Chromatin Remodeling Complexes and Histone Modification Complexes.
Histones are acetylated
Nucleosomes are displaced from the promoter region
all other organs, PAX6 remains in heterochromatin
What happens 10.5 days after fertilization after the displacement of nucleosomes:
Mediator and GTFs communicate
GTFs recruit RNA polymerase II
The CTD of RNA polymerase II is phosphorylated by the TFIIH kinase
The TFIIH helicase activity opens the DNA helix and the RNA polymerase starts transcribing
Between 10.5 to 13.5 days after fertilization:
: enhaceosome exchange
The Lens/cornea enhancer stops working
The pancreatic enhancer continues to operate
The retina enhancers start operating. They will bend DNA and come in the proximity of the promoter.
After day 13.5:
telencephalon and rhomboencephalon will start working in the brain
What is the difference between the transcripts of the different promoters of PAX6
PAX6 can produce three different transcripts from its three promoters
Alternative splicing of these transcripts produces proteins with different function.
Transcript a) is missing exon 1 and the alternative exon α (alpha)
Transcript b) is missing exon 0 and the alternative exon α (alpha)
Transcript c) is missing exons 0 and 1-4 but retains the alternative exon α (alpha)
Exons 1-4 encode a part of the DNA-binding domain of PAX6
Hence, transcripts a) and b) produce similar DNA binding proteins that differ in their DNA-binding domains and bind to different DNA elements
Transcript c) has an entirely different function
Enhancers
Range ~50 to 200 bp.
Binding sites for several transcription factors.
Interacting proteins distally bound can produce loops in the DNA between their binding sites
β-Interferon enhancer:
Six overlapping binding sites bind two heterodimeric factors:
Jun/ATF-2, p50/RelA (NF-κB), and 2 copies each of the monomeric transcription factors IRF-3 and IRF-7.
Hydrophilic hormone pathway
Hydrophilic signaling molecules – (small molecules [adrenaline, acetylcholine], peptides [yeast mating factors, glucagon], and proteins [insulin, growth hormone])
Cannot diffuse across the cell membrane
Step 4: Bind to specific cell-surface receptor proteins – triggers receptor conformational change that activates the receptor
Step 5: Activated receptor activates one or more downstream signal transduction proteins or small-molecule second messengers.
Step 6: Signal transduction proteins or small-molecule second messengers activate one or more effector proteins.
Step 7a: Effector – stimulates modification of specific cytosolic proteins; short-term (sec-min) changes in cellular function, metabolism, or movement
Step 7b: Effector – moves into the nucleus; triggers long-term (hours-permanent) changes in gene expression
Termination or down-modulation of the cellular response –
Step 8: Negative feedback/feedback repression from intracellular signaling molecules
Step 9: Destruction of the extracellular signal
Hydrophobic signalling pathway
Hydrophobic signaling molecules – (steroids and related molecules)
Step 1: Diffuse through the plasma membrane
Step 2: Bind to cytosolic receptors
Step 3: Receptor-signal complex moves into the nucleus – binds transcription-control regions in DNA to activate or repress gene expression
Types of extracellular signalling
- endocrine
- paracrine
- autocrine
- membrane protein signals
Endocrine signaling
endocrine hormones affects target cells some distance from site of synthesis
transported through circulatory system
ex. epinephrine and insulin
Paracrine signaling
paracrine hormones affect target cells in close proximity
some may bind to ECM
affect only nearby target cells expressing the receptor
ex. neurotransmitters, growth factors
Autocrine signalling
cells respond to signals that they produce
target sites are on the same cell
ex. growth hormones
Signalling by plasma-membrane attached proteins
signal neighboring cells by direct contact with surface receptors.
What is Dex?
Dexamethasone, activates the Glucocorticoid Receptor
What are the three domains of the glucocorticoid receptor?
Activation domain, DNA binding domain and ligand binding domain
How does Dex activate the receptor and such…?
Dex = Dexamethasone, activates the Glucocorticoid Receptor
In the absence of Dex the Glucocorticoid Receptor is in the cytoplasm
In the presence of Dex the receptor-hormone complex moves to the nucleus
The control ꞵ-galactosidase is cytoplasmic regardless of the presence of Dex
In more detail:
(–) Hormone – receptor retained in the cytoplasm by interaction between its ligand-binding domain (LBD) and chaperone proteins
(+) hormone –
1) Hormone complex diffuses through the plasma membrane and binds to the receptor ligand-binding domain.
2) Causes a conformational change that releases the chaperone proteins
3) Receptor-hormone complex translocates into the nucleus.
4) DBD binds to response elements
Ligand-binding domain and an additional activation domain (AD) at the N-terminus stimulate transcription of target genes.
Membrane receptors
bind only a single type of hydrophilic signaling molecules
All hormone receptors are highly specific for their ligands
High affinity of the hormone-receptor interaction
Each receptor binds only a single type of hormone
The hormone changes the conformation of the receptor sending a signal to the cascade
Examples of hydrophilic signaling molecules
small molecules: adrenaline, acetylcholine
peptides: glucagon
proteins: insulin, growth hormones
Growth hormone coupled to two receptors findings:
Basically - found that only 8 DISTANTLY SPACED AMINO ACIDS CONTRIBUTE 85% BINDING ENERGY
As determined from the three-dimensional structure of the 1 growth hormone:
2 growth hormone receptor complex, 28 amino acids in the hormone are at the binding interface with the first receptor protein.
To determine which amino acids are important in ligand-receptor binding, researchers mutated each of these amino acids one at a time to alanine and measured the effect on receptor binding.
From this study, it was found that only eight amino acids on growth hormone (green) contribute 85 percent of the energy that is responsible for tight receptor binding; these amino acids are distant from each other in the primary sequence but adjacent in the folded protein.
Similar studies showed that two tryptophan residues (blue) in the receptor contribute most of the energy responsible for tight binding of growth hormone, although other amino acids at the interface with the hormone (yellow) are also important
What do receptors do?
Hormone receptors activate Effector Proteins
Often the effector proteins are enzymes that produce small molecules called second messengers
Secondary messengers carry and amplify the signals from activated receptors/hormone complexes to the effector molecules
What are secondary messengers?
- small short-lived molecules
- diffuse rapidly
- allow enzymatic amplification
What are the water-soluble secondary messengers?
Ca2+ ions, cAMP, and cGMP
What are the examples of lipid-soluble hormones?
DAG, IP3
cAMP
- type of water-soluble secondary messenger
- Generated from ATP by adenylyl cyclase
- Activates PKA
cGMP
- type of water-soluble second messenger
- Generated by guanylyl cyclase
- Activates PKG and specific cation channels
IP3 and DAG
- type of lipid-soluble second messengers
- Both made from PIP2 by phospholipase C
- IP3 – opens channels to release Ca2+ from the ER
DAG – with Ca2+ activates PKC
Ca2+ ions
Released from intracellular stores or transported into the cell
Activates calmodulin, specific kinases (PKC), and other regulatory proteins
What are the two types of kinases in animal cells?
- tyrosine (Y) kinases
- serine (S) / threonine (T) kinases
GTPase switch proteins - simple mechanism
Hydrophilic hormones bind to membrane receptors.
These so-called G-protein coupled receptors (GPCR) activate G-proteins (GTPase switch proteins).
G-proteins exchange GDP to GTP leading to a conformational change and activation.
Active G-protein sends a signal
G-protein has intrinsic GTPase activity, which can be accelerated by GTPase-accelerating proteins (GAPs)
Once the signal is sent, the G-protein inactivates itself by hydrolyzing GTP to GDP
How is G-protein activity regulated?
G-protein activity is regulated by exchange of GDP to GTP
Association with GTP switches the protein “ON”
After sending the signal, GTP is hydrolysed and the protein is switched “OFF”
GTP hydrolysis rate
time G protein remains in the active conformation
What is the switching mechanism of monomeric G proteins?
Conformation of the G protein is altered by GTP/GDP binding
Inactive (GDP-bound) G protein is capable of interacting with upstream activators
Active (GTP-bound) G protein is capable of interacting with downstream effector proteins
The downstream effector (often an enzyme) produces SECOND MESSENGER MOLECULES
Switch domain conformation – can bind and activate specific downstream effector proteins
IN THE ACTIVE/ON STATE, THE SWITCHES ARE PULLED IN AND BOUND TO GTP through interactions with the backbone amide groups of the conserved Thr and Gly residues (blue and green switches in diagram)
IN THE INACTIVE/OFF STATE - SWITCH i AND ii ARE RELAXED INTO OFF CONFORMATION - inhibits interaction with downstream effectors
What are the 4 elements GPCR signal transduction pathways have in common?
(1) A receptor that contains seven membrane-spanning α helices
(2) A receptor-activated heterotrimeric G protein cycling between GTP-active and GDP-inactive forms
(3) A membrane-bound effector protein
(4) Proteins that desensitize the signaling pathway
What are the 7 Human GPCR of pharmaceutical importance?
H2 receptor (histamine is ligand) - affects acid secreting cells of stomach
H1 receptor (histamine is ligand) - causes symptoms of allergy
Serotonin 5Ht 2a and 1a (serotonin is ligand) - causes synaptic transmission btw neurnons in CNS
Angiotensin - that is also ligand - increases blood pressure
B-adrenergic receptor - (epinephrine is ligand) - facilitates respiration
CysLT - (Leukotrienes are ligand) - contracts smooth muscle
Synthesis and hydrolysis of cAMP by adenylyl cyclase and phosphodiesterase (PDE)
> 30 different mammalian GPCRs activate Gαs activation of adenylyl cyclase production of cAMP as a second messenger. (Most cell types express one or more such GPCRs.)
Adenylyl cyclase (AC) – catalyzes formation of cyclic cAMP (second messenger) bond from ATP precursor
cAMP phosphodiesterase (PDE) – catalyzes hydrolysis of cyclic bond – AMP (not second messenger)
Similar reactions occur for production and destruction of cGMP second messenger.
AC and PDE
adenylyl cyclase - catalyzes FORMATION of cyclic cAMP (a second messenger) from ATP precursor
then, PDE (phosphodiesterase) takes that cAMP and catalyzes HYDROLYSIS of the cyclic bond AMP (not second messenger)
What are the steps in the transduction and AMPLIFICATION initiated by epinephrine?
(Amplification of an extracellular signal by a signal transduction pathway involving cAMP and PKA)
Amplification:
Single epinephrine molecule binding to one G protein–coupled receptor – activates up to hundreds of G proteins (amplification)
Each G protein activates 1 AC until G protein hydrolyzes GTP (not amplification).
AC catalyzes the synthesis of a large number of cAMP molecules while activated (amplification).
Two cAMPs activate two PKA catalytic subunits (not amplification).
Activated PKA phosphorylates and activates multiple target proteins (amplification).
Activated target proteins:
Other kinases – kinase cascade (may be multiple steps of amplification; the more steps in such the cascade, the greater the signal amplification)
Enzymes – multiple products (amplification)
Cellular responses to hormone-induced rise in cAMP
Epinephrine activation of the fight-or-flight response:
Liver and skeletal muscle – increase in glucose for energy production
Adipose – breakdown of triglycerides for energy production
Cardiac muscle – increased heart rate – O2 and glucose transport to peripheral tissues
Smooth muscle cell relaxation (not shown) – increase blood flow and air intake
Intestine – fluid secretion (use your imagination)
What are the 6 steps of the Induction of gene expression by MAP kinase?
Steps 1) MAP kinase in the cytosol phosphorylates and activates the kinase p90RSK,
2) p90RSK then moves into the nucleus and
3) once in the nucleus - phosphorylates the SRF (serum-response factor) transcription factor.
4) MAP kinase translocates into the nucleus and
5) directly phosphorylates TCF transcription factor already bound to the promoter of the c-fos gene.
6) Phosphorylated TCF and SRF together stimulate transcription of c-fos and other genes that contain a promoter SRE
(serum response element) sequence.
MAP kinases phosphorylate over 175 target proteins in the nucleus.
General mechanism of the activation of effector proteins by heterotrimeric G proteins:
Step 1: Ligand binding induces receptor activation conformational change.
Step 2: Activated receptor binds to trimeric G protein.
Step 3: Activated receptor GEF activity stimulates Gα subunit release of GDP.
Step 4: GTP binding changes Gα conformation
Dissociates Gβγ
(Gβγ subunit activates other effector enzymes in some pathways (b))
Activates Gα
Step 5: Gα·GTP activates effector enzyme.
Step 6: Gα intrinsic GTPase activity hydrolyzes GTP to GDP – dissociates Gα and turns off effector enzyme. (G protein active for minutes or less)
What is the map kinase pathway steps simplified:
1) RTK activation leads to the auto-phosphorylation of the RTK intracellular domain
2) The phosphorylated intracellular domain interacts with adaptor proteins (GRB2, Sos) through the so called SH2 and SH3 domains (standing for Src-Homology Domains)
3) Sos activates Ras (binding of GTP)
4) Ras activates Raf (MAP Kinase Kinase Kinase)
5) Raf activates MEK (MAP Kinase Kinase)
6) MEK activated ERC (MAP Kinase)
What are the 3 steps in the activation of Ras?
Receptor tyrosine kinases activate Ras via adapter proteins.
1) Binding of FGF hormone causes receptor dimerization, kinase activation, and phosphorylation of cytosilic receptor tyrosine kinases
A specific phosphotyrosine on an activated, ligand-bound receptor provides a specific site for binding the cytosolic adapter protein GRB2 SH2 domain.
2) GRB2 SH3 domains specifically bind proline domains on SOS, recruiting SOS to the membrane to interact with Ras∙GDP (tethered to the cytosolic surface of the plasma membrane by a hydrophobic farnesyl anchor).
(Binding of GRB2 and Sos couples receptor to inactive Ras)
(3) The SOS guanine nucleotide exchange factor (GEF) activity promotes release of GDP and formation of active Ras∙GTP.
Sos promotes dissociation of GDP from Ras; GTP binds and active Ras dissociates from Sos
SH2 domain
Part of Grb2 protein involved in Ras activation
SH2 domain associates with phospho-tyrosine present on ligand-activated receptors of the RTK
SH3 domain
part of Grb2 in the Ras activation
domain binds specifically to proline-rich sequence in Sos
Grb2
is a protein involved in ras activation
- Has SH2 and SH3 domains
- Joins active receptor (RTK) to cytoplasmic Sos (brining it closer to the plasma membrane)
What is Sos
Sos is a GEF involved in Ras activation
Sos gets recruited by SH3 domain of Grb2 to be close to the plasma membrane, where it can promote dissociation of GDP from Ras
Ras/MAP kinase pathway steps
Stimulated cell:
Step 1: Active RTK receptor activates formation of active Ras·GTP.
(Ras activated by the exchange of GDP to GTP)
Step 2: Ras·GTP triggers the downstream kinase cascade by interacting with the Raf N-terminal regulatory domain, which results in dephosphorylation of one of the serines, release of 14-3-3, and activation of the Raf kinase activity.
(Active Ras recruits, binds, and activates Raf)
Step 3: Ras GTP hydrolysis to Ras·GDP releases active Raf.
Step 4: Raf phosphorylates and activates MEK.
Step 5: MEK, a dual-specificity protein kinase, phosphorylates MAP kinase (and ERK kinase) on both tyrosine and serine/threonine residues.
Step 6: MAP kinase phosphorylates many different proteins in different cells, including nuclear transcription factors, that mediate cellular responses.
What is the only other signal transduction pathway that is activated by RTK that we are studying?
PI-3 kinase! aka Phosphoinositide Signaling Pathway
Phosphoinositide Signal Transduction Pathways
RTKs and cytokine receptors can initiate the IP3/DAG signaling pathway by activating phospholipase Cγ.
RTKs and cytokine receptors can initiate another phosphoinositide pathway by activating PI-3 kinase formation of PI 3-phosphate, which bind PH domains in various proteins to form signaling complexes that activate PKB.
Activated PKB promotes survival of many cells by directly inactivating pro-apoptotic proteins and synthesis of pro-apoptotic proteins.
What are the names of the produced molecules in the phosphoinositide signalling pathways?
PhosphoInositol 3,4,- biphosphate (IP2)
PhosphoInositol 4,5,- biphosphate (IP2)
PhosphoInositol 3,4,5- triphosphate (IP3
How is full activation of PKB achieved?
Full activation is achieved by phosphorylation of the membrane-bound PKB by two other kinases, 3-phosphoinositide dependent protein kinase 1 and 2 (PDK1 and PDK2)
What are the steps in the PI3 pathway?
Unstimulated cells: PKB is in the cytosol with its PH domain bound to and inhibiting its catalytic kinase domain.
Hormone stimulated cells:
Inactive PKB is in the the cytosol of unstimulated cells
Step 1: Hormone stimulation leads to activation of PI-3 kinase, which forms PI 3-phosphates
(phosphorylates 3 position of PI to form PI 3,4 bisphosphates)
Step 2: The 3-phosphate group binds the PH domains of PKB (partially active) and PDK2, docking both to the membrane and partially activating PKB
Step 3: PDK1 phosphorylation of the PKB activation loop serine and PDK2 phosphorylation of a PKB C-terminus serine fully activates PKB activity, which induces many cellular responses.
General structure of G protein coupled receptors
All G protein–coupled receptors have:
The same orientation in the membrane – N-terminus outside, C-terminus in cytosol
Contain seven transmembrane α-helical regions (H1–H7)
Have four extracellular segments (E1–E4)
Four cytosolic segments (C1–C4)
Structure of the β2-adrenergic receptor in the inactive state and in the active state with a bound ligand and with its associated heterotrimeric G protein
Ligand binding causes movement of the receptor TM5 and TM6 helices and changes in the C3 loop to create a surface that forms extensive interactions parts of Gαs
Gαs small conformational change
Lengthening the α5 helix creates a large surface consisting of the N-terminal α-helical segments αN and α5 that bind mainly to receptor TM5 and
TM6.
Triggers release of GDP and Gβγ
(c) Binding of epinephrine to the side chains of multiple amino acids in the interior of the β2-adrenergic receptor
What’s the FRET technique?
Forster resonance energy transfer (FRET) technique:
Wavelength of emitted fluorescence changes when two fluorescent proteins interact.
Reveals kinetics of G protein activation
Mechanism of detection of protein interactions IN VIVO
One protein is tagged with Gα-cyan fluorescent protein (CFP)
Another protein is tagged with Gβ-yellow fluorescent protein (YFP, excited by CFP (cyan fluorescent protein) when alpha subunit is bound to beta subunit
On the left: The two proteins are close to each other (INACTIVE RECEPTOR) Then the light emitted by CFP (blue) excites YFP and WE OBSERVE YELLOW LIGHT
On the right: The two proteins are away from each other (ACTIVE RECEPTOR) We observe only the light emitted by CFP (blue) WE ONLY OBSERVE CYAN LIGHT
What are inhibitory hormones of cAMP and how do they work?
PGE and adenosine are inhibitory hormones and they bind to the inhibitory receptor which activates G protein complex and binds and causes the inactivation of adenylyl cyclase which causes the down regulation/lack of production of cAMP
(seesaw looking diagram)
What are stimulatory hormones of adenylyl cyclase
Epinephrine and Glucagon
these are stimulatory hormones that bind to receptors causes activation of a stimulatory G protein complex that binds and activates adenylyl cyclase and causes production of cAMP
(seesaw looking diagram)
Receptor Tyrosine Kinases
Hydrophilic signaling molecules (hormones or peptides) activate Receptor Tyrosine Kinases (RTKs), which phosphorylate downstream targets and initiate a signal transduction cascade
RKTs have a different mechanism of action as compared to GPCRs
Usually the hormone or peptide leads to dimerization of two kinase polypeptides
The dimerization of RTKs leads to cytoplasmic domain autophosphorylation, which recruits proteins that initiate intracellular signaling pathways
What are the types of cell-surface receptors?
1) receptor associated kinases
2) cytosolic kinases
3) protein subunit dissociation pathways
4) protein cleavage pathways
Receptor-associated kinases (type of cell-surface receptor and signal transduction pathway)
Many receptor cytosolic domains are protein kinases or tightly associate with a cytosolic kinase.
(1) Ligand binding causes receptor dimerization which activates kinase activity:
Kinases directly phosphorylate and activate transcription factors or other signaling proteins.
(2) Many receptors also activate small GTP-binding “switch” proteins such as Ras.
The Ras signal transduction pathway and others activate a kinase cascade, in which one kinase phosphorylates and thus activates (or occasionally inhibits) the activity of multiple copies of another kinase for signal amplification.
G-protein pathway kinases phosphorylate multiple protein targets, including cell type-specific transcription factors.
Cytosolic kinases (type of cell-surfacr receptor and signal transduction pathway)
Seven membrane-spanning segments receptors activate GTP-binding Gα proteins.
Gα-GTP activates specific kinases or other signaling proteins.
Protein subunit dissociation pathway (type of cell-surface receptor and signal transduction pathway)
Several signaling pathways cause disassembly of a multiprotein complex in the cytosol, which releases a transcription factor that translocates into the nucleus.
Protein cleavage pathways (type of cell surface receptor and signaling pathway)
irreversible signaling
Proteolytic cleavage of a receptor releases an active transcription factor.
What conditions does the ability of a transcription factor to bind depend on?
The binding sites for the factor and
the gene’s epigenetic state
and on the presence of master transcription factors and other nuclear proteins
Signaling molecules that activate RTKs:
are soluble or membrane-bound peptide or protein hormones:
Growth factors:
NGF - nerve GF
PDGF - platelet-derived GF
FGF - fibroblast GF
EGF - epidermal GF
Steps of RTK activation:
RTK activation mechanism:
1) (–) ligand: RTKs generally exist as monomers with poorly active kinases.
(2) (+) ligand: Two ligands binding to two RTK extracellular domains homodimerizes the receptors,
bringing together two poorly active cytosolic domain kinases,
which phosphorylate each other on an activation loop tyrosine residue.
The phosphorylated loop moves out of the kinase catalytic site, increasing ATP and/or the protein substrate binding.
(3) The activated kinase phosphorylates several tyrosine residues in the receptor’s cytosolic domain, which provides docking sites for SH2 and other binding domains on downstream signal-transducing proteins.
Ligand-induced dimerization of HER1, a human receptor for epidermal growth factor (EGF).
(a) (–) EGF: β-hairpin from domain II that forms the “dimerization arm” binds to domain IV of the same receptor molecule, inhibiting dimerization.
(b) (+) EGF: EGF binds domains I and III, inducing an extracellular domain conformational change that exposes the domain II dimerization arm.
(c) Dimerization arms interactions dimerize two identical ligand-bound receptor monomers.
What does the activation of the EGF receptor by EGF result in?
The formation of an asymmetric kinase domain dimer
The EGF receptor kinase domain is activated by phosphorylation of an activation loop tyrosine.
Inactive, monomeric state: activation loop binding in the kinase active site inhibits kinase activation.
Receptor activation mechanism:
The dimer is stabilized by interactions between the juxtamembrane segments of the two receptors.
Dimerization generates an asymmetric kinase dimer in which the donor kinase C-terminal C-lobe binds the acceptor kinase N-terminal N-lobe.
Interactions cause a conformational change that removes the activation loop from the acceptor kinase site, activating its kinase activity.
The active acceptor kinase phosphorylates cytosolic domain tyrosine residues on both receptors
What is the cell cycle?
a sequence of ordered events
these events are controlled by CDKs
consists of G1, S, G2 and mitosis phases
What regulates the cell cycle?
Cyclin Dependent Kinases (CDK)
The key regulatory subunits of CDKs are called Cyclins
Other regulatory mechanisms involve synthesis and degradation of CDK, inhibitors or activating or inhibitory phosphorylation of the CDK
Positive regulators of the cell cycle
Each Cyclin acts at a specific stage of the cell cycle and sets up the transition to the next stage, then gets degraded
Negative regulators of the cell cycle
there are surveillance mechanisms that check for the proper progression through the cell cycle.
If anything goes wrong, these mechanisms, called checkpoints, hold the cycle until repair is done
Why is the end of G1 so important?
The decision to divide is tightly controlled at the end of G1. Once this decision is made, the cell must divide or die.
Interphase
G1+S+G2
Interphase checkpoints
Make sure that the stage of the cell cycle is completed
act as brakes on cell cycle progression when events are not completed or when DNA is damaged
Budding or Fission Yeasts
models for studying the cell cycle
unicellular organisms, easy to grow and synchronize
rapid cell cycle (2-4 hrs)
cell cycle stage visualized by the morphology of the cell
can be grown as haploid cells (amazingly simple genetics)
conditional loss of function (ts) mutants display distinct morphology
Budding yeast cell cycle
G1 – growth to size large enough to form viable bud; irreversible commitment to progression through cell cycle is made at the G1 START point; bud begins to emerge
S – DNA replication (1n to 2n in haploid organism)
G2 – less distinct stage in yeast than in higher eukaryotes
M – intranuclear spindle; nuclear envelope does not break down in yeast
Fission yeast cell cycle
cell cycle mutations are easily generated and recognized by differences in cell size
G1 grow longer before cell division.
G1 START point – irreversible commitment to cell division
S − nuclear envelope does not breakdown
What have model organisms revealed?
Model organisms have revealed that:
A few master cell cycle regulatory proteins control and coordinate processes such as DNA replication initiation and entry into mitosis.
Master regulators and the proteins that control them are highly conserved across eukaryotic cells
Progesterone stimulation of Xenepus oocytes
progesterone stimulates maturation of Xenopus oocytes
Model cell − large enough for biochemical characterization of cell extracts
Progesterone treatment causes G2-arrested Xenopus oocytes to enter meiosis I, a modified form of mitosis.
Homologous chromosome segregation and asymmetric cell division expels one set of chromosomes into the first polar body.
Oocyte arrests in metaphase II to yield an egg.
When fertilized by sperm, egg proceeds through meiosis II anaphase and a highly asymmetric cell division expels one chromatid of each chromosome into a second polar body.
The haploid female and sperm pronuclei fuse to produce a diploid nucleus.
Diploid zygote proceeds through cell cycle to divide by mitosis into two cells.
11 more synchronous divisions produce blastula.
Human cells undergoing mitosis visualisation
HeLa Kyoto cells seen under phase contrast microscopy were filmed as they underwent nitosis.
HeLa Kyoto cells − are flat during interphase but round up during mitosis and cell division. Daughter cells flatten again.
Normal primary human cells and other mammalian cells have a finite life span when cultured in vitro.
Normal human cells divide 25–50 times before undergoing replicative senescence.
Immortalized cell lines, some of tumor origin such as the HeLa cells, harbor genetic alterations that enable them to escape replicative senescence and become immortal.
What agents are used to synchronyze cells in YEAST
Mating pheromone arrests the cells in late G1
Nocodazole disrupts mitotic spindle – cells arrest in pro-metaphase
What agent is used to synchronyze cells in mammals?
Hyproxyurea and nocodazole
What is nocodazole
Anti mitotic agent used in yeast and humans that reversibly interferes with polymerization of microtubules
it disrupts the mitotic spindle
What is mating pheromone?
a cell synchrony agent used in yeast cells that arrests the cells in late G1
What is hydroxyurea
aka thymidine is an antimetabolite medication that inhibits the synthesis of dNTPs -
CAUSES CELL AREST IN EARLY S PHASE
USED IN MAMMALIAN CELL CULTURE
What does flow cytometry measure?
the amount of cellular DNA
Flow cytometry procedure:
Fix cells by Ethanol (Permeabilizes and preserves cells)
Degrade all RNA (RNAse)
Soak the cells with a DNA staining dye (propidium Iodine)
Wash the cells
Run cell suspension through a capillary tube, one cell at a time
Use lasers to detect the amount of stain (indirect measure of the amount of DNA) in each cell
Plot the results
Analysis of DNA content by flow cytometry
Researchers use multiple tools to study the cell cycle.
DNA in yeast cells stained with propidium iodide fluorescent dye can be quantified by flow cytometry.
DNA content analysis shows three populations of cells: cells with unreplicated DNA (1C), cells with replicated DNA (2C), and cells in the process of DNA replication (S phase).
Isolation of ts mutants in S. cerevisiae
Isolated temperature sensitive (ts) mutants not growing at higher temperature (arrest with certain morphology)
Treat yeast w mutagen
Grow colonies (23*C)
Check which colonies do not grow at higher temperature (these are called TS - temperature sensitive mutants)
Check morphology of TS mutants
(the morphology tells you at which state of the cell cycle the mutated gene works)
Isolate and analyze the gene
ts cdc mutants
temperature sensitive cell division cycle mutants
In ts mutants grown at restrictive temperature, cell morphology (presence and size of bud) indicates at which stage of the cell cycle the mutants gene works. Position in the cell cycle can be determined by morphology
cdc mutants in S. pombe
mitosis at size larger than normal
delayed progress through cell cycle
(fission yeast)
wee mutants
mitosis at size smaller than normal
advanced progress through cell cycle
How do cyclin levels differ at each stage of the cell cycle?
Cyclin levels rise during interphase
peak at mitosis
and drop before anaphase
Discovery of cyclin in dividing eggs
samples collected at different times post-fertilization (every 10 min)
identified a protein that was continuously synthesized and periodically destroyed
cyclin destruction occurred just prior to onset of mitotic cleavage (anaphase)
Which genes that regulate the cell cycle are homologous?
cdc28, cdc2, Cdk1 and MPF (cdk1)
Overview of how CDKs regulate the cell cycle
Early G1 − no cyclin − CDKs are active
Mid-G1 − G1/S phase CDKs activate transcription of genes required for DNA replication
S phase − initiated by SCF ubiquitin-protein ligase ubiquitinylation of S phase CDKs inhibitors, marking them for degradation by proteasomes. S phase CDKs activate DNA replication.
Late G2, mitotic CDKs trigger entry into mitosis.
M Anaphase − anaphase-promoting complex (APC/C) ubiquitinylates:
The anaphase inhibitor protein securin, marking it for degradation by proteasomes, activating securin to degrade cohesin complexes linking the sister chromatids. Separation of sister chromatids initiates anaphase.
Mitotic cyclins, targeting them for degradation by proteasomes. The resulting drop in mitotic CDK activity and protein phosphatase activity results in chromosome decondensation, reassembly of nuclear membranes around the daughter-cell nuclei, and cytokinesis.
CDKs are active only in…
CDKs are active only in the stages of the cell cycle they trigger:
G1/S phase CDKs are most active at the G1–S phase transition to trigger entry into the cell cycle.
S phase CDKs are most active during S phase.
Mitotic CDKs are most active during mitosis.
APC/C cyclosome
The anaphase promoting complex
ubiquitin protein ligase
- ubiquitinylates proteins, including cyclins, targeting them for degredation by proteosomes
- catalyzes two key cell cycle transitions: anaphase and exit from mitosis
What do the graphs showing activity of CDKs and cyclins show?
The activity of the different cyclin-CDK complexes varies in a manner that is very similar, but not exactly identical to the accumulation of the corresponding cyclins:
Conclusion:
There are other regulators of CDK activity
What are the 3 main types of regulators of CDK activity?
1) Kinases and Phosphatases
2) inhibitory proteins
3) ubiquitin-protein ligases
CDK inhibitors
CDK inhibitors (INKs, CIPs, KIPs) associate with CDK/Cyclin complexes and inhibit them until the previous stage of the cycle is completed.
After the completion, the active CDK/Cyclins phosphorylate the CDK inhibitors. These inhibitors are then degraded. The inhibitor degradation provides a clean molecular trigger of the next stage of the cycle.
Next, the cyclins from the previous stage are degraded via protein ubiquitination.
The protein-ubiquitin ligases are:
SCF - works at the transition from G1 to S phases
APF - works at the onset of mitosis (anaphase)
SCF
A protein ubiquitin ligase
works at the transition from G1 to S phases
Degrades phosphorylated S phase inhibitor
APC/C proteosome
a protein ubiquitin ligase
used during metaphase/anaphase transition AND at the mitotic exit/end of anaphase
degrades securin
degrades mitotic cyclins
What are the regulators of cyclin/CDK activity?
Kinases and Phosphatases
Inhibitory proteins
ubiquitin-protein ligases
and
Phosphoserine/Threonine Binding proteins and modular domains
Wee1 kinase
Wee1 kinase phosphorylates Y (tyrosine) and T(threonine) residues on CDK and inhibits the mitotic kinases in S-phase.
The dual-specificity Wee1 kinase inhibits mitotic cyclin-CDK activity by phosphorylation of CDK T14 and Y15 until DNA replication is completed
Cdc25 phosphatase
removes the inhibitory phosphorylation of Y and T residues previously phosphorylated by Wee1 and activates Mitotic CDKs
When DNA replication is completed, the dual-specificity Cdc25 phosphatase is activated (by an unknown mechanism) and dephosphorylates CDK T14 and Y15 , reversing the inhibitory effect of Wee1 and activating cyclin-CDK activity.
WHat do mitotic cyclins synthesized during S and G2 do?
They activate mitotic CDKs
Mitotic cyclin-CDK positive eedback on its own activity:
Phosphorylates Cdc25, activating its CDK-activating phosphatase activity
Phosphorylates Wee1, inhibiting its CDK-inhibiting kinase activity
Cdr2
inhibits Wee1 activity when a cell has reached an appropriate size to divide
When is the decision to divide made?
The decision whether to divide or not is made in late G1 phase: RESTRICTION (or START) point
This decision is tightly regulated
Cells that never divide (highly differentiated cells) arrest before this START point in G0
What proteins and stuff are used to regulate G1-S phase CDK activation?
Whi5
SBF
Rb
E2F
How is the G1 to S phase transition regulated in S. cerevisiae/budding yeast?
Whi5 and SBF!
Whi5 is on top of SBF and it is a transcriptional repressor - inhibits SBF activation of the CLN1/2 cyclin gene transcription
Nutrient availability allows translation of Cln3 mRNA, activating Cln3-CDK activity.
Cln3-CDK phosphorylates Whi5, promoting its export from the nucleus.
When 50 percent of Whi5 has been exported, cells transition through START.
Active SBF induces transcription of CLN1 and CLN2 and other genes whose products are needed for DNA replication.
G1/S phase CDKs further phosphorylate Whi5, promoting further CLN1 and CLN2 transcription.
High levels of G1/S phase CDKs initiate DNA replication, bud formation, and spindle pole body duplication
Metazoan/vertebrate regulation of G1/S phase transition
G1 − The E2F transcription factor and its regulator Rb control the G1–S Phase transition
Rb binds and inhibits E2F activity.
Growth factors (mitogens) stimulate G1 CDK activity.
Cyclin D–CDK4/6 complexes phosphorylate Rb, releasing E2F.
E2F stimulates transcription of early response genes encoding cyclin E, CDK2, and E2F.
Cyclin E–CDK2 further phosphorylates Rb, initiating a positive feedback loop that leads to a rapid rise in the expression and activity of both E2F and cyclin E–CDK2.
G1/S phase cyclin E/A-CDK2 CDKs support cell passage through the restriction point and initiation of DNA replication and centrosome duplication.
Are there cyclins at the end of mitosis?
No! All cyclins have been degraded at the end of mitosis by the APC
In yeast, presence of nutrients leads to…
to increased synthesis of cyclin Cln3 in early G1
Rb
regulator of E2F transcription factor
binds to E2F and suppreses the transcription of cyclin E or cyclin A in mammals in early G1
What phosphorylates Rb?
Cyclin D (CDK4/6)
What’s the Sic 1 protein and what process is it involved with?
Sic1 is an S phase inhibitor (it’s attatched to S phase CDK/cyclin) it prevents initiation of DNA replication ununtil the cells have completed all G1 events
1) G1/S phase CDKs assembled in late G1 phosphorylate Sic1 at multiple sites, marking it for degredation
2) SCF ubiquitin-protein ligase ubiquitinylation of SCF and subsequent proteasomal degradation
3) The active S phase CDKs trigger initiation of DNA synthesis by phosphorylating and recruiting MCM helicase activators to DNA replication origins.
How many sites do we phosphorylate on Sic and why?
G1/S phase CDK phosphorylation of a single optimal Sic1 site would cause a sluggish G1/S phase transition. G1/S phase CDKs accumulation during G1 would cause slow progressive degradation of Sic1 and initiation of S phase.
slow Slc1 degradation
slow S phase CDK activation
sluggish G1 – S phase transition
(c) Six suboptimal phosphorylation sites ensure that Sic1 is fully phosphorylated and targeted for destruction by SCF only when G1/S phase CDKs have reached high levels. This ensures that Sic1 degradation and DNA synthesis initiation occurs rapidly, when G1/S phase CDKs have accomplished all their other G1 tasks.
Six sub-optimal sites of phosphorylation of Sic1
fast Sic1 degradation
fast S phase CDK activation
optimized G1 – S phase transition
What are the molecular mechanisms governing DNA replication
Step 1: Exit from mitosis and early G1 the origin recognition complex (ORC) is recruited and associated with all DNA replication origins
Step 2: Late G1 CDK activity is still low and the Cdc6 and Cdt1 MCM loading factors associate with ORC and load the INACTIVE MCM replicative helicase complex onto each DNA replication origin.
The Pre-replication complex is formed
Step 3: DDK and S phase CDKs are activated. They phosphorylate:
MCM → activation of helicase fires up the origin of DNA replication
Cdc6 and Cdt1 → degradation (after firing there will be no loading of MCM on the newly replicated origins)
CDC45, Sld2 → Recruit GINS to MCM to facilitate elongation
replication.
Step 2: Late G1 - DDK and S phase CDKs are activated.
DDK phosphorylates two subunits of the MCM helicase.
S phase CDKs phosphorylate Sld2 and Sld3.
These phosphorylation events promote recruitment of Cdc45-Sld3 complex and the GINS complex MCM helicase activators to sites of replication initiation, which leads MCM helicases to unwind DNA.
(S phase CDKs prevent reloading of MCM helicases by phosphorylating Cdc6 and Cdt1, promoting their release from the replication origins and degradation by SCF.)
S phase CDKs also phosphorylate MCM helicases, causing the helicases to disengage from the DNA when replication is complete and export from the nucleus.
Step 3: DNA polymerases recruited to origins initiate DNA replication
What do Cdc6 and Cdt1 do?
They load MCM onto the origin of replication
Why are checkpoints of the cell cycle important?
Checkpoints ensure that the next stage of the cell cycle does not initiate prior to the completion of the preceding one.
Checkpoint pathways arrest the cell cycle progression in response to DNA damage or improper spindle assembly.
so we don’t make bad cells
What are the three main parts of checkpoint pathways?
- event sensors
- a signaling pathway
- and effector that halts cell cycle progression
What are the 4 steps of DNA damage response system (short answer w the 4 arrows) what’s next…
1) Damaged DNA activates ATM, ATR (protein kinases) and DNA-PK
2) DNA-PK, ATM and ATR phosphorylate and activate the chicks! (Chk1, Ck2) and MK2
(Chk1 and Chk2 are checkpoint kinases)
3) Chk1 and Chk2 inhibit/block cdc25, which induce DNA repair machinery!
(cdc25 normally would activate mitotic CDKs but now that it’s inhibited, it can’t)
4) This inhibits mitotic CDKs and causes cell cycle arrest
What do cells require to repress apoptosis for surviva(that binds to surface receptors)
Cells require TROPHIC FACTORS! that bind to surface receptors to repress apoptosis for survival
Caspase proteases in apoptosis
they degrade cellular proteins and apoptosis involves the activation of caspase protease
BMPs
Bone morphogenic proteins
- expressed by interdigital cells
- induce apoptosis
(Dominant negative type I BMP receptor blocks BMP signal and apoptosis (like in the diagram with the hands and one’s webbed)
(BMPs – not expressed in duck interdigital cells)
Conclusion: BMP signaling mediates interdigital cell death in the embryonic limb.
What’s Necrosis?
MESSY CELL DEATH
Different than apoptosis
cell rupture, cellular contents pouring out leading to inflammatory response, auto-immunity (messy cell death)
What is difference between necrosis and apoptosis?
Necrosis: cell rupture, cellular contents pouring out leading to inflammatory response, auto-immunity (messy cell death)
Apoptosis: cell membrane remains intact, cell is dismantled and packaged into apoptotic bodies, cellular contents within vesicles, absorbed by macrophages and recycled (clean cell death)
What are the main signs of cell death by apoptosis
Mild convolution
Chromatin campaction and margination
Condensation of cytoplasm
Breakup of nuclear envelope
Nuclear fragmentation
Blebbing
Cell fragmentation
INTER_NUCLEOSOMAL DNA FRAGMENTATION IS A FEATURE OF APOPTOTIC CELLS
Apoptotic bodies are produced
Phagocytised
Apoptotic stimuli triggers DNA degrading nucleases that cleave DNA between nucleosomes
Degraded chromatin is packaged into apoptotic bodies
The apoptotic bodies contain less that 2n DNA as compared to normal living cells
What are the two assays for the detection of fragmented chromatin that we study?
- Flow cytometry and COMET assay
How can you detect apoptotic bodies on a flow cytometry graph?
You detect these apoptotic bodies as a broad peak to the left of the C1 peak since they have much LESS DNA THAN NORMAL CELLS
What is the COMET assay process?
Cells are imbedded in agarose
Electric current is passed through the agarose
Similarly to gel electrophoresis, DNA fragments will move because of their charge
Large fragments of DNA (intact chromosomes) do not move
Fragmented chromosomes leave the cell and produce a “comet-like” tail.
The detection of such tails is indicative of apoptosis.
What did S. Brenner find?
S. Brenner: introduced Caenorhabditis elegans as a model organism to study development
Small nematode roundworm, ~1.2 mm long, transparent
Consists of 1090 somatic cells produced during development
Of these, 959 cells survive, 131 “doomed” cells die by apoptosis
What did John Sulston find?
determined complete cell lineage map
CED genes
cell death genes
What did R. Horvitz find?
R. Horvitz isolated > 4000 cell lineage (lin) mutants that do not kill some of the 131 cells
He postulated that the mutant genes must be responsible for the killing
Conclusion: Cell death is genetically controlled
- Led to search for cell death (CED) genes
Mutations in ced-3 picture meaning
ced-1 mutant: apoptotic cells are not engulfed and digested (arrows)
ced-1/ced-3 double mutant: apoptotic cells do not appear
They do not die!!!
ced-3 is necessary for apoptosis (pro-apoptotic gene)
What are the two classes of caspases?
Initiator caspases: process and activate effector caspases
- casp9 (intrinsic)
- casp8 and 10 (extrinsic)
Effector (executioner) caspases: cleave specific cellular proteins leading to apoptosis
- caspase 3, 6, 7
these caspases digest the proteins of the cell and activate nucleases that degrade DNA and chromatin
What regulates the release of Cyt C
The release of Cyt C is regulated by pro-apoptotic and anti-apoptotic proteins
Pro-apoptotic proteins: Bak, Bax, Boc
Pro-survival proteins: Bcl-2, Bcl-xl
All these proteins are homologous to Bcl-2 and contain Bcl-2 Homology domains (BH1, BH2, BH3, BH4)
BH3-only proteins: (Bad, Bim, Puma) Regulate pro-survival proteins
BH3-only proteins
Bad, Bim and Puma - regulate pro-survival proteins (Bcl-2)
Bcl-2 family proteins
Play central anti-apoptotic (pro-survival) or pro-apoptotic roles in apoptosis
Contain one or more functional Bcl-2 homology domains (BH1–4)
All participate in oligomeric interactions.
Three classes – one anti-apoptotic and two pro-apoptotic:
Only some BH3-only proteins contain a transmembrane (TM) domain that anchors other Bcl-2 family proteins in the outer mitochondrial membrane.
Pro-apoptotic proteins Bax and Bak form pores in the outer mitochondrial membrane large enough for cytochrome c escape to the cytoplasm.
Difference between intrinsic and extrinsic death pathway
Intrinsic Death Pathway
activated by proteins that reside in the mitochondria (Bak, Bax, Bcl-2, Cytochrome C (CytC)
activated in response to DNA damage, cell stress, lack of adhesion, lack of trophic signals
Extrinsic Death Pathway
activated through cell surface death receptor
activated by direct contact with other cells that direct them to self-destruct by apoptosis
How does DNA damage induce apoptosis
DNA damage or ultraviolet irradiation stimulates apoptosis:
- Induces synthesis of Puma BH3-only protein
- Puma binds to Bak, Bax, and Bcl-2.
- Bak and Bax form oligomeric pores that release Cyt c.
Cell lack of adhesion to substrate -a
Cell lack of adhesion to substrate – apoptosis in adhesion-dependent cells:
- Disruption of integrin signaling
- Release of the Bim BH3-only protein from the cytoskeleton
- Bim binds to Bak and Bax to promote pore formation and Cyt c release.
What’s more abundant, Bcl-2 or Bak proteins?
Pro-survival proteins (Bcl-2, Bcl-x) are abundant. They bind to pro-apoptotic proteins (Bak or Bax, less abundant), prevent oligomerization and close pore
What do Bad, bim and puma do?
Bad, Bim, Puma, when activated, displace Bcl-2 from Bak or Bax and allow to open the pore
What does Cyt C do when it’s released?
Cyt C is released, binds to Apaf-1 and activates Caspase 9, which activates the executioner caspases
(in intrinsic pathway)
General framework for the evolutionary progression of cancer
The progression of cancer from a normal somatic cell to a full-grown tumor can be thought to take place in four stages.
Step 1: Initiation.
Except in rare cases in which cancer is caused by a singular genetic event, such as infection by a tumor virus, we usually cannot pinpoint the initiating event but a somatic mutation is usually suspected.
Step 2: Cancer progression.
Precancerous cells acquire mutations that dysregulate growth-control pathways, causing inappropriate cell proliferation.
Most cancers progress by a process of natural selection, sequentially acquiring multiple such mutations in different pathways.
Step 3: Evasion of cancer cell elimination.
Precancerous cells are normally removed from the body by either apoptosis or immune-surveillance processes.
To progress beyond this stage, tumor cells must acquire additional somatic mutations that allow them to evade these systems.
Step 4: Tumor growth and dispersal.
Continued growth of a tumor requires a blood supply and tumor cells must acquire yet more changes that promote angiogenesis.
To spread throughout the body by metastasis, cells from a solid tumor must acquire the ability to migrate from their original site and adhere at a new location in the body.
DNA repair and surveillance molecules
Genome destabilization
P53
Inhibitors of CDKs
Sensors of DNA damage (ATM, ATR)
DNA repair genes
Cell cycle regulatory proteins
- G1/S Cyclins or CDKs
- Regulators of G1/S (Rb protein)
- Growth Factor Receptors and Signal transduction proteins
Metastatic cancer cells:
Degrade the basement membrane
Migrate on extracellular matrix (ECM) fibers away from the primary tumor to reach the blood vessels
Can be attracted by signals such as epidermal growth factor (EGF), which can be secreted by macrophages
Penetrate the blood vessel endothelial cell layer that forms the vessel walls and enter the bloodstream
Carcinoma cells how do they penetrate the ECM and blood vessel wall?
Carcinoma cells penetrate the ECM and blood vessel wall:
Use the actin cytoskeleton to extend invadopodia
Produce matrix metalloproteases and other proteases that degrade basement membranes to open a path for metastasis
What is extravasation?
circulating tumor cell adheres to the blood vessel lining in a new location and migrates through it to colonize a new tumor in the underlying tissue.
Fewer than 1 in 10,000 cells that escape the primary tumor survive to form a secondary metastatic tumor
Cancer cell genetic characteristics?
Individual chromosome number is altered
Composite chromosomes composed of pieces from different chromosomes
Colon mutation on APC tumour - consequence of this mutation
- causes the mutated cell to divide to form a localized polyp of benign tumour cells
Events that lead to formation of malignant tumours
- Loss of APC tumor-suppressor gene (chromosome 12)
- activation of K-Ras oncogene (chromosome 12)
- Loss of p53 tumor-suppressor gene (chromosome 17)
- Other changes that in combination can lead to tumor cells invading blood vessels, allowing metastasis to occur
TGF-beta receptor. Two points the effect of this active receptor has on the Smad transcription factor
1) The active receptor phosphorylates inactive SMAD in the cytosol, causing it to oligomerize and activate
2) Active Smad translocates into nucleus where it binds to the following genes to induce their transcription
What genes are activated by smad?
Smad stimulates expression of:
1) p15 (which inhibids CDK4) - this causes cells to arrest in G1
2) An inhibitor of a protease that degrades extracellular matrix
- cells do not migrate.
Secondary sex traits
changes in the body not a part of reproduction (ex. growth of pubic hair, muscle in boys and breasts in girls)
menarche
onset of menstruation (usually 12)
Cognitive reframing
strategy of dealing with emotions where we learn to look at our experience through a different frame - like turning something negative into a learning experience
Delaying gratification
putting off immediate temptations in order to focus on long term goals
formal operational thinking
abstract reasoning and logic and higher cognitive abilities (starts to go up after age 12)
Lawrence Kohlberg
moral development
learning right from wrong
Levels of reasoning:
preconventional
conventional
postconventional
Preconventional reasoning
- people reason based on self-interest, avoiding punishment
Conventional reasoning
people reason based on social conventions (traditions), can involve authority figures “Because mom said so”
Post conventional level
Level of reasoning that considers justice and fairness
Sensitive periods
windows of time during which exposure to a specific type of environmental stimulation is needed for normal development of a specific ability
ex. becoming fluent in a language –> should be introduced to that language in really early years
assimilation
fitting new information into the belief system you already posses
ex. all girls have long hair - that person has short hair but is a girl! (azlan)
accomidation
creative process where people modify their belief structures based on experience ex. that girl has short hair, so girls can have short hair