tumour biology/signalling/genomic instability Flashcards

lecture 37

1
Q

what does Rb control

A

the G1/S checkpoint.

must be phosphorylated by CDK4,6/cyclin D to release E2F which acts as a TF for S phase proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

what does E2F do

A

activates transcription of itself and cyclin E/cdk2 which further stimulates Rb phos and more E2F.
overall the E2F coordinates regulation of genes that together promote entry into S phase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

what happens when Rb/E2F binds DNA

A

recruits hisotne deacetylases and chromatin remodelling factors to the E2F responsive promoters to repress them

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

how many cancers does Rb disregulation occur?

A

almost all. but different players in the pathway in different cancers.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

whats the pathway inc Rb and how can it be compromised at each stage

A

p16INK4a inhibits cyclinD+CDK4/6. it can be mutated, deleted or promoter hypermethylated (silences).

  • cyclinD+CDK can be amplified
  • Rb can be mutated or deleted
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

how does p53 work

A

short half life so normally low concs.
- DNA damage stablises it so conc rises.
-p53 acts as a TF for the CDKI p21CIP which inhibits almost all cdk/cyclin complexes.
- leads to G1 and G2 arrest.
if DNA damage is severe it can activate pro-apoptotic genes eg BAX

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

how many cancers have mutant p53

A

about half of all human cancers.

the bioactive form is a tetramer so mutating one allele sig reduces prob of a normal tetramer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

what does the INK4a gene encode

A

2 potent tumour suppressors
- p16INK4a
-p14INK4a
the exon combination is different.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

how is p53 activated in oncogene activation

A

ARF accumilates and activates p53 in response to abnormal proliferative signals such as oncogene activation.
- ARF accumilation sequesters mdm2 which would normally have targeted p53 for degredation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

what activates p53 in DNA damage

A

AT protein kinase inhibiting mdm2 and activating p53.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

so what happens in general when p53 is mutated

A

cell can survive with oncogenes on and gene amplification.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

what is essential to overcoming replicative senescence

A

deregulation of both Tb and p53.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

phenotype of APC

A

100s of adenomatous polyps in the large intestine following inherited mutation of APC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

describe the APC protein

A

cytoplasmic. mutated in most colonic adenomas and carcinomas.
- regulates Wnt signalling pathway, crucial in development and maintenance of tissue organisation. cell to cell signalling.
- control beta catenin levels by promoting its phos and degredation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

what is beta catenin

A

a bi functional molecule.

  • forms complex with e cadherin at plasma membrane in adherens junctions.
  • in cytoplasm it is phosphorylated by GSK3beta and rapidly degraded by the proteasome.
  • other regulatory proteins also act on beta catenin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Wnt signalling pathway

A

Wnt ligand binds receptors. intermediates inhibit GSK3beta.
- unphos beta catenin is stabilised, translocated to the nucleus, promotes transcription of growth genes (eg c-myc) via the TF TCF-4 (which is complexes with)
TCF-1 inhibits TCF-4

17
Q

what is Wnt

A

a paracrine growth factor

18
Q

what is the beta catenin/TCF complex

A

a master switch in intestinal crypts controlling prolif vs differentiation via c-myc etc

19
Q

why do cells fail to die

A
  • p53 inactivation

- telomerase activation

20
Q

how do cancer therapies, radiation and chemo work in general

A

by inducing apoptosis.

in tumour cells at least one death pathway remains intact.

21
Q

cancer mutation to block apoptosis targets what

A
  • death receptors (fas, TNFR)
  • activation of anti apoptotic protiens eg Bcl-2
  • downregulation of proapoptotic proteins eg Bax and p53 (which activates Bax, PUMA, NOXA)
  • doesnt target the caspases
22
Q

why is genetic instability important in oncogenesis

A

normal to cancer reuires probably at least 6 mutations in a single cell, several in tumor suppressor genes hence needing to be in both alleles. for this to happen within a lifespan the mutation rate has to be sped up by acquisition of genetic instability, called by some ‘the mutator phenotype’.

23
Q

what is the evidence for genetic instability is malignant cells

A
  • most solid tumours are aneuploid. abnormal numbers and arrangements.
  • most have deregulated p53 which normally arrests the cell cycle to allow DNA repair
  • mutations in DNA repair genes gives an increased susceptability to cancer.
24
Q

the 4 known major DNA repair mechanisms

A

1 - DNA mismatch repair
2 - nucleotide excision repair
3 - DNA strand break repair
4 - fanconi anaemia repair

25
Q

what does mismatch repair correct

A
  • mismatched bases like C-t instead of C-G.
  • insertion/deletion loops, commonly occur where short sequences are repeated ie CACACACA. microsatellite sequences. these occur often in the human genome.
26
Q

what happpens when mismatch repair fails and when does it do so.

A

mutation rates rise 100-1000 times.

  • genes: MLH1, MSH2. mutated in hereditary non-polyposis colorectal cancer (HNPCC). and 2-5% of all colorectal cancers such as lynch syndrome.
  • also sporadic changes such as MLH1 silencing due to promoter methylation, found in about 15% of colorectal cancer.
27
Q

name 2 things that NER repairs

A

crosslinked adjacent thymines crosslinked by UV light. or a carcinogen attached to a base.

28
Q

what happens when NER fails

A

xeroderma pigmentosa

29
Q

what does DNA strand break repair do

A

fixes double and single strand breaks which can be induced by ionising radiation, chemical carcinogens and sometimes in virally infected cells.

30
Q

genes in DNA strand break repair

A

BRCA1 and BRCA2 in double strand break repair

31
Q

whats fanconi anaemia repair

A

used for the repair od DNA intrestrand crosslinks eg by acetaldehyde (ethanol metabolite) or formaldehyde (smoking) which would otherwise block both transcription and replication.
mutations in the mechanisms lead to a range of cancers and fanconi anaemia

32
Q

where do you find chromosomal rearrangements

A

in solid tumours, particularly extensive in carcinomas. the changes are not characteristic of a specific cancer except in the case of some leukaemias.
- rearrangements often involve tuour genes

33
Q

how do aneuploidies arise

A

poorly understood but the loss of a functional p53 pathway is usually critical

34
Q

what’s an aneuploidy

A

an abnormal number of chromosomes

35
Q

what are karyotypic rearrangements

A

structural rearrangements of the chromosomes.

36
Q

overview of possible damage that leads to genetic instability

A

1 - damage sensors
2 - cell cycle checkpoint controllers
3 - dna repair mechanisms
4 - apoptosis

cancer is a multistep process, reflecting the accumulation of multiple genetic changes to oncogenes and tumour suppressor genes.