Oncogenes and Tumour Suppressor Genes Flashcards

1
Q

Which distinct genetic changes are involved in tumour development?

A

Activation of oncogenes (from proto-oncogenes) and inactivation of tumour suppressor genes

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

What is a proto-oncogene?

A

A normal gene that becomes an oncogene after being mutated - usually promotes cell growth.

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

What is an oncogene?

A

A protein coding gene, that when activated/overexpressed/mutated has the potential to promote oncogenesis. Accelerator of a car that gets stuck.

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

What is oncogenesis?

A

The process by which healthy cells become transformed into cancer cells

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

What is an oncovirus?

A

A cancer causing virus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is a retrovirus?

A

An RNA virus which produces dsDNA copies of their genome that integrate into the host cell chromosome.

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

How are retroviruses grouped?

A

On their structure and replication method within the host

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

What is a tumour suppressor gene?

A

A protein coding gene that can slow cell growth/division, repair DNA mistakes and promote appropriate apoptosis. Absence/repression/inactivation of the gene can promote oncogenesis. Brakes of a car that don’t work.

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

What type of mutations occurs in tumour suppressor genes?

A

Loss of function mutations (inactivation) - usually “reccessive” ie. two copies of the mutation needed to cause cancer.

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

Describe the two hit hypothesis

A

As loss of function mutations are “reccessive”, two hits are needed to cause cancer. A child with an inherited mutation is more likely to get cancer as they only need one extra “hit” - patients usually younger and have multiple tumours. A child with no inherited mutations is less likely to get two hits in the same gene, as a result the patients are older and only have one tumour usually.

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

How common is inheritance of tumour suppressor gene mutations?

A

They’ve been found in some familial cancers, causing them to run in the family. APC gene mutations are associated with increased colon cancer risk in people with familial adenomatous polyposis. Most tumour suppressor gene mutations aren’t inherited but acquired - acquired TP53 mutations been found in >50% of human cancers

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

What are the two main cellular functions of tumour suppressor genes?

A

Caretakers and gatekeepers

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

Describe the caretaker function of tumour suppressor genes

A

They maintain chromosomal integrity (genome stability). Includes DNA repair genes eg. TP53 -> p53 which also has gatekeeper properties

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

Describe the gatekeeper function of tumour suppressor genes

A

Negative regulation of cell cycle. Inhibition of proliferation, induction of apoptosis, inhibition of angiogenesis. eg. Retinoblastoma protein

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

What is retinoblastoma?

A

A rare cancer usually affecting children under 5. Starts in the light sensitive lining in the retina and can occur in one or both eyes. Affects 50-60 kids in the UK per year. If caught early, 95% treatment success but metastasis can be fatal.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are the causes of retinoblastoma?

A

40% of cases - mutations in RB1 gene, usually presenting as bilateral retinoblastoma. Mutations can be inherited or acquired in the womb during early development. One functional copy of RB1 gene produces sufficient RB protein.
60% of cases - Unknown causes, no mutant gene. Usually unilateral

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

How does RB protein function?

A

When a cell reaches the G1 checkpoint, E2F (a TF) lets S phase proceed. RB protein inactivates E2F by binding to it and not allowing the cell cycle to proceed (stops replication. RB protein is inactivated by phosphorylation, so S phase proceeds. Mutant RB protein cannot bind to E2F regardless of phosphorylation state, so S phase always proceeds - uncontrolled replication.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

How can tumour suppressor genes also act in a dominant manner?

A

In terms of causing cancer, tumour suppressor genes are recessive in that two mutations are required. But in terms of causing predisposition to cancer, they are dominant, as an inherited mutation makes cancer more likely to occur

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Describe p53

A

It’s a TF that acts in many pathways to prevent mutations and stabilise the genome. It’s encoded by TP53 gene on chr 17. Expressed in nearly all cells

20
Q

What is the role of p53?

A

It responds to stresses such as hypoxia, DNA damage, inhibition of transcription, oncogene signalling, radiation. Causes apoptosis, senescence, autophagy, cell cycle arrest etc. that lead to tumour suppression.

21
Q

What are the two mechanisms by which p53 stops tumours?

A

Response to DNA damage/stress is activation of a gene that stops the cell cycle.
Triggers apoptosis in damaged cells

22
Q

Describe the relation between p53 mutations and cancer.

A

50% of cancers have TP53 mutations. Rarely, the mutations are inherited, which leads to Li Fraumeni syndrome, where 50% of patients get cancer by age 30.

23
Q

How does p53 result in cell cycle arrest?

A

p53 induction activates cell cycle checkpoints - the highest level being at G1/S checkpoint, so damaged genomes don’t replicate. Here, p53 upregulates p21 which binds to CDK1 or CDK2 complex, preventing entry into S phase. This is either reversible or not (senescence).

24
Q

How does p53 induce DNA repair?

A

p53 regulates NER. It’s role is best characterised in reponse to UV, which upregulates p53 expression (also other carcinogens like H202 and radiation). p53 then promotoes expression of genes in the repair pathway. p53R2 codes for ribonucleotide reductase and Gadd45 which recognise and bind DNA damage. p53 may also interact with BER proteins like AP endonuclease and DNA Polymerase

25
Q

How is p53 involved in angiogenesis?

A

p53’s activity as a transcription factor allows it to regulate angiogenesis genes. Loss of p53 -> increased tumour vascularisation.

26
Q

How is p53 involved in apoptosis?

A

p53 switches on BAX in healthy cells, which initiated drilling of the mitochondrial membrane and release of cytochrome C -> apoptosis. p53 loss results in evasion of apoptosis

27
Q

How is p53 regulated in healthy cells?

A

Mdm2 E3-ubiquit keeps cellular p53 levels low by binding to the transactivation domain, preventing its TF ability. This also promotes movement of p53 to the cytosol where it’s bound by ubiquitin and degraded by proteasomes.

28
Q

How is p53 regulation different in stressed cells?

A

DNA damage triggers kinases to phosphorylate p53 at various sites. This results in Mdm2 not being able to bind to p53, so it isn’t degraded.

29
Q

How else is p53 stabilised, apart from phosphorylation?

A

ARF protein can act as a decoy, binding to Mdm2, making it unable to bind to p53 and cause its degredation, leading to p53 build-up in the nucleus. Also, mutations in p53 don’t allow p53 to bind to Mdm2

30
Q

Describe the discovery of the viral oncogene

A

Rous showed that a filtered cell extract from a chicken’s spontaneous spindle cell sarcoma causes tumours in healthy chickens. The fine filter proves that the cancer doesn’t come from the cell transplant but from a small infectious agent - Rous Sarcoma Virus. Cells infected in vivo with RSV develop characteristics commonly found in cancer cell cultures, and the cause was found to be v-src gene. Retroviral oncogenes help us to understand cancer as a genetic disese

31
Q

How do oncogenes arise?

A

By the mutation, or overexpression of a proto-oncogene (usually promotes cell growth). This can lead to constitutive activation of the proto-oncogene, turning it into an oncogene, leading to uncontrolled growth -> cancer. Mutations can be inherited, usually acquired.

32
Q

In which ways are proto-oncogenes turned into oncogenes?

A

Chromosomal rearrangements - Change in chromosome structure: deletion, inversion, translocation
Gene reduplication - Additional copies of a gene -> too much protein
Point mutation - Increasing protein activity of affecting normal regulation

33
Q

How can a chromosomal rearrangement of a proto-oncogene result in an oncogene?

A

Translocation of the proto-oncogene to control under a new promoter leads to overproduction of a normal protein

34
Q

How can gene amplification of a proto oncogene result in an oncogene?

A

Gene amplification results in overproduction of a normal protein

35
Q

How can point mutations result in the transformation of a proto-oncogene to an oncogene?

A

A point mutation within the gene can result in a hyperactive/degredation resistant protein. Mutation within a control element can result excessive amounts of normal protein.

36
Q

How are Ras proteins regulated?

A

Ras bound with GDP is inactive. A GTP exchange factor activates Ras by swapping GDP for GTP. Ras then activates a number of TFs that activate genes associated with promoting cell cycle activity and cell division. Ras’ intrinsic GTPase activity changes GTP to GDP, inactivating itself. This is aided by GTPase activity accelerating protein.

37
Q

How can mutations in Ras proteins lead to constitutive activation?

A

If the GTPase part of Ras is mutated, it won’t be able to inactivate itself.

38
Q

How do Ras mutations lead to cancer?

A

If Ras cannot inactivate itself, there will be uncontrolled cell division/growth. This is a gain of function mutation, so only one copy is needed.

39
Q

What are Ras proteins and list the main different types

A

GTPases that have roles in transmission of cellular signals via their molecular switch action. HRAS, KRAS, NRAS most important types and mutants are found in about 20-30% of tumours. The genes code for HRAS, NRAS, KRAS4A and KRAS4B - highly homologous

40
Q

Describe the role of the different Ras proteins

A

They have identical effector binding domains and can interact with the same set of downstream effectors, but differences in their post translational modifications mean that they are part of different signalling pathways

41
Q

What is Burkitt’s lymphoma?

A

Cancer resulting from a chromosomal rearrangement of most commonly a region of 8 to 14 and less commonly a region of 8 to 2 and 22.

42
Q

Describe the genetic changes that occur to lead to Burkitt’s lymphoma

A

At the chr 8 breakpoint there is a gene called MYC (the cellular equivalent of an RNA virus that causes sarcomas in chickens). Chromosome 14, 2 and 22 breakpoints contain Ig heavy chain, kappa light chain, and lambda light chains respectively. The translocation puts Myc (codes a TF usually under strict regulation) under the Ig promoters, which causes continuous Myc expression, leading to TF activating patheways that shouldn’t be permanently activated, causing cancer. Translocations frequently found in leukaemias and lymphomas.

43
Q

What is the Myc family of oncogenes?

A

A TF that regulates many genes involved in functions such as cell cycle, protein biogenesis, cell adhesion, metabolism, signal transduction, transcription, translation. In >50% of cancers, MYC is deregulated - poor prognosis and unfavourable patient survival

44
Q

What are double agent genes?

A

Proto-oncogenes with tumour suppressor function. Have been identified in 12 major cancer types. Their mutations types are consistent with those of oncogenes, but their incidence is similar to tumour suppressor genes. A single mutation event could release oncogenic function and eliminate tumour suppressive function - one hit cancer

45
Q

Give an example of a double agent gene and explain its effects

A

NOTCH receptors. Have an ongogenic role in T-lineage acute lymphoblastic leukaemia, and a tumour suppressor function in squamous epithelia. Also TF TP53, kinase BCR, and FAS are all double agents too.

46
Q

Give an example of some oncogenic gain of function mutations in p53

A

Some p53 mutants exert a tumorigenic gain of function effect by downregulating AMPK (a usually tumour suppressive energy sensor). Also they can stimulate the Warburg effect in cancer cells.