Mod8 - Cycles of Division and Growth in Cell Populations Flashcards

1
Q

Name 4 reasons why cells divide (mentioned)

A

Sustain life, propagate heritable traits, generate mass, generate diversity

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

Approximately how many cells - and cell types - are in the human body?

A

37 trillion (200 types)

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

What is the difference between embryonic and adult cell cycles?

A

Embryonic cell cycles can be simply S->M oscillators; adult stem cells have Gap phases to separate alternate S and M phases

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

What is G0 (name and function)?

A

QUIESCENCE: A dormant state which some cells remain in for years until they are ready to divide

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

Which Cdk is active in G1 phase, and which cyclin partner activates it?

A

Cdk4 AND Cdk6 (Cyclin D)

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

Which Cdk is active in G1->S phase, and which cyclin partner activates it?

A

Cdk2 (Cyclin E!)

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

Which Cdk is active in S phase, and which cyclin partner activates it?

A

Cdk2 (Cyclin A)

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

Which Cdk is active in M phase, and which cyclin partner activates it?

A

Cdk1 (Cyclin B)

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

What happens to Cdk and Cyclin levels throughout the cell cycle?

A

Cdk levels remain constant throughout, but Cyclin levels rise and fall to activate various Cdks as required

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

How do Cyclins actually activate Cdks?

A

Without a bound cyclin, the Cdk is inactive because the T-loop (or activation loop) occludes the active site;
When a cyclin binds to its partner Cdk, a conformational change pulls the T-loop away from the active site

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

Name 4 ways in which cyclin-Cdk complexes are regulated

A
  1. Phosphorylation (can be activating OR inhibitory)
  2. Ubiquitin-mediated proteolysis (provides directionality as you can’t un-degrade a cyclin)
  3. Localisation (e.g., cyclin B imported into nucleus JUST before mitotic entry
  4. CKI binding (Cdk inhibitors UPregulated in response to damage)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What could happen if the cell cycle progressed without the cell cycle checkpoints?

A

Progression without accurately duplicating and separating the chromosomes could lead to a damaged genome

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

Describe the 1st cell cycle checkpoint

A

Before entering S phase (end of G1) - “is environment favourable?” if not, Cdk4/6 activation is blocked

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

Describe the 2nd cell cycle checkpoint

A

Before entering mitosis (end of G2) - is all DNA replicated/is all DNA damage repaired? If not, Cdk1 activation is blocked

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

Describe the 3rd cell cycle checkpoint

A

Before duplicated chromosomes are pulled apart - are all chromosomes properly attached to mitotic spindle? If not, Cyclin B degradation is blocked

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

State the 4 steps required for “getting into and out of mitosis”

A

Step 1 - Switch off DNA damage checkpoints

Step 2 - Activate Cdk1 to get into mitosis

Step 3 - Silence the spindle assembly checkpoint (SAC)

Step 4 - Inactive Cdk1 to get out of mitosis

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

Describe how Cdk1 is activated (in step 2 of GIAOOM)

A
  • First, Cyclin B must bind to Cdk1 (obvs)
  • Then, it must be activated by Cdk-Activating Kinase (CAK), by phosphorylation
  • THEN, it is INactivated by Wee1 kinase via phosphorylation elsewhere on the enzyme
  • Finally, it is activated by Cdc25 which removes the inhibitory phosphates (Mitosis can now begin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Why is there such an apparently convoluted mechanism for Cdk1 activation?

A

In reality, it is more of an equilibrium than linear, due to feedback loops; the active form of Cdk1 further activates Cdc25 (positive feedback) and inactivates Wee1 until eventually, all the Cdk1 is in the active form

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

How does Cdk1 actually trigger mitosis

A

It phosphorylates various target proteins, including other kinases, which activate mitotic pathways and inactivate interphase pathways; this brings about the morphological changes associated with mitosis (e.g., spindle assembly, chromosome condensation)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

How do cells have “permission” to pass the G2/M checkpoint (in Step 1 of GIAOOM)?

A

Unreplicated or damaged chromosomes prevent entry into Mitosis, as repair pathways activate Wee1;
Once damage is all repaired, this pathway becomes silenced and equilibrium is balanced, so cell can enter mitosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

How is Cdk1 inactivated (in Step 4 of GIAOOM)?

A

Ubiquitylation and subsequent proteolysis of Cyclin B inactivates Cdk1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Why is a “less straightforward” method used in Step 4 to inactivate Cdk1, rather than just inhibitory phosphorylation?

A

Because DIRECTIONALITY! (Destruction of Cyclin B via proteolysis is irreversible)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Which protein is mainly responsible for Ubiquitylation and Proteolysis of Cyclin B to exit mitosis

A

APC acts as the E3 ubiquitin ligase (which actually joins ubiquitin to Cyclin B)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Why must the APC be carefully regulated (i.e., why can’t it be active all the time)?

A

If it were always active, it would constantly degrade Cyclin B, and mitosis would never occur

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Describe the Silencing of the SAC (in Step 3 of GIAOOM) - and the role of the APC

A

The APC becomes active during mitosis, however the Spindle Assembly Checkpoint (SAC, which is activated by unattached kinetochores) blocks FULL activation of APC until chromosomes are all aligned;

Once kinetochores are attached, SAC is silenced, APC is activated

APC degrades Cyclin B to trigger mitotic exit (APC also degrades securin, triggering separation of sister chromatids -> so anaphase onset and mitotic exit are coupled)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is erythropoietic (EPO) and what does it do?

A

It is a secreted cytokine that stimulates RBC production by activating the JAK/STAT pathway

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Name the three (mentioned at start) diseases which can result if proliferation is not controlled

A

Colon cancer; leukemia; gingival hyperplasia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Are proliferation and growth the same thing?

A

NO - proliferation is the division of cells to increase in number; cells can grow without proliferating (e.g., large neurons) due to increased protein synthesis and decreased degradation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Name the four main types of external factors that affect proliferation/growth etc in cells

A

Mitogens - stimulate proliferation (by driving cell cycle)
Growth factors - stimulate growth
Survival factors - suppress apoptosis
Differentiation factors - control cell lineage

30
Q

Name the four main types of external factors that affect proliferation/growth etc in cells

A

Mitogens - stimulate proliferation (by driving cell cycle)
Growth factors - stimulate growth
Survival factors - suppress apoptosis
Differentiation factors - control cell lineage

31
Q

What is the restriction (R) point?

A

The “point of no return” in the cell cycle - up until this point, cells are responsive to mitogenic GFs and to TGF-ß, but past the R point, the cell is committed to completing the cycle

32
Q

What is the R point in terms of Cyclins?

A

R marks the handover from Cdk4/6 to Cdk2 (driven by the rise of Cyclin E)

33
Q

How does Cyclin D rise in early G1?

A

Mitogens activate RTKs -> Ras GTPase -> MAPK pathway (Mitogen activated protein kinase) -> transcription factors -> Cyclin D

34
Q

How does the rise of Cyclin D lead to the rise of Cyclin E in late G1?

A

Cyclin D-Cdk4/6 complex phosphorylates Rb - a transcriptional repressor normally bound to E2F - causing it to release E2F

E2F then drives synthesis of Cyclin E, which then activates Cdk2, driving the G1/S transition

35
Q

Name 3 different pathways that feed into Cyclin D (note that there are other examples too)

A

Growth factors in breast -> Herceptin -> pathway

Wnts in intestine

EPO cytokine pathway

36
Q

Describe how E2F drives its own synthesis

A

Positive feedback: Cdk2 phosphorylates Rb, driving further rise in Cyclin E, until the G1/S transition

37
Q

What is one (mentioned early) example of how CyclinE-Cdk2 activity drives entry into S phase

A

Phosphorylating replication origins, thus triggering DNA synthesis

38
Q

Describe the role of proliferation in the small intestine

A

Paneth cells at the base of the crypt secrete Wnt signals, which are received by stem cells, causing them to induce cyclin D and proliferate

39
Q

Why do not all cells in the small intestine proliferate?

A

Precursor cells migrate up and out of the crypt (away from the Wnt) signals

Once out of range, they stop proliferating and then differentiate

40
Q

What are the two main types of carcinomas?

A

Adenomas (benign) and adenocarcinomas (malignant)

41
Q

State the stages from normal epithelium to metastasis (cancer)

A

Normal Epithelium -> Hyperplastic Epithelium -> (early/intermediate/late) adenomas -> carcinoma -> invasion and metastasis

42
Q

Describe what is meant by ‘Clonal Expansion’

A

One cell in a layer mutates, giving it a proliferating advantage
Then, a specific clone within this layer becomes dominant due to a second mutation
Eventually, there may be mutations which increase the mutation rate, leading to multiple parallel clonal expansions

43
Q

Name the 4 hallmarks of cancer focused on in this lecture

A
  1. Sustaining Proliferative Signalling
  2. Evading Growth Suppressors
  3. Resisting Cell Death
  4. Genome Instability + Mutation
44
Q

What is the fundamental cause of cancer?

A

Mutations in oncogenes and tumour suppressor genes (which together control proliferation, differentiation and survival of cells), leading to abnormal cell behaviour

45
Q

How do mutations in oncogenes and tumour suppressor genes “manifest themselves” in tissues?

A

They manifest as deregulated tissue homeostasis, in turn disrupting normal tissue architecture

46
Q

What was the first oncogene to be discovered, and how did it come about?

A

src -> first observed in Rous Sarcoma Virus (RSV)

Most retroviruses only had 3 genes, but RSV also had v-src, which had a cellular counterpart c-src.
c-src regulates a signalling pathway for the cell cycle; at some point, a pro-virus got accidentally integrated next to c-src and then got taken up with it, causing src to become part of the viral genome

47
Q

Define viral oncogenes

A

Mutated versions of cellular genes that regulate proliferation, found in viral genomes

48
Q

State the common Ras mutation commonly associated with cancer, and how this works

A

Ras G12->V (this blocks Ras’ ability to inactivate itself via GTP hydrolysis to GDP. Therefore, Ras stays active and drives Cyclin D synthesis even in the absence of mitogens (NOTE - ONLY A SINGLE MUTATED RAS ALLELE REQUIRED FOR THIS)

49
Q

Why does mutation of (both) Rb alleles lead to Retinoblastoma?

A

In the absence of Rb function, E2F is constitutively active, and will drive entry into S-phase even in the absence of mitogens

50
Q

Why is familial retinoblastoma far more common than sporadic?

A

Both Rb copies must be mutated for disease; if one copy is already mutated (inherited), then it is quite possible for the other to mutate as well. However, it is very unlikely for both copies to mutate by chance

51
Q

How does Neurofibromatosis arise?

A

If both copies of the NF1 tumour suppressor gene are mutated and NF1 function lost, Ras is constitutively active, causing cancer

52
Q

What happens inside cells in the small intestine when Wnt signals are not present?

A

A 3-protein complex forms [active GSK-3ß + APC + ß-catenin]. Active GSK-3ß phosphorylates ß-catenin, causing it to be degraded and no Cyclin D to be produced

53
Q

What happens inside cells in the small intestine when Wnt signals ARE present?

A

GSK-3ß is inactivated, so ß-catenin is NOT phosphorylated and degraded; ß-catenin translocates to the nucleus, where it stimulates Cyclin D synthesis, causing proliferation

54
Q

What are APC and ß-catenin examples of, respectively?

A

Tumour suppressor gene + Oncogene

55
Q

Why do ß-catenin mutations lead to colon cancer?

A

(NOTE: ONLY 1 DOMINANT MUTATION REQUIRED)

This can prevent phosphorylation + degradation of ß-catenin, so Cyclin D is continuously produced even in the absence of Wnt signals, leading to continuous proliferation of non-differentiating cells
(leading to Hyperplasia -> Polyp -> Adenoma -> Adenocarcinoma)

56
Q

What is TFG-ß and what can happen if it is mutated?

A

It is an important tumour suppressor (“a brake on the cell cycle”) - mutation can permanently “release the brake” on the cell cycle

57
Q

Name 3 examples of apoptosis in normal cell physiology

A

Tadpole tails during embryonic development; separating digits in mouse paw; eliminating immune cells which recognise “self”

58
Q

Describe p53 concentration under normal conditions and how this is maintained

A

Normally low, because Mdm2 ubiquitinates p53, so it is proteolysed

59
Q

How does p53 normally pause the cell cycle when the cell is under stress?

A

Stress inhibits Mdm2, so p53 is stabilised, p53 then activates p21, which inhibits Cdk2, thus preventing the transition to G2 phase

60
Q

Distinguish between the intrinsic and extrinsic apoptosis pathways

A

Intrinsic: internal signals
Extrinsic: external signals

61
Q

How do both the intrinsic and extrinsic apoptosis pathways actually cause cell death?

A

Activation of CASPASES, a family of proteases which lead to protein and DNA digestion

62
Q

How are the mitochondria involved in apoptosis

A

Normally, cytochrome c is localised within the mitochondria. However, the formation of pores in the Outer Mitochondrial Membrane (due to pro-apoptotic signals) can allow cytochrome c to be released into the cytosol, leading to apoptosis

63
Q

How does the release of cytochrome c into the cytosol lead to apoptosis?

A

Cyt c is a part of the APOPTOSOME that is assembled, with active caspase 9 at the centre

Active caspase 9 then causes proteolysis of death substrates (e.g., ICAD, lamin, vimentin and actin)

64
Q

How does proteolysis of ICAD (one of the death substrates) lead to apoptosis?

A

ICAD is an inhibitor of CAD (Caspase-Activated DNase); when caspase degrades ICAD, CAD is free to digest DNA

65
Q

How is p53 involved in the “fight to control the pore” between survival and death factors?

A

p53 inhibits pro-survival factors, and brings the balance more in favour of pro-death factors (p53 mutations can prevent apoptosis as pro-survival factors are not inhibited)

66
Q

What is the role of BcI-xL and BcI-xLi in apoptosis?

A

BcI-xL is a pro-survival factor; the drug BcI-xLi inhibits it, thus enhancing Taxol activity and sensitising cancer cells to chemotherapy

67
Q

Briefly describe the extrinsic apoptosis pathway

A

Death ligands bind to extracellular death receptors, which then activate initiator pro-caspases to active Caspase 8 and Caspase 10, which then activate executioner procaspases, which leads to proteolysis of death substrates

68
Q

Name 3 death signals (extrinsic)

A

FasL, Tnf-alpha, Trail

69
Q

What is the function of the Fas extrinsic apoptosis pathway?

A

Eliminating T cells that recognise “self”
If a T-cell binds an antigen-presenting cell recognising “self” then FasL is expressed, which then binds to its own Fas receptor, inducing apoptosis

70
Q

What can happen if there are defects in the Fas pathway?

A

Autoimmune syndrome

71
Q

Describe the impact of the MYC oncogene on cancer incidence in mice

A

Overexpressing the MYC oncogene alone did not cause cancer, as apoptosis balanced out the hyperproliferation

Only when BcL-xL is also activated does cancer occur, as hyperproliferation is no longer balanced by apoptosis