Cancer Biology Flashcards

1
Q

Which genetic alteration is pancreatic cancer commonly a/w?

A

K-RAS at codon 12

Mnemonic: Pankreas

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

Which genetic alterations are cutaneous melanomas commonly a/w?

A

BRAF

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

Which genetic alterations are basal skin carcinoma and medulloblastoma commonly a/w?

A

PTCH

Mnemonic: BiTCcH

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

Which genetic alterations are thyroid cancers commonly a/w?

A

RET proto-oncogene

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

Which genes are a/w HNPCC?

A
  • Mismatch repair genes (most common ones in bold)
    MLH1
    MSH2, MSH3, MSH6
    – PMS1, PMS2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

How does APC lead to polyps?

A

During differentiation from stem cells to crypt cells, APC-gene (aka β-catenin) gets turned on. This leads to a progenitor-like phenotype, causing abnormal, continued proliferation → polyp formation

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

Which famous syndrome is a/w p53 mutation?

A
  • Li Fraumeni syndrome
    – a/w breast, lung cancers, brain tumors, sarcomas
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is APC gene?

A

It is a tumor suppressor gene a/w colon (FAP), stomach, and intestine tumors.

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

Which CDKs phosphorylate RB1 protein?

A

CDK4/6

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

What other cancers are patients with familial retinoblastoma at risk for?

A
  • Osteosarcomas
  • Lung Ca
  • Kidney Ca
  • Bladder Ca
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Is p53 inactivated by mutations only?

A
  • No, it can be inactivated in two other ways:
    – Viruses with proteins that inactivate p53
    – Deletion in INK4A-ARF locus (common in cancers). The ARF (aka p19) region makes proteins that control p53 activation through MDM2 inactivation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Where on DNA does p53 bind?

A

It binds to single-strand regions generated in damaged DNA, including those at telomeres.

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

Is p53 activated or inactivated by DNA damage?

A

Activated

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

Which downstream proteins does p53 activate?

A

p21, GADD45A, Bax, MDM2, PCNA, NFκB, and others.

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

How does p53 cause apoptosis?

A

It activates BAX, BID, and PUMA.

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

What is the function of MDM2?

A
  • Negative self-regulator of p53
    – Produced by p53 itself
    – MDM2 binds to p53 to cause p53 degradation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What’s the first step for ATM after radiation damage to DNA?

A
  • Auto-phosphorylation
  • It then Phosphorylates a number of effector proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What is the function of ATM as it related to DNA repair?

A

It coordinates DNA repair by activating repair enzymes, including p53 through phosphorylation.

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

How does CHK2 interact w/ CDC25C to regulate the cell cycle?

A
  • ATM activates CHK2
  • CHK2 phosphorylates CDC25C phosphatase, inactivating it and inhibiting cell cycle
    – CDC25C phosphatase normally removes inhibitory phosphates from cyclin-CDK complexes promoting cell cycle progression
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

How does p53 affect the G2-M transition?

A

p53 inhibits CDC25C → prevents G2-M transition.

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

What is the main mechanism of lymphoma cell death following radiation?

A
  • p53-mediated apoptosis
  • p53 mutations make them radioresistant
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

How do p53 mutations affect lymphocyte and thymocyte radiation sensitivity?

A

This mutation makes them more radioresistant.

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

What is the function of p53?

A

It is a tumor suppressor gene and plays a role in cell cycle regulation and apoptosis.

It also plays a vital role in DNA damage surveillance and almost all forms of DNA repair, including NHEJ, HRR, MMR, BER, and NER.

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

What is the function of BRCA1/2?

A
  • Tumor suppressor genes.
  • Play a role in homologous recombination.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Which cancer is a/w ABL1 mutation?

A

CHRONIC myeloid leukemia

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

What is the function of ABL?

A

It is an oncogene activated through chromosome translocation, forming BCR-ABL (9:22)

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

What’s the function of the PI3K-Akt-mTOR pathway? What’re the steps?

A
  • Function:
    – Anti-apoptosis
    – Pro growth
    – Pro angiogenesis
    – Pro ribosome biogenesis
  • Some activators:
    – VEGF
    – IGFR
    – HIF1
  • Steps:
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What is the function of PTEN?

A
  • Tumor suppressor gene
  • Counteracts PI3K
    – PI3K: PIP2 → PIP3
    – PTEN: PIP3 → PIP2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What’re some drugs that target the mTOR pothway?

A
  • Everolimus
  • Temsirolimus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What is the function of NF1?

A

It is a tumor suppressor gene a/w neurofibroma, and sarcoma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is the function of WT1?

A
  • Tumor suppressor gene and a transcription factor
  • When mutated or absent, it leads to the development of Wilms tumor.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Which main proteins regulate p53?

A
  • ATM
  • ATR
  • DNA-PK
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Approximately what % of cancers have p53 inactivation?

A

≥ 50%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Which domain of the p53 protein is acted on by viral inactivating proteins?

A

DNA-specific binding domain, the same region affected by mutations in the majority of cancers.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Which domain of the p53 protein is mutated in the majority of cancers?

A

DNA-specific binding domain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Which viruses contain proteins that inactivate p53?

A
  1. HPV (E6)
  2. Adenovirus (E1B55K).
  3. SV40 (large T)

Such proteins are absent in EBV virus.
Mnemonic: HAS proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Which kind of viruses contain proto oncogenes?

A

Retroviruses contain altered/mutated proto-oncogenes, usually acquired through their vertebrate hosts.

They were instrumental in the study of oncogenes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are exons? What are introns?

A

Exons → coding regions
Introns → non-coding regions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What are the most common ligands for EGFR?

A
  • EGF
  • TGF-α
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What is EGFR? What are the different types of EGFR, its ligands, and its function?

A
  • Cell surface tyrosine kinase receptor family. Members:
    – EGFR (Her1; ErbB)
    – EGFR2 (HER2; ErbB2)
    – EGFR3
    – EGFR4
  • Activated by the epidermal growth factor (EGF) ligand and TGF-α.
  • Enhances cell proliferation
    – Pathway: See figure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What’re the molecules involved in the EGFR pathway?

A
  • In order:
    – EGFR
    – Ras/Raf
    – MAPKK (Mek)
    – MAPK (Erk)
  • Figure:
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Does EGFR exist as a homo or a heterodimer?

A

Homodimer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What mechanism normally inactivates tumor suppressor genes?

A

Loss of heterozygosity, since they act in a recessive manner.

Also inactivated by epigenetic silencing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What mechanism(s) activate oncogenes?

A
  • Deletion
  • Point Mutation
  • Retroviral integration
  • Gene amplification
  • Chromosome rearrangement
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What is the function of the promoter region?

A

Determines whether a gene is transcribed or not.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What is the function of BRCA1/2?

A

They are tumor suppressor genes.

They play a role in homologous recombination.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Why are levels of p16 elevated in HPV-driven cancers?

A
  • Normal p16 negative feedback loop fails
    – RB is inactivated by E7 and becomes insensitive to p16 levels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

Which cancers are a/w p16-INK4A mutations?

A
  • Pancreatic
  • Esophageal
  • Malignant Melanomas

PEMs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What is the role of p16-INK4A?

A
  • Protein product of the p16 tumor suppressor gene
  • Inhibits CDK 4/6:cyclin D/E complex
  • Prevents RB phosphorylation
  • Limits cellular proliferation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

How is p53 usually modified?

A
  • Phosphorylation
  • Acetylation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What is the role of p14-INK4A?

A
  • Inhibits MDM2-mediated degradation of p53
  • ↑ p53 → cell cycle inhibition
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Which cancer is a/w ALK translocation?

A

NSCLC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Which cancer is a/w amplified C-MYC?

A
  • Burkitt Lymphoma
  • Leukemias
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

Which cancer is a/w amplified L-MYC?

A

SCLC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Which cancer is a/w mutated Notch1?

A

T cell acute lymphoblastic leukemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

Which cancer is a/w KIT and/or PDGFR receptor kinase activations?

A

≥ 90% gastrointestinal stromal tumor (GIST)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

Which drug targets the KIT and/or PDGFR receptor kinase activations?

A

Imatinib (Gleevac)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What is the oncogene addiction model?

A

Some tumors rely on the continued activity of a single oncogene for growth and survival.

Inactivation of this gene can halt the progression of such cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What kind of enzymatic activity does telomerase exhibit?

A
  • Reverse transcriptase.
  • Telomerase complex has an RNA template from which a repeating TTAGGG sequence is reverse transcribed and added to the ends of the DNA.
  • Mnemonic: Telomere adds TAGs to chromosome ends
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Do all tumor cells have deficiences in DNA repair?

A

No

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Why are levels of p16 elevated in HPV-driven cancers?

A

The p16 negative feedback loop fails. RB is inactivated by E7, and becomes insensitive to p16 levels.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Which notable chromosomal instability disorder is not a/w cancer?

A

Cockayne’s syndrome: stunted growth, impaired nervous system development, photosensitivity, and premature aging.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What causes Cockayne’s syndrome?

A

Deficiency in nucleotide excision repair, akin to what happens in XP.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

Which cancer is a/w Bloom’s syndrome?

A
  • Leukemia
  • Lymphoma

Mnemonic: Bloom’s

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

Which cancer is a/w Fanconi’s anemia?

A

Leukemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

Which cancer is a/w Nijmengen breakage syndrome?

A

Leukemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

Which cancers are a/w ataxia telangiectasia?

A
  • Leukemia
  • Lymphoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

How is ATM activated following radiation?

A
  • Autophosphorylation
    – Inactive dimer to an active monomer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

How does ATM interact with MDM2?

A

It phosphorylates MDM2, preventing it from inhibiting p53.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

How does ATM interact with H2AX?

A

It phosphorylates H2AX, resulting in γH2AX formation, which is a marker of DNA damage.

71
Q

How does ATM regulate the cell cycle?

A

It activates CHEK2, which phosphorylates CDC25 phosphatase, which can now not dephosphorylate CDK1 (a step necessary for G2-M progression)

72
Q

How does the MRN complex interact with ATM?

A

MRN activates ATM.

73
Q

What is the function of NF-κB?

A

Irradiation → activation
Transcription factor → inhibits apoptosis
Activation → tumor progression

74
Q

How do mutations in the Ras family affect the Ras proteins?

A
  • Prevent GTPase activity.
  • Ras proteins are active and inactive when bound to GTP and GDP respectively. By preventing GTPase activity, mutations can lead to continuous activation of the RAS protein and its downstream signalling.
75
Q

Which cellular compartment is a/w the RAS enzyme?

A

RAS enzymes are located on the inner surface of the plasma membrance

76
Q

Are mutations in genes of the Ras family oncogenic or tumor suppressing?

A
  • Oncogenic
  • Mutated in almost 25% of all cancers
77
Q

What kind of protein is RAS?

A

A small GTPase

78
Q

What are the phenotypic characteristics of people with ataxia-telangiectasia?

A
  • Progressive ataxia
  • Telangiectasias of the sun-exposed regions
  • Dysphagia
  • Dysarthria
  • Chronic lung diseases
  • 24% increased risk for leukemias and lymphomas
  • Hypogammaglobulinemia
79
Q

What is the inheritance pattern of a loss of function mutation in tumor suppressor genes on a pedigree level?

A
  • Autosomal dominant
  • One copy of the mutated gene inherited from the parents is present in all body cells.
    – During life, this person will very likely lose the normal gene copy (loss of heterozygosity) in at least a few cells in their body.
    – This will lead to cancer, giving the impression of dominance
80
Q

What’s the inheritance pattern of a loss of function mutation in tumor suppressor genes on a cellular level?

A
  • Autosomal recessive
  • One copy of a tumor suppressor gene is enough to suppress tumor growth
81
Q

What is the inheritance pattern of a gain of function mutation in oncogenes on a pedigree and cellular level?

A

Dominant in both

82
Q

What is DCC and what’s its function?

A

DCC (disseminated colon carcinoma) is a tumor suppressor gene
Often altered in colon cancer

83
Q

How do levels of p21 change in irradiated cells?

A

↑ 2/2 ↑ p53 expression

84
Q

Is neurofibromatosis a/w inactivation of tumor suppressor genes or activation of oncogenes?

A

Inactivation of tumor suppressor genes

85
Q

What is the function of BCL2?

A
  • Anti-apoptotic protein
  • Counters the release of cytochrome c from mitochondria
86
Q

Which virus produces E6 and E7 proteins?

A

HPV

87
Q

What does E6 do?

A

Inhibits p53

88
Q

What does E7 do?

A

Inhibits RB

89
Q

What happens to tumor cells (or normal cells) with wt p53 after irradiation?

A

Genotoxic insult → Senescence

90
Q

What defines a stem cell?

A
  • Ability to self renew
  • Ability to produce daughter cells that have limited mitotic potential and are destined to differentiate
91
Q

What is chromothripsis?

A

A catastrophic event is where portions of chromosomes break apart and join together randomly. It can result in many oncogenic changes in a single event.

92
Q

What is a driver mutation?

A

A mutation that occurs early on in the development of cancer.

Usually occurs in a recessive or dominant oncogene.

93
Q

What is clonal evolution?

A

Changes in the genetic makeup of tumors over time

94
Q

What drives clonal evolution?

A

Mutations, such as through imperfect DNA repair.

Mutations that are disadvantageous are selected out.
Mutations that are advantageous become enriched.

95
Q

What are mutational signatures?

A
  • Distinct patterns of alterations in DNA bases.
  • Only considers point mutations.
  • Signature refers to the fact that these patterns can be specific to certain mutagens, such as aging, smoking, etc
  • By knowing the pattern, you can deduce the identity of the mutagen
96
Q

What influences the mutational signatures of DNA?

A

Specific mutagen
Bases surrounding the base of interest
DNA damage repair processes at work

97
Q

How do mutations in the RAS family affect the proteins?

A

They usually prevent GTPase activity.

RAS proteins are active and inactive when bound to GTP and GDP respectively. By preventing GTPase activity, mutations can lead to continuous activation of the RAS protein and its downstream signaling.

98
Q

What are some hallmarks of cancer?

A

Sustained proliferative signaling
Replicative immortality
Resisting cell death
inducing angiogenesis
activating invasion and metastasis

99
Q

What are some characteristics of cancer stem cells?

A
  • Small subset of cancer cells
  • Can give rise to cancer stem cells or non-stem cells
  • More radioresistant and chemoresistant than proliferative tumor cells
  • Reside in specialized niches that impede chemo and radiation activity, eg hypoxic regions
  • Often increase after radiation
  • Have high anti-oxidant levels
  • Slow cycling
  • High MDR1 expression
  • Reprogramming
100
Q

How is the tumorigenic capacity of cancer cells quantified?

A

By injecting them in mice and determining the number required to cause tumors in 50% of sites (TD50).

It inversely correlates with TCD50

101
Q

What are Yamanaka factors? What’s their significance?

A

They can induce pluripotent cells from mature cells, and include:
1. c-Myc
2. Oct3/4
3. Klf4
4. Sox2

Mnemonic: Yamanaka MOKS

102
Q

How did cancer stem cell theory gain fame?

A

When it was discovered that AML patients have tumor-initiating cells that grew tumors in NOD/SCID mice.

103
Q

Why are Till and McCulloch famous?

A

They discovered that bone marrow-derived colonies arose within 10 days in the spleens of WBI mice.

104
Q

Which stem cells reside in the intestinal crypts?

A
  1. Rapidly active intestinal stem cells (LGR5+)
    – radiosensitive
  2. Quiescent, reserve intestinal stem cells
    – Can re-enter the cell cycle to help repopulate the crypt
105
Q

How long does it take for the stem cell compartment in the intestinal crypts to regenerate (aka crypt turnover time)?

A

3-5 days

106
Q

How does the radiosensitivity of cancer stem cells compare to non-stem cells?

A

More radioresistant

107
Q

Why are cancer cells more radioresistant?

A
  • A higher level of free radical scavengers
  • Abnormal developmental pathways
  • Hypophosphorylation of checkpoint kinases
  • Hyperactivation of anti-apoptosis pathways
108
Q

Can differences in metabolism be used to improve the diagnosis or treatment of cancer with radio-chemotherapy?

A

No

109
Q

What cancer is a/w RET/PTC1 type of rearrangement?

A

Papillary Thyroid Cancer

110
Q

Mutations in what gene has been consistently linked to radioresistance?

A

RAS

111
Q

Oncogenes are a/w what kind of epigenetic change?

A
  • Hypomethylation
  • Acetylation
112
Q

Which epigenetic therapy is being investigated for cancer treatment?

A

Decitabine, which can re-activate silenced genes

Mnemonic: De-silences genes

113
Q

Tumor suppressor genes are a/w which epigenetic change?

A
  • Hypermethylation
  • Deacetylation
114
Q

What is Provenge?

A

It is a cancer vaccine, used to treat metastatic, castrate-resistant prostate cancer. Very costly.

115
Q

What is the advantage of using nanodevices?

A

They can be engineered to avoid biological or biophysical barriers

116
Q

Which cancer is a/w N-myc (MYCN) amplification?

A

Neuroblastoma
Lung carcinoma

117
Q

Which cancer is a/w MET amplification?

A

Renal cell carcinoma

118
Q

Which syndrome and cancer is a/w RET amplification?

A
  • MEN 2A and B
  • Papillary thyroid cancer
119
Q

How does p53 cause G1-S arrest?

A
  • ↑ p53 → ↑ p21
  • p21 → cyclin E/CDK2 (G1-S checkpoint) inhibition
120
Q

How does RB1 control the cell cycle?

A
  • RB is normally complexed w/ E2F
  • For division, CDK 4/6 w/ Cylins D/E phosphorylate RB1, causing it to detach from E2F
  • E2F is a transcription factor that activates genes responsible for the G1-S transition
121
Q

What is an HSR?

A
  • Homogeneously staining region
  • HSR is a region within a chromosome where an oncogene has multiplied several times.
122
Q

What is a double minutes?

A
  • Extrachromosomal DNA
    –“Mini-chromosomes”
    – Contain copies of oncogenes in large numbers
  • Detected in many tumors
123
Q

What is erb? Which cancers are a/w erb?

A

It is an oncogene (of the EGFR family) a/w GBMs and SqCC

124
Q

Which cancers are a/w K-Ras amplification?

A
  • Ca:
    – Lung
    – Ovarian
    – Bladder
    – Colon
    – Pancreatic
  • Commonly mutated at codon 12 → constitutive K-Ras activation
125
Q

What cancer is a/w N-RAS?

A

H&N Ca

126
Q

What is an allograft or syngeneic animal model?

A

Transplant tumors from one mouse to another

127
Q

What is a xenograft animal model?

A

Transplant human tumors into immunodeficient mice

128
Q

What is a transgenic mouse?

A

A mouse that can rapidly develop tumors 2/2 some genetic changes.

129
Q

What is a PDX mouse?

A

A human tumor and surrounding tissue are harvested from a patient and transplanted onto a mouse (PDX mouse).

130
Q

What is the downstream RAS on Jun kinase?

A
  • ↑ Ras → ↑ Jun kinase (JNK)/ Stress kinase (SEK-1) → apoptosis inhibition, ↑ proliferation
131
Q

What is the downstream RAS pathway affects chromatin remodeling?

A
  • Ras → Raf → MEK → ERK → Msk1 → chromatin remodeling
    – remodels chromosomes MSK (musculoskeletal system)
132
Q

What is the downstream RAS pathway affects protein synthesis?

A
  • Ras → PI3K → Akt → mTOR → protein synthesis
    – Mnemonic; mRNA → mTOR
133
Q

What are the downstream effects of RAS activation on cellular proliferation?

A
  • Ras/Raf → Mek (MAPKK) → Erk (MAPK) → cellular proliferation
134
Q

What is the downstream effects of RAS activation on survival transcription?

A

RAS → PI3K → survival transcription

135
Q

What is a homodimer?

A

A receptor made of two identical subunits (proteins)

136
Q

What is a heterodimer?

A

A receptor made of two distinct subunits (proteins)

137
Q

Is TGF-β Receptor a homo or heterodimer?

A

Heterodimer (two different protein subunits)

2 → β

138
Q

What is the receptor for TGF-β?

A

TGF-β Receptor

139
Q

Are dominant oncogenes tumor-specific?

A

Not really, the same oncogenes can be activated in many different tumors.

140
Q

Are tumor suppressor genes tumor-specific?

A

Yes!

141
Q

What is an autocrine tumor?

A

A tumor that makes its own growth factors

142
Q

What are the functions of myc?

A

It has multiple functions, many of which cause immortalization, proliferation, and apoptosis.

143
Q

What are collaborating oncogenes?

A

Two oncogenes, acting on different pathways, act together to create a tumor.

144
Q

What is the collaborating oncogene for Ras?

A

Ras-Myc

145
Q

What is the collaborating oncogene for erbB?

A
  • erbA
    – erbA-erbB
146
Q

How can NFκB be activated?

A
  • NFκB and IκB are bound together as a dimer
  • Mutation in either can lead to constitutive activation
  • Constitutive IκB activation can also lead to constitutive NFκB activation
  • Radiation (stressor) can induce IκB inactivation, causing NFκB activation (temporary activation)
147
Q

What is loss of heterozygosity (LOH)?

A

Cells start out heterozygous for a gene. However, the normal variant of the gene is lost, leaving the cell with the mutant copy only → loss of heterozygosity (LOH)

148
Q

Where on DNA does p53 bind?

A

It binds to single-strand regions generated in damaged DNA, including those at telomeres.

149
Q

What genes are most commonly mutated in colon cancer?

A
  • MLH1
  • MSH2, MSH6
  • PMS1, PMS2
150
Q

Which syndrome/cancers is/are a/w ATM mutations?

A
  • Ataxia telangiectasia
  • Leukemia, lymphoma
151
Q

Which syndrome/cancer is a/w XP?

A
  • Xeroderma pigmentosum
  • Skin cancers
152
Q

What is the downstream effect of caspase 9?

A
  • Caspase 9 → Caspase 3 activation
  • Caspase 9 → Caspase 7 activation → PARP inhibition → DNA repair inhibition
153
Q

What are the downstream effects of caspase 3?

A
  • Caspase 3 → DNA-PKcs inhibition → DNA repair inhibition
  • Caspase 3 → DNA fragmentation (contributes to blebs)
  • Caspase 3 → caspase 6 → Lamin A (cell shrinkage)
  • Caspase 3 → Lamin B (cell shrinkage)
154
Q

How does stress response (radiation, etc) lead to apoptosis?

A
155
Q

How do cyclins and CDK levels vary throughout the cell cycle?

A
156
Q

What is the role of Chk1/2 in cell cycle control?

A

They control the G2 → M transition

157
Q

What is the human telomere repeat sequence?

A

TTAGGG

158
Q

How does telomere length vary with cell division in germ cells, stem cells, normal cells, and cancer cells?

A
159
Q

What is a molecular marker for senescence?

A

SA-β-gal

160
Q

What is miRNA (micro-RNA)?

A
  • Hairpin-like RNA’s that are produced in the nucleus
  • Move into the cytoplasm and regulate messenger RNA
    – inhibit mRNAs → block translation
161
Q

What is a passenger mutation?

A
162
Q

What is a driver mutation?

A
163
Q

Which cancers are most a/w microsatellite instability?

A
  • Endometrial (>25% MSI)
  • Colon & gastric (10-20% MSI)
  • Rectal (5-10% MSI)
  • Adrenocortical, ovarian, esophageal, prostate, cervical (2-5%)
164
Q

Does the number of driver mutations required for a particular cancer vary by cancer type?

A

Yes!

165
Q

What’s the function of Von Hippel-Lindau (VHL) protein?

A
  • VHL regulates HIF
  • In the presence of O2, 4prolyl hydroxylases (PHDs) add hydroxyl groups to HIF leading it to binds to VHL → HIF ubiquitination and destruction
  • Defects in VHL lead to ↑ HIP levels, which can cause malignant development
166
Q

If a cancer staging system changes in such a way that the cancers previously characterized as II are now III, how will the prognosis of stages II and III change?

A

The prognosis of both will improve!

167
Q

Changes in which genes lead to a loss of cancer cell adhesion?

A
  • E-CAD
  • N-CAM
168
Q

What two functional changes are required for cancer invasion and metastases?

A
  • Loss of adhesion
  • Extracellular matrix degradation
    – via Matrix Metalloproteinases (MMPs)
169
Q

What’re the functions of Chk1/2?

A
  • Cell cycle checkpoint molecules
  • Promote cell cycle arrest
170
Q

What cells or cellular features underlie the abscopal effect?

A

T-cells and dendritic cells

171
Q

What cells or cellular features underlie the bystander effect?

A
  • Cell-to-cell gap junction communication
  • Factors secreted by neighboring irradiated cells
172
Q

How does HIF impact cellular metabolism?

A
  • Increases glycolysis & inhibits oxidative phosphorylation
    – Up-regulates GLUT-1 transporter (↑ cellular glucose uptake)
    – Induces pyruvate dehydrogenase kinase 1 (PDK1), which Inhibits pyruvate dehydrogenase (PDH), preventing Actyl Co A formation and initiation of Krebs cycle (↓ oxidative phosphorylation)
    – Downregulates e- transport chain (mitochondrial respiration)
    – Increases lactic acid formation (↑ anaerobic metabolism)
    – Increases lipid metabolism (alternative to glucose)
173
Q

Why are mammalian cells more radiosensitive than lower eukaryotes or prokaryotes?

A
  • 2/2 larger DNA content (genome)
    – Larger target for radiation, making it more susceptible to damage
174
Q

Which proto-oncogenes are usually activated by point mutations?

A
  • RET
  • Ras (N-, K-, and H-)
  • neu