APP Cancer I (part 2) Flashcards

Lecture 2: APP

1
Q

Carcinomas originate in what cell line?

A

Carcinomas are cancer of epithelial origin

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

If the cells are well differentiated, is it most likely a benign or malignant tumor?

A

Benign - well differentiated cells, specialized features of the parent cell is preserved (e.g. hormone release)

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

What kind of cells proliferate in adults?

A
  • Bone marrow myeloblasts - Immune Cells - Epidermal cells - Epithelial cells (e.g. gut) - Regenerating Tissues
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

If a tumor is well demarcated, it is probably….. (benign or malignant)?

A

Benign - usually well demarcated/encapsulated masses - no invasion of the surrounding tissue

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

lack of differentiation indicates that a tumor is ____

A

malignant (Lack of differentiation = anaplasia)

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

Proto-oncogenes encode for proteins that normally___ cell proliferation

A

stimulate

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

In cancers, the oncogenes have sustained gain-of-function alterations resulting from 3 possible events….

A
  • point mutations - chromosomal rearrangements - gene amplification
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Protooncogenes are affected by ____ factors

A

Growth Factors

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

Proto-oncogene mutations usually arise somatically in tumor cells and are _____ (dominant or recessive?)

A

dominant

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

What are the mechanisms of oncogene activation?

A
  • Point mutation - Gene amplification - Chromosomal Translocation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Describe protooncogene assoication with growth factor receptors?

A
  • Mutated or truncated forms of the receptors with constitutive activity - Epidermal growth factor (EGF) receptor - ERBB1 is truncated in glioblastoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

2 types of cancer caused by overexpression of autocrine growth factors:

A
  • Platelet-derived growth factor (PDGF) in glioblastomas. - Transforming growth factor α (TGF-α) in sarcomas.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Overexpression of growth factor RECEPTORS in 2 types of cancers:

A
  • ERBB1 in squamous cell carcinomas of the lung - ERBB2 (HER2) in breast cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

RAS encodes?

A

p21 G-protein

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

_____ mutations are the most common abnormalities in human cancer, particularly high incidence in colon and pancreatic cancers.

A

RAS

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

How is p21 G-protein involved in GF signaling?

A
  • p21 transmits mitogenic signal from an activated GF receptor to the nucleus - (through phosphorylation cascade of transducing proteins)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

_____ mutations changing amino acids in the pocket binding GTP and region essential for GTP hydrolysis lead to constitutive activation of RAF-MAPK mitogenic cascade

A

Point mutations (RAS pathway)

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

Describe ABL and what it promotes

A

Non-receptor tyrosine kinase, promotes apoptosis;

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

The BCR-ABL fusion protein retains in cytoplasm and, due to its high _______ activity, stimulates several pathways, including RAS-RAF mitogenic cascade.

A

Tyrosine kinase activity

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

What do nuclear transcription factors stimulate?

A

Stimulate expression of several growth-related genes, such as cyclin-dependent kinases (CDKs)

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

Describe the nuclear transcription factor MYC…

A

The most commonly involved in human cancer

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

In burkitt lymphoma, what is overexpressed due to a translocation to the chromosome 14, in close proximity to an Ig gene.

A

MYC nuclear transcription factor

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

In breast, lung and other cancers, what is overexpressed due to gene amplification?

A

MYC nuclear transcription factor

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

Amplification of related genes _____ and _____ are common in neuroblastoma and small cell cancer of lung, respectively.

A

N-MYC.

L-MYC.

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

Dysregulation of cyclin and ____ expression or their mutations occur often in cancer cells and promote proliferation

A

CDK

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

The most common perturbations affect proteins involved in G1-S transition?

A

Overexpression of cyclin D (breast, esophagus, liver cancers, lymphomas) Amplification of CDK4 (melanomas, sarcomas, glioblastomas)

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

Tumor supressor genes encode proteins, which normally ____ cell proliferation or stimulate apoptosis upon cell damage

A

inhibit cell proliferation

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

Tumor Suppressor genes are _____ in cancer cells

A

inactivated

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

TSGenes are inactivated in cancer cells by _______ leading to uncontrolled growth. (4)

A

mutations, truncation, deletions or methylation

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

Mutations of TSGs are usually _____ - two alleles must be altered to lose their function

A

Recessive

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

Inherited mutations of the TSGs contribute to _______ cancers

A

Familial cancers

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

_______ cancers develop earlier in age than sporadic malignancies and often arise in multiple locations

A

Hereditary

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Mechanisms of Tumor suppressor gene inactivation? (list 3)

A

Point mutations, deletions, epigenetic changes (methylation of the promoter)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

RB (Tumor suppressor gene) encodes DNA-binding protein, which controls ______ checkpoint

A

G1 - S checkpoint

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

In quiescent cells, an active hypophosphorylated RB prevents activation of ______ genes

A

S-phase

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

Upon growth factor stimulation, Rb protein is ________ by cyclin-dependent phosphorylation, which allows progression of cell cycle

A

Inactivated

36
Q

Deregulation of _____ checkpoint by mutations in one of these tumor suppressor genes has been found in majority of cancers

A

G1-S checkpoint

37
Q

Retinoblastoma - pediatric tumor developing in retina due to deletion of ______ gene.

A

RB tumor suppressor gene.

38
Q

What are the two forms of Retinoblastoma?

A

Sporadic and Familial

39
Q

In the sporadic form of retinoblastoma, both mutations in RB gene are acquired ____ birth, thus frequency of the tumor is relatively ____.

A

acquired after birth; frequency is low.

40
Q

In the familial form, one mutation in RB protein is ______, therefore only one additional mutation has to occur in one of the retinal cells. Due to this, frequency of retinoblastoma is very ______ and the tumors often arise bilaterally.

A

inherited; high frequency

41
Q

APC gene stands for?

A

Adenomatous polyposis coli

42
Q

APC gene encodes a cellular protein regulating ______ and adhesion

A

cell proliferation

43
Q

APC protein interacts with ______, a signaling molecule in WNT pathway

A

Beta-catenin

44
Q

In the absence of _____, APC binds beta-catenin and stimulates its degradation

A

WNT

45
Q

Upon WNT stimulation, APC releases Beta-catenin, which translocates to the ______ and activates genes _______ cell cycle

A

nucleus; promoting

46
Q

Germline mutations of APC gene lead to development of multiple benign tumors (polyps) in colon, which in almost 100% of cases transform to the malignant cancer; this trasformation is associated with loss of the ______

A

second APC allele

47
Q

APC mutations also occur in majority of _________ colorectal cancers.

A

sporadic

48
Q

p53 is a _____ _____ gene

A

tumor suppressor gene

49
Q

p53 controls cell proliferation and apoptosis; it “_________” cellular stress and prevents propagation of damaged cells

A

“detects”

50
Q

Under normal conditions, p53 is bound to MDM2 gene, which causes its ________ and short half-life;

A

degradation by MDM2 gene

51
Q

Upon cellular stress (hypoxia, DNA damage, overexpression of mitogenic factors), p53 is released from the complex with MDM2, which increases its half-life and activates its ______ _____activity;

A

transcription factor

52
Q

Active p53 stimulates transcription of CDK inhibitor - _______, which leads to G1 growth arrest; simultaneously DNA repair systems are activated (GADD45);

A

CDK inhibitor - p21,

53
Q

_____ is the one of the most commonly mutated genes in human cancer (over 70%)

A

p53

54
Q

Mutations or loss of p53 leads to accumulation and propagation of mutated and damaged cells; it also allows survival of the cells with _______ or ________ mitogenic factors;

A

overexpressed or deregulated mitogenic factors

56
Q

Germline mutations in the p53 gene cause _____, associated with high risk of multiple tumors.

A

Li-Fraumeni Syndrome (LFS-dominant)

57
Q

Tumors associated with p53 induced LFS?

A
  • Soft-tissue sarcomas, osteosarcomas, brain tumors, breast cancer. - tumors develop at a younger age than sporadic and often in multiple locations.
58
Q

p53 protein

A
  • a transcription factor - binds to DNA in promoter region of genes encoding proteins responsible for cell cycle arrest and apoptosis
59
Q

p53 in its functional form is a ____

A

tetramer

60
Q

tumor-associated mutations of P53 genes often affect which domain?

A

DNA-binding domain (DBD)

61
Q

What is loss of function (due to p53 mutation)?

A

mutant p53 is not functional, but does not interfere with actions of normal p53 allele.

62
Q

What is dominant negative mutant (due to p53 mutation)?

A

mutant p53 forms a complex with the wild type allele and prevents it from binding to target gene promoters

63
Q

What is gain of function due to mutated p53?

A

mutant p53 binds to and activates different target genes, can lead to stimulation of cell proliferation, instead of cell cycle arrest and apoptosis

64
Q

What other factors contribute to Li-Fraumeni Syndrome? (mutations of what gene and polymorphism of what?)

A

Mutations of CHK2 Gene. Polymorphism of WT p53.

64
Q

Mutations of BRCA1 have been found in 40 - 50% of families with multiple ______ cancer cases

A

breast cancer

65
Q

BRCA1 gene (TSG)

A

Germline mutation (truncation, inactivating frameshift mutations) of this gene is associated with increased risk of breast cancer and ovarian cancer (85% and 50%)

66
Q

Germline mutations (truncation, inactivating frameshift mutations) of what gene can lead to increased risk of breast cancer (80%), while ovarian cancers are not as common (~10%)

A

BRCA2 gene (TSG)

67
Q

BRCA2 mutations are associated with increased risk (6%) of male _____ cancer

A

male breast cancer

68
Q

Both BRCA1 and BRCA2 encode nuclear proteins involved in response to DNA _____ and in DNA ______

A

DNA damage and DNA repair

69
Q

____ ______ genes (TSG) usually are not directly involved in cell cycle regulation. However, lack of DNA repair activity leads to genetic instability and facilitates mutations in other genes, including oncogenes and tumor suppressors.

A

DNA repair genes

70
Q

Hereditary nonpolyposis colorectal cancer (HNPCC) is associated with defects of?

A

Defects of DNA mismatch repair genes

71
Q

Xeroderma pigmentosum is increased risk of UV induced skin cancers due to defects in the _____ _____ repair system responsible for removal of UV-crosslinked residues

A

Nucleotide excision repair system

72
Q

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase preferentially expressed in ______ and _____ nervous systems

A

central and peripheral nervous systems

73
Q

Germline activating mutations of ALK have been associated with familial ________, which segregates as autosomal-dominant diseases with limited penetrance

A

Neuroblastoma

74
Q

The mutations and amplification of _____ occur also in sporadic cases of neuroblastoma

A

ALK (anaplastic lymphoma kinase)

75
Q

oncogenic miRNAs target _____ ____

A

tumor suppressors

76
Q

Tumor Supressor miRNA target _____

A

oncogenes

77
Q

Alteration of the balance of Micro RNAs can trigger/facilitate ______ transformation

A

Malignant transformation

78
Q

Deregulation of apoptosis leads to propagation of _____, _____ cells

A

damaged mutated cells due to deregulation of apoptosis

79
Q

Deregulation of apoptotic mechs: What can happen at the receptor level?

A

Reduced levels of CD95

80
Q

Deregulation of apoptotic mechs: What can happen with death-induced signaling?

A

Inactivation of death-induced signaling complex by FLIP protein

81
Q

Deregulation of apoptotic mechs: What can happen to anti-apoptotic molecules of mitochondrion?

A

Up-regulation of anti-apoptotic BCL2 (e.g. due to translocation near Ig genes in B-cell lymphoma)

82
Q

Deregulation of apoptotic mechs: Down regulation of what is due to lack of p53

A

Proapoptotic BAX (downregulation of this is due to lack of p53)

84
Q

Deregulation of apoptotic mechs: Loss of what can occur by cytochrome C.

A

APAF-1 loss Deregulation of apoptotic mechs: Loss of what can occur by cytochrome C.

85
Q

methylation of Caspase 8 gene causes what?

A

methylation inactivates caspase 8 gene, resulting in deregulation of apoptotic mechanisms.

86
Q

most normal cells only replicate 60-70 times; after that they enter _________. Why?

A
  • non-replicative senescence. - replicative potential due to shortening of telomeres.
87
Q

Telomerase in stem cells?

A

telomerase maintains telomere length, preventing senescence (aging)

88
Q

Telomerase is responsible for what feature of cancer cells?

A
  • up-regulation of telomerase enzyme allows unlimited division of cancer cells.