Midterm 4 Flashcards

1
Q

Genomic instability and evolution

A

there needs to be a certain amount of genomic instability for mutations to occur–this powers evolution. But not so much so that you win dup having a decrease in viability

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

g2/m checkpoint

A

last chance to check before mitosis

did we duplicate properly did we correct for errors

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

compartmentalization of tissues & proliferation

A

some cells in tissues/organs have different levels of ability to proliferate based on their function

so only some cells we are worried about their genome integrity. we only care about cells that are sticking around in regard to cancer

PROLIFERATIVE CELLS ARE DANGEROUS

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

Stem cells

A

divide rarely and are anatomically shielded, preserve genomic integrity

maintain ability to proliferate

so they are the ones we want to protect because of they will always be able to proliferate

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

stem cells divide _____

A

asymmetrically

one daughter cell stays in the stem compartment and one goes to eventually differentiate.

in regard to genome integrity we only worry about the one in the stem cell compartment

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

how do we protect stem cells?

A

we anatomically shield them (ex. hide them in the crypts in the intestine. crypts also secrete mucins that trap digestive acids)

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

how do the crypts specifically serve as an example of the anatomical shielding of stem cells?

A

they physically keep the stem cells away from digestive acids and toxins. they also secrete mucins that capture toxins and digestive acids which makes them unable to wreak havoc

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

how does pulse chasing work?

A

you radioactively label cells and cells that have stopped replicating would have a lot of radioactively labelled dna whereas the proliferating cells would have way less become diluted, and it wouldn’t be labelled

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

how can you tell transit amplifying cells move from the crypts?

A

radiolabelling pulse chasing

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

how do tissues protect themselves from mutant stem cells?

A

apoptosis
pumps
asymmetric dna strand allocation

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

stem cells and apoptosis (what is different about stem cells when they are damaged in comparison to a normal differentiated cell?)

A

stem cells undergo apoptosis rather than cell cycle arrest and DNA repair, after DNA damage

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

stem cells and pumps:

A

stem cells over express protein pumps (MDR-1) which effectively remove toxins from cells, preventing dna damage

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

stem cells and asymmetric dna strand allocation:

A

preserves template strand in stem cell lineage, gives newly synthesized strand to transit cell

basically new strand goes to differentiate

template strand stays in the stem compartment (immortal)

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

transit amplifying cells

A

undifferentiated cells that are sort of the transition between stem cells and truly differentiated cells

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

how can you see the migration of transit amplifying cells in the crypts (lab technique)?

A

pulse chasing/radio-labelling they keep the radioactive labels because they do not replicate as much as they head toward becoming fully differentiated cells

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

cell genomes are threatened by ________

A

errors made during DNA replication

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

for an error to become permanent during DNA replication what needs to happen?

A

that cell would need to replicate twice. because if it goes AC in the first mistake it would need to replicate again to GC to become a permanent mutation that could stay in the cell without alerting a problem

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

DNA polymerase has __________

A

proof reading exonucleolytic activity

means that as it adds nucleotides on the 3’ end it checks its work. if one is wrong, it undergoes a conformational change to cut out the mistake nucleotide

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

proof reading function of DNA polymerase detects ________errors

A

99%

1 in every 10^7 nucleotides will be wrong with just dna polym alone

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

1 in ________ nucleotide errors happen which corresponds to _____ errors per cell genome replication

A

10^9

6

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

mutation in exonuclease activity in DNA polymerase results in ________

A

tumor phenotype

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

what nucleotide scenarios give dna polymerase a hard time when its trying to exercise its endonuclease activity?

A

mono and di nucletoide repeats. super easy for them to misalign and result in insertions and deletions

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

MMR

A

repairs the mono and dinucleotide repeats that dna polymerase usually misses

scans genome after dna polymerase is done. looks for nicks and mismatches.

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

How does MMR work?

A

MutS scans for mismatch, then determine which strand is the newly synthesized strand, then repair

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

How does MMR know which strand is the new strand?

A

methylation or looking for nicks (places where removing the primer and binding together okazaki fragments hasn’t happened yet)

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

why is the single stranded DNA in the replication fork more susceptible to breakages than double stranded ?

A

only one phosphodiester bond has to break no ligase to hold together. this can cause deletions inversions etc.

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

depurination

A

purine is cleaved off of the sugar phosphate by spontaneous hydrolysis.

endogenous biochemical attack

RESULTS IN SINGLE BASE DELETION

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

deamination

A

cytosine –> uracil

which causes c to t

this leads to a change in the base pair

called a transition because both bases are from the same class pyrimidines and purines

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

why is the deamination of 5 methylcytosine really bad?

A

turns into thymine which legit belongs in dna so its a really bad mutation. very common endogenous

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

frameshift mutations usually result in ______

A

premature stop codons and wrong amino acids which result in nonfunctional proteins

lead to gene loss if it happens early on

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

transition mutation

A

stay in the same class of nucleotide

pyramidine –> pyramidine

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

tranversion mutation

A

switch nucleotide class

pyramidine –> purine

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

endogenous oxidation

A

free radicals bounce around cells and oxidize stuff around it

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

what can endogenous oxidation lead to?

A
  • oxidize bases themselves
  • cleave the bases away from deoxyribose (abasic sites)
  • cross link proteins to DNA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

oxidation can result from _______

A

the immune system!!!

it uses oxidants to consume infected cells so we can clear them out. this can damage bystander cells

inflammation can be an initiator and a promoter

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

most dangerous thing that can happen as a result of oxidation

A

the oxidation of G

oxo-g if it is not repaired prior to dna replication it will base pair with an A

this would result in a transversion mutation (G GOES TO T)

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

why are transversion mutations worse than transition mutations

A

the amino acid changes are more profound because the amino acid class (polar, charged etc) changes. the protein product would then be radically different

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

MTH1

A

degrades oxo-gs

mice that do not have MTH1 have a tumor phenotype

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

cancer genes break down into the following

A
oncogenes (gas)
tumor suppressors  (cut breaks)
genome maintenance/care-takers (mechanics)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

exogenous mutations

A

come from outside of the cell/the environment

ex. ionizing radiation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

ionizing radiation

A

ionizes water causes base mutations, deletions, and chromosomal translocations

these ions break bonds to get electrons. can cleave off the base, cleave phosphodiester bonds, and can alter the base,.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

UV radiation

A

NOT STRONG ENOUGH TO BREAK PHOSPHODIESTER bonds

but can excite electrons in the bases and can cause pyrimidines
to dimerize. when this happens it distorts the appearance of the DNA double helix

results in transitional mutation.

turns all into TT
CT, TC, CC would then be AA which results in a TT

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

alkylation

A

many chemos are alkylating agents

destabilizes linkage between base and deoxyribose so base can be lost

creates mutations

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

ENU

A

alkylating agent of guanines.

MGMT (methyl-guanine-DNA transferase) is important in reversing alkylating damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

MGMT (methyl-guanine-DNA transferase)

A

important in reversing alkylating damage

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

how do some cancers become resistant to chemo?

A

upregulating MGMT

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

how do we fight alkylating agent resistant chemo?

A

combine it with DNA repair inhibitors that act as a substrate for MGMT and basically if we can keep the bad DNA damaged them we promote a checkpoint and hopefully the cell dies there

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

MGMT wt mice versus MGMT super physiological mice

A

Excess mgmt reduces tumor incidence in mice exposed to alkylating agent

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

some potent mutagens are formed when ________

A

we ingest or inhale compounds that are altered by cellular metabolic processes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

large and nonpolar molecules

A

cannot be eliminated through the blood and the urine

so detoxification must occur

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

detoxification

A

make nonpolar molecules into polar in xenobiotics so that we can eliminate it in the urine

this process can actually make things more carcinogenic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

PAHs (polycyclic aromatic hydrocarbons)

A

products of combustion

they go through detoxification and become carcinogenic by forming adducts

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

adduct

A

something covalently attached to nucleotide

DNA transcription hits a wall because it can’t happen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

cytochrome p450 enzymes

A

involved in biosynthesis of steroid hormones, cholesterol, bile acids, and degradation of fatty acids and steroids.

they also oxidize xenobiotics aka pahs

the goal is to detoxify, solubilize and excrete but the result often mutagenic

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

pahs/cytochrome p450 products result in _______ that result in _______

A

adducts that result in G to T transversion mutations

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

G12V mutation

A

G to T transversion mutation in codon 12 inactivates ras GTPase activity. Ras is GTP bound, with deregulated mitogenic signaling

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

more than ______ of human tumors have mutated (activated) ras

A

20%

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

tp53 mutations

A

hotspots in lung tumors of smokers coincide with experimentally induced B(A)P adducts

G to T transversion mutations in DNA binding domain region prevent p53 mediated cell cycle arrest, apoptosis, and DNA repair

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

hupki

A

human p53 knock in mice–part of the p53 gene in the mouse is human p53 (DNA binding region)

produce huf cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

what do huf cells do?

A

expose huf cells or the mice to carcinogens and see what patters of mutations you see

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

carcinogen fingerprint

A

mutation associated with a carcinogen

want to find that a characteristic mutation pattern occurs in exposed individuals vs unexposed individuals

see that suspected carcinogen induces similar mutations in experimental model systems

evidence that mutations occur in early tumor development

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

uv radiation cause a ________

A

CC to TT mutation in tp53

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

aflatoxins cause a ________

A

G to T mutation in tp53

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

tobacco smoke causes a _________

A

G to T mutation in tp53

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

why is liver cancer elevated in asia?

A

aflatoxin is an initiator and when liver attempts to detoxify it, it causes mutagenesis in the liver

hepatitis B is the promoter

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

what are the initiator and promoter in asia liver cancer example?

A

aflatoxin is the initiator

hepatitis B is the promoter

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

genome sequencing allows for ____________

A

scientists to compare normal genomes from a person to the cancerous genomes from the same person to understand the diverse patterns of mutagenesis that can occur

in adults usually from carcinogens

pediatric usually random

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

G2/M checkpoint

A

CHECKS FOR DNA DAMAGE

  • stop entry into mitosis if damage is detected
  • repair damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

how is G2/M checkpoint activated?

A

CDK1 drives entry into mitosis,

so CDK1 is tightly regulated by cyclins by inhibitory and activating phosphorylations by inhibitors and by subcellular localization

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

budding yeast experiments to determine what happens in a defective G2 checkpoint

A

yeast are haploid so no need to worry about dominant or recessive.

expose yeast to radiation:

wt-phenotype rad9, the cell buds but doesn’t go into mitosis

mutant rad9 goes through mitosis no delay

cells with defects in the checkpoint continued into mitosis even though they had DNA damage and died with fragmented chromosomes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

what does the cdk at the g2/m checkpoint do when activated?

A
allow for assembly of mitotic spindle
ensure chromosomes are attached to the spindle
condenses chromosomes
nuclear envelope breakdown
actin cytoskeleton rearrangement
reorganization of golgi and ER
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

fission yeast experiments to determine function of G2 check point

A

identified wee1 kinase and cdc25 phosphatase.

wt- cell expands and divides normally

wee1 deficient- the cell divides prematurely (produced smaller daughter cells WEEEEE DAUGHTER CELLS BRITISH)

cdc25 deficient- the cell does not divide

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

wee1 deficient cells ______

A

divide prematurely

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

cdc25 deficient cells ________

A

do not divide

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

wee kinase adds _________

A

an inhibitory phosphorylation that stops premature cell division and overgrowth

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

cdc25 ________

A

removes an inhibitory phosphorylation that allows the cell to continue to grow

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

How do we activate the cdk in the G2/M checkpoint?

A

cdk1 associates with M cyclin as M cyclin levels rise.

The complex formed is phosphorylated at the active site by a cdk activating kinase (CAK) and phosphorylated inhibitory sites by Wee1 kinase.

ONLY WAY TO ACTUALLY ACTIVATE IS CDC25 NEEDS TO REMOVE INHIBITORY PHOSPHATE:

The resulting inactive M-cdk complex is then activated at the end of G2 by the phosphatase cdc25.

Cdc25 is further stimulated by active M-CDK resulting in positive feedback. this feedback is enhanced by the ability of M-cdk to inhibit Wee1.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

in order to hold a cell at the g2/m checkpoint _______ needs to be inactivated

A

cdc25

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

what are cdc25s regulated by?

A

stress signals (ATM/ATR) that cause stimulatory and inhibitory phosphorylation, sequestration, and ubiquitlation/degradation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

ATM and ATR

A

protein kinases that become activated following specific types of DNA damage. they act through checkpoint signaling pathways that lead to cell cycle arrest

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

ATM is activated in response to ________

A

double stranded DNA breaks

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

ATR is activated in response to ___________

A

protein coated ssDNA that forms when replication forks become stalled or the DNA is being repaired after various types of damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

DDR kinases _______ CDC25

A

inactivate

they buy time to do DNA repair

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

CDK1/cyclin B (mitotic cyclin) are _______ by CDC25

A

activated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

types of DNA repair

A
reversal of chemical damage
nucleotide excision repair
base excision repair
mismatch repair
double strand break
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

reversal of chemical damage repair is done by ________

A

alkylating agents. alkyl transferase enzymes remove methyl and ethyl groups (gene is silenced in many cancers). other enzymes oxidize the alkyl groups

restoration of normal base structure by deakylating repair enzymes (like MGMT)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

nucleotide excision repair

A

removes damaged base and adjacent bases, usually during transcription, repaired by pol gamma

used for more profound damage such as when something disrupts the helix shape. happens when there is a covalently attached adduct from carcinogens or UV light.

88
Q

base excision repair

A

DNA glycolase cuts the base from the sugar, endonuclease removes the base free sugar, DNA pol beta repairs.

typically endogenous DNA damage. used for when depurination occurs, and for bases that have been oxidized, reduced, alkylated, or deaminated

89
Q

MMR = mismatch repair

A

uses poly gamma as well and repairs any mismatched based we went over this

90
Q

Double stranded break repair

A

Nonhomologous end joining in G1 and by homology dependent repair in S and F2

91
Q

MGMT has been shown to be _______ by a promoter Ch3’n in a variety of tumor types

92
Q

tumors that express high levels of MGMT have _____ response to chemotherapy and a ____ outcome

A

poor; worse

it helps dealkylate the DNA during chemo

93
Q

DNA glycosylase

A

cleaves covalent bond between base and deoxyribose.

ex. uracil glycosylase removes uracil created by deaminating cytosine; t:g glycosylases remove the ts that are opposite gs

94
Q

genetic disorders that impact NER repair:

A

XP - xeroderma pigmentosum: 1000x increase in skin cancer. cant repair uv light induced pyramidine dimers. onset is significantly earlier age.

CS: Cockayne’s syndrome: short stature, premature aging, photosensitivity

PIBIDS: photosensitivity, ichtyosis, brittle hair, impaired intelligence, decreased fertility, short stature

95
Q

HNPCC is due to:

A

mutations in MMR enzymes

HNPCC: accerlerated progression from adenoma to carcinoma due to germline mutations in mismatch repair enzymes

96
Q

homo-polymeric sequences (stretches of the samenucleotide) are susceptible to ____________

A

mismatches. will need MMR to fix the mutations

97
Q

a mutation in tgf-beta results in _________

A

a defect in MMR. the frameshift mutation means a loss of the anti-mitogenic signaling.

TGF-beta normally is responsible for anti-mitotic signalling. it turns on CDK inihibitors

98
Q

BRCA risks

A

50% familial breast cancer have this mutation

70-80% familial ovarian cancer have this mutation

99
Q

BRCA – what does it do?

A

plays a role in fixing double stranded breaks

protein transits to the site of the double stranded break.

100
Q

how did we discover that BRCA transits to the double strand breaks?

A

In the experiment HU inhibits the production of dNTPs which causes the replication fork to stall which leads to breaks and you see it transit to the breaks

PCNA is always at replication fork
BRCA is only at the fork when there is DNA damage

101
Q

H2AX

A

variant histone H2A. it gets phosphorylated by ATM and ATR kinases and then becomes yH2QX which recruits BRCA1 and other repair proteins

102
Q

two kinds of double strand break repairs

A
non homologous end-joining (less accurate)
homologous recombination (more accurate)
103
Q

NHEJ

A

looks for ends and sticks them together. could be a problem when some of the DNA is not cut back. can lead to translocations, deletions, inversions etc.

104
Q

HR

A

same as HDR. happens in G2 the damaged DNA cheats of sister chromtid that isn’t damaged as a template to fix itself.

BRCA1 enables the entire complex that does this to form. it is the park bench it comes to sit on.

105
Q

homologous recombination how it works

A

broken dna is in proximity to unbroken homologous dna which serves as a template in G2

they do a strand dance where the damaged strand uses the complementary strand as a template for repair

resection, strand exchange, extension, displacement, ligation

106
Q

BRCA’s role in double strand breaks and forming complexes

A

it sits on where the break is and recruits the complex. rad50 can sit on it and initiate HR. then it gets phosphorylated which is key to its ability to function. a lot of mutations in brca cause a frame shift there to make it not function

107
Q

BRCT

A

c terminal domains common to proteins with genome maintenance functions. interacts with Rb, HDAC

it supports chromatin remodeling and transcriptional regulation

108
Q

BRCA1 controls ____ and promotes _____

A

expression, phosphorylation, and localization of cdc35 and cdk1/cyclin b kinase

BASICALLY REGULATES KEY EFFECTORS OF THE G2 CHECKPOINT

promotes mono-ub of histone H2A which is associated with constitutive heterochromatin formation (gene-poor regions such as centromeres)

THEREFORE LOSS OF BRCA 1 PROMOTES WIDESPREAD GENETIC DESTABILIZATION

109
Q

PARP inhibitors

A

kill cells that have defects in BRCA1 or BRCA2

parp is a pathway to repair DNA – so if it is inhibited you can’t repair DNA with double stranded breaks if you also dont have BRCA so that cell will die.

110
Q

myriad genetics

A

story of a researcher. he thought he could get exclusive rights to sequence the brca1 gene and patent it and refuse to license it to anyone else

ACLU sued and won in the supreme court

111
Q

why is BRCA related cancer limited to certain tissues?

A

its also involved in sex hormone signaling so if it goes to shit that means that we won’t have it and its why you see this related to breast and ovarian cancers

112
Q

Folkman

A

THE MANS who was convinced that angiogenesis, the ability of cancers to promote their own vascularization, was the key to treating it

during WW2 was asked to develop a way to freeze dry blood and preserve it.

113
Q

Folkman experiment set up

A

normal thyroid from a rabbit was grown in glass chamber with a cancer cell. they grew to a certain size and then stopped every single time. but if you took those cancer cells and put them into an animal they grew like crazy

REASON FOR THIS in the small thyroid gland they didnt have enough blood vessels to link up to so they didn’t grow!

114
Q

Folkman eye experiment set up

A

tumor cells transplated into the anterior chamber of the eye of a rabbit form tiny tumors that soon stop growing. BUT when they are transplanted into the iris area, which has a blood supply the tumors grow CRAZILY because they are infiltrated by blood vessels

115
Q

angiogenesis

A

cancer can tell the organism to make new blood vessels. it normally happens in us when we are developing and growing. it continues throughout our life, pregnancy also involved! but its all tightly regulated

116
Q

angiogenesis activators

A

vascular endothelial growth factor VEGF

fibroblast growth factor (FGF)

117
Q

when cells are ________ vegf goes crazy

A

oxygen deficient

118
Q

how does vegf and fgf have a role in cancer?

A

cancer cells release vegf and fgf into surrounding tissue. they bind receptors on endothelial cells lining the blood vessels. this binding activates a signaling pathway. in response the endothelial cells divide and secrete matrix metalloproteinases (MMPs). the MMPs break down the extracellular matrix allowing endothelial cells to migrate into the surrounding tissues. they organize into hollow tubes and develop into new blood vessels.

119
Q

t or f: angiogenesis can occur prior to invasion

120
Q

what is the association between angiogenesis and disease outcome

A

the more vascularized the tumor the worse the outcome because it has progressed further

121
Q

t or f: there are natural activators and inhibitors of angiogenesis

122
Q

anti-angiogenesis therapies

A

angiostatin & endostatin- naturally occurring. effective because it only goes after neoangiogenesis. difficult and expensive to produce.

interferons- suppress bFGF and IL-8 (both angiogenic) which leads to tumor regression

drugs-
neutralizing anti VEGF monoclonal antibody (avastin) it blocked growth of human sarcoma and glioblastoma in mice. used in combo with conventional chemo

also some tyrosine kinase inhibitor for VEGF and PDGF

123
Q

do angiogenesis inhibitors have side effects?

A

cancer doesn’t advance. but are there side effects? people would need to be on these for decades

can cause problems with bleeding, clots in the arteries, hypertension, and protein in the urine.

likely possible complications are still unknown

124
Q

HIF-1alpha pathway

A

turns on in hypoxia conditions. recent studies have found a strong correlation between elevated levels of HIF-1 and tumor metastasis, angiogenesis, poor patient prognosis, and tumor resistance to therapy.

tumors turn on survival pathways to adapt to hypoxic stress

HIF is a transcription factor so theres many ways to attack it like sequestering it in cytoplasm or targeting it for destruction

125
Q

metastasis

A

first cancer cells invade surrounding tissues and gain access to bloodstream

then they get transported throughout the body

finally they leave the bloodstream and establish new metastatic tumors in various organs

126
Q

angiogenesis happens ______ metastasis

127
Q

what changes make cancer cells able to metastasize?

A

cell-cell adhesion proteins that cause cells to adhere to one another are often missing or defective.

E-cadherin is an important one.

cancer cells can also produce proteases (plasminogen, mmps) that degrade protein containing structures such as the basal lamina and the ECM

they also get increased motility from stimulating signaling molecules from the surrounding tissues

128
Q

EMT

A

epithelial mesenchymal transition.

in order to acquire motility and invasiveness, epithelial (carcinoma) cells change their characteristic phenotype, morphology, and pattern of gene expression, and become more invasive and motile. happens naturally during developmental morphogenesis (like gastrulation neuralation etc)

129
Q

MET

A

mesenchymal epithelial transition. revert back to epithelial once you get to where you were going so that you can proliferate

130
Q

MMPs

A

extracellular proteases that all cancer cells to remodel their tissue environment by creating a passageway through the EXM.

MT-1 MMP on the cancer cell membrane cleaves cell surface adhesion molecules such as cadherin and integrins

131
Q

how do cancer cells become more motile?

A

cell locomotion requires changes in the actin cytoskeleton and the making and breaking of contacts with the underlying substrate. Lamellopodia extend the leading edge, stress fibers contract the trailing edge. these are controlled by rho family gtpases, downstream targets of ras

132
Q

how does EMT happen?

A

programmed by transcription factors that orchestrate key steps in embryogenesis.

  • loss of e-cadherin
  • stromal proteases cleave off ecm domain of e-cadherin
  • mmps chew through the basal lamina
133
Q

twist

A

twist (transcription factor) turns genes on and off, if you introduce it into the epithelial cells you will see less epithelial markers and more mesenchymal markers

134
Q

association of EMT-inducing transcription factors with cancer

A

more tf more cancer

135
Q

t or f: proliferation is required for metastasis

A

f

you could metastastize and not be crazy proliferative. converse also true.

136
Q

how does EMT impact drug sensitivity in the treatment of cancer?

A

when epithelial tumor cells undergo emt they become mesenchymal cells in order to metastasize. in the process we lose sensitivity to many drugs that target epithelial tumors.

137
Q

t or f: most cancer cells can survive in the blood

A

f

not hospitable for cancer cells. flow usually kills them. but some get stuck to the capillary beds and thats how they metastasize

138
Q

blood flow model

A

first capillary bed cancer cell lands is where it will go. so lung or liver

139
Q

seed and soil model

A

cancer cells grow where there is optimal growth for its kind.

140
Q

Coley

A

Could an infection prompt the immune system to attack tumors?

Saw 25% of his patients cured

141
Q

is cancer foreign or self?

A

based on failed implants of tumors in mice we think foreign

142
Q

humoral versus cell mediated response in adaptive immunity

A

humoral - antibodies

cell mediated - t cells

143
Q

helper t cells

A

activate both humoral and cell mediated responses

antigen presenting cell hits up helper t cell they release cytokines. then they hit up b cells to make antibodies and and t cytotoxic cells

144
Q

cytotoxic t cells

A

put peforin into the bad cell and then it causes apoptosis

145
Q

cancer immunoediting

A

3 phases:
elimination
equilibrium
escape

how cancer still winds up developing in bodies even though it is seen as foreign

146
Q

elimination

A

tldr; immune system takes care of the lowest hanging fruit – the cells that it has easy access to / look the most foreign

recognition of tumor cells by innate immune cells and their limited killing.

migration of antigen presenting cells and priming of t lymphocytes

generation of tumor-antigen-specific t lymphocytes and activation of cytotoxic mechanisms

homing of tumor-antigen-specific t lymphocytes to the tumor site and elimination of tumor cells

147
Q

equilibrium

A

cancer evolves and the immune system is selecting against the ones it can kill.

continuous sculpting of tumor cells and selection of those with reduced immunogenicity. promoting production of resistant variants.

can go latent until a mutation comes that increases resistance to immune pressure

148
Q

escape

A

tumor stops expressing antigens, or they are resistant to cell death, or the cancer cells can keep t cells from killing them

149
Q

checkpoint inhibitors

A

target receptor-ligand interactions that negatively regulate immune response.

drugs inhibit the cancer cells ability to inhibit the t cells.

150
Q

PD-1 receptor

A

PD-1 programmed cell death checkpoint.

long term exposure to antigen causes t cells to express the pd-1 receptor which prevent chronic, prolonged, excessive activation. most cancer cells express ligands that bind to it.

this protects against auto immunity but is an immune suppressive pathway that makes it difficult to mount and sustain anti tumor t cell responses

151
Q

What does the MAb to PD-1 do to bring back t cell immunity to cancer cells?

A

it blocks the interaction between PDL1 ligand on the tumor cells and PD-1 receptor on the t cells, which reactivates them

152
Q

CTLA-4

A

stimulatory signals on t helper cells become replaced by inhibitory signals normally. CTLA4 suppresses T cell activation

drugs can be made to inhibit CTLA4

just keeps your own t cells active for longer.

153
Q

CTLA-4 Antibody and Tumor growth

A

tumors don’t grow with CTLA-4

154
Q

Adoptive Cell Transfer (ACT)

A

try to find something specific to the tumor and prime our t cells with it and then put them back into the human

problems with this: you can have too much at once. tumor lysis syndrome can occur.

155
Q

there has been an overall ______ in cancer mortality

A

decrease

due to better food storage, testing, etc.

156
Q

what does it mean for a cancer to be refractory?

A

they don’t respond to conventional therapy or respond and then come back and progress to a highly malignant metastatic stage

usually in cancers that the first symptoms are vague like abdominal pain

157
Q

incidence vs detection

A

is there actually increased incidence or do we just have better detection or are more people going to get more testing ?

can compare to global populations where there is less detection but then lifestyle factors confound

158
Q

indolent

A

slow growing cancer

do we treat it or do we watch it

159
Q

aggressive cancers

A

should we treat them because it will ruin quality of life :/

160
Q

conventional treatments

A

surgey/radiation/chemo

161
Q

rational therapies

A

new precision medicines that are all in pre-clinical and clinical trials

162
Q

radiation

A

ionizing radiation removes electrons from water and other molecules producing free radicals that damage DNA

these cells cant undergo apoptosis because they lost p53. radiation kills cells through chromosomal damage so severe that they wouldn’t survive mitosis

rapidly growing cancers respond better to radiation than slower growing ones

163
Q

why do you want multiple small doses of radiation rather than a few big ones?

A

want to minimize damage to normal cells. the time gives the normal cells a chance to repair damage prior to replication

164
Q

chemotherapy

A

kills all dividing cells and are therefore not tumor specific

more likely to make apoptosis go down

are mutagens and therefore are carcinogens so there is risk of second-site cancers when under chemo

165
Q

classes of chemo

A

alkylating agents:

platinum compounds:

anti-metabolites

topoisomerase inhibitors

mitotic inhibitors

166
Q

anti-metabolites:

A

interfere with metabolic pathways. methotrexate inhibits dihydrofolate reductase which is important to making bases in DNA. it could slow cancer growth

also analogs of purine and pyramidine. can competitively inhibit some of the enzymes in dna synth

167
Q

platinum compounds

A

discovered by accident. can form covalent bonds with nitrogens in protein so when you cross link to them it disrupts ability to DNA to be transcribed and synthesized

168
Q

alkylating agents:

A

bind directly to DNA and alkylate it.

169
Q

topoisomerase inhibitors

A

prevent religation of single strand dna breaks during replication. so the replication fork will fall apart and there will be no replication.

these drugs irreversibly bind dna so transcription is disrupted as well.

170
Q

mitotic inhibitors

A

mess with the mictrobules in spindle assembly so that mitosis can’t happen properly.

prevents spindle disassembly and promotes abnormal MT bundles

171
Q

resistance to conventional therapies

A
  • increased drug efflux (MDR pumps out wide range of molecules)
  • decreased drug uptake
  • increasing target molecules
  • alterations in drug metabolism
  • increased DNA damage response efficiency
  • drugs are not killing the cancer stem cells which eventually regrow the tumro
172
Q

problem with single drug vs multi drug treatment

A

single drug - resistance

multi drug- MUY toxic could hurt people / kill them

173
Q

prevention

A

decrease initiators and promoters in our lives

ex. diet!
obeseogens (chemicals that alter metabolism and promote adipogenesis)

174
Q

prevention in diet

A

fruits and vegetables contain vitamins minerals and dietary fibers that protect against cancer

175
Q

SFN

A

inhibits histone deacetylase. derepresses epigenetically silenced genes in cancer cells and activates genes in normal cells

176
Q

vitamin D

A

dominant negative ligand for EGFR and it upregulates BRCA1 and p21, downregulates Bcl-2 and IAPs

tldr vitamin D blocks ras signaling and supports p53 mediated arrest, repair, and apoptosis

177
Q

hormones as ______

A

promoters

lifetime estrogen exposure contributes to risk

oral contraception, hormone replacement therapy, and obesity increase exposure while pregnancy, lactation, and physical activity decrease exposure

178
Q

hormone therapy

A

some cancers are estrogen or testosterone dependent. looked for molecules that can antagonize hormones in their role in cancer (usually competes to bind to receptor)

179
Q

functional genomics

A

classify biopsies in microarrays of 300 tumor samples to see which genes were upregulated or downregulated in breast cancer

created prognosis genes

180
Q

signature genes

A

genes associated with proliferation

took the prognosis genes from the previous experiment to predict the outxome of 150 women on 10 year follow up

181
Q

targets

A

enzymes with well defined catalytic cleft and easily assayed

182
Q

drugs ______ biological function

A

inhibit.

such as remove tumor suppressors or gate keepers or shut off oncogenes

183
Q

____ are the most attractive targets for drug development

A

oncoproteins

184
Q

valid target for drug k-ras example

A

if a mutation early on (say in kras) is what causes tumor progression but then subsequent things happen such as loss of p53 or p16, could the tumor still survive if you inhibit kras? that would mean it wouldnt be a good target for the drug

185
Q

oncogene addiction

A

if you need the oncogene to be on for the tumor to survie

186
Q

how do we find a drug?

A

compound libraries- collection of compounds

high throughput screening- test the function of a drug on the target

187
Q

how are high throughput screens done?

A

FRET- fluorescence resonance energy transfer. interaction between two proteins is detected by FRET, and HTS looks to see what compounds disrupt the interaction

188
Q

how do we turn a hit into a drug?

A

preclinical testing- tissue culture and animals! test there and see if it works in physiology.

THEN you submit an IND to the FDA.

THEN you move into clinical trials in humans

THEN you apply for an NDA and when it is accepted it is cleared as a drug

189
Q

phase 1 clinical trial

A

toxicity trial. what is the maximum tolerated dose?

small group of people not statisically significant. monitor side effects

also pharmacokinetics and pharmacodynamics

190
Q

therapeutic window

A

minimum tolerated dose to maximum tolerated dose

191
Q

phase 2 clinical trial

A

efficacy

test in a larger groups, come up with indications for the drug and define efficacy. does it work in patients

192
Q

phase 3 clinical trial

A

is the clinical response statistically significant? is the therapeutic benefit greater than the current standard of care

very expensive

193
Q

phospholipase D

A

lipid signaling enzyme superfamily that has been studied for its roles in cell communication

PLD1&2 converts phosphatidylcholine to phosphatidic acid (PA) which can also become diacylglycerol etc.

194
Q

____ & _____ activate PLD1&2

A

GPCR & RTK

195
Q

what is the main role of PLD in cancer?

A

progression including growth, metabolism, angiogenesis, and mobility

196
Q

matrigel plug assay

what happened to mice pld knock out

A

allows you to study vascularization

double knock out pld mice showed no vascularization

wt mice showed vascularization dependent on VEGF

197
Q

platelets and pld1

A

found absence of pld1 reduces tumor cell and platelet interaction

198
Q

pld knockout mice have _______ tumors than regular

199
Q

FIPI

A

pld inhibitor in humans

showed decreased platelet interaction and decreased metastasis

200
Q

current research ta1

A

is there a biological pathway being altered by pdl1&2 that we could narrow to focus in on

working on mouse breast cancer

201
Q

MAMPs

A

microbial associated molecular patterns

202
Q

immune checkpoint inhibitors

A

basically the body has this downreg t cell after a while so tumor cells take advantage of this machinery to program t cells to not hurt it. but gut microbiome can prevent this

203
Q

gut microbiome can ________

A

impact response to different therapies that are meant to stim immune cells

204
Q

herceptin

A

targets EGFR receptor and did well in all clinical trials

205
Q

tarceva & iressa

A

tyrosine kinase inhibitor egfr

lead to discovery of egfr addicted cancers in certain people who responded even better to treatment

206
Q

nutlins

A

keep mdm2 from binding to p53

207
Q

if p53 is _____ it can’t be ______

A

acetylated; ubiquitylated

208
Q

onyx 015

A

mutated adenovirus that kills only cells with mutant p53. E1B (normally shuts down p53 to keep the virus host alive) is defective in it so that only cells with wt p53 can fight it off

209
Q

velcade

A

proteosome degradation

210
Q

why did scientists want to improve from velcade and what did they do ?

A

velcade was non-specific would impact all ubiquitin mediated proteasomal degradations

so they began to target E3 ubiquitin ligases that were specific to their substrate

211
Q

kras targeting one ad10

A

basically this plus the pd-1 immunotherapy showed crazy results

212
Q

Sulforaphane (SFN)

A

a. n isothiocyanate found in cruciferous vegetables such as broccoli, is a potent anticarcinogen that may act through epigenetic mechanisms.
b. SFN has been shown to inhibit histone deacetylase (HDAC) activity in human colon and prostate cancer lines, with an increase in global and local histone acetylation status, such as on the promoter regions of p21 and bax genes.

i. So, SFN can inhibit HDAC and allow suppressor genes to be expressed

213
Q

deamination: biggest issue

A

either form u which isnt usually in dna

i. Biggest issue is last one – doesn’t raise red flags
• C to T transition (deamination of 5-me-cytosine)
⇒ Transition = staying in same class of base (purinepurine, v/v/)
⇒ Transversion: purinepyrid or v/v

214
Q

oxidation of G causes

A

G to T tranversion

215
Q

DNA adducts cause

A

a. When DNA poly comes along, it’s like slamming into a wall – adducts are huge molecules
b. Causes collapse of replication fork
c. Introduces GT transversion mutations, as well as others

216
Q

BRCA enables ______ to happen