Cancer BIO Exam 1 Flashcards

You may prefer our related Brainscape-certified flashcards:
1
Q

As mutations happen during cell divisions, More cell divisions lead to

A

More chances of DNA fidelity (replication) errors

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

Research: increased usage of tobacco (smoking) can lead to

A

Increased chances of getting cancer (lung)

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

Somatic DNA changes

A

-Happens in non reproductive cells of an individual
- acquired/present in single cell
-cannot be inherited
-can lead to cancer

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

Germline DNA changes

A

-present in all cell of individual including sperm and egg
-can increase susceptibility to cancer
-can be inherited

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

Autosomal dominant

A

One copy of gene from either parent is enough to present/ express the trait

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

Autosomal recessive

A

Two copies from one and the other parent to present/ express the trait ( meaning both parents must have the gene)

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

variable penetrance

A

(refers to the proportion of individuals carrying a specific genetic variation (allele) associated with a trait or condition who actually express the trait or develop the condition)

the cancer gene is present in the person, but it doesn’t mean the person will have cancer

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

variable expressivity

A

(The expression of the genetic trait can vary among individuals carrying the same genetic alteration.)

is when a cancer gene is present, but expresses itself differently.
ex: brca gene can be present in two people, but can manifest as ovarian cancer for one, and breast cancer for the other

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

common variants (low penetrance

A

individuals carrying a specific genetic variation associated with a risk of developing cancer may not necessarily go on to develop the disease
ex:
even if someone has the gene linked to cancer, their likelihood of actually developing cancer is relatively lower compared to individuals with a higher penetrance

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

rare variants (moderate penetrance) gene variants like BRCA 1 & BRCA 2

A

individuals with this genetic variation have a higher risk of developing cancer compared to those without the mutation, but the association is not absolute

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

rare variants (high penetrance) gene variants like BRCA 1 & BRCA 2

A

if an individual carries a specific genetic mutation associated with cancer, there is a high likelihood that they will develop the associated cancer
ex:
individuals carrying that mutation are at a significantly increased risk of developing breast cancer compared to those without the mutation

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

The chance of getting cancer increase when mutations in these genes (brca 1 and 2) happen. Why?

A

BRCA 1 and BRCA 2 are DNA repair enzymes. If mutation happens— that means there is less Dna repair, more Dna damage (leading to cancer)

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

what causes cancer?

A

-hereditary (lowest chance): inherited genes passed by parents to child
-familial (mid-chance): genes with lifestyle and environment
-sporadic (highest chance): happens by chance from somatic mutations

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

Primary site like:
1. Breast
2. Skin
3. Lung
4. Prostate
5. Colon
6. Pancreas
… and many, many more … is the?

A

The organ location in the body where the cancer first developed.

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

Histological type: Tissue type of origin, which groups hundreds of types of cancer into six general categories:
1. Carcinoma-in epithelial cells like skin, body cavities, organs
2. Sarcoma- in bones and soft tissues
3. Myeloma-in plasma cells
4. Leukemia-blood cells which stem from bone marrow
5. Lymphoma- in immune system like lymph nodes, spleen, stomach, testicles
6. Mixed Types-other cell/tissue types

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

carcinoma

A
  • most common (80-90% of cancer)
    *malignant neoplasm of epithelial origin (means cancer of internal or external lining of the body)
  • affects organs and glands responsible for secretion like breasts, lungs,, colon, prostate, bladder
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

subtypes of carcinoma:

A

*adenocarcinoma–Originates in an organorgland, and generally occurs inmucusmembranes. They often spread easily through thesoft tissuewhere they occur.
*squamous cell carcinoma–Originates in the squamousepithelium, many areas of the body.

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

Leukemia, Lymphoma, and Myeloma

A

White blood cells (WBCs) accumulate in the bloodstream.
Depending on the subtype, WBCs can also accumulate in the spleen or lymph nodes

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

Key Types of Leukemia:
Acute Myeloid (or Myelogenous) Leukemia (AML)
Chronic Myeloid (or Myelogenous) Leukemia (CML)
T-cell Acute Lymphocytic (or Lymphoblastic) Leukemia (T-ALL)
B-cell Acute Lymphocytic (or Lymphoblastic) Leukemia (B-ALL)
Chronic lymphocytic leukemia (CLL)
More subtypes….

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

Key Types of Lymphoma:
Non-Hodgkin’s Lymphoma (NHL)
Diffuse Large B-cell Lymphoma (DLBCL)
More subtypes….

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

Key Types of Myeloma
Multiple myeloma
More subtypes…

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

signs of cancer:

A

*A palpable lump under the skin can be either the tumor itself or a swollen lymph node
*see google notes

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

*mammography-screening for breast cancers
*Prostate Specific Antigen (PSA) screening for prostate cancers
**prostate gland releases PSA molecules in blood (low PSA levels are normal, but high PSA levels can be a sign of prostate cancer–but it could also be just UTI, rigorous exercise, ejaculation, enlarged prostate)

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

*Pap smears can detect abnormal cells from the cervix and are recommended every 3 years.

*Human papilloma virus (HPV) is a primary cause of the majority of cervical cancers.
*However, HPV is considered as a risk factor and many women with HPV never develop cervical cancer.
*HPV is a common sexually transmitted disease whose risk can be mitigated by prophylactics.
*The risk of transmission can be mitigated by HPV vaccines. Both men and women can be vaccinated against HPV to decrease spread. The CDC recommends HPV vaccines for people ages 9-15.

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

Benign nevus (mole) or melanoma. How do we know it’s melanoma?

A

Check for:
*Asymmetry-the two halves of mole look different
* Border-is poorly defined or irregular
*Color- varies from one area to another
*Diameter-bigger than a pencil eraser

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

Colonoscopy to screen for colorectal cancer

A

Colonoscopies are recommended every 10 years for people age 45 to 75 by the U.S. Preventative Services Tas Force

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

A tumor biopsy

A

is a procedure where a sample of tissue is taken from a suspicious mass or lump to examine it for the presence of abnormal cells. It helps diagnose and determine the nature of the tumor.

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

H&E stain, or Hematoxylin and Eosin stain

A

, is a common histological staining technique used in pathology. Hematoxylin stains cell nuclei blue, while Eosin stains cytoplasm and extracellular matrix pink, providing contrast and helping to visualize tissue structures under a microscope (to see if theres abnormal cell growth and loss of tissue organization)

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

Immunohistochemistry (IHC)

A

is a technique used in pathology to detect and visualize specific proteins in tissue samples. It involves using antibodies that bind to the target proteins, followed by a chemical reaction that produces a visible stain. IHC is valuable in identifying specific markers associated with various diseases, including cancer. It provides additional information about the protein expression patterns within tissues, aiding in diagnosis and understanding the biological behavior of diseases.

  • IHC is very flexible because the primary antibody can be against any cell surface marker (that someone has made a good antibody against)-meaning it wont detect non cancerous cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Positron Emission Tomography (PET) scan

A

PET scans use a slightly radioactive tracer to identify internal regions that have unusual metabolism, can pick up cancer very early

*if there’s an increased metabolism in certain area which usually doesn’t have organ positioned there or even with normal organ that has abnormal metabolic activity, it may reveal a possible cancer cells because cancer cells have high metabolism

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

Flow cytometry

A

*flow cytometry can indicate the presence of cancer when specific immune markers on cells glow brightly. Abnormalities in the expression of surface markers, such as CD antigens, can be detected through increased fluorescence during flow cytometry analysis. These changes may suggest the presence of cancerous cells or abnormal immune cell populations.
* a patient’s blood sample is stained with fluorescently labeled antibodies targeting specific immune cell markers. The fluorescence patterns generated when these antibodies bind to their respective markers help identify and characterize different immune cell populations, including cancerous or abnormal cells.

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

Liquid biopsy

A

is a non-invasive diagnostic technique that involves analyzing components of bodily fluids, such as blood, to detect and monitor various conditions, including cancer.
* often used when there’s a suspicion of cancer but obtaining a tissue biopsy may be challenging or invasive
1.Blood Sample Collection: A blood sample is taken from the patient.
Isolation of CTCs: 2.CTCs shed from tumors into the bloodstream are isolated from the blood sample.
3.Analysis of CTCs: The isolated CTCs are then analyzed to provide information about the cancer, such as its type, genetic mutations, and potential for metastasis.

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

Cancer onset can be slow

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

Stages of breast cancer
Stage 0: ductal carcinoma in situ
Stage I: Small, invasive, has not spread Stage IIA: ≤20 mm and 1-3 lymph nodes or ≤50 mm
Stage IIB: 20-50 mm and 1-3 LN or >50 mm without spread to LN
Stage IIIA: Spread to 4-9 LN or >50 mm with 1-3 LN
Stage IIIB: Spread to the chest wall or caused swelling or ulceration of the breast, or it is diagnosed as inflammatory breast cancer
Stage IIIC: Spread to 10+ LN but nowhere else
Stage IV (metastatic): Spread to other organs

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

HER2 (positive and Negative)

A

HER 2 positive means:
-overexpression of HER2 protein (increased number)
-cancer is more aggressive–more growth of cancer cells
-targeted therapies may work
HER2 negative means:
-no overexpression of HER2 protein
-targeted therapies may not work

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

ER (positve and negative)

A

ER+: cancer grow because receptor are present that binds to estrogen.
-also means therapies can be used for this type
ER-: no receptor present
-therapies that work to inhibit cancer growth may not work because of no presence of receptor

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

PR (+ and -)

A

PR+:may respond to hormonal therapies because of the presence of receptors that may be inhibited
PR-: hormonal therapies may not work

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

Rank greatest prognosis to worst prognosis (1-8, 8 as the worst)

A

1.HER2-,ER+,PR+
2. TRIPLE POSITIVE
3.HER+,ER-,PR+
4.HER2+,ER+, PR-
5.HER2+,ER-,PR-
6.HER2-,ER+,PR-
7.HER2-,ER-,PR+
8. TRIPLE NEGATIVE

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

Metastasis Process

A
  1. Primary tumor formation (still in tissue or organ)
  2. Local invasion (break off from boundaries of organ/tissue)
  3. Intravasion (cancer cells have invaded/entered the circulatory system/blood vessels)
  4. Survival in the circulation (cancer cells continue to travel through blood/lymphatic vessels- possibly successful in evading immune system response)
  5. Arrest at a distant organ site (cancer cells may not immediately start growing and forming tumors. Instead, they might enter a phase of dormancy or latency,”lingering” at the new organ without actively invading or forming detectable tumors. This period of arrest allows the cancer cells to adapt to the new microenvironment and conditions in the distant organ before resuming growth and colonization)
  6. extravasation (cancer cells escape from blood vessels or lymphatic vessels and invade the surrounding tissues)
  7. micrometastasis formation (established in a new location, these cancer cells can start to grow and divide)
  8. metastatic colonization (more advanced stage where these cancer cells have successfully established themselves- proliferated in the new site)
  9. clinically detectable macroscopic metastases (presence of secondary tumors that have grown to a size that can be visually identified or detected through clinical examination, imaging techniques, or other diagnostic methods)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

COMMON ROUTES OF METASTASES

A

*STARTS IN BREAST-CAN GO TO liver,lung, bone, brain
*starts in colon-can go to lungs, liver
* from stomach- to esophagus, liver
*from lung- to adrenal gland, brain, liver
* from pancreas- to lung, liver
* from prostate-to bone

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

progression to colon cancer

A

benign:
1. hyperproliferation
2. small adenomatous polyps
3. large adenomatous polyps
4. severe dysplasia (precancerous polyp)
malignant:
5. adenocarcinoma
6. cancer

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

stages of colon cancer

A
  1. cancer cells only found in innermost lining of the colon (hasn’t spread)
    I.tumor has spread beyond the inner layer but remains in colon
    II. cancer has grown outside the colon, but not yet spread to lymph nodes
    III. cancer has grown outside the colon, has spread to lymph nodes
    IV. cancer has spread to other parts of the body
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Rate of progression of colon cancer with or without surgery

A

*greatest progression of cancer–without surgery
*least–with surgery

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

lung cancer

A

Small-cell lung cancer (SCLC):
-typically initiates in the bronchii
-more tightly associated with smoking
Subtypes include:
-Small cell carcinoma
-Combined small cell carcinoma (mixed SCLC and NSCLC)

Non-small cell lung cancer (NSCLC):
-cells lining the surface of the lung airways. These include the bronchi, bronchioles, and alveoli.
Subtypes include:
1. Adenocarcinoma in situ
2. Adenocarcinoma (begins in glands in the alveoli, slow-growing, most common)
3. Squamous cell carcinoma (second most common overall, most common in smokers, begins in squamous cells, slow-growing)
4. Large cell (undifferentiated) carcinoma (least common, begins in large cells in lungs, fast growing)

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

lung cancer progression

A

Stage 0 – the tumor hasn’t grown outside of the lungs and is also called “in situ” disease.
Stage IA: tumors are ≤ 3 cm, no spreading to lymph nodes
Stage IB: tumors are 3-4 cm, no spreading to lymph nodes
Stage IIA: 4-5 cm, no spreading to lymph nodes
Stage IIB: 4-5 cm, spread to lymph nodes
Stage III: Spread outside of the lung to nearby structures including lymph nodes, but not to distant parts of the body.
Stage IV: Metastatic spread to more than 1 area in the other lung, the fluid surrounding the lung or the heart, or distant parts of the body through the bloodstream.

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

prostate cancer stages

A

Stage I:Slow growing, low PSA, cannot be felt and involves ≤ one-half of 1 side of the prostate. The cancer cells look like healthy cells.
Stage II:PSA medium or low, no spreading.
Stage III:PSA levels are high, the tumor is growing, or the cancer is high grade. These all indicate a locally advanced cancer that is likely to grow and spread.
Stage IV:The cancer has spread beyond the prostate.

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

stages of pancreatic cancer

A

Stage 0: Carcinoma in situ
Stage IA: ≤2 cm, no spreading
Stage IB: ≤ 4 cm, no spreading
Stage IIA: >4 cm, no spreading
Stage IIB: Any size, spread to ≤3 nearby lymph nodes but no distant sites
Stage III: Any size, spread to 4+ nearby lymph nodes OR nearby blood vessels, but no distant sites
Stage IV: Tumor is any size and has spread to distant sites

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

pancreatic cancer Pathological tumor grades

A

Pathological tumor grades:
-Grade 1 (G1) means the cancer looks much like normal pancreas tissue.
-Grade 2 (G2) falls somewhere in between.
-Grade 3 (G3) means the cancer looks very abnormal.

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

stages of melanoma

A

0: melanoma confined to epidermal region of skin
I:localized disease, only in skin but very thin
II:localized disease, thicker than stage I
III:spread to lymph nodes
IV:spread to other organs

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

stages of brain tumors:

A

I: slow growing cells, almost normal appearance, least malignant, usually long-term survival
II: relatively slow growing cells, slightly abnormal appearance, can invade nearby tissues, sometimes recur as higher grade
III: abnormal cells proliferation, abnormal appearance, infiltrate normal tissue, recur as a higher grade
IV:rapidly reproducing abnormal cells, very abnormal appearance, area of necrosis in center, angiogenesis

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

Peyton Rous
Can the cancer be transmitted without the cancer cells being injected

Rous Sarcoma Virous

A

Carcinoma from one chicken was isolated, grind up, and injdcted into another chicken. Observed was the chicken who got the carcinoma developed cancer.
Conclusion: cancer can be caused by virus (virus containing oncogene)

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

Focus forming assay

A

Used to identify oncogene

Injected cancer DNA to normal cell in the petri dish, then Analyzed the foci developed when cancer cells transformed the cell in dish, and analyzed even more which gene caused the transformation

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

Where does the cancer-causing part of tumor viruses come from?

Experiment by J Michael Bishop and Harold Varmus

A

1975 Experiment: Compared Rous Sarcoma Virus genome vs. non-foci forming variant to identify the cancer-causing gene. ( i assume the cancer causing agent was from rsv genome)
Then they compared the cancer-causing gene to animal genomes.
Result: The gene in the virus that caused cancer matched a nearly identical gene in animal cells! ( meaning that normal cells have the same gene, but only mutated in cancer cells)

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

Retrovirus can inject its genetic material into us( host cell),

Recombination event can alter retrovirus into an oncogneic retrovirus

Once the oncogenic retrovirus inject its genetic material into or near the protooncogene of the host cell, it can activate the host cell’s gene to become oncogene(increased proliferation of normal cells)

Retrovirus can pick up cancer causing genes inside one host cell and transmit it into another host cell

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

C src (cancer causing gene present in the normal cell)

V src (dsDNA provirus from avian leukemia virus + C src)

How does C src turn into V src

A

A harmless virion (avian leukemia,virus),
through infection and reverse transcription, creates dsDNA provirus. When dsDNA provirus gene is accidentally injected in the host cell DNA near C src gene and enclosed in capsid, the RSV now contains mutated gene of src gene-which js an oncogene)

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

Protooncogenes

A

Proto-oncogenes are a group of genes that cause normal cells to become cancerous when they are mutated

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

Oncogene

A

Mutated protooncogene that causes an increased proliferation of normal cell( now abnormal)

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

So can virus really cause cancer?

A

Tumor promoting virus were important in the discovery that cancer are caused by mutation in protooncogene which result in oncogene

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

Homolog

A
  1. Homologs:
    derived from a common ancestor gene through speciation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

A. Paralogs:

A

genes within the
same species that have arisen through gene duplication events

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

Ortholog

A

B. Orthologs: genes found in different species that evolved from a common ancestral gene through speciation
(genes which evolved from a common ancestral gene by speciation)

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

Analog

A
  1. Analogs: Genes with a similar function that do not share a common ancestor, resulting from convergent evolution (structures or traits are those that have similar functions or purposes but evolved independently in different lineages and do not share a common evolutionary origin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Coding mutation in protooncogene results in

Why?

A

Abnormal protein

changes in the DNA sequence of the gene affect the amino acid sequence of the protein it encodes. These mutations can include substitutions, insertions, deletions, or rearrangements of nucleotides within the gene. altered amino acid sequence of the protein, can lead to changes in its structure, stability, or function

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

Regulatory mutation in protooncogene results in

How?

A

Excessive amount of protein

occur in regulatory regions of the gene, such as promoters or enhancers, or in transcription factor binding sites. These mutations can alter the normal regulation of gene expression, leading to increased transcription of the proto-oncogene and ultimately higher levels of the corresponding protein.

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

Translocation in protooncogene results in

How?

A

Novel protein

The protooncogene becomes fused with another gene, transcribed and translated. The resulting fusion protein may have abnormal activity compared to the original protein. Then that promotes abnormal activity

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

Gene amplification iof protooncogene results in

How?

A

Excessive amount of protein

multiple copies of the gene are present within the cell. So, there is an increased abundance of mRNA transcripts derived from the amplified gene, which are then translated into protein by the cellular machinery. With more copies of the proto-oncogene being transcribed and translated, the production of its corresponding protein is elevated.

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

Point mutations in coding regions

A

Silent- a change in both DNA and mRNA level, but the translated amino acid sequence isn’t affected so the mutation had no effect

Nonsense-introduce a premature stop codon into the coding sequence

Missense (conservative): does not completely alter the function of the amino acid. substituted amino acid has similar chemical properties to the original amino acid

Missense (nonconservative):
the substituted amino acid has different chemical properties from the original amino acid. may significantly alter the structure or function of the resulting protein.

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

Point mutations in (blank) drive 15-30% of all cancers

How?

A

KRAS

Point mutations in the KRAS gene can lead to constitutive activation of the KRAS protein, meaning that it remains in its active GTP-bound state even in the absence of upstream signaling stimuli

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

RAS isoforms are:

A

KRAS, HRAS, NRAS

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

P-loop:

Switch I:

Switch II:

A

( G domain contains P-loop): which forms part of the nucleotide-binding site. (Where GDP and GTP binds to)

Effector lobe: interacts with downstream effector proteins, transmitting signals from activated Ras to intracellular signaling pathways.

Switch I:undergoes conformational changes upon nucleotide binding. (GDP bound means closed conformation, while GTP bound means open conformation- means ras is activated)- also more focused on exposing the binding site of p loop

Switch II: undegoes the same conformational changes, but it is more invovled in stabilizing the binding between ras and nucleotides

Hypervariable region HVR contributes to the diversity of Ras isoforms.

Even when switch i and switch ii are disruoted, as long as the p loop is able to bind gtp, ras activation can still happen. Not in its full extent capacity, but ras activation can still help in progression of cancer

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

Recurring RAS point mutation in cancer

What amino acid gets mutated at position 12 of the sequence that results in uncontrolled cell growth-leading to cancer

A

glycine is replaced with valine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

KRAS cycles between “on” and “off” states

(RTK, GTP, GDP, RASGEF, RBD, KRAS)

steps:

A
  • When a ligand binds to a receptor tyrosine kinase (RTK), it triggers a signaling cascade that ultimately leads to the activation of RAS proteins like KRAS. RTK recruits RASGEF.
  • RASGEF like SOS promote the exchange of GDP to GTP (for activation “on”)
  • GAPs like NF1 and P120 RASGAP stimulate GTPase activity of KRAS (which is responsible for hydrolysis of bound GTP to GDP-inactivation), as GTP is hydrolyzed into GDP, an inorganic phosphate is released.
  • when active GTP is bound to KRAS, effectors like RAF and P13K kinases bind to RBD to enhance proliferation and growth
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

what happens when there’s a mutation in codon 12 and codon 61? what is likely the result of this mutation?

A
  • Mutations in codons 12, 13 and 61 disrupt the GTP hydrolysis and guanine exchange rates of RAS proteins
  • mutations in codon 12 disrupt the GTPase activity of RAS —which decreases the rate of GTP hydrolysis, so the mutant protein accumulates in the GTP-bound state (increased activation)
  • Mutations in codon 61 accelerate the rate of GDP–GTP exchange and simultaneously decrease the rate of GTP hydrolysis, so codon 61 RAS mutants also accumulate in the GTP-bound state (increased activation)

***Mutations in codons 12 and 61 of the RAS gene are associated with decreased GTP hydrolysis activity. So, RAS proteins with these mutations tend to remain in the active (GTP-bound) state for longer periods of time. REsult: prolonged activation of downstream signaling pathways involved in cell proliferation, survival, and differentiation contribute to the development and progression of various types of cancers.

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

how can Alterations in noncoding regions of the genome cause oncogenesis?

A

enhancers: DNA sequences that increases transcription of genes (help regulate which genes gets turned on or off)-like bookmarking which pages needs to transcribe

Cohesin and CTCF: help organize chromatin structure and regulate gene expression (create loops to make dna accessible)—book page holder make it easier to copy pages

RNAPII: responsible for transcribing DNA into RNA during the process of gene expression— the real transcriber

Transcription factors: regulate the transcription of genes (ensure that the copying is right)— like the person double checking the info

**if something goes wrong with the way genes are being copied or there’s too much genes being copied, that leads to cells growing incorrectly and wrongly which leads to cancer

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

Mechanisms of genomic amplifications (extra copies) of an oncogene in cancer:

A
  • double minutes–extrachromosomal fragments of DNA that contain amplified sequences like oncogenes. They replicate independently. Can arise through errors and chromosomal breakage
    *Homogeneously Staining Regions (HSR)–appear as intensely stained bands due to high DNA amplification. Amplification can happen due to unequal crossover events during meiosis
    *scattered–extra copies of oncogene are dispersed across multiple chromosomes rather than being concentrated in specific regions

***genomic amplifications of oncogenes play a significant role in driving cancer progression by increasing the expression levels of genes that promote abnormal cell growth and survival.

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

N-myc in neuroblastoma

which mechanism of amplification is it associated?

A

This amplification is associated with the formation of either double minutes (DMs) or homogeneously staining regions (HSRs)

  • The HSRs, which contain multiple copies of the genomic region encompassing the N-myc gene

*often have low prognosis

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

How many copies of each gene?

identify using Array Complete Genomic Hybridization (aCGH)

steps:

A
  1. label differently the tumor and reference (normal) DNA sequence
  2. add unlabeled blocking DNA to prevent repetitive DNA sequence
  3. all three components are added together and hybridized into a microarray containing millions of DNA probes of the entire genome.

result: the ratio of fluorescent signals will be the same if there is no difference between the tumor and reference sequence.
If the ratio is altered, that means there must be amplification or deletion that happened in the tumor sequence, which can help identify the genomic location of the genes that undergone amplification/deletion

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

Mechanisms of gene amplification and rearrangement

A
  1. normal crossing over–genes are on their normal lines, with no rearrangement, and getting amplified normally
  2. genes do not match (one gene gets behind, so the other gene gets lined up with a different gene)

result: either complete gene duplication happens (normal genes still together) or crossing over results with partial gene duplication (abnormal gene)—gene rearrangement

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

How is the Amplification of extrachromosomal double minutes harmful?

A

Double minutes are replicated during S phase to form paired structures. In mitosis, double minutes attach to chromosomes at metaphase and are found in proximity of the chromosome tips in anaphase. Sister double minutes remain paired during G2 and mitosis, and their mitotic nondisjunction results in unequal distribution of double minutes over daughter cells

Double minutes aren’t connected
to spindle fiber machinery, so
copies are distributed randomly.
After mitosis these two daughter
cells could have received 0-8
copies, leading to fast genomic
amplifications and deletions.

conclusion: double minutes undergo replication and distribution during the cell cycle in a manner that is independent of the cell’s normal chromosomal machinery—this leads to random/rapid distribution of genes–contribute to cancer

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

ways cell may create a double minute (circular DNA)

A

a)replication>re-replication> DSB formation> circularization
b) replication> DSB fformation> circularization > continued replication
c) DSB formation> circularization> homologous recombination
d)DSB formation> circularization> non-homologous end joining

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

How can translocation activate oncogenes?

A

translocation– fuse a region from one chromosome with a region from a second, unrelated chromosome

***When the c-myc gene ends up next to IgH, it starts following the rules of IgH, which tell it to grow a lot all the time. This makes the c-myc gene produce too much of a protein called Myc, which tells cells to grow really fast. This extra growth can cause Burkitt’s lymphoma.

Myc translocations cause ~70% of Burkitt’s lymphoma cases
(through reciprocal translocation—myc protooncogene becomes myc oncogene)

The aberrant expression of the c-myc oncogene leads to the production of structurally normal Myc protein but in abnormally high amounts.

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

can Oncogenic amplification of HER2/Neu lead to a worse prognosis after being diagnosed with breast cancer?

A

it suggests that the presence of amplified erbB2/HER2 gene and elevated protein expression correlates with a more aggressive form of breast cancer and poorer prognosis compared to cases where the gene is not amplified and protein expression is normal

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

which chromosomal translocation cause fusion protein?

A

Reciprocal chromosomal
translocations between human
Chromosomes 9 and 22, which carry the abl and bcr genes (BCR-ABL translocations) turn out to be the single cause of virtually all chronic myelogenous leukemia

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

phosphorylation role in cellular signaling

A
  • Phosphorylation involves the addition of a phosphate group to a protein molecule, by enzymes called kinases—this helps regulate function and activity of the cell
  • Phosphorylation serves as a molecular switch, turning proteins “on” or “off” in response to specific cues
    *phosphatase reverse phosphorylation (releasing inorganic phosphate)

**Phosphorylation is a cell’s way of fine tuning on and off signals
depending on what other signals it receives and its environment

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

what are the ways cell signaling can be regulated

A

-phosphorylation
-upregulation
-direct binding to change conformation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

A acivates B, B inhibits C, C inhibits E.

If you have more A, what happens to E?

A

If you have more A, which activates B, it would likely lead to an increase in B activity. Since B inhibits C, this increased B activity would result in decreased C activity. As C inhibits E, a decrease in C activity would mean less inhibition of E, possibly resulting in increased E activity. So, in summary, having more A could lead to an increase in E activity

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

A acivates B, B inhibits C, C activates D.

If you have more A, what happens to D?

A

If C is being inhibited due to increased B activity, then there would indeed be less activation of D, resulting in decreased D activity. So, with more A leading to increased inhibition of C, the likely outcome would be a decrease in the activity of D.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

A acivates B, B inhibits C, C inhibits E, and E inhibits A.

If you have more A, what happens to A over time?

A

With more inhibition of C, there’s more E being produced. And with more E being produced, more E inhibits A, leading to less A over time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

Proto-oncogenic kinases examples

A

***Raf
*MEK
*ERK
*mTOR
*AKT
PDK1
**
P13K

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

types of proto-oncogene examples:

A

*Rheb
Raptor
**
Ras

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

tumor suppressors examples:

A

***PTEN
*TSC1 and TSC2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

Ras P13K signaling

A

Ras signaling:

  • Ligand binds to RTK.
  • Signaling is initiated then by IRS
  • IRS activates Ras
  • Ras activates Raf
  • Raf activates MEK
  • MEK activates ERK
    Result: cell cycle progression and proliferation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

P13K signaling

A

P13K signaling:

  • Ligand binds to RTK.
  • Signaling is initiated then by IRS
  • IRS activates Ras
  • Ras activates P13K
  • P13K activates PIP2
  • PIP2 activates PIP3

PTEN can inhibit PIP3 from activating downstream protein (if cell cycle has to be stopped when damage is detected in DNA)

BUT IF NO PTEN PRESENT:

  • PIP3 can activate AKT directly
    Or
  • PIP3 activates PDK1 and PDK 1 activates AKT
  • Activated AKT prevent inhibition of TSC1/ TSC2
  • inhibited TSC1/ TSC2 activates mTOR/Raptor complex (mTORC1) through Rheb

Result: protein synthesis and Cell growth

94
Q

2 activated oncogenes > 1 activated oncogene

How is having one activated oncogene compared to having two activated oncogene?

A

The study found that the collaboration of myc and ras oncogenes led to an increased incidence of mammary carcinomas compared to mice carrying either oncogene alone, demonstrating their synergistic effect in promoting cancer development.

95
Q

function of tumor suppressor?

A

serves as a break. When there’s DNA damage and the cell cycle must be stopped—tumor suppressor functions as the break, that stops the cell cycle and allow for DNA repair

96
Q

how does mutation in proto-oncogene lead to cancer?

A

mutation causes proto-oncogene to have too much gas—meaning it is proliferating and dividing really fast that the cell is losing control

97
Q

how does mutation in tumor suppresor genes lead to cancer?

A

mutation in tumor suppressor genes causes the cell to have no break—meaning instead of stopping the cell cycle because of DNA damage and repairing it, the cell ignores the repair signal and goes on to continue dividing and synthesizing, so many damaged DNA is being copied and leading to multiple mutated cell

98
Q

How many hits needed for Retinoblastoma?

A

2—one hit was from inherited mutated gene and one more hit from a somatic mutation that happens in the gene

99
Q

Types of pediatric retinoblastoma:

What are they?

A

Sporadic retinoblastoma
* Typically single focus in one eye
* Relatively less risk of additional cancers
* 2 hits must happen independently on the same cell (or if not independently, there must be Loss of heterozygosity or when a tumor suppressor is mutated and the mutated cell is left)

Familial retinoblastoma:
* Typically multiple foci in both eyes (bilateral).
* Inherited from a parent who survived childhood retinoblastoma or mutation
in a parent’s sperm or egg cell.
* Rarely observed before the onset of modern medicine that allowed affected kids to survive to breeding age.
*&raquo_space;> risk of additional cancers
throughout life.
* all cells in the body have first hit (so more likely to get 2nd hit)

100
Q

How to identify the number of tumor suppressors:
Knudson’s Two-Hit Hypothesis

A

the rate of appearance of familial tumors was consistent with a single random event (single hit), while the sporadic tumors behaved as if two random events were required for their formation (two hit)

It turns out that two hits of one gene are
not entirely independent events because of loss of heterozygosity

101
Q

How do cells go from one hit to two hits? (LOH involvement)

Loss of heterozygosity (LOH) can happen via at least four
common mechanisms: what are they?

A
  1. Mitotic recombination—when wild type Rb allele is replaced with mutant Rb allele, so now cell lack functional Rb gen copies
  2. Gene conversion–when a mutant Rb allele is transmitted from one chromosome to its homolog and replaced by the wild-type allele as the replication jumps back and forth to both chromosome and homologous templates
  3. Chromosomal nondisjunction–when one daughter cell retains both chromatids of a chromosome rather than its usual allotment of a single chromatid–result: descendants of the cell may contain chromosomes that is mutant
  4. Gene deletion– when the wild type gene is deleted, so now you don’t have a functional tumor suppressor gene because the normal one got deleted and the mutant one is present
102
Q

Haploinsufficiency?

Haploinsufficient proteins

A

State in which the presence of only a single functional copy of a gene yields a mutant or partially mutant phenotype.

  • don’t need 2 hits sometimes. 1 hit is enough to cause cancer because that one hit has generated enough mutation to the gene that complete function is not enough to function normally
103
Q

Haplosufficient proteins

A

retain their tumor-suppressing function even when one copy of the gene is lost or mutated

*2 wild type copies is enough to function
*1 mutant and 1 wild type is too little but is enough for the protein to act normally
*2 mutant proteins is too little and insufficient to allow functional proteins

104
Q

difference between haploinsufficient and haplosufficient proteins

A

haploinsufficient—1 hit is enough to deem the protein dysfunctional–protein cant function normally
haplosufficient—2 hits generate little functional proteins but are enough for protein to function normally

105
Q

How to identify a tumor suppressor:
Single nucleotide polymorphisms (SNPs

A

a SNP may replace the nucleotide cytosine (C) with the nucleotide thymine (T) in a certain stretch of DNA.

can provide insights into the potential presence or activity of tumor suppressor genes in cancer patients by comparing the nucleotides from cancer patients to normal genes

106
Q

haplotype block mapping

A

a technique used in genetics to identify regions of the genome where genetic variation, such as Single Nucleotide Polymorphisms (SNPs), tends to be inherited together in blocks or segments. These blocks are referred to as haplotype block

107
Q

Linkage analysis using single nucleotide polymorphisms (SNPs)
can map cancer genes to sites on the genome

A

linkage analysis with SNPs involves studying families with a history of cancer to identify regions of the genome that are inherited along with the disease phenotype.

By genotyping SNPs across the genome in affected and unaffected family members, researchers can look for patterns of co-inheritance between certain SNP alleles and the presence of cancer.

108
Q

Genetic mapping to identify a heritable tumor suppressor gene

A

Linkage within well-documented family trees identified the gene Adenomatous Polyposis Coli (APC) as a major causative gene of FAP (familial adenomatous polyposis)

109
Q

Tumor suppressor inactivation by methylation and gene silencing

A

In normal cells, the CpG islands of promoter regions are open chromatin and generally unmethylated. Gene transcription is activated by transcriptional factors with H3K27ac.

In cancer cells, the CpG islands in the promoter regions are hypermethylated,
constructing heterochromatin with H3K27me3 and H3K9me3, which suppresses gene transcription
(methylation of CpG islands can interfere with binding of transcription factors–so it prevents gene expression of tumor suppressors, without tumor sup. cancer can develop)

110
Q

Deep sequencing of tumors (Whole Exome Sequencing)

A

allows variations in the protein-coding region of any gene to be identified, rather than in only a select few genes. Because most known mutations that cause disease occur in exons, whole exome sequencing is thought to be an efficient method to identify possible disease-causing mutations.

111
Q

Mutation patterns in oncogenes vs. tumor suppressors

A
  • Oncogenes tend to have recurring hotspot mutations because gain-of-function changes drive cancer
  • Tumor suppressor genes tend to have mutations spread out without a clear pattern because of loss-of-function changes drive cancer
112
Q

How many oncogenes and/or tumor suppressors are mutated in a tumor?

A

Average of 4-8 known driver
mutations per tumor

113
Q

amplification—associated with oncogene
deletion–associated with tumor supressor

A
114
Q

RNA interference to “knockdown” a messenger RNA using permanent short hairpin RNAs (shRNAs) or temporary small interfering RNAs (siRNAs)

What’s the purpose?

A

RNA interference can be used to “knock down” or reduce the expression of specific genes by introducing short hairpin RNAs (shRNAs) or small interfering RNAs (siRNAs) into cells. These molecules target and bind to the complementary sequences of the mRNA molecules, leading to their degradation and ultimately reducing the production of the corresponding proteins. This approach can be used for experimental purposes to study gene function or as a potential therapeutic strategy for diseases caused by overexpression of certain genes.

115
Q

Clustered regularly interspaced short palindromic repeats (CRISPR)

CRISPR can introduce new mutations, many of which cause nonsense-mediated decay (NMD), how?

A
  1. you have the target sequence
  2. guide RNA (gRNA) binds to target sequence
  3. Cas9 enzyme binds to guide RNA
  4. Cas9 enzyme then cuts both strands of DNA (up and bottom)
  5. when the cut is repaired, the mutation is introduced
116
Q

Mass functional characterization of deleted genes using genetic screens

deleted genes can result in either depleted or enriched: what are they?

A
  • enriched–is when deletion of genes can provide an advantage to the cancer cells (proliferating better and surviving)
    ex: deletion of tumor suppressor can lead to cancer cells having the ability to proliferate without instructions

*depleted–is when deletion of genes leads to decrease cell proliferation/ survival
ex: if the deleted gene is an oncogene–then its deletion would be better for the individual as there would be less proliferation

**enriched—applies to tumor suppressors—bad for individual—good for cancer

**depleted–applies to oncogene–good for individual–bad for cancer

117
Q

p53: The guardian of the genome

*Germline p53 deletion in mice causes many cancers
*p53 is the most commonly mutated gene in cancer

A

p53 protein doesn’t function to transmit signals that control cell growth and stop it. Instead, it appears to have a specific role in stopping the formation of abnormal cells, particularly those with the potential to develop into tumors.

*having 1 functional p53 and 1nonfunctional p53=longer survival compared to having 2 nonfunctional p53=shorter life span since there is no more helping stop the cell cycle (no control)

118
Q

p53 functions:

A
  1. cell cycle arrest—senescence—return to proliferation
  2. DNA repair
  3. Block of angiogenesis
  4. apoptosis
119
Q

what signals p53 to function:

A
  • lack of nucleotides
  • UV radiation
  • ionizing radiation
  • oncogene signaling
  • hypoxia
  • blockage of transcription
120
Q

p53 is a transcriptional regulator

How does P53 regulate MDM2 and vice versa?

A

**MDM2’s main function is to ubiquitylate (attach ubiquitin molecules to) p53, marking it for degradation by the proteasome
*MDM2 attach ubiquitin to p53–p53 gets degraded (happens when p53 is not needed)

**during times of stress or when p53 is needed to respond to cellular damage, MDM2 is inhibited
*stress–p53 is needed—MDM2 is not functioning to inhibit p53 functions

121
Q

Is p53 always a tumor suppressor?

A

hotspot mutations in the p53 gene can result in gain-of-function activities that contribute to tumor development and progression

Because if mutation keeps happening in p53– you risk it to be deleted- and deleting it can result to oncogenic activity

122
Q

p53 point mutations have complex functions:

A
  • Protein Stability: point mutations in the p53 gene can destabilize the protein structure and may lead to increased degradation of p53
  • Non-specific DNA Binding: Some mutations may cause p53 to bind non-specifically to DNA sequences that it would not normally recognize–leading to abnormal transcription
  • Modulation of Stability of P53-DNA Complex: Certain mutations can alter the stability of the p53-DNA complex–affect the ability of p53 to regulate gene expression properly.
  • Point mutations in p53 may affect its interactions with these cofactors (help in transcription activity), leading to dysregulated gene expression patterns (can be less interaction with cofactors–repress transcription or more interaction with cofactors –which lead to unnecessary transcription)
123
Q

Neurofibromatosis type I symptoms:

A
  • Cafe-au-lait spots
    *Neurofibroma nodules
    *Lisch nodules in Iris
124
Q

NF1

A
  1. ligand-activated GM-CSF receptor causes GTP-bound Ras levels to rise rapidly
  2. having Nf1 (-/-) means that there is more Ras bound to GTP (Ras is activated)
  3. having Nf1 (+/+) means that there’s less Ras bound to GTP (Ras is in a less activated state)

explanation:
NF1 catalyze hydrolysis of GTP to GDP—thus inactivating RAS—but with mutated NF1 (-/-), Ras remains active (more GTP is bound)

125
Q

CDKN2A / ARF what is it and its function?

A
  • The CDKN2A gene encodes multiple tumor suppressor proteins like p14^ARF (or p19^ARF in mice)
  • P19 ARF activates p53 and promtoes cell cycle arrest and DNA damage, etc. and P19 ARF inhibits MDM2 function
126
Q

What is P16 Ink4a and its function?

A
  • p16^INK4a protein, encoded by the CDKN2A gene, functions as a cyclin-dependent kinase inhibitor (CDKI) that specifically inhibits the activity of cyclin D1-bound cyclin-dependent kinase 4 (CDK4)
  • When cyclin D-CDK4/6 complexes are inhibited by p16^INK4a, Rb remains unphosphorylated (or hypophosphorylated). Hypophosphorylated Rb is in its active form and exerts its tumor-suppressive function
127
Q
A
  1. Ligand binds to RTK
  2. PI3K binds to the phosphorylated RTK and becomes activated.
  3. Activated PI3K phosphorylates (PIP2) at the plasma membrane, which gets converted to (PIP3)
  4. PIP3 can activate PDK1, which activates AKT or directly activate AKT and other targets—which all lead to proliferation

*PTEN is a tumor suppressor and a phosphatase that acts as a negative regulator of the PI3K signaling pathway. It opposes the action of PI3K by dephosphorylating PIP3 back to PIP2.

128
Q
A

quasi-sufficiency + obligate haploinsufficiency is what happens when even more than 1 hit but less than 2 hits is enough to cause cancer

129
Q

genetic progression of colorectal cancer

A
  1. Normal Colon Epithelium: The APC gene regulates cell proliferation and migration by controlling the Wnt signaling pathway.
  2. Early Adenoma Formation: Loss-of-function mutations in the APC gene lead to dysregulated Wnt signaling and the formation of early adenomas.
  3. Late adenoma formation: K-ras/B-raf activation leads to dirsruption of MAPK/P13K/TGF beta signaling
  4. disruption of p53 and PTEN lead to carcinoma
130
Q

APC loss hyperactivates Wnt signaling pathway

A

Mutant APC is unable to form a
stable complex with β-catenin,
leaving β-catenin free to
translocate to the nucleus and
activate pro-growth transcriptional
targets (c-myc and Cyclin D1)

  • so normal APC (tumor suppressor) form a complex that deregulates Beta-catenin by putting it in the garbage by ubiquitin. Non-functional beta-catenin leads to less function of c-myc and cyclin D1, which results in less cell growth (means there is controlled cell growth)
131
Q

Mitosis
*Interphase (G2)
* Prophase
* Prometaphase
*Metaphase
* Anaphase
*Telophase & cytokinesis

Describe important events at each step

A

*Interphase (G2)
-not mitotic yet. There are centrosomes with centrioles. Chromosomes are uncondensed
* Prophase
-early mitotic spindle forms. There are 2 sister chromatids present
* Prometaphase
-There’s a kinetochore microtubule and kinetochore. Fragments of the nuclear envelope
*Metaphase
-metaphase plate aligns
* Anaphase
-daughter chromosomes separates
*Telophase & cytokinesis
-nuclear envelope forms and cleavage furrow forms

132
Q

interphase:

*G1
*S
*G2
*M (mitosis)
*G0

what happens at each step

A

*G1–cell grows
*S–cell continues to grow as it duplicates its chromosomes
*G2–Cell grows more and prepares for mitosis
*M (mitosis)– cell divides into two cells, Chromosomes
condense, attach to spindles, and pull to opposite ends of the cell. Those two cells restart the cycle.
*G0–a cell can leave the cycle and stop growing

133
Q

G0: Quiescence, terminal differentiation, and senescence

what happen here:
*reversible quiescence
*Terminal differentiation
*Senescence

A

*reversible quiescence
–enter G0 and can also exit G0
*Terminal differentiation
–selective reversing of cells that enter G0
*Senescence
—cannot be reversible and is a response to cellular stress like aging

134
Q

what causes senescence?
Give 3
how is senescence triggered?

A

1.DNA damage response
2. telomere shortening and damage
3.oncogene activation

*When these 3 features are detected–they lead to cell cycle arrest (because when damage is detected, one way to respond is to arrest the cell cycle)
*Prolonged Dna damage response triggers senescence, shortening of ends of chromosomes (super short), and oncogene activation are senescence triggers

135
Q

senescence features:

A

*prolonged cell cycle arrest
*oxidative damage
*resistance to apoptosis
*SA-beta-gal
*SASP
*SAHF

136
Q

*SA-beta-gal
–how is it activated?
–what is its role?

A

–In “young” or non-senescent cells, lysosomal β-gal is activated at low pH = 4, but in “old” or senescent cells, lysosomal β-gal is activated at pH = 6
–SA-β-gal is not required for senescence, but is a common biomarker used to identify senescent or aging cells

137
Q

cell cycle checkpoints in:
*G2
*M
*G1
*S

A

*G2–if DNA replication is not completed, the cell won’t continue to M phase
*M– anaphase is blocked, if chromatids are not properly assembled on the mitotic spindle,
*G1–the entrance to the S phase is blocked if DNA damage or genome damage is detected
*S–DNA replication is stopped if the genome is damaged

138
Q

what is the cell cycle clock?

A

a network of interacting proteins—a signal-processing circuit—that receives signals from various sources both outside and inside the cell, integrates them, and then decides the cell’s fate.

139
Q

G1 is the period during which the cells are responsive to what?

A

mitogenic growth factors and to inhibitory factors (TGF-beta)
–otherwise, if GFs are removed, the cell wont make it to complete G1. Similarly, if inhibitory factors are more effective in stopping G1 than if cell made it to the R point—at which the cell makes it through the next round

140
Q

what is the R point?

A

–the point within G1 at which the
cell is committed to dividing. It no longer requires growth factors to complete the cell cycle
–also the point in time when the cell must commit to advance through the remainder of the cell cycle through the M phase, to remain in G1, or to retreat from the active cell cycle into G0.

141
Q

Cyclins and Cyclin-Dependent Kinases (CDKs) regulate cell cycle progression

A

*Each type of cyclin pairs with a specific cyclin-dependent kinase
*all cells in the embryo go through S and M at the same time
*seeing in cyclin B—t the end of the M phase, the cyclin increases as it is prepared to be degraded
*after G1 phase, cyclin D1 can no longer influence the cell cycle

142
Q

Cyclin E associated with poor breast cancer survival

A
  • abnormally high levels of cyclin E in the cancer cells of breast carcinoma patients are strongly predictive of aggressive malignancy and poor patient outcome, while low levels indicate long-term, disease-free survival
  • lower molecular-weight forms of cyclin E that are found in many aggressive tumors function abnormally to drive the formation of multiple centrosomes, which results in genetic instability and acceleration of tumor progression
143
Q

Cyclins and Cyclin Dependent Kinases (CDKs) alterations in cancer

A

*mutations in CDKs tend to co-
occur more frequently than expected by chance
*alterations in cyclins and CDKs can mess up the cell’s “growth and division” room, causing the cell to grow and divide uncontrollably.

144
Q

Co-occurrence and mutual exclusivity

A

Co-occurring mutations happen together
more frequently than would be expected by chance. These drivers typically either:
A. Activate collaborating oncogenic
pathways (cdk collaborate on activating oncogenic pathways)
B. Activate the same pathway in multiple
ways that are synergistic (meaning:they activate the same pathway through different ways, but together they make the effect more powerful)
Mutually exclusive alterations (happens exclusively, not in group)tend to be
either:
C. Located in the same pathway in
sequence, such that having one
mutation makes the other one not
helpful (one mutation is doing more damage than the other)
D. In a synthetic lethal relationship with one another, where either mutation alone is OK but both together are lethal (mutation alone is ok, as one is still functional, but a mutation that together happened is powerful or worse because it leads to a nonfunctional gene)

145
Q

how does synthetic lethality work in genes?

A

*when two mistakes happen in the gene, and together, they make the genes stop working
*both gene ok=viable
*gene A ok, but not gene B=viable
*Gene B ok, but not gene A=viable
*Gene B and Gene A not ok= lethal

146
Q

Internal and external growth signals
When do cells listen to extracellular signals and when do they stop listening and start listening internally?

A

the cyclin D–CDK4/6 complexes are capable of ushering a cell from the beginning of the G1 phase up to the R-point gate. After the cell has moved through the R point, the remaining cyclins—E, A, and B—behave in a pre-programmed fashion, executing the fixed program that begins at the R point and extends all the way to the end of the M phase. once the cell has passed through its R point, its cell cycle machinery takes on a life of its own that is quite autonomous and no longer responsive to extracellular signals.

147
Q

Upstream regulation of CDKs/Cyclins

A
  • Cyclin–CDK complexes are also regulated by CDK inhibitors at varous points in the cell cycle
  • these CDK inhibitors affect cell cycle progression, transcriptional regulation, apoptosis, cell fate determination, etc
148
Q

DNA damage and cell cycle regulation
2 example of cdk inhibitors?

A
  • When TGF-β is applied to epithelial cells, it antagonizes cell proliferation
  • ex: increase in the levels of p15INK4B, which block the formation of cyclin D–CDK4/6 complexes and inhibit the formed complexes
    *Without active D–CDK4/6 complexes, the cell is unable to advance through early and mid-G1 and reach the R point
    *p21Cip1–a cdk inhibitor
    *damage to cellular DNA causes strong, rapid increases in p21Cip1, which in turn can shut down the cyclin–CDK complexes that are active in the phases of the cell cycle after the cell has passed through the R point in the late G1 phase
    *ensuring that the cell does not progress into the S phase and inadvertently copy still-damaged DNA sequences

Tgf beta- before passing r point
P21- after passing r point

149
Q

P21 andP27 role incyclin CDK complexes (cyclin cdk 4/6 and cyclin E/cdk2

A
  • p21Cip1 and p27Kip1 inhibit the actions of cyclin E–CDK2, cyclin A–CDC2, and cyclin B–CDC2 (cyclin-cdk complexes), but they actually stimulate the formation of cyclin D–CDK4/6 complexes.
  • cells in G0 quiescent state, p27kip1 is present in high concentrations—suppress cyclin-cdk complex involved
  • growth factors—sends signals to cells to grow—so cyclin D-CDK4/6 increases
    *cyclin D–CDK4/6 forms=p27kip1 reduces
    *p27kip1 reduces=allows cell to move to the R point (importantly: less p27kip1 means cyclin E-CDK2 is less inhibited, so they’re able to pass through the R point)
  • as cycle keeps progressing to the R point—p27kip1 is degraded (more activation of cyclin E-CDK2)
150
Q

what controls Rb phosphorylation, and how does it affect E2Fs?

A

*G1 phase–pRb is weakly phosphprylated
*during R point–pRb is hyperphosphorylated until the end of mitosis
*end of mitosis—pRb is dephosphorylated to prepare for another round of cell cycle

*when pRb is weakly phosphorylated, pRb binds E2F (E2F is inactive)
*when hyperphosphorylated, pRb unbinds to the E2Fs (E2F is active)

*pRb can control cell cycle advance—-Rb phosphorylation is controlled by cell cycle progression (certain phase hypo or hyper phosphorylates Rb, which affects how E2F behaves

151
Q

*CDK2 and cyclin E hyper-phosphorylate Rb

*but what controls CDK2 and cyclin E?

A
  • hyperphosphorylated pRb leads to its own inactivation (unattached to E2F)
    *Inactivation of pRb leads to increased cyclin E levels, which in turn further inactivate pRb (positive reinforcement)
    *free E2F produces more cyclin E, which forms a complex with CDK2, which further inactivate pRb, so we have more E2F (positive feedback)
152
Q
  • p27 regulates CDK2 and cyclin E

*but what regulates p27? cyclin E-CDK2 complex

ASK professor!!!

A

*p27Kip1 molecules, which normally inhibit E–CDK2 activity
*Cyclin E–CDK2 complexes phosphorylate p27Kip1, leading to the destruction of p27Kip1 molecules–Cyclin E-CDK2 is active= (positive reinforcement)

153
Q

*Upstream regulation of p27 by Myc/Max

*What regulates Myc/Max?

could it be dephosphorylation of myc itself or just the lost of presence of growth factors itself? (ask)

A

*Myc/max boosts the expression of cyclin D2 and CDK4 genes, which form complexes that help regulate the cell cycle
*increasing CDK4 = increased p27kip1 (an inhibitor)
*Myc/max enhances production of Cul1 = Cul1 inhibits p27kip1
*inhibited p27kip1 = activates cyclin E-CDK2 complex = cyclin E-CDK2 complex inactivates pRb = inactivated pRb activates E2F =activated E2F means cell can push through the cell cycle (reached the R point–after which cell can continue until end of mitosis)=CELL GROWTH

154
Q
A
  • TGF-β signals through its receptor to phosphorylate Smad proteins, including Smad2 and Smad3
    *Phosphorylated Smad2/3 form a complex with Smad4 and move to the nucleus, where they team up with Miz-1 to activate p15INK4B and weakly p21Cip1 (who are CDK inhibitors)
    *if cdk complexes are inhibited=myc levels collapse=cell cycle wont pass G1 level
    *if myc team up with Miz-1=they can stop cdk inhibitors from inhibiting cdk complexes
    *but TGF-β can block Myc’s action by sending Smad3 to form a complex with E2F4 or E2F5 plus p107 =so myc will be kept inhibited
  • This leads to a collapse in Myc levels, ensuring that TGF-β successfully induces the expression of CDK inhibitors

result: shuts down cell cycle progression in the early/mid G1 phase

155
Q

sterile technique

A

*Sterile technique is a way to perform tissue culture procedures (like from tissue from a tumor) without introducing contaminating microorganisms from the environment

156
Q

Tissue culture

A

Cancer cells are typically grown in a sterile incubator at human body temperature of 37°C and 5% CO

157
Q

model organisms to study cancer

A
  • tissue culture–fast
    *organoids–tissue have to grow fresh, hard to get
    *drosphila
    *zebrafish
    *mice–more similar to humans

*all of these example can be engineered

158
Q

tissue culture

A

A method of growing cells in an incubator

159
Q

endogenous

A

Naturally made by the cell

160
Q

exogenous

A

Artificially added to the cell

161
Q

Complementary DNA (cDNA)

A

*Encodes the sequence of Your Favorite Thing (no introns)
*synthesized from or complementary to mRNA

162
Q

short hairpin RNA (shRNA):

A
  • Decreases the amount of Your Favorite Thing (temporarily)
163
Q

small interfering RNA (siRNA):

A

Temporarily decreases the amount of Your Favorite Thing for ~1-3 days

164
Q

CRISPR (gRNA):

A

*Permanently deletes or genomically edits Your Favorite Thing

165
Q

Molecular cloning:

A

*Creates a DNA plasmid that will express Your Favorite Thing

166
Q

Restriction enzyme:

A

Cuts purified DNA at a specific sequence

167
Q

Inducible promoter:

A

Controls where and when Your Favorite Thing is expressed

168
Q

Transient transfection:

A

Temporarily expresses Your Favorite Thing for ~1-3 days

169
Q

Transduction:

A

Can permanently express Your Favorite Thing by integrating it into a cell’s genome

170
Q

How do you construct something that expresses more of Your Favorite Thing?

steps?

A

through Molecular Cloning

  1. at ecori site where ecori enzyme attaches, cut at top and bottom (creating an L shape like for both top and bottom genome)
  2. the L-shape overhang has a sticky ends
  3. target gene (YFT) will be put in the middle—so when it forms a circle, the sticky ends attach to each other
171
Q

How do you control when Your Favorite Thing is on or off?

types:

*Inducers that activate or repress transcription can be added manually by researchers, or researchers can choose inducers that are naturally expressed at a known time and place (you can pick which gene or part of organ is active or inhibited)

A

through Inducible promoters (ex. scenario: genes in lungs are on, but off for liver)

*not a type but, Constitutively Active Promoter–where transcription of YFT gene is always on
**positive inducible–activator + inducer=transcription
**negative inducible– repressor + no inducer = transcription inhibited
**negative inducible– repressor + inducer=transcription activated

*repressor job is to repress gene, so when inducer is not bound, repressor bind to dna prohibiting transcription. But when inducible is bound to repressor, repressor cant bind to dna, so dna can be transcripted
*activator job is to activate gene. So when activator along with inducer can enhance full transcription of the gene

172
Q

How do I make more of the plasmid containing Your Favorite Thing?

steps:

A

through Bacterial cloning

  1. take a gene from one place (ex: from molecular cloning)
  2. stick molecularly cloned gene into a plasmid vector
  3. insert your gene
    *antibiotic resistance gene is also present (so your gene won’t be killed)
  4. stick the vector into bacteria—bacteria will make copies (TRANSFORMATION)
  5. (SELECTION)–let the ones selected from the plasmid grow on the plate, which is antibiotic-containing media (antibiotic kill bacteria that do not have a plasmid)
  6. pick colonies and let them make more copies in a tube
    *result would be tons of bacteria with plasmid
173
Q

To understand what Your Favorite Thing does, what should you do?

ways you could do this:

A

*artificially make more or less of it, then measure what happens
–Make cells create more YFT:
through Complementary DNA (cDNA) encoding YFT to overexpress it
–Remove most of the YFT cells already have: use Short hairpin RNA (shRNA) to knockdown YFT permanently OR Small interfering RNA (siRNA) to temporarily knockdown (~1-3 days) YFT
Result: decrease endogenous YFT protein
–Remove half or all of the YFT cells already have:
using CRISPR (will destroy genome as gRNA binds to DNA and cas9 cuts the bound dna in gRNA) using a guide (gRNA) to knockout one or both copies of YFT from the genome
Result: no endogenous YFT protein

174
Q

Modern applications of CRISPR in NHEJ and HR:

A

*CRISPR in NHEJ
—(GENE KNOCKOUT)without donor dna, insertion and deletion can happen which can knockout the gene
—(GENE INSERTION) with donor dna–the donor DNA in big amount can be inserted alone with the other dna
*CRISPR in HR
—(GENE MODIFICATION) with donor dna–donor dna in small amount with its complete genome can be added to the other dna
—(GENE INSERTION) with donor dna–the donor dna in big amount with its all genome is added to the other dna

175
Q

How do you make cells temporarily express Your Favorite Thing ?

steps:

A

*through Transient transfection

  1. Original DNA is silenced by siRNA
    ** plasmid contains silenced DNA and YFT
  2. transfection reagent (to express YFT) is added to the plasmid forming transfection complex
  3. complex is enclosed by endosome
  4. plasmid survives (means it contains YFT) and now enters the nucleus
176
Q

How do you make cells forever
express Your Favorite Thing?

A

*Transduction for stable integration

  1. (TRANSFECTION)—YFT + VSVG (virus glycoprotein) + Gag-Pol (viral packaging components and reverse transcriptase)=virus
  2. virus endocytose–preparing to go inject the virus into the target cell
  3. transduction—to cells that can’t make virus
    *virus is used as a way to deliver YFT to the host cell’s genome
    *YFT now becomes permanent part of the host cell’s genetic material
177
Q

Competition experiment test what?

mechanisms?

possible result?

A

–Test whether YFT provides a selective advantage (oncogene if has advantage and tumor suppressor if doesn’t)

*plasmid (containing a GFP and YFT) is transduced in the IL3-dependent cell line—we will observe whether YFT will show selective advantage due to removal of IL3 for 2 days and addition of IL3 for 3 days

possible Result:
*a 90% GFP is expressed =means YFT is oncogenic (removal of IL3 and addition was both an advantage as YFT was an oncogene
*a 50% GFP+ is expressed = means YFT has no result as both removal and addition of IL3 generated the same result
* a 10% GFP+ is expressed =means YFT is tumor suppressive because when IL3 was removed for 2 days, the cell was also inhibited from growing (it depended on IL3)— so yft is TS because it’s keeping the grwoing of cell in check

178
Q

Subcellular localization:

A

Immunofluorescence visualizes where YFT is expressed (is YFT localized or not)

  • direct immunofluorescence–primary antibody specific to YFT is labeled with dye—so if it binds YFT, we can locate it
    *indirect immunofluorescence–when a primary antibody binds YFT and a secondary antibody containing the fluorescent bind to the primary= together, they will help light up and locate the gene (YFT)
179
Q

Apoptosis of YFT: (Is Your Favorite Thing Killing Cells?)

A

Annexin V or Cleaved Caspase-3 measures programmed cell death (did cells die or not)

*early apoptosis= phosphatidyl serine translocates to outer from the inner leaflet, cell membrane still intact so there’s programmed cell death
*secondary necrosis= phosphatidyl serine is in inner and outer, cell membrane not intact, and there’s unprogrammed cell death which means YFT is killing cells

180
Q

Viability/Proliferation:

A

Trypan Blue or CellTiter-Glo count viable cells over time,
Ki67 reflects proliferative state

181
Q

Secretion of YFT:

A

Enzyme-linked immunosorbent assay (ELISA) for local or ELISA array for global (some proteins leave the cell—YFT change where they’re secreted)

182
Q

test metastasis:

A

Migration assay or scratch test

183
Q

test DNA changes:

A

Sanger sequencing for local, exome sequencing for global

184
Q

RNA expression: this test which gene changed

A

qPCR for local, RNAseq or single cell RNAseq for global (some genes change how they’re expressed)

185
Q

Protein expression and post-translational modification:

A

Immunoblot (Western blot) for a few, protein array for many

186
Q

binding to DNA:

A

Chromatin immunoprecipitation (ChIP-seq)

187
Q

Binding to RNA:

A

Cross-linking immunoprecipitation (CLIP-seq)

188
Q

Cell surface markers:

A

Flow cytometry

189
Q

Cytokine dependency

A

–IL3-dependent cell line + IL3 =cell growth
–IL3-dependent cell line - IL3 = no cell growth
–IL3-dependent cell line - IL3 + oncogenic YFT= supposed to be cell growth but oncogenic YFT drives the cell growth

*FL5-12 cells are an example of an IL3-dependent B-cell lymphoma cell line

190
Q

Does Your Favorite Thing cause cells to grow faster and/or be more viable?

what can we use to know this?

A

*use trypan blue live/dead stain

trypan blue stains dead cells because they can penetrate compromised cell membranes. But try pan cannot penetrate live cells.

with this, we can count the number of dead cells, and find out if cells did grow and become viable or did they die?

191
Q

How do we find out how many viable cells are there?

A

Viability assay (CellTiter-Glo)

the cells are the source of ATP in the luciferase reaction—when luminescence is produced, that tells us the number of viable cells. How many cells light up=tells us how many of the cells are viable

192
Q

edge effect

A

“edge effect” refers to the phenomena in which contents of the wells on the outside of the plate evaporate during incubation.

193
Q

Does Your Favorite Thing change what cells secrete?
what assay to use to find that?

A

use Enzyme-linked immunosorbent assay (ELISA)

the binding of the target protein with the enzyme-labeled antibody in ELISA provides a quantitative measurement of the amount of the target protein secreted by cells.

if YFT change what cells secrete, we can detect it through elisa, which monitors the different target protein that binds to enzyme

194
Q

How to tell if metastasis happen

A

Migration assay (scratch test)

we can observe the cancer has metastasized, if we scratch the middle part of the cell in the dish, if the cell fills up quickly to cover the middle part, means that cancer is present as it didn’t follow contact inhibition

195
Q

Does Your Favorite Thing change RNA expression?

A

RNAseq—can tell how much RNA is expressed

196
Q

Does Your Favorite Thing change RNA in some cell types differently than others?

which assay to use?

A

Single cell RNAseq for heterogeneous samples (like tumors)

each cell is put on a separate wells—we can see that each cell type has a distinct expression—then we can observe heterogeneity (difference) and variablity of thousands of cells

197
Q

Does Your Favorite Thing change the expression or modification of another specific protein? (you can overexpress YFT or tweak it)

test how?

A

use Immunoblots (Western blots)—immunoblots to measure protein

  1. lyse the sample and heat the sample (induced change applied here)
  2. gel electrophoresis—proteins separate
  3. membrane transfer–
  4. immunodetection—use protein stain to check transfer efficiency—

*beta-actin–loading control (no changes)
*downstream protein in YFT (one you’re testing in YFT)

*They show whether a particular protein is present in a sample and can provide information about its abundance or changes in expression levels under different conditions.

198
Q

Does Your Favorite Thing change protein expression or modification of many things?

A

Protein arrays
*forward phase–is you stick protein on plate, then stick the antibody on it
*reverse phase is you stick antibody on array, and it catches the protein specific to it

**both mechanisms allows us to see protein expression and modification

199
Q

If Your Favorite Thing binds to DNA, where does it bind?

A

Chromatin immunoprecipitation (ChIP-seq)

the sequence at the end creates a map sequence that shows which sequence bind to YFT and match it to certain genes to identify it

200
Q

If Your Favorite Thing binds to RNA, where does it bind?

A

Cross-linking immunoprecipitation (CLIP-seq)

irradiation induce cross linking, and at the end we can see if YFT bind to the binding site

201
Q

*genetic engineering–permanent change in genome and inheritable (slow)

A
202
Q

xenografts–take human cell line and put on immunocompromised mice

A
203
Q

patient dervied xenografts—more cell types of cell line that is put on immunocompromised mice (can test whether YFT is a tumor supressor

A
204
Q

adoptive transfer—for immune malignancies—wipe out mice immune system temporarily and replace it with YFT for example (can test whether YFT is oncogene)

A
205
Q

syngeneic system—recipient mice wont reject tumor—Researchers can implant tumor cells from a genetically identical mouse strain into the recipient mouse and observe how the immune system responds to the tumor.

A
206
Q

modification using CRISPR/Cas9—means next generation will be the same (permanent change, that can be inherited by next gen)

A
207
Q

transgenic cas9 + sgRNA virus=noninherited genes (2 step

A
208
Q

cas9 with sgRNA virus (1 step)=noninherited genes

A
209
Q

Tumor xenografts in immunodeficient mice

*if the YFT is injected with human cancer line, and it lead to no cancer=means YFT is tumor supressor, since it was bale to supress development of cancer
*
*if the YFT is injected with human cancer line, and it lead to cancer, means YFT is an oncogene and it drove the further development of cancer

A
210
Q

Is the effect of Your Favorite Thing on cancer impacted by the normal immune system?

A

Syngeneic xenograft models (immunocompetent mice with intact immune system)

*in vivo tumor from donor mice is grown in a bottle/dish, then transferred to the recipient mice from the same type of mice==test if immune system of different mice but of the same type can respond to the YFT (is YFT oncogene and bypass immune system or tumor supressor and be subjected to the immune system

211
Q

Where is Your Favorite Thing located within a solid tumor sample?)

A

using Pathology and immunohistochemistry (IHC)—we can stain yft and see where it goes

212
Q

EZH2 and other PRC2 members act as oncogenes by silencing the expression of multiple tumor suppressors

A
213
Q

Chromatin is regulated by histone acetylation

*acetylation—HAT opens chromatin, whih allows RNAPII to activate transcription
*deacetylation—HDAC makes chromatin condensed–which turn off transcription

*On balance, HDACs tend to be oncogenic because they repress expression of multiple tumor suppressors

A
214
Q

alternative splicing of mRNAs gives rise to different proteins containing different mRNA from a single gene

A
215
Q

microRNA

A

—short endogenously expressed RNAs
—-Downregulation of target messenger RNAs by degradation and inhibiting their translation
—-6-8 nucleotide seed sequence provides target specificity

216
Q

oncomiR-1 cluster cooperates with myc to drive b-cell lymphoma (example of adoptive transfer model)

A

—The miRNAs in the oncomiR-1 cluster might suppress the expression of genes that normally inhibit cell proliferation or induce apoptosis, thereby promoting cell survival and proliferation. —Myc, on the other hand, could further stimulate cell growth and division by activating the expression of genes involved in these processes

217
Q

Testing individual miRNAs for effects on lymphocyte survival

A
  • a cell line can grow in the absence of cytokine (IL3) through application of oncogenes
    *miR-19 protects lymphocytes from growth factor depletion—even without IL3, cells were still generated (because of mir-19)
218
Q

*mir-19 cooperates with Notch in T-ALL—powerful combination to drive acceleration of T-cell leukemogenesis
*miR-19 regulate its predicted target genes
*mir-19 activates P13K signaling–

A
219
Q

PTEN and the PTENP1 pseudogene

A

*ptenp1 does not contain the methionine that inititates pten. ptenp1 is not translated into a protein
* all proteins are started in methionine
*PTENP1 overexpression led to increased PTEN mRNA and PTEN protein levels, decreased cell proliferation and induced apoptosis, and inhibited migration and invasive ability (more ptenp1 leads to more tumor suppressor)

220
Q

Competing endogenous RNA hypothesis

unsure (slide 20)

A

*Noncoding RNAs “sponge” up RNA binding factors like miRNAs or RNA binding proteins
decreasing their availability to bind messenger RNAs

221
Q

long non-coding RNA function (master regulators of gene expression)

  • can act as an oncogene and tumor suppressor
A
  1. LncRNAs can recruit different protein components of the chromatin remodelling complex to change the chromatin organizational patterns
  2. They can act as ‘sponges’ by base pairing with their complementary miRNAs and reducing their effects
  3. LncRNAs can play scaffolding roles by providing docking sites for proteins that function together in the same biological pathway
  4. They activate transcription of certain genes by guiding transcription factors to their promoters.
  5. LncRNAs are capable of suppressing transcription by sequestering transcription factors and keeping them away from their promoters. They can modulate mRNA functioning through base pairing with them
  6. inhibiting their translation
  7. altering their splicing patterns and (H) subjecting them to degradative pathways.
222
Q

protein translation in cancer

need more info

A

–keep growing and forming protein

223
Q

cap dependent translation–meaning translation depends on external factors for initiation to happen—like EIFs

A

*EIFs help the ribosome get loaded and moving
*initiation complex scans for the start codon (AUG)
eifs are located at the 5’ cap of mrna

224
Q

Upstream regulation of cap-dependent translation

A

*ligand-activates ras-ras activates raf- raf activates mek-mek activates erk-erk activates mnk1/2and EIFs to inittiate translation
*ligand activates p13k-p13k activate akt-akt inhibits tsc-tsc inhibits mtor complex (translation initiation is inhibited)

*p13k is an oncogenic activity—so pten inhibits p13k activity
* mtorc1 complex on—more proteins get made

225
Q

less eif4e limits translation, but overexpression of eif4e leads to lymphoma

*Adoptive transfer model of lymphoma driven by MYC + Your Favorite Thing

A
226
Q

cellular stressors can affect translation—

stressors can lead to phosphorylated EIF2 alpha —which means EIF2 is inhibited and will also inhibit translation

A
227
Q

IRES can recruit translation machinery without a 5’ cap—can initiate translation if 5’ cap is not present (needed to start translation)

A
228
Q

how do you test what factors affect cap-dependent translation?

A

Dual luminescent assay

229
Q

elongation Initiation Factor 4A (eIF4A)

A

eIF4A, the helicase responsible for unwinding RNA structures, can act as an oncogene

*silvestrol inhibit eif4a
*eif4a selectively inhibits translation of mrnas with specific 5’ utr sequence elements

*Inhibition of eIF4A can affect translation initiation by preventing the unwinding of these secondary structures, particularly G-quadruplexes, which can hinder ribosome recruitment and scanning along the mRNA molecule.

230
Q
A