Molecular, carcinogenesis, Familial disorders, viruses, DNA repair, telomeres, Cell cycle Flashcards
What is the definition of RNA interference?
RNAi = The process of mRNA degradation that is induced by dsRNA in a sequence specific manner
Can specifically silence the expression of any gene for which the sequence is available
Large set of specific cellular responses to RNA called RNA silencing
Group of proteins degrade ds mRNA, meant to protect genome from viruses that use dsRNA in their life cycles
What is the process of RNAi?
First, Drosha and DGCR8 complex process miRNA in the nucleus to form pre-miRNA. This is exported to the cytoplasm by Exportin 5. Dicer then processes it into mature ds-miRNA (siRNA), which is 18-25 nucleotides in length. RNA strands are separated
miRNA is incorporated into an RNA-induced silencing complex (RISC). miRNA guides RISC to the 3’ untranslated region of the complementary mRNA and represses expression. (such association can result in either the inhibition of translation or the mRNA or its degradation, or both)
What are miRNAs? Where are the transcribed from?
RNA silencing plays a critical role in regulation of cell growth and differentiation using endogenous small microRNAs (miRNAs)— mostly transcribed from introns or other noncoding areas of the genome; +/- exons of other genes
What is siRNA?
Long dsRNA molecules are digested by Dicer into 19-21(18-25) nucleotide fragment (siRNA= mature miRNA)
T/F? Loss of Dicer enzyme has been associated with cancer progression.
True
How do you interpret Western blots?
No bands present= Negative
Bands present= Positive
Bands present, but pattern does not meet criteria for positivity= Indeterminate
What is FISH?
What does it require to run?
What chromosomal abnormalities can it commonly detect? List examples of this.
What phase of the cell cycle is it performed on?
form of cytogenetic analysis (detects chromosomal abnormalities)
o Requires knowledge of DNA sequence to create a specific probe
o Can assess chromosomes for amplifications, deletions, or other abnormalities
o Uses: determine HER2 status, detection of N-myc amplification, etc.
o Can be performed on interphase nuclei
What is comparative genomic hybridization (CGH)?
What chromosomal abnormalities can it commonly detect?
What chromosomal abnormalities can it NOT detect?
How does it differ from FISH?
CGH was more recently replaced by what test, which can better detect abnormalities of a smaller magnitude?
Also a form of cytogentic analysis (detects chromosomal abnormalities)
o Detects increases or decreases in cancer DNA relative to controls (changes in copy #)
Cannot determine the cause (amplification vs. translocation, etc.)
Balanced rearrangements (inversions, translocations) may escape detection
o If sequence is unknown, cannot select a probe for FISH – CGH is a more generalized technique
o More recently replaced by microarrays, which can detect abnormalities of a smaller magnitude
What is the difference in what annexin dye vs propidium iodide stains for or detects in regards to apoptosis in cells?
o Annexin dye = stains cells that have initiated early apoptosis; detects PS bound by annexin V protein
Binds phosphatidyl serine on the outer leafleft of cells undergoing early cell death
o Propidium iodide = detects cells in late apoptosis because it requires permeability to the nucleus
Binds DNA of cells if is able to enter owning to damaged membranes, hence indicating late cell death
Dot plot with annexin on x axis and PI on y axis – determine which answer choice is correct with percentage of cells in necrosis/late apoptosis, early apoptosis, or overall apoptotic
Sorry no picture- use your imagination
* see example pictures in Phoenix
Y axis= PI Necrosis
Viable cells Apoptosis
X axis= Annexin
In regards to the process of carcinogenesis, describe from the list below which does NOT and which DOES lead to formation of a tumor?
- A single low dose of initiator
- Promoter is applied only or promoter is applied before initiator
- Low doses of initiator followed by repeated doses of promoter
- Application of promoter is delayed for several months after initiator
- A single low dose of initiator does not give rise to a tumor
- No tumor develops if only promoter is applied or if promoter is applied before initiator
- Low doses of initiator followed by repeated doses of promoter gives rise to a tumor
- Tumor can develop if application of promoter is delayed for several months after initiator
As part of the steps of carcinogenesis, what is tumor initiation?
Tumor initiation: involving the interaction of a reactive chemical species with DNA to produce damage, which if not repaired before the next cell division, would lead to erroneous DNA replication resulting in fixation of mutations within the genome of individual cells
Initiator: generates mutation but, by itself, does not cause tumor formation
As part of the steps of carcinogenesis, what is tumor promotion?
Tumor promotion: clonal expansion of an initiated cell as a consequence of events that alter gene expression, so as to provide the cell with a selective proliferative advantage
o Promoter: exerts nongenetic effects, but triggers proliferation
Increased division increases likelihood that a second mutation will occur
As part of the steps of carcinogenesis, what is tumor progression?
Tumor progression: the stage whereby benign lesions acquire the ability to further grow, to invade adjacent tissues, and to establish distant metastasis
o Progression: development of a 2nd mutation
Increased genomic instability and karyotypic alterations
Acquires ability to grow, invade, metastasize
Which of the steps of carcinogenesis is reversible
tumor promotion
Which proteins or mutations that occur as cancer associated genetic changes are responsible for regulation of cell proliferation, death, and differentiation?
T/F: Mutations of these proteins correlate with the highest risk of cancer?
Gatekeeper mutations
o Typically tumor suppressor genes
o Mutations of these proteins correlate with the highest risk of cancer= TRUE
Which proteins or mutations that occur as cancer associated genetic changes are responsible for guarding the integrity of the genome?
Loss of function increases risk of what?
These mutations often involve what part of cell functions?
Care taker mutations
Loss of function increases the risk of acquiring gatekeeper mutations that would lead to oncogenic growth
o Often involve DNA checkpoints and repair factors (i.e., BRCA1/2 mutation)
Which proteins or mutations that occur as cancer associated genetic changes alter the tumor microenvironment?
landscaper mutations
List the original 6 Hallmarks of cancer.
- Sustained proliferative signaling
- Evading growth suppressors
- Activating invasion and metastasis
- Enabling replicative immortality
- Inducing angiogenesis
- Resisting cell death
List the enabling Hallmarks of cancer.
- Genomic instability and mutation
2. Tumor promoting inflammation
List the emerging Hallmarks of cancer.
- Reprogramming of E metabolism
2. Evading immune destruction
The following are examples of which of the hallmarks of cancer?
Upregulation of EGFR or HER2/neu
Activating mutations in BRAF
Activating mutations in catalytic subunit of PI3K
Upregulation of CDK4
Loss of negative feedback via:
• Activating mutations in RAS
• Loss of function mutations in PTEN
• Activation of mTOR
Sustained proliferative signaling
The following are examples of which of the hallmarks of cancer?
Loss of E-cadherin
Upregulation of N-cadherin
Mutation/inactivation of B catenin genes
Increased ezrin
Upregulation of TFs involved in EMT: Snail, Slug, Twist, Zeb 1/2
Upregulation of protease genes
Activating invasion and metastasis
The following are examples of which of the hallmarks of cancer?
RAS and c-Myc can upregulate expression of angiogenic factors
Loss of VHL (results in upregulation of VEGF)
Upregulate VEGF and/or FGF expression
Loss of TSP-1 or IFN-B
Inducing angiogenesis
The following are examples of which of the hallmarks of cancer?
Defects in caretaker genes (DNA repair machinery)
Loss of telomeric DNA
BUB1
Genomic instability and mutation
The following are examples of which of the hallmarks of cancer?
Warburg effect (aerobic glycolysis)
Upregulation of GLUT1 to include Glc delivery to cytoplasm
Increased HIF-1a- upregulates glycolysis
Gain of mutation mutations in isocitrate dehydrogenase (IDH1/2)
Reprogramming of E metabolism
The following are examples of which of the hallmarks of cancer?
Loss of RB (gatekeeper)
• Down-regulation of TGF-B receptors (TGF-B prevents phosphorylation of RB)
• Loss of SMAD4 (transmits signals from TGF-B)
• Deletion in p15INK4B
• CDK4 may be unresponsive to inhibitory signals from p15INK4B
o Therefore, Cyclin D-CDK4 complex can inactivate (hyperPhos) pRb
• Mutation in pRb itself
• Sequestration of pRb by viral oncoproteins (e.g., HPV E7)
Loss of P53 (another gatekeeper)
• Li-Fraumeni syndrome
• HPV E6 oncoprotein
Avoid differentiation
• Overexpression of c-Myc
Loss of contact inhibition
• Loss of Merlin, the cycoplasmic NF2 gene product
• Suppression of LKB1, which maintains epithelial polarity (normally overrules effects of Myc)
Redirection of TGF-B pathway (antiproliferative activates EMT)
Evading growth suppressors
The following are examples of which of the hallmarks of cancer?
TERT expression or upregulation of ALT – resistance to senescence and crisis/apoptosis
Loss of P53 – survive telomere erosion and BFB cycles
Enabling replicative immortality
The following are examples of which of the hallmarks of cancer?
• Upregulate factors that inhibit apoptosis = Bcl-2, BCL-XL, BCL-W, MCL-1, A1
o Activation of PI3K-AKT/Pkb pathway
• Downregulate factors that trigger apoptosis = Bax, Bak, Bim, Bid
o Loss of P53 (normally upregulates Bax expression)
o Loss of caspases
o Loss of Fas
Upregulation of survival factors (i.e., IGF-2, IL-3)
Evasion of autophagy
• PI3K, AKT, and mTOR inhibit autophagy
• Loss of Beclin-1, a BH3-only protein (normally triggers autophagy)
Necrosis may trigger inflammation to further tumor progression
Resisting cell death
The following are examples of which of the hallmarks of cancer?
Bioactive molecules supplied to the microenvironment, including GFs that sustain proliferative signaling, survival factors that limit cell death, proangiogenic factors, ECM-modifying enzymes to facilitate angiogenesis, invasion and metastasis, and induction signals that lead to activation of EMT
Release of ROS that are actively mutagenic
IL-1B production
Tumor-promoting inflammation
The following are examples of which of the hallmarks of cancer?
Deficiencies in development or function of CD8+ or CD4+ T cells, or NK cells
Secretion of TGF-B, which is immunosuppressive
Recruitment of Tregs, MDSCs, TAMs
ICAM-1 production
Evading immune destruction
Which of the following mechanisms of action is/are associated with loss of heterozygosity?
- Gene conversion
- Mitotic recombination
- State of possessing two identical forms of a particular gene
- Loss of chromosomal segment resulting in hemizygosity
- Degradation of a set of short-lived chromosomes, enabling cellular regulation at the level of chromosome instability
- Nondisjunction- loss of chromosome
o (1) Gene conversion: when the DNA strand is elongated, it temporarily switches templates and forms a hybrid with the cDNA strand of the homologous chromosome. It then reverts back to its original template (copy choice). If the cDNA strand contains an inactive allele, then LOH has occurred.
More frequent that mitotic recombination.
o (2) Mitotic recombination (recombination between sister chromatids)
o (3) Loss of chromosomal segment, resulting in hemizygosity (only single copy)
o (4) Nondisjunction: inappropriate chromosomal segmentation during mitosis may result in loss of an entire chromosome: If resulting daughter cell is triploid, then it will shed the extra chromosome (may result in LOH).
At the chromosomal level, what are the exact mechanisms of loss of heterozygosity?
Loss of heterozygosity (LOH) is defined as the loss of one parent’s contribution to the cell, can be caused by direct deletion, deletion due to unbalanced rearrangements, gene conversion, mitotic recombination, or loss of a chromosome (monsomy).
Which of the following is the most carcinogenic? aflatoxin B1
benzo[a]pyrine
4-aminobiphenyl
2-naphthylamine
o Alfatoxin B1 is produced by Aspergillus sp. and causes HCC with concurrent p53 mutations
o Characteristic G-to-T transversion
What are common mechanisms that occur with hypermethylation of CpG areas within the cell?
• DNA methylation specifies which genes are actively transcribed or silenced
•CpG dinucleotides are abundant within the 5’ ends of some genes, typically including the promoter and first exon (CpG islands)
o DNA methyltransferase (DNMT) attaches methyl group to 5-carbon of cytosines within this sequence results in gene silencing
Choose which of the following statements is true regarding methylation of CpG islands.
- Cancer cells exhibit global hypermethylation, which results in genomic instability.
- Local hypermethylation of gene promoters silences tumor suppressor promoters.
- Oncogenes can be activated through hypomethylation
These statements are true:
- Local hypermethylation of gene promoters silences tumor suppressor promoters.
- Oncogenes can be activated through hypomethylation
This was false and should read as follows:
1. Cancer cells exhibit global hypomethylation, which results in genomic instability.
What are the physical effects of methylation of the CpG islands?
- Physically blocks binding of transcriptional activators to the promoter
- Binds methyl-CpG binding domain proteins (MBDs), which recruit HDACs to form heterochromatin (inactivate, tightly coiled)
T/F: Normally, methylation occurs in multiple sites throughout the genome to block activation/movement of transposable elements to prevent chromosomal instability.
True
Which is the type of covalent adduct that is induced by UV light?
> 60% of pyrimidine dimers are TT
__________________________________________
•UV light damages DNA by forming pyrimidine dimers (covalent bonds between two pyrimidines: TT, CC, or TC)
• 30% are CC
• Very stable, mutagenic compounds
o TT is more able to be repaired by DNA enzymes and therefore only weakly mutagenic
o CC dimers are harder to repair and often result in CC –> TT substitutions during replication
• XP = lack of NER enzymes, sensitivity to UV radiation because of pyrimidine dimers
Which molecular diagnostic tool are used to study loss of heterozygosity or copy neutral LOH (uniparental disomy or gene conversion) whereby one allele or whole chromosome is missing and the other allele is duplicated with pathological consequences?
SNP arrays
The following describes which SNP inheritance pattern in a pedigree chart?
Males affected almost exclusively
Single recessive gene on X chromosome will cause dz
Gene alteration transmitted from mother to son
Affected males cannot transmit to sons
X linked recessive
*see Phoenix for picture examples
The following describes which SNP inheritance pattern in a pedigree chart?
Pattern of transmission of the mutant phenotype from an affected male parent to ALL female offspring, and from an affected heterozygous female to 50% of ALL offspring
X linked dominant
*see Phoenix for picture examples
The following describes which SNP inheritance pattern in a pedigree chart?
an affected individual has one copy of a mutant gene and one normal gene on a pair of autosomal chromosomes
Autosomal dominance
*see Phoenix for picture examples