final exam Flashcards

1
Q

endosymbiont hypothesis + evidences in mitochondria & chloroplasts

A

suggests organelles were once free-living bacteria that were engulfed by ancestral cells

  • overtime formed symbiotic relationship
  • proposed by Lynn Margulis and was controversial at first

evidences, both mitochondria & chloroplasts:
- have their own chromosomes
- divide by fission (similar to bacteria)
- have circular chromosomes
- their genomes look more like bacteria than eukaryotes
- also encode tRNA and ribosomes that look much more like prokaryotes than eukaryotes

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

3 genes that are not part of the main genome

A
  • mitochondria
  • transposons
  • retrotransposons
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

transposons & retrotransposons

A

transposons: “jumping genes,” DNA pieces that can move from one place to another within the main genome, “copy and paste genes”
- importance: can influence function of other genes, sometimes causing mutations or helping w/ evolution by adding new DNA

retrotransposons: type of transposon that move different, instead of jumping around, they copy themselves into RNA and use reverse transcription to insert the copy back into DNA at a new spot
- importance: make up significant part of genome and can create new genetic material, but can also disrupt important genes if they jump into the wrong spot

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

extranuclear inheritance

A

inheritance of genes that are not located in the nucleus

  • primarily uniparental, almost exclusively maternal inheritance (mitochondrial inheritance)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

mitochondrial-inherited diseases

A

diseases caused by mutations in mitochondrial DNA
- passed from mother to children since maternal mitochondria is passed

  • is dose-dependent (depends on how many normal mitochondria are given): ex. 80% defective mitochondria = lethal, whereas <20% means healthy still
  • UK approves 3 parent IVF to prevent mitochondrial disease (basically egg has 2 moms- nuclear and mitochondrial)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Barbara McClintock

A

discovered “jumping genes” (transposons) & won the Nobel peace prize

  • worked with corn plants (maize) and studied the colors of kernels
  • noticed unusual patterns in the kernel colors- like spots or streaks- and wanted to figure out why

experiment:
- saw that some kernels changed color over generations in ways that didnt follow menders laws
- found certain pieces of DNA could move to different locations in the genome

  • ex. gene responsible for kernel color could be “turned off” when jumping gene landed in it, causing patches of different color
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Barbara McClintock’s experiment + findings

A
  • noticed a maize strain that frequently showed chromosome breakage at a specific location on chromosome 9
  • this strain produced kernels with unusual spotting patterns = some were purple, while others were a mix of purple & yellow
  • she identified 2 genetic factors associated with this phenomenon: Ds (dissociation) and Ac (Activator)
  • when she mapped them, Ds always mapped to the break but Ac was in a different spot every time
  • Ds gene WAS the fragile location (it was a nonautonomous transposon, while Ac encodes transposase that can move Ds around)
  • proposed that Ac was located elsewhere in the genome and triggered breaks at the Ds site = purple mostly but has a little bit of yellow
  • when Ac activated movement of Ds in and out of C gene, pigment production was restored= purple spots

C = purple pigment made
c = no pigment, yellow

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

F-Factor Mediated Gene Transfer + Hfr strains

A

process where genetic material is transferred from one bacterium to another through direct contact

  • made possible by the F-factor (plasmid that can exist independently)

Hfr strains: have the F factor integrated into their main chromosome
- during conjugation, can transfer portions of their chromosomal DNA along with the F factor

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

how do transposons move around + transposase

A

they excise themselves from the genome, find a new location, and splice into it

transposase: enzyme encoded by some transposons (like Ac) that can cut and paste transposon into new locations
- DNA sequenced called inverted repeats (IR) is recognized by the transposase enzyme and thats where it cuts and pastes

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

integrase

A

enzyme that inserts viral DNA into host cell’s genome

  • process is crucial for retroviruses like HIV to establish permanent infection

mechanism of retrotransposon integration is very similar to that of retroviruses

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

retrotransposons + example

A

like retroviruses, move by first being transcribed into RNA

  • RNA is then reverse transcribed into DNA, which is integrated into new location in the genome = like sending copies of your genome everywhere, leaving copy behind wherever you go

example: classic white mutation found in TH Morgan’s flies is a result of retrotransposon insertion (retrotransposon went in and broke the red gene = white eyes)

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

2 main types of transposes in the human genome

A

LINEs (long interspersed sequences): can encode transposase and reverse transcriptase, type of retrotransposon, repeated many times in the human genome

SINEs (short interspersed sequences): non autonomous (cant move on their own), require LINE enzymes for movement
- Alu family

significance: create genetic diversity by moving around genome, serve as evolutionary markers to trace genes (transposition in germline), but ARE MUTAGENIC

genes that came from transposon insertion can be detected by a region of altered %GC content

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

somatic transposition

A

transposons insert themselves into non-reproductive cells (somatic cells) leading to mosaics, where different cells within the same organism can have different genetic makeups

  • could be useful (neural progenitor cells have higher levels of LINE transposon activity) or VERY BAD (rett syndrome)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

VDJ recombination

A

process that generates antibody diversity in the immune system

  • involves rearranging gene segments (V, D, and J) in a way that is similar to how transposons move (inverted sequence repeats)

enzymes that do the genomic editing are RAG-1 and RAG-2 that look just like transposons

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

TBXT gene

A

TBXT in all primates has an Alu SINE (transposon) in intron 5 but in tail-less primates, they have a second Alu element in intron 6 (AluY) causing a hairpin structure to form = skipping exon 6 during splicing = heterozygote (short/no tail mice)

  • homozygotes for this are just DEAD (highly variable phenotype)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

introns vs exons

A

introns: non-coding regions of the DNA that are spliced out

exons: coding parts of the DNA that are in the mature mRNA

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

Complete dominance, Co-dominance, & Incomplete Dominance

A

genes can come in multiple alleles, not just a single dominant or recessive

Complete Dominance: occurs when phenotypes of heterozygote and dominant homozygote are identical (Mendel)
- ex. purple flower is dominant so all the kids have purple flowers

Co-dominance: both alleles are expressed equally in the heterozygote
- ex. AB blood group, where both groups are present

Incomplete dominance: heterozygotes display intermediate phenotype b/w the 2 homozygotes
- ex. cross b/w red (CrCr) & white (CwCw) flowers produces pink (CrCw) showing neither is completely dominant

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

hypomorph allele (reduction-of-function) vs null alleles

A

hypomorph allele: mutation with reduced activity (works but not as well as the normal version)

null alleles (Lf- loss of function): mutations that eliminate activity (insertions, deletions, or frameshift usually result in LF alleles)

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

haploinsufficiency + example

A

one one functional copy of the gene is not enough for normal function

  • usually, if one copy of gene is mutated, the other can compensate and everything functions normally but in haploinsufficiency, one working copy isn’t enough

example: cri du chat syndrome (cry of the cat): results from deletion on chromosome 5
- babies have intellectual disabilities, smaller head, and a high-pitched cry due to problems with larynx
- kids usually die early

still have intact copies of the deleted gens on the normal chromosome 5, but the phenotype is NOT the wild type

example: polydactyly (6 fingers) - several transcription factor loss-of-function alleles

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

dominant-negative mutation + example

A

mutation in one copy of a gene produces a defective protein that actively interferes with the function of the normal protein from the other copy

example: p53 tumor suppressor gene is a tetramer (meaning it works as a complex of 4 protein subunits so even 1 single mutant protein causes the entire thing to not work)

different from haploinsuffiency which is when 1 chromosome isn’t enough

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

blood groups & phenotypes

A

both A and B are co-dominant and they are both dominant over O

to be A you need to be: IAIA or IAi

to be B you need to be: IBIB or IBi

IAIB: AB blood type

ii: O blood type (double recessive)

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

trillium clover

A

follows incomplete dominance pattern in flower color

plant produces gradient of phenotypes depending on the levels of enzyme produced by Cr gene

  • single gene w 7 possible alleles results in 22 combinations with diff phenotypes!!

more Cr enzyme = more cyanidin = deeper red

Cw coded enzyme is non-functional so CrCw have half the Cr enzymes as homozygotes for Cr!!

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

F2 ratio in incomplete dominance patterns

A

1:2:1 instead of the classic 3:1 mendelian ratio (neither allele is dominant)

2 is the intermediate color (like pink b/w red and white)

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

pleiotropy + 4 examples

A

pleiotropy: one gene can have multiple phenotypic effects (basically one gene gets deleted and theres a lot of symptoms of the disease)

examples:
- phenylketonuria (PKU): autosomonal recessive disease that leads to brain damage AND light skin, lack of phenylalanine hydroxylate → buildup of phenylalanine → damages neurons
- also less try available → less melanin made → light skin

  • marfan’s syndrome: dominant deletion allele, result: tall stature, curved spine, elongated fingers, heart valve problems
  • sickle cell anemia: heterozygotes for E6V mutation have normal looking cells and are not anemic but are resistant to malaria (two traits - anemia AND resistance to malaria but same gene)
  • lamin A gene: single mis-spliced gene results in at least 4 distinct diseases, depending on the mutation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Complementation test

A

used to determine whether 2 mutations that produce the same phenotype are in the same gene or in different genes

“are the different dumpy strains mutated in the same gene or different genes?”

  • if 2 mutations are in different genes = the 2 non-mutated copies (one from each parent) can “complement” each other to restore the normal phenotype
  • if on same gene → no normal copy to compensate → mutant phenotype remains
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

corn snake skin example + what it demonstrates

A

corn snake skin color is determined by 2 genes:
R locus: orange pigment enzyme (R allele makes enzymes)
B locus: black pigment enzyme (B allele makes enzymes)

“In some cases, multiple genes act independently, and the observed phenotype is the sum of the products, similar to incomplete dominance at a single locus

both R & B = orange + black = red
both r and b = no orange and no black = albino

same with black and orange

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

additive effects of multiple genes

A

in some cases, multiple genes contribute to a single trait, acting independently and additively
- phenotype depends on sum of effects of individual genes, similar to incomplete dominance
- ex. corn snake skin color

different from epistasis which is when one worker paints over the work of another worker while additive effect is they both work together to contribute to wall without interference

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

epistasis + example

A

one gene masks or modifies the effects of another gene on particular phenotype
- often because genes are in the same pathway

example: coat color in Labrador retrievers
- B locus (determines pigment black or brown) = make black pigment from brown pigment
- E locus (determines pigment deposition, color visible or yellow coat) = make brown pigment from yellow pigment

  • when epistasis occurs, observed phenotypic ratios in offspring will deviate from expected mendelian ratios (F2 = 9:4:3)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

when is 9:3:3:1 classical mendelian ratio expected in additive relationships

A

IF the genes R and B in corn snake skin act independently and additively

  • but this assumes no interaction between the genes
  • but in additive inheritance, there is still some interaction because the effects are cumulative so the relationship is more complex (ex. 1:4:6:4:1)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

continuous traits

A

additive effects of multiple genes can also be used to model continuous traits
- in these cases, phenotype is determined by the combined effects of many genes, each with a small effect

example: trait controlled by 3 genes, each w 2 alleles (one for brown and one for white). individuals with more brown have darker phenotype and more white have lighter phenotype

  • result of continuous distribution of phenotypes from very dark to very light
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

MCR1R and TYRP1 (epistasis)

A

TYRP1 (B Locus): encodes enzyme involved in production of brown pigment
- B allele codes functional enzyme (black pigment), while b allele is non-functional (brown pigment)

MCR1R (E Locus): encodes receptor protein that plays crucial role in determining whether any pigment (black or brown) is produced at all
- E allele codes functional receptor (allowing pigment production), while e allele codes non-functional receptor (blocks pigment synthesis regardless of genotype at B locus)

therefore, e allele of MCR1R is epistatic to alleles at TYRP1 (B) locus

so 2 copies of e allele (ee genotype) = no pigment so yellow color irrespective of whether B or b

(labrador retrievers)

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

supressor mutations

A

mutation in another gene that reduces the effect of the first mutation
- often encode proteins that interact with the protein encoded by the first gene

  • ex. gene m mutation so proper complex cannot be formed but mutation in s compensates, permitting complex formation
  • but mutation in s without mutation in m cannot form active complex

suppressors in same pathway: reduce effect of original mutation by acting in the same pathway
- ex. gain of function mutation in suppressor gene can compensate for loss-of-function mutation in original gene, restoring normal phenotype

suppressors in parallel pathways: can also occur in genes that function in parallel pathways
- ex. mutation in gene A leads to mutant phenotype, suppressor mutation in gene B in parallel pathway can compensate and restore normal phenotype

opposite manner of synthetic mutations

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

3 applications of suppressor mutations

A

identifying genes in pathways: mutagenize organisms w/ mutations of interest and screen for suppressor mutations that restore the wild-type phenotype → identify genes w/ suppressor mutations that reveal other genes involved in the same pathway

drug target identification: suppressor screens are powerful ways to identify drug targets for genetic disease
- inhibit drug in parallel pathway → possible to suppress effects of a disease-causing mutation

understanding human disease: individuals found that have strong mendelian genetic disease but they’re very healthy due to unknown suppressor alleles
- investigation into these suppressor alleles could lead to novel therapeutic strategies for treating these diseases

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

synthetic mutants

A

2 mutations, which individually cause no or minor defects, together produce a severe phenotype
- mutant phenotype observed only when both genes are mutated

  • act in manner opposite to the suppressor mutations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

synthetic alleles and suppressor alleles working together in researched

A

Drosophila suppressor and synthetic alleles were used to map signaling pathways used by growth factors and the Ras oncogene (mutated causes cancer) to regulate cell division

C. elegans - both were used to study apoptosis

these pathways are conserved in humans so understanding them provides insight into diseases like cancer

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

phenotypic variability

A

even with identical genomes, individuals can exhibit range of phenotypes due to factors like penetrance, expressivity, conditional alleles, and genetic buffering

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

quantitative traits

A

these traits are determined by the combined effects of multiple genes, each with a small effect

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

what reasons can phenotypic variability come from?

A

sex differences: same genotypes for most genes, but different phenotypes
- ex. voice pitch, behaviors, and other secondary sexual characteristics are heavily sex-influenced

  • variable penetrance
  • variable expressivity
  • environmental effects
  • conditional alleles
  • developmental noise
  • somatic events: things like X activation
  • transcription/translation variability: randomness in gene expression
  • gene expression differences in heterozygotes: cells might express more of one allele than the other
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

3 examples of variable penetrance

A

osteogenesis imperfecta: autosomal dominant disorder, is 100% penetrant (everyone with the mutation exhibits some phenotype), but the severity of the phenotype can vary greatly.
- ranges from blue eyes to severe skeletal abnormalities or anything in between

BRCA 1 - heterozygote mutant women have 50% higher risk of breast cancer

neurodibromatosis: autosomal dominant disorder, exhibits incomplete penetrance (50-80%), meaning not everyone with the mutation develops symptoms
- but even among those who have it variable expressivity - ranges from skin splotches to massive tumors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

variable expressivity vs variable penetrance

A

variable penetrance: not everyone with a particular genotype shows the associated phenotype (some individuals have the trait, others do not)
- ex. BRCA 1- heterozygote women have 50% higher risk of breast cancer, osteogenesis imperfecta, neurofibromatosis

variable expressivity: the extent to which a phenotype is expressed varies among people (all individuals have trait, but different amounts of it)
- ex. deleting exon 6 from mice TBXT results in heterozygote short/no tail mice (all have short tails but some have shorter tails than others)

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

environmental effect in phenotypic variability

A

genetical identical individuals in different environments have different phenotypes
- ex. risk of breast cancer alters by lifestyle
- himalayan rabbits have temp sensitive alleles of melanin producing enzyme (if temp less than 25ºC, extremities turn black while rest of rabbit is white)

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

conditional alleles in phenotypic variability

A

conditional alleles: alleles whose phenotype depends on environment

  • auxotroph mutations are conditional alleles: genetic mutation that causes an organism to lose ability to produce vital nutrient for growth (in bacteria prevent growth in minimal media unless required nutrient is supplied)

ex. phenylketonuria (PKU): leads to brain damage but can be avoided is homozygous recessive individuals avoid eating phenylalanine

ex. (ts) alleles in Drosopholia- ts means temp sensitive, neurotransmission at high temperatures but no effect at lower temps - temp sensitivity makes them useful tools for studying the nervous system

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

development noise in phenotypic variability + paddington’s landscape

A

developmental noise: random fluctuations during development can affect phenotype

examples: inverted sides of face, identical twins with different fingerprints

waddington’s landscape: metaphor for development and phenotypic variability
- ball starts same but hills and valleys so rolling down ends up at different endpoint
- THE HILLS AND VALLEYS: explains things like mosaics (like McClintock’s maize), transcriptional/translational variability

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

X-inactivation in phenotypic variability

A

X-inactivation: process in female mammals where 1 of 2 X chromosome in each cell is randomly inactivated during embryonic development
- ensures that only 1 X chromosome is active per cell, preventing overdose of X-linked gene products

creates Barr bodies (the inactivated X copy)

example is calico coat color in cats, O allele is the inactivated one, so either orange or black fur expressed

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

genetic buffering & heat shock protein 90 (hsp90)

A

genetic buffering: ability of some genes to buffer (“mask”) the effects of mutations in other genes → reduces phenotypic variability

hsp90: highly conserved molecular chaperone that helps proteins fold correctly, stabilizes them, and assists in their proper functioning; some mutations lead to them misfiling and HSP90 rescues them, ensuring they still function properly

  • under stressful conditions (like heat shock) → hsp90 ability reduced → previously masked genetic mutations are exposed → phenotypic changes

ex. FANFA (fanconi anemia - autosomal recessive anemia) protein interacts with hsp90 increasing severity of mild FANCA mutations

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

feedback & GFP expression in phenotypic variability

A

GFP: green fluorescent protein - used as marker to measure gene expression levels
- cells with more GFP glow brighter

TetR repressor: TetR is a protein that can bind to DNA and inhibit transcription of specific genes
- negative feedback loop: when TetR levels are high, suppresses its production and when TetR levels are low → resumes production

feedback reduces phenotypic variability because without feedback, the TetR gene (and GFP indirectly) is expressed freely and levels fluctuate randomly (“noise”)
- but when feedback is there, TetR levels are regulated, stabilizing GFP levels

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

qualitative vs quantitative traits

A

qualitative: either-or (binary traits) like yellow or green

quantitative: composed of multiple underlying distributions
- ex. height, “intelligence,” heart disease, hair loss, diabetes, personality traits

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

GWAS (genome-wide association study) + SNPs

A

GWAS: used to identify genes or genetic variants associated with specific traits or diseases
- involves genotyping thousands of people & comparing their genotype at different locations in the genome
- useful for genes that are variable and have small effects

SNPs (single nucleotide polymorphisms): common genetic variations that can be used as markers (should use markers that don’t have any effect on the gene itself and are put in b/w gene regions that do not bind transcription factors)
- microarrays & PCR are techniques used to genotype SNPs

  • SNPs are crucial for GWAS = ex. if specific SNP is common in diabetes patients than those without, it might point to a gene related to diabetes risk
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

positional cloning

A

method for finding genes associated with a particular trait when the gene’s function is unknown

  • relies on principle that genes located near each other on chromosome tend to be inherited together
  1. identify DNA markers that are known (polymorphic sites) - like SNPs or Microsatellites
  2. narrow down region to pinpoint approximate location of gene on chromosome by finding markers closely linked to the gene
  3. fine tune using markers near that gene
  4. candidate gene analysis

has to do with GWAS I think

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

why are mutations with subtle, quantitative effects harder to map?

A

because they affect protein activity or levels, not whether the protein is made or not

  • hard to identify since individuals are more-or-less OK, but at increased or decreased risk of a disease when averaged over large population
51
Q

whats the difference between QTL and GWAS?

A

QTL mapping and GWAS are related but not the same

QTL mapping:
- uses experimental crosses (breeding plants or animals)
- requires controlled genetic background
- explores large chromosomal regions and focuses on lineage

GWAS:
- uses natural population, like humans, without experimental crosses
- explores SNPs across genome
- focuses on detecting associations at a fine scale, often identifying much smaller regions

52
Q

False Discovery Rate (FDR) in GWAS

A

statistical method in GWAS to control for false positives

false positives: associations that appear significant by chance

  • basically computer finds the optimal p-value based on the data that you have - FDR is data-set dependent

different from Bonferroni’s Correction method which basically lowers the threshold for significance but issue is that you miss true associations (false negatives

53
Q

Bayes Factor (BF)

A

statistical measure used in GWAS to evaluate strength of evidence supporting association between SNP and trait

  • used in Bayesian GWAS to complement or replace traditional p-value thresholds (directly compares likelihood of data under 2 hypothesis of association and no association and provides measure of evidence for each)

whereas p value tests probability of observing data if theres no association

54
Q

effect size in GWAS

A

effect size: “how important is this gene?”
- with large populations, GWAS can detect genes that are statistically significant yet have tiny effect that might not even be biologically meaningful

effect sizes matter!!

55
Q

polygenic score

A

combines effects of multiple alleles to predict an individual’s phenotype for a complex trait

  • once computational model is trained, can predict phenotype of new individuals based on their genotypes
  • not a very accurate prediction, but on average does well
  • reflect genetic predisposition but environmental factors play a big role (ex. height)

R^2 value = 0.11
means 90% of variation due to factors not measured by GWAS (not genetic)

56
Q

why do most mutations have such small effects?

A

because in order to be widespread, needs to be small otherwise everyone dies and the mutation doesnt even exist down the line

57
Q

DNA shuffling

A

mixing & matching pieces of DNA to create new combinations

new DNA molecule might have new or improved traits

  • screen/select for improved variants using cheap high-throughput assay
58
Q

Direct Evolution

A

method scientists use to speed up the natural process of evolution in the lab

  • like giving nature a little push to create something specific

involves creating variations, identifying better variants, repeating process to create highly optimized molecules

  • farmers have been doing this for a long time without even knowing

example in the slides: artificial Diels-Alderase - computer based enzyme that was 10000x better

59
Q

transfection (gene therapy) + tools used

A

process of introducing foreign material (like DNA) into mammalian cells, with goal of cells taking up material and using it to produce proteins or alter their genetic makeup

  • common step in gene therapy, where scientists try to fix or replace faulty genes

tools used:
- lipid nanoparticles: tiny fat-like bubbles that can carry genetic material (DNA or RNA) into cells

-viral vectors: modified viruses used to deliver genes into cells; harmful parts of virus are removed

  • microinjection: directly injecting genetic material into single cell
  • calcium phosphate (Ca3(PO4)2) mediation: forms small particles with DNA that can stick to cell surface and be taken up by the cell
60
Q

Cas9

A

CRISPR-associated protein 9

protein that cuts DNA and is used in genetic engineering and genome editing

61
Q

hematopoietic stem cells

A

blood cells derived from bone marrow

62
Q

base editing + example

A

developed by David Liu

attaches a base deaminase enzyme to deactivated cas9 = way of precisely changing just 1 letter in the entire genome

  • successfully used to inactivate PCSK9 in monkey liver cells (PCSK9 loss of function allele causes lower LDL cholesterol basically hijacking liver)
  • seems to be successful in human trials but businesses don’t wanna fund it because its too much of a quick fix = not a good business model
63
Q

what did the dolly experiment prove?

A

dolly the cloned sheep: used a sheep skin cell to genetically clone a sheep named Dolly

  • proved existence of egg/ES (embryonic stem cells) cell factors that can drive “de-differentiation” (reboot)
64
Q

ES cells

A

embryonic stem cells

65
Q

iPS cells

A

induced pluripotent (can become an cell type in the body) cells

  • generated from differentiated cells by expressing specific transcription factors (*notably KLF4, SOX2, c-myc, Oct-3/4)
66
Q

ethical considerations of genetic engineering + controversial case

A
  • scientific community restricted embryo research to the first 14 days and prohibiting implantation (embryo discarded after 14 days)

controversial case of Jiankui He: edited CCR5 gene in human embryos to confer HIV resistance

67
Q

preimplantation genetic testing (PGT)

A

way to check the genetic health of embryos before they are implanted into the uterus during IVF (in vitro fertilization)

  • embryo screening to test only the healthy embroys and implanting those
68
Q

the cell cycle is highly regulated and driven by what molecules?

A

cyclin proteins

  • are there and then go away during some parts and come back - why they’re called “cycle”
69
Q

CDKs (cyclin-dependent kinases)

A

proteins that act like traffic controllers for the cell cycle

need to partner up with cyclins to become active

once active, CDKs add chemical tags to other proteins (phosphorylate them) to signal the cell to move to the next stage of the cell cycle

different CDKs are active at different times to ensure cell cycle works in the right time and order

70
Q

what phases are the 3 major cell cycle checkpoints found?

A

G1 checkpoint: monitors for DNA damage and sufficient cell size before allowing cell to progress to S phase (DNA replication)

G2 checkpoint: checks for DNA damage and complete DNA replication before cell enters mitosis (M phase)

M checkpoint: ensures that chromosomes are properly attached to the spindle fibers before proceeding to anaphase (sister chromatids are separated)

  • if any of these things are not done, then cell cycle is halted until the damage is fixed. if not fixed, then apoptosis
71
Q

how are CDK’s regulated?

A

1. positive regulation by cyclin protein levels
- when cyclin levels increase, CDKs pair with them & become active
- ensures that CDKs are only active when needed, like during specific stages of the cell cycle

2. inhibitory phosphorylation of the kinase subunit
- even when CDKs are paired with cyclins, can be temporarily turned off by adding chemical tag called phosphate group
- acts like a brake stopping the CDK from working until cell is ready

72
Q

3 external factors that regulate the cell cycle

A

telomere length: cells stop dividing when the telomeres shrink (protective caps at the end of chromosomes)

anchorage dependence: cells needs to be attached to a solid surface to divide

density-dependent inhibition: cells stop dividing when they start to touch other cells

cancer cells hallucinate all of this and think its not happening

73
Q

metastasis & angiogenesis in cancer cells

A

metastasis: get out of membranes by making enzymes and chewing their way out (what makes a tumor malignant)

angiogenesis: feed themselves by secreting growth factors to promote blood vessel development

74
Q

tumor suppressor genes + examples

A

inhibit cell growth

  • once they’re mutated, cells can divide unconditionally = cancer
  • ex. Rb protein that blocks E2F transcription factor required to drive DNA synthesis
  • also p53 (“defender of the genome”)
  • BRCA1 (also tumor suppressor, if it gets mutated then can get breast cancer)
75
Q

tumor suppressor genes vs oncogenes vs proto-oncogenes

A

tumor suppressor: loss of function mutations

oncogenes: gain-of-function mutations
- promote the cell cycle

proto-oncogenes: also promote cell division, but normal version (not yet mutated)
- can become oncogenes

76
Q

growth factors, growth factor receptors, growth factor signaling molecules

what happens when you activate these inappropriately?

A

growth factor: proteins released by certain cells that stimulate other cells to divide

growth factor receptors: required for responding to growth factors

growth factor signaling molecules: transmit signals from growth factor receptors to promote cell division (machinery inside cell that send message to nucleus from receptor)

activated inappropriately = cancer (uncontrolled cell growth)

77
Q

peto’s paradox

A

highlights observation that larger animals w/ more cells do not get cancer more frequently

explained by:
- increased apoptosis in elephant cells
- multiple copies of p53 tumor suppressor gene in elephants

78
Q

2 proto-oncogenes + their mechanisms

A

Ras: “molecular switch” in signal transduction pathway that sends signals to cells that growth factor is present
- single amino acid change in Ras protein can cause stimulation of pathway, even in the absence of growth factors = cells hallucinate growth factor = become cancerous

abL: kinase that mediates growth factor signals
- translocation outs abL under control of the bcr promoter = over expression of gene = driving cell division and contributing to chronic myelogenous leukemia (CML)

bcL: protein that prevents apoptosis
- translocations can lead to bcL over expression = prevent cells from undergoing apoptosis when they should = B-cell lymphoma

79
Q

4 genetic changes that can turn proton-oncogenes into oncogenes

A

translocation: gene moved to new locus, under new controls
- new fusion gene created that can drive uncontrolled cell growth

point mutation within the gene: random change in DNA sequence when a base pair is added, deleted, or substituted
- hyperactive or degredation-resistant protein

gene amplification: normal growth-stimulating protein in excess

point mutation in a control element: normal growth-stimulating protein in excess

80
Q

“two-hit” model for tumor suppressors + multi hit model

A
  • explains how mutations in tumor suppressor proteins can lead to cancer

hets (who inherit one copy of tumor suppressor gene) are more susceptible to cancer b/c they require only one additional mutation in remaining copy to lose tumor suppressor gene’s function

  • the longer you live = the more cell divisions you have = greater chance of cancer

multi-hit model: cancer development is a multi-step process, multiple mutations accumulated over periods of many years

81
Q

multi step development of cancer is similar to what?

A

stepwise evolution of antibiotic resistance

82
Q

retrovirus relation to cancer

A

retroviruses often carry oncogenes

  • good for virus if the host cell just keeps on making copies

ex. Rous’s sarcoma virus & cervical cancer caused by HPV virus

83
Q

myeloid white blood cells vs white blood cells (B and T lymphocytes)

A

myeloid white blood cells: innate immune system, pre-programmed in all individuals to engulf or destroy pathogens
- phagocytes and macrophages

lymphoid white blood cells: part of the adaptive immune system which learns and adapts to pathogens to reduce change of second infection from the same pathogen
- B cells and T cells

84
Q

B cells vs T cells

A

B cells: produce antibody molecules with exquisite specificity and affinity for pathogens

T cells: can recognize and kill virus infected cells to prevent the spread of infection

85
Q

parts of antibodies

A

consist of 2 heavy chain and 2 light chain proteins covalently linked by disulfide bonds

Fab: variable region that binds to a specific antigen

Fc: constant region; binds cells and molecules of the immune system

86
Q

phagocytes & macrophages

A

terms are usually used interchangeably but there are some differences

phagocytes: more general term for any cell that can engulf and destroy foreign particles, including pathogens.

macrophages: specific type of phagocyte that are larger and more long-lived
- also more specialized in their function and play a role in both the innate and adaptive immune responses

87
Q

3 ways that antibodies interact with antigens

A

agglutination: antibodies can clump together multiple antigens, making it easier for phagocytes to clear them.

precipitation: antibodies can bind to soluble antigens and cause them to precipitate out of solution.

complement fixation: antibodies can activate the complement system, which is a group of proteins that can lyse bacterial cells (basically drill into them and pop them from the inside)

88
Q

Fab region + CDRs

A

binds to the specific antigen (like lock and key)

determines:
- specificity: Fab region determines which specific antigen the antibody will bind to
- affinity: Fab region also determines how strongly the antibody will bind to the antigen (higher affinity means tighter bond)

CDRs (complementary-determining regions): hypervariable loops within the antibody’s Fab region that directly contact the antigen
- contribute significantly to the antibody’s ability to bind a specific antigen with high affinity

89
Q

VDJ recombination

A

VDJ recombination: mechanism for generating antibody diversity (crucial outcome is formation of B-cell receptors)

  • antibody genes in the genome are made of multiple V (variable), D (diversity), and j (joining), and constant region exon modules - VDJ shuffles these regions
  • molecules are edited by recombination at the DNA LEVEL
  • this process occurs in B cells during their development in the bone marrow
  • results in unique combination of V, D, and J segments in mature antibody gene, leading to vast array of possible antibody specificities
  • heavy chain and light chain rearrangements
90
Q

allelic exclusion

A

allelic exclusion: mechanism for ensuring that each B cell produces only one type of antibody

  • each B cell has 2 alleles for each antibody chain (maternal and paternal) just like other genes
  • however, only one allele for each chain undergoes rearrangement
  • ensures that each B cell produces only a single type of antibody with a unique specificity
91
Q

RAG-1 and RAG-2

A

enzymes that do the genomic editing in VDJ recombination - essential for development of B and T cells

also are similar to transposons!!

  • phylogenetic analysis suggests that our porto-vertebrate ancestor acquired these enzymes through horizontal gene transfer (transfer of genes to unrelated organisms)
92
Q

B cell journey from birth to right before maturation

A
  • originate from hematopoietic stem cells in the bone marrow

begin as Pro B cell → RAG-mediated heavy chain rearrangement → Pre B cell → light chain rearrangement → Immature B-cell

  • throughout this process, VDJ recombination is taking place → unique blueprint for the antibody the B cell will eventually produce
  • most importance outcomes: B cell receptor (BCR): this is what determines which cells will be killed using negative selection and which ones will proceed to the spleen for further maturation
  • if they recognize self-antigen → apoptosis

all happening in the bone marrow

93
Q

B cell Maturation in the Spleen (3 things)

A

affinity maturation: B cells encounter foreign antigens and undergo affinity maturation → process that enhances binding strength between BCR and antigen
- involves introducing mutations in the antibody genes (using AID - cytidine deaminase), particularly in the CDRs, the regions that directly contact the antigen
* like fine-tuning the lock to perfectly match the key*

ways of affinity maturation:
Somatic Hypermutation: involves introducing additional mutations in the antibody genes, further increasing the diversity and affinity of antibodies

Class Switching: Initially, B cells produce IgM antibodies (first line of defense)
- However, can switch to producing other antibody classes like IgG (most prevalent in bloodstream)
- IgA (mucosal)
- IgE (Fc stimulates allergic response)
- class switching depends on the type of immune response required.

94
Q

how are the mutations made during B cell maturation?

A

somatic hypermutation: mutations are introduced in the antibody genes (specifically in the VDJ region)
- process is triggered by activation of the B cell receptor (BCR)

activation-induced cytidine deaminase (AID): key enzyme responsible for generating these mutations
steps:
1. AID binds to RNA poly II (enzyme responsible for transcribing DNA into RNA)
2. as RNA poly II transcribes the antibody gene, AID hitches a ride and introduces mutations in the DNA sequence
3. AID’s primary action is deamination (removing amino group from a molecule) - AID specifically dominates C to U → mismatch → body tries to fix it but it causes mutations
4. mutations lead to more affinity for antigen in the mutated antibodies

95
Q

how are antibodies used as a powerful diagnostic tools?

A

because of antibodies remarkable specificity and affinity, they are powerful tools in diagnostics and therapeutics

ELISA (enzyme-linked immunosorbent assay): widely used, uses antibodies to detect presence of specific proteins or antigens in a sample
- relies on ability of antibodies to bind tightly to their target antigens

their high affinity allows for detection even at low temperatures, enhancing sensitivity of diagnostic tests

96
Q

how are antibodies used as therapeutic agents?

A
97
Q

what are monoclonal antibodies and why are they important?

A

monoclonal antibodies (mAbs): antibodies produced from a single clone of B cells → results in population of antibodies that bind to the same epitope of the same antigen

  • different from polyclonal antibodies: mix of different antibodies that recognize various epitopes on the antigen

importance:
- target a single, well-defined epitope → minimizes cross reactivity w other molecules & potential side effects
- therapeutic potential: can block receptors (bind to receptors on surface of cell and prevent them from interacting with ligands →stop cell division), can flag cells for destruction (bind to target cells and mark them for destruction), can neutralize pathogens

  • any medicine using mab’s name ends with mab!!
98
Q

how are monoclonal antibodies produced (mab)?

A

hydridoma technology: traditional method, fuses antibody-producing B cell with immortalized myeloma cells → hybridomas can be grown in culture & produce large quantities of mAbs

phage display: more recent technology, involves creating libraries of Fab genes displayed on bacteriophages, phages are then screened against target antigen to identify those carrying the desired antibody

99
Q

MHCs vs HLAs

A

MHCs: found in all vertebrates

HLA (human leukocyte antigens): human version of MHCs (only specific to humans)
- term “HLA” was first coined b/c these molecules were first identified on white blood cells (leukocytes) and were recognized for their role in tissue rejection during transplantation

100
Q

2 main classes of MHC molecules and what they do

A

MHC Class I: main function is to present endogenous antigens (typically derived from intracellular pathogens like viruses) to CD8+ killer T cells → kills infected cell
- expressed on surface of all nucleated cells in the body

MHC Class II: main function is to present exogenous antigens (typically derived from extracellular pathogens or foreign substances that have been engulfed by APCs) to CD4+ helper T-cells → T cells secretes cytokines→ stimulate B cells to produce antibodies and activate other immune cells
- primarily expressed on surface of specialized immune cells called antigen presenting cells (APCs) like macrophages, dendritic cells, and B cells

101
Q

TCR (T-cell receptors) and their roles

  • CD8+ and CD4+ T cells
A

protein complexes found on surface of T cells that recognize and bind to specific MHC complexes

  • each T cell expresses a unique TCR that can recognize a specific combination of MHC molecule & peptide antigen

have dual specificity: must recognize both MHC molecule & the peptide antigen being presented
- ensures they only respond to infected cells and not to healthy cells displaying self-antigens

structually similar to antibodies and also generated through VDJ recombination

102
Q

3 key features of MHCs

A

polymorphism: highly polymorphic → many different alleles (variants) within population
- high degree of polymorphism ensures that different individuals present wide array of peptides → more ability to respond to diverse pathogens

co-dominance: both MHC alleles are inherited from each are expressed equally→increases diversity even more b/c each individual can express up to 6 MHC I molecules and up to 12 MHC II molecules

role in transplantation: major targets of immune system during organ transplantation

linkage and haplotypes: MHC genes are located close to each other on chromosome 6 → often inhertied together as a unit called haplotype → more possiblities of MHC genes

103
Q

positive & negative selection that T cells undergo

A

occurs in the thymus

negative selection: T cells that recognize self-antigens are killed

positive selection: makes sure T cells that can bind to MHC molecules

104
Q

tasmanian devil facial tumor disease (DFTD)

A

contagious cancer that affects Tasmanian devils, spread through bites and causes tumors on face and neck

  • cancer cells evade immune system bc they express MHC molecules similar to host → difficult for immune system to recognize them as foreign
105
Q

PD-1 receptor

A

protein found on surface of T cells

when binds to PD-L1 ligand (expressed on tumor cells) → inhibits T-cell’s activity to kill the tumor cell → mechanism used by cancer cells to evade immune system

Anti-PD-L1 and anti-PD-1 antibodies are anti-cancer drugs (they block interaction between the PD-L1 ligand and the PD-1 receptor)

106
Q

T cells & killing cancer cells

A

T-cells can kill cancer cells by recognizing antigens presented on MHC I molecules on surface of cancer cells, however they can fail sometimes because of:

  • negative selection: T cells that recognize self-antigens were eliminated during T-cell development & cancer cells often present self-antigens
  • low antigen expression: some cancer cells express tumor antigens at very low levels → difficult for T-cells to detect them
  • immune evasion mechanism: cancer cells can evolve mechanisms to evade the immune system, expressing PD-L1 to inhibit T-cell activation
107
Q

2 main strategies for immunotherapy

A
  • engineer T-cells to recognize specific cell types
  • engineer the dendritic cells that activate T-cells
108
Q

CAR-T

A

type of immunotherapy that involves engineering T cells to express chimeric antigen receptors (CARs)

CARs: antibody that binds a cell-surface receptor expressed on tumor cells to a T-cell activating receptor

  • gene encoding CAR is inserted into T cells harvested from patient and engineered T cells are re-injected back into the patient
  • these engineered T cells can now recognize tumor cells via the CAR-T receptor and kill them
  • was first used on Emily Whitehead (12 year old leukemia patient) at age 6 who has been cancer free for 5 years now*
109
Q

coronavirus structure and function

A

coronaviruses: enveloped viruses with crown-like appearances

  • possess single stranded RNA genome (ssRNA) that can be directly translated into proteins
  • key structural components: spike protein, nucleocapsid, membrane, and the envelope

spike protein is crucial for viral entry by binding to the host cell’s ACE2 receptor

110
Q

how is the presence of COVID-19 detected?

A

using RT-qPCR

111
Q

polyprotein ORF1ab in coronaviruses

A

large protein encoded by the coronavirus genome that is essential in the coronavirus life cycle

  • poly protein is one single lone polypeptide chain but is cleaved into 16 smaller proteins called non-structural proteins (NSPs)

key NSPs:
- NSP3 and NSP5: act as proteases, enzymes that break down other proteins
- responsible for cleaning the poly protein into individual NSPs

  • RNA directed RNA polymerase: enzyme crucial for replicating virus’s RNA genome
112
Q

TMPRSS2 protease

A

protease enzyme that breaks down proteins, for entry, SARS-Cov-2 needs to be cleaved by this protease

interestingly, omicron variant bypasses this need = why it can efficiently infect cells in the upper respiratory tract (making it more widespread but less deadly)

113
Q

whats inside of a vaccine

A

antigens in vaccines are packaged with preservatives, stabilizers, and immune stimulants (adjuvants)

114
Q

traditional vaccines + drawback

A

use weekend or dead pathogens to elicit immune response

  • major challenge: need to grow lots of virus and kill it or inactivate it without losing immunogenicity AND without harming people
115
Q

vaccines using parts of the virus: protein vaccines, viral vector vaccines, and mRNA vaccines

A

protein vaccines: directly inject recombinant viral proteins to trigger antibody production
- since proteins do not invade cells, may not get a strong T-cell response

viral vector vaccines: put SARS-Cov-2 spike protein gene into crippled adenovirus genome
- stimulates both antibody AND strong T-cell responses

mRNA vaccines: deliver synthetic mRNA encoding the spike protein, prompting cells to produce the antigen and trigger an immune response
- considered safer and faster than traditional approaches.

116
Q

mRNA vaccine challenges + breakthrough

A

RNA is fragile AF!

fix by Dr. Katalin Kariko: to use modified RNA where the U base is modified → RNA translates correctly but is resistant to degradation

117
Q

optimizing spike protein for vaccine efficacy

A

researchers discovered that the spike protein exists in different conformations (shapes)

-stabilizing spike protein in pre-fusion conformation, using mutations like S-2P → elicit antibodies to closed spike → prevent binding to ACE2 → neutralize virus

118
Q

vaccine escape

A

pathogen can evade antibodies by mutating the binding site → even one amino acid change can prevent antibody binding (b/c antibodies have very high specificity)

  • acquire mutations near ACE2 binding site so reduce binding to neutralizing antibody, but maintain binding to ACE2 receptor
  • examples are the emergence of variants like Alpha, Gamma, and Omicron
119
Q

convergent evolution example in covid virus evolution

A

independent emergence of similar mutations in different viral lineages, such as the E484K mutation

  • N501Y and K417N bind ACE2 even better than wild-type!

highlights the selective pressure exerted by the immune system and the virus’s ability to adapt

120
Q

how was Omicron able to escape vaccines?

A

it possessed numerous mutations in its spike protein, esp in the region that binds to the ACE2 receptor

  • allowed it to evade neutralizing antibodies

also had altered cellular tropism: infected cells in the upper respiratory tract

121
Q

T-cell responses in Omicron infections

A

neutralizing antibodies primarily block viral entry, but T-cells eliminate infected cells (control spread of virus & prevent severe illness)

omicron retained most of the classic spike protein sequence: meant that T-cells that had memory before (from vaccination) could still recognize and target Omicron-infected cells

122
Q

what do non-neutralizing antibodies do? and how is it different from neutralizing antibodies?

A

neutralizing antibodies: directly block viral entry into cells

non-neutralizing antibodies: agglutination & precipitation
- can bind to multiple virus particles, causing them to clump together = makes it easier for phagocyte white blood cells to clear the virus from the body (agglutination)
- can also cross-link viral proteins, forming large insoluble complexes that precipitate out of solution

123
Q
A