NBME Review Flashcards

1
Q

what is a telomere?

A

Nucleotides found at the end of chromosomes; contain TTAGGG sequences

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

why is telomerase needed?

A

Lagging strand has no place for RNA primer, so telomerase needed

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

how does telomerase work?

A

Telomerase recognizes telomere sequences and adds them to new DNA with RNA template -> “RNA-dependent DNA polymerase”

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

where is telomerase activity especially important?

A

cells that need controlled indefinite replications (hematopoietic stem cells, epidermis, hair follicles, intestinal mucosa —> esp affected by chemotherapy)

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

general process of base excision repair

A

damaged base removed, phosphate backbone removed, new nucleotide added

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

when does base excision repair happen

A

all phases of cell cycle

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

what is base excision repair for

A

Specific base errors recognized (ie deaminated bases, oxidized bases, open rings)

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

DNA glycosylase

A

removes damaged bases in base excision repair

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

AP endonuclease

A

attacks 5’ end and creates 3’ -OH in base excision repair

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

AP lyase

A

attacks 3’-OH end in base excision repair

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

when is nucleotide excision repair active

A

G1 phase (prior to DNA synthesis)

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

what is nucleotide excision repair for

A

For damage that involves multiple bases

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

What is nucleotide excision repair especially important for

A

repair of pyrimadine dimers caused by UV damage

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

process of nucleotide excision repair

A
  1. Endonucleases remove damaged bases
  2. DNA polymerase adds back new bases
  3. DNA ligase seals it
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

problem leading to xeroderma pigmentosa

A

defective nucleotide excision repair

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

signs and symptoms of xeroderma pigmentosa

A

extreme sensitivity to sun, dry skin, changes in pigmentation, HIGH risk of skin cancer

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

when does mismatch repair (MMR) occur

A

Occurs in S/G2 phase (after DNA synthesis)

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

what is mismatch repair (MMR) for

A

incorrectly placed bases (insertion, deletion, incorrect matches)
*KEY: the base itself is not damaged

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

mechanism is mismatch repair

A

Newly synthesized strand compared to template strand, errors removed, then resealed

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

why is mismatch repair (MMR) important

A

needed for microsatellite stability

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

what can occur if MMR is faulty

A

DNA slippage can occur at microsatellites -> insertions/deletions + possible frameshift

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

HNPCC is a problem with

A

HNPCC = hereditary non-polyposis colorectal cancer = lynch syndrome: germline mutation of MMR enzymes -> microsatellite instability and colon cancer

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

homologous end joining (HEJ)

A

for double stranded DNA damage:

Uses sister chromatids as template

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

Non-homologous end joining (NHEJ)

A

for double stranded DNA damage:
Proteins used to re-join broken ends (DNA pol lambda and mu)
KEY: no template -> highly error prone

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

defected NHEJ can lead to

A

Ataxia telangiectasia: ATM gene on chromosome 11, DNA sensitive to ionizing radiation. CNS, skin, immune system affected. Usually 1st year healthy then slow dev, progressive motor coordination problems. High risk cancer

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

major CFTR trafficking deficit

A

In the ΔF508 mutation, the CFTR protein is still made, just laking the 508th a.a. (Phenylalanine)

It is still a functional protein, but is misfolded in ER which causes ubiquination (rather than transport to golgi) and then degredation by proteasomes

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

what is a dominant negative effect

A

“A mutation whose gene product adversely affects the normal, wild-type gene product within the same cell. Usually occurs if product can still interact with the same elements as the wild-type product, but block some aspect of its functions”

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

ex of dominant negative effect

A

Nonsense mediated decay -> quality control mechanism that eliminates mRNA transcripts that have premature termination codons (PTCs)

Beta-thalassemia depends on NMD pathway; many mutations can alter splicing and/or result in PTC -> triggering mRNA decay and loss of protein

Other mutations are NMD resistant and result in truncated products that act in dominant negative manner

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

point mutation

A

1 base switched for another
Transition=purine to purine or pyrimadine to pyrimadine

Transversion = purine to pyrimadine or vice versa

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

silent mutation

A

nucleotide substitution codes for same aa, often a change in the 3rd position of codon (“wobble”)

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

nonsense mutation

A

early stop codon

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

missense mutation

A

codes for different aa

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

frameshift

A

insertion/deletion that’s not multiple of 3; can cause a early stop codon or loss of stop codon

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

mechanism of retinoblastoma formation

A

Mutation in rb protein, which normally binds to E2F until rb is phosporylated
Phosphorylation of rb by G1-S-CDK releases inhibition

rb regulates cell growth -> “tumor suppressor”

Abnormal rb -> unregulated cell growth via E2F

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

two-hit origin of cancer

A

mutation of tumor suppressor genes
Heterozygous mutation -> no disease

Loss of heterozygosity

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

huntington gene abnormality

A

HTT gene located at 4p16.3; CAG expansion in Exon 1

CAG codes for glutamine (Q) -> PolyQ tract

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

hypothesized cause of expansion in huntington

A

Meiotic instability in sperm -> unequal crossing over

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

huntington protein has high expression in

A

testes and brain

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

function of huntington protein

A

Found in nucleus and cytoplasm and regulates intracellular transport of many proteins, including shuttling TFs in and out of nucleus or sequestering them

Required for normal embryonic dev and neurogenesis

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

CAG expansion in huntington leads to

A

aggregation of mutant protein into inclusion bodies

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

inheritance of huntington

A

AD

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

symptoms in huntington caused by

A

degeneration in basal ganglia (striatum)

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

presentation of huntington

A

Characterized by dementia, chorea, ataxia, and dysarthria

Death usually 10-20 years after diagnosis

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

results of meiosis I

A

“Reductive division” -> diploid to halpoid

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

results of meiosis II

A

Chromatids separate -> 4 daughter cells (haploid)

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

spermatogenesis

A

Begins at puberty
Spermatogonium (2n) -> Mitosis -> 1° spermatocyte (2n) -> meiosis I -> 2° spermatocyte (1n) -> meiosis II -> spermatid (1n) -> spermiogenesis -> spermatozoa

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

oogenesis

A

1° oocytes (2n) formed in utero -> arrested in prophase I

At puberty, 1° oocytes begin completing meiosis I each cycle -> 2° oocytes (1n) and polar bodies

2° oocytes arrested in metaphase II until fertilization

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

meiotic NDJ

A

Failure of chromosomes to separate; most common cause of aneuploidy

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

Meiosis I NDJ

A

homologous chromosomes fail to separate -> games have chromosomes from both parents

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

Meiosis II NDJ

A

sister chromatids fail to separate (ie XXY males)

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

maternal NDJ

A

common cause of trisomy; higher risk because meiosis 1 is so drawn out

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

mitochondrial diseases typically refer to defects in

A

aerobic metabolism (electron transport chain)

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

how many proteins encoded by mtDNA

A

13 polypeptide protein subunits

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

systems most affected by mitochondrial diseases

A

neurologic, muscular, cardiac

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

heteroplasmy

A

Mitochondria have multiple copies of mtDNA

Cells have multiple mitochondria
Heteroplasmy occurs when there is a mixture of normal and abnormal

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

implication of heteroplasm

A

uncertain chance of passing on mitochondrial diseases from mother

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

chimerism

A

2 genomes present in 1 individual -> usually result of fusion of 2 zygotes

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

mosaicism typically occurs

A

as result of a post-fertilization mitotic error

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

somatic mosaicism

A

in the body, usually develops post-conception

ie Congenital hyperpigmentation- male with mental retardation and swirling pigmentation. Diagnosed by chromosome study of skin cells

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

germline mosaicism

A

confined to germ cells. The individual will not have any symptoms, but may have multiple offspring with a mutation frequently thought of as sporadic

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

inheritance of CF (and chromosome)

A

AR, chromosome 7

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

mutations in CFTR gene cause

A

abnormal chloride transport -> thick mucous due to lack of water equilibrium

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

PKU inheritance pattern

A

AR

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

presentation of PKU

A

Normal neonate, dev delay beginning around 3-4 months

65
Q

problem in PKU

A

Phenylalanine hydroxylase (PAH) deficiency -> phenylalanine cannot be converted to tyrosine

66
Q

inheritance of marfan syndrome

A

AD, 25% are de novo

highly penetrant

67
Q

gene and protein in marfan

A

FBN-1 gene -> fibrillin protein

68
Q

associated problems with marfan

A

dilated aortic root
ectopia lentis
skeletal changes
dural ectasia

69
Q

hemophilia A

A

XR

caused by reduced factor VIII -> excessive bleeding

70
Q

Tay-sachs inheritance

A

AR

71
Q

what is tay sachs

A

Neurodegenerative lysosomal storage disorder

72
Q

mutant enzyme in tay-sachs

A

β-Hexosaminidase (A isoenzyme) -> critical role in brain and spinal cord; buildup of fatty substance GM2 ganglioside

73
Q

clinical features in tay-sachs

A

hypotonia, spasticity, seizures, blindness

74
Q

hardy weinberg equations

A

P+Q=1

P^2 +2PQ + Q^2 = 1

75
Q

hardy weiberg assumptions

A
Large population
Random mating
No effect of recurrent mutation
No selection against any phenotype
No migration
Autosomal locus
76
Q

exceptions to hardy weinberg

A

Nonrandom mating: stratification, assortative mating, consanguinity

Small populations: inbreeding, genetic drift, founder effect

77
Q

what is klinefelter syndrome

A

A male with 1 or more extra X chromosomes; 1/1,000 males

78
Q

presentation of klinefelter syndrome

A

Tall with long limbs, small firm testes with hyalinization of seminiferous tubules and azospermia

Gynecomastia: breast cancer risk = women

79
Q

causes of klinefelter (XXY)

A

maternal and paternal NDJ are equal

80
Q

most common trisomy

A

16, but never seen in liveborns

81
Q

mechanism of trisomy

A

NDJ in mother or father results in trisomy (fusion of a 2n gamete with a 1n gamete)

82
Q

trisomy 21, 18, and 13 and XXX most commonly caused by

A

maternal MI NDJ

83
Q

cause of XXY

A

equally caused by maternal and paternal NDJ

84
Q

45,x caused by

A

mainly by paternal NDJ

85
Q

importance of X-chromosome inactivation

A

AKA lyonization

Random from cell to cell which X chromosome will be inactivated (“functional mosaicism”)

Skewed lyonization can result in females having x-linked recessive expression

86
Q

inheritance of achondroplasia

A

AD
complete penetrance
80% de novo

87
Q

genetic abnormality in achondroplasia

A

Mutations in the fibroblast growth factor receptor (FGFR) 3 gene

FGFR3 is negative regulator of bone growth -> mutation activates the gene -> inhibiting bone growth -> gain of function mutation

88
Q

presentation of achondroplasia

A

Short limbed (rhizomelic), macrocephaly, skeletal and CNS complications, normal IQ, clinically and genetically homogeneous

89
Q

complications in achondroplasia

A

Compression of spinal cord and/or upper airway obstruction increaces risk of death in infancy

7-8% of infants die from obstructive or central apnea, which can be due to brain stem compression

90
Q

most common pathogenic variant in achondroplasia

A

G1138A (98%)

91
Q

features of prader willi syndrome

A

Hypthalamic dysfunction -> lack of satiety -> obesity

Hypogonadotropic hypogonadism

Growth hormone deficiency -> short stature and diminished muscle
Cognitive/behavioral impairment

92
Q

cause in prader willi

A

lack of expression of paternal genes at 15q11.13

93
Q

mechanism frequencies in prader willi

A

microdeletion of on paternal 15q11.13 (70%), maternal UPD (25%), imprinting defect on paternal 15q11.13 (5%)

94
Q

key of prader willi in infant

A

hypotonia, feeding problems, cryptorchidism, may be hypopigmented

95
Q

key of prader willi in children

A

obesity, oppositional behaviors, learning problems, short stature

96
Q

key of prader willi in adults

A

type 2 DM, obstructive sleep apnea, hypogonadism

97
Q

fragile x inheritance

A

X-linked dominant

98
Q

genetic abnormality in fragile x

A

Expansion of CGG in 5’ UTR of the FMR-1 gene that encodes FMRP

99
Q

FMRP implicated in

A

dendritic spine maturation, synapse formation, and synaptic plasticity

100
Q

high FMRP expression in

A

brain and testes

101
Q

presentation of fragile x

A

Males have characteristic appearance: large head, long face, prominent forehead and chin, protruding ears

Associated with CT findings -> joint laxity and large testes after puberty, hypotonia

Behavioral abnormalities common

102
Q

associated problems with fragile x

A

Can have mitral valve prolapse, HTN, seizures, strabismus

103
Q

mechanism of genomic imprinting

A

Parent-of-origin difference in gene expression due to epigenetic modification

Usually done by methylation or changes in chromatin structure

Most imprints erased and restored each new generation

104
Q

genetic testing can lead to (counseling)

A

detection of false paternity
Stigmatization - including survivor guilt

Loss of employment or insurance
Psychological harm

105
Q

therapeutic index formula

A

TI=TD50/ED50

106
Q

what is bioavailability (F)

A

the fraction or percent of unchanged drug that reaches systemic circulation from a site of administration

107
Q

calculate bioavailability (F) from graph

A

graph concentration vs. time for 2 methods of administration (ie IV and PO) and compare the areas under their curves

F=(AUCoral/AUCiv)

108
Q

factors that affect absorption of a drug

A

bioavailability and first-pass metabolism

109
Q

factors that determine bioavailability

A

physiology (ie first-pass metabolism), physicochemical (ie drug ionization), and biopharmaceutical (ie table dissolution, particle size)

110
Q

majority of drugs use what mechanism of permeation

A

passive diffusion through cell membrane lipid

111
Q

key relationship in passive diffusion through cell membrane lipid

A

rate of absorption ∝ unionized [drug] at site of admin

112
Q

henderson hasselbach

A

HAH+ + A-
BH+ H+ + B

pKa - pH = log (protonated/unprotonated)

pKa is dissociation constant

pH is the pH of surroundings

When pHpKA, unprotonated forms predominate (A- and B)

113
Q

key relationship in carrier mediated transport (active transport of facilitated diffusion)

A

RATE OF ABSORPTION ∝ DRUG CONC ONLY WHEN CARRIER NOT SATURATED

114
Q

carrier mediated transport can be affected by

A

competitive and noncompetitive inhibition

115
Q

drug distribution

A

only unbound drug can penetrate cell membranes

Many drugs bind to albumin, basic drugs bind to globulins, binding usually reversible, nonselective, and competitive

116
Q

phase 1 metabolism

A

introduce or unmask polar functional group (-OH, -NH2, -SH); if sufficiently polar, will be excreted, if not -> phase II

117
Q

phase II metabolism

A

conjugation and synthetic reaction addition of acid or amino acid ie Glucuronidation

enterohepatic recycling

118
Q

partition coefficient directly proportional to

A

amount that gets absorbed

119
Q

mixed function oxidases (MFOs)

A

involved in phase I metabolism
AKA monooxygenases require reducing agent and molecular oxygen
(NADPH is reducing agent)

120
Q

hepatic sites of metabolism

A

Microsomal: vesicles enriched in ER membranes; contain enzymes catalyzing oxidation reactions and glucorinide conjugation

Non-miccrosomal: primarily in liver

121
Q

2 key microsomal enzymes

A

NADPH-cytochrome P450 recutase

cytochrome P450

122
Q

zero order drug elimination

A

a constant amount eliminated per time

123
Q

first order drug elimination

A

a constant fraction (or percentage) of drug is eliminated per unit of time

dD/dt= -Ke * D

124
Q

oral drug administration

A

(PO)

most convenient; may have significant 1st pass metabolism

125
Q

IV drug administration

A

100% bioavailability; most rapid onset of action

126
Q

IM drug administation

A

may be painful

127
Q

SC drug administrtion

A

smaller volumes than IV; may be painful

128
Q

rectal drug administration

A

less first-pass effect than oral

129
Q

inhalation drug administration

A

often rapid onset of action

130
Q

sublingual drug administration

A

rapid onset; minimal first-pass effect

131
Q

intrathecal drug administration

A

bypass blood-CSF barrier and blood-brain barrier; risks of infection & HA

132
Q

transdermal drug administration

A

slow absorption; longer duration of action; lack of first-pass effect

133
Q

equation for volume of distribution

A

Vd= D/C, where D is amount administered and C is concentration of drug

134
Q

what is volume of distribution

A

(Vd) = volume of fluid that would be needed to contain the administered amount of drug at the concentration measured in plasma

135
Q

calculate dose

A

D=Vd*C

If bioavaliability not 100%, D = (Vd * C)/F

136
Q

calculate drug clearance rate from blood

A

(CL=Vd * Ke)

Note: only unbound (free) drug can be cleared by an organ

137
Q

steady state drug clearance proportional

A

Css ∝ 1/CL

138
Q

first order clearance kinetics

A

Occurs at relatively low substrate conc.

Generally, when V is less than or equal to 10% Vmax

V∝ [D]

139
Q

zero order clearance kinetics

A

Occurs when [D] is relatively high

V = Vmax

140
Q

compare effectiveness of drugs

A

Therapeutic index = TD50 / ED50

Margin of safety = TD1/ED99

141
Q

calculate drug loading dose

A

Loading dose = (Vd * C)/ F

142
Q

calculate maintenance dose

A

Maintenance dose = (CL * Css)/ F

143
Q

how to maintain steady state

A

drug administration = drug elimination = CL * CSS

144
Q

agonist

A

a drug that mimics the effects of the endogenous ligand for a receptor

intrinsic activity >0friedreich

145
Q

antagonist

A

a drug, which does not itself have intrinsic activity, but which interferes with the binding of the endogenous ligand (or an agonist) to a receptor

146
Q

friedreich’s ataxia inheritance

A

AR

147
Q

genetic defect in friedreich’s ataxia

A

GAA repeat in first intron on FXN gene, chromsome 9

148
Q

GAA repeat in friedreich’s ataxia causes

A

transcriptional repression -> less frataxin

149
Q

compound heterozygotes in friedreich’s ataxia

A

4%

expansion on one allele and other mutation of FXN in other allele

150
Q

frataxin protein function

A

removes iron in the cytoplasm and around mitochondria

iron buildup causes free radical damage (oxidative stress) to mitochondrial membrane, esp affecting nerve and muscle cells

151
Q

iron buildup in friedreich’s ataxia causes

A

spinal cord becomes thinner and nerves lose part of their myelin sheath

152
Q

signs and symptoms of friedreich’s ataxia

A

muscle weakness in arms and legs, loss of coordination, vision impairement, hearing impairment, slurred speech, scoliosis, pes cavus, diabetes, hypertrophic cardiomyopathy, afib->tachycardia

153
Q

steroid receptor function

A

Steroid hormones are lipid soluble/intracellular and can cross plasma membrane

Receptors are in the cytoplasm or nucleus

These hormones travel through blood bound to a protein

154
Q

histone structure

A

H2A, H2B, H3 and H4 make up an octamer that DNA wraps around

H1 special histone outside the nucleosome core; larger and more basic; ties “beads on string” together

155
Q

HAT=histone acetyltransferase

A

Acetyl groups can be added to lysine residues on histone -> relaxes chromatin -> transcription

156
Q

HDAC=histone deacetylase

A

If acetyl groups are removed -> condenses chromatin -> blocks transcription

157
Q

insulin receptor signaling pathway

A

Insulin uses receptor tyrosine kinase (RTKs)

Insulin binds -> RTK autophosphorylates -> IRS-1 is phosphorylated -> gene transcription

No second messenger

158
Q

growth factor signaling pathway

A

Growth factors use RTKs

GF binds RTK -> dimerization -> autophosphorylation -> Ras -> Raf -> MEK -> ERK -> TFs

GTP->GDP as ras phosphorylates raf