exam 1 review Flashcards

1
Q

relevant features of the infant/toddler exam:

A

delayed growth
development
abuse/neglect
Immunizations

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

relevant features of the child exam (3-12 years):

A

visual/hearing deficits
accidents
abuse/neglect
immunizations

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

relevant features of female youth (3-12 years) exam:

A

rubella immunization, contraception, STDs, substance abuse

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

relevant features of the male youth (13-24 years) exam:

A

MVA, STDs, contraception, substance abuse

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

relevant features of the adult exam (25-44 years):

A

Female: CA, BP, substance abuse, eating disorders
Males : MVA, BP, cholesterol, DM, substance abuse, family violence

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

relevant features of middle aged exams (45-54 years):

A

Females: osteoporosis, DM; lung, colon, skin, and breast CA
Males: prostate/lung/skin/colon CA, ischemic heart, DM

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

relevant features of senior citizen exams:

A

Elder abuse, falls, drug related interactions, nutrition, CA

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

which vaccine is given to newborns immediately following birth? (AKA first vaccine received)

A

Hep B

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

In addition to Hep B, which other vaccines are received starting 1-2 months post-birth?

A

RV, DTaP, Hib, PCV13, IPV, yearly influenza (starting at 6 months)

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

key milestone at 6 months:

A

begins to sit without support

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

key milestone at 12 months:

A

says “mama” and “dada”

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

key milestone at 18 months:

A

begins to walk around towards the end of 18 months

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

minors who are allowed to consent to services on their own, without parental consent:

A

Minor who has graduated from HS
Minor who has been pregnant
Minor who has been married
Minor who has received court’s decree of emancipation

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

what are some treatments/diagnostic tests that minors can receive without parental consent:

A

Can provide informed consent for diagnostic tests and treatments related to a “reportable disease”:

Contraception, pregnancy, and STD testing
Health services to treat pregnancy, STDs, and prenatal care, “not including abortions”
Inpatient mental health treatment (voluntary consent, 14+ years)
Outpatient mental health (any minor over 14 years)
Substance abuse treatment
Emergency situations

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

How do USPSTF grade guidelines?

A

Grade A through Grade I

Grade A - recommends the service as there is a high certainty that the net benefit is substantial. Suggest to provide the service.

vs.

Grade I - concludes that current evidence is sufficient to asses the balance of benefits and harms, evidence is lacking, is of poor quality, or is conflicting. Balance of benefits and harms cannot be determined. Suggestion is that if the service is offered, patients should know about the uncertainty.

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

explain basic concepts of XR imaging:

A

XRays shot towards the patient
There is either absorption, partial absorption, scatter, or penetration
XRs that pass through the patient and penetrate the detector film, create an image

Few pass through - white
Many pass through - black

Differential tissue absorption based on:
Atomic weight (density)
Tissue depth (thickness)

2D representation of 3D structures - superimposition

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

explain the concept of CT imaging:

A

XRs used but in narrow slices
Generator and detector rotate completely around the patient
Computer reconstructs data to create a 2D image slice
Scan = series of multiple slices
Cross sectional anatomy

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

explain the concept of US imaging:

A

uses sound waves, based on the density of the tissue

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

explain the concept of MRI imaging:

A

magnetic and radiographic waves, non-ionizing radiation

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

explain the concept of nuclear medicine:

A

injects radioactive substance into the body to visualize function (PHYSIOLOGY), uses ionizing radiation

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

what does radiopaque/radiodensity mean:

A

few XRs pass through - appears white - metal

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

what does radiolucent mean?

A

many XRs pass through - appears black - air

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

Categorize different tissues from least to most opaque on x-ray including: air, fat, soft tissue, bone, and metal.

A

(least opaque) Air - fat - soft tissue (solid organs and fluid) - bone - metal (most opaque)

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

define hypodense/hypoattenuating:

A

CT terminology
appears dark
e.g. air

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

define isodense/isoattentuating:

A

CT terminology
two structures appear the same

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

define hyperdense/hyperattenuating:

A

CT terminology
appears bright
i.e. metal

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

what imaging modalities use ionizing radiation?

A

CT, XR, nuclear med

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

define “screening”:

A

presumptive identification of previously unrecognized condition

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

how are screenings conducted?

A

Conducted via procedures, exams, tests
Preliminary in design
Confirmation of diagnosis required (additional tests, exams, evaluations)

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

define sensitivity:

A

Correctly identify as diseased as those who actually have the disease
Sn=1-false negative rate
Sn= (true positive)/(true positive)+(false negative)
SNOUT - high sensitivity to rule out
If it is 100% sensitive, all negatives are true negatives

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

define specificity:

A

Correctly identify as non-diseased as those actually without the disease
Sp=1-false positive rate
Specificity=(true negative)/(true negative)+(false positive)
SPIN - high specificity to rule in
If it is 100% specific, positives are true positives

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

define positive predictive value:

A

Proportion of true positive of all those testing positive
PPV (+) = (true positive)/(true positive)+(false positive)

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

define negative predictive value:

A

Proportion of true negative of all those testing negative
NPV (-) = (true negative)/(true negative)+(false negative)

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

how does prevalence affect PPV, sensitivity, NPV, and specificity?

A

As prevalence increases, PPV and sensitivity increase (more common in population, more likely to be a true positive)
As prevalence increases, NPV and specificity decrease (more common in population, less likely to be a true negative)

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

what is a dietary reference intake (DRI)?

A

estimates of the amounts of nutrients required to prevent deficiencies and maintain optimal health and growth; expand the recommended daily allowances and establish upper limits on the consumption of some nutrients. Focuses on lifelong health - goes beyond just dietary deficiencies

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

what is an EAR?

A

EAR - estimated average requirement
Average daily nutrient requirement estimated to meet the requirement of ½ of the healthy individuals in a particular life stage and gender group
Intakes below the EAR need to be improved because the probability of inadequacy is 50%

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

what is an RDA?

A

RDA - recommended dietary allowance
Average daily nutrient intake level sufficient to meet the requirements of NEARLY ALL individuals in a particular life stage and gender group
Intakes between the EAR and RDA likely need improvement because the probability of adequacy is <98%
Intakes at or above the RDA can be considered adequate

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

what is an adequate intake (AI)?

A

Set if sufficient scientific evidence is not available to calculate an EAR or RDA
Based on nutrient intake by a group of apparently healthy people
Intakes above AI can be considered adequate

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

what is the tolerable upper intake level?

A

Highest average daily nutrient intake level that is likely to pose no risk or adverse health effects to almost all individuals of the general population
Intakes between the UL and RDA can be considered to have no risk for adverse effects

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

what is an estimated energy requirement (EER)?

A

amount of food energy (calories) needed to balance energy expenditure to maintain body size, body composition, and a level of necessary and desirable physical activity consistent with long term good health

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

what is the EER for women?

A

2100 kcals

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

what is the EER for men?

A

2900 kcals

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

what are the 3 main energy needs in humans?

A

resting metabolic rate (RMR) - 60%
thermic effect of food - 10%
physical activity - 30%

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

what are some factors that can affect resting metabolic rate (RMR)?

A

-growth
-hospitalization during recovery from serious injuries such as infection or burns
-pregnancy
-lactation

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

what is BMI?

A

body mass index
used as an index of an ideal body weight
may not be appropriate for muscular individuals

BMI= weight (kg)/height squared (m2)

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

BMI and body weight ranges for adults:

A

BMI <18.5: underweight
BMI 18.5 to <25: healthy
BMI 25 to <30: overweight
BMI 30 or higher: obesity

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

describe MyPlate:

A

MyPlate: designed by the US Department of Agriculture to graphically illustrate its recommendations as to what food groups and how much of each should be consumed daily

5 food groups: vegetables, grains, protein, fruit, and dairy

number of servings depends on age and sex

cons: does not include fats, people need to have access to the graphic

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

what were the changes made to the nutrition facts label on July 1, 2021?

A

Servings/calories noted with larger, bolder type
Added sugars, vitamin D, and potassium are included
Vitamins A and C, total fat, and calories from fat removed
Serving size adjusted to reflect amounts people are now consuming
Updated daily values
Proposed addition/removal of certain micronutrients is based on newer data on the risk for underingestion

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

summarize the key steps during pre-initiation of eukaryotic DNA replication:

A

TIME SETTING: in the final phases of mitosis

  1. ORC (origin replication complex) recognizes eukaryotic origins of replication/identifies epigenetic modifications; works in conjunction with HBO1
  2. Cdt1 and Cdc6 (origin licensing factors) use ATP to load the replicative helicase complex (MCM) onto the DNA next to the origin
  3. phosphorylation via DDK –> DDK dependent complex (G1 phase - pre-replicative complex)
  4. polymerase E added —> helicase activated (CMG complex/pre-initiation complex/S phase)
  5. polymerase alpha and delta added —> complete replisome/ready for DNA replication
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

how does the MCM helicase complex “melt” into DNA?

A

hydrolyzes ATP to hydrolyze DNA

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

when do the pre-replicative and pre-initiation processes take place?

A

pre-replicative - G1 phase
pre-initiation - S phase

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

compare and contrast the pre-replicative and pre-initiation phases?

A

pre-replicative:
DDK is phosphorylating, allowing other proteins to bind
occurs in G1 phase

pre-initiation:
polymerase E added and activates helicase (CMG complex)
followed by polymerase alpha and delta
complete replisome is formed
occurs in S phase

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

what is the function of polymerase alpha?

A

-reads the template strand and synthesizes RNA primer from 5’ to 3’ on the complementary strand
-the primer contains a free 3’ -OH end upon which DNA polymerase can add nucleotide monomers

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

what is the function of replication protein A?

A

RPA

prevents re-annealing

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

in what direction does DNA replication occur?

A

nucleotides are added in the 5’ to 3’ direction

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

what is the function of polymerase E?

A

reads parental strand and synthesizes the complementary strand in a CONTINUOUS fashion

lEading - Epsilon

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

what is the function of polymerase delta?

A

reads parental strand and synthesizes the complementary strand in a DISCONTINUOUS fashion
therefore, needs numerous primers and numerous polymerases
Okazaki fragments and DNA ligase

deltA - lAgging

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

what is proliferating cell nuclear antigen (PCNA)?

A

fits around DNA primer and template strand
DNA polymerase attaches to it
acts as a progressivity factor and increases speed

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

what is the function of Fen 1?

A

flap endonuclease that removes DNA overhang (DNA sequences that overlap with the primer)

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

what is the function of RNAse H1?

A

removes RNA primer fragments

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

what is involved in DNA replication termination?

A

-destruction of the MCM (CMG) complex
-activity of topoisomerase IIA
-activity of Holiday Junction Resolvase (helps merge the replication bubbles together)

replication forks/bubbles merge as they expand
form larger replicons until the chromosome is replicated

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

what is the function of topoisomerases?

A

superhelical tension is created by the helicase, in front of the replication fork
may prevent replication from occuring further
tension relieved by topoisomerases

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

contrast activities of Type 1A, 1B, and IIA topoisomerases:

A

1A: single DNA strand is nicked and then the opposite strand is passed through to relieve superhelical tension, happens fast

1B: introduces a single strand nick and the strand is rotated around, happens fast

IIA: introduces a double strand break, using homodimeric enzymes in eukaryotes (the things holding the spaghetti), slower process, USED IN DNA REPLICATION

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

what is common between all topoisomerase types?

A

break DNA via a transesterification reaction using an active site tyrosine as a nucleophile to attack the sugar phosphate backbone

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

explain the “telomere end replication problem”

A

-eventually, run out of space to put primers at the 3’ end of the lagging strand
-some DNA will be lost
-cells undergoing rapid synthesis/growth will lose DNA especially
-overall loss of telomere length

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

what is the telomere repeat?

A

(TTAGGG)n

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

what is the result of the telomere end replication problem?

A

loss of telomere sequences leads to cell senescence
AKA permanent shit down of the cell cycle within a cell
prevents CA (turning off replication)

**cell senescence is linked to premature aging - secretes factors that cause stem cells to age prematurely

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

what is the function of telomerase?

A

replicates single stranded telomere DNA

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

what are the 2 main subunits of telomerase?

A

nucleoprotein complex of multiple protein subunits combined with an RNA molecule.

TERT: RNA dependent DNA polymerase
TERC: RNA molecule that serves as a template to synthesize the telomeric sequence repeats

“teRt Reads”

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

when is telomerase known to be activated during?

A

embryogenesis (fetal tissue) and in stem cells

research says during other times too

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

describe the telomerase process:

A
  1. telomerase binds to the 3’ end of the telomere sequence, along with an RNA template
  2. telomerase catalyzes the addition of bases, restoring the telomere length
  3. DNA polymerase extends and seals the DNA strands
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

effects of genotoxic agents:

A

XRs, oxygen radicals, alkylating agents, spontaneous reactions
UV light, polycyclic aromatic hydrocarbons
Replication errors

etc.

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

what are the cellular consequences of exposure to genotoxic agents?

A

-transient cell cycle arrest
-inhibition of transcription, replication, and chromosome segregation
-apoptosis
-senescence (cell cycle arrest)
-mutations or chromosome aberrations (cancer, aging, inborn disease)

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

what is genomic instability?

A

-a process by which the cell accumulates mutations or chromosome damage
-DNA repair mechanisms may not be able to cope with or recognize the changes

may lead to:
-replication fork stalling
-loss of polymerase proofreading ability
-defects in DNA repair mechanisms
-translesion DNA synthesis over DNA damage
-reduced dNTPs selectivity

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

how does DNA polymerase proofreading work?

A

-3’ to 5’ exonucleotide ability
-removes incorrect bases
-polymerase “chews back” and fixes the 3’ end incorrect base

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

describe the Mismatch Repair (MMR) system:

A
  1. recognition by Msh2 and Msh6
  2. recruitment of endonuclease (Mlh1, Pms2) complex
  3. Exo1 exonuclease removes the mismatched base
  4. DNA synthesis (polymerase E or D) and ligation (DNA ligase)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

what is Lynch Syndrome? What causes it?

A

genetic disorder characterized by early onset non-polyposis colorectal CA

caused by inherited mutations that effect Mismatch Repair System (MMR)

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

what are microsatellite DNA?

A

regions of small repetitive DNA stretches (2-6 bases)
tandemly repeated multiple times
also known as “short tandem repeats” (STR)
due to their repetitive structure, they are prone to strand misalignment during DNA replication - INDELS (insertions and deletions)

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

which repair system recognizes INDELS?

A

Mismatch Repair (MMR)
prevents micro-satellite instability that leads to triplet expansion genetic disorders: Fragile X and Huntington’s Chorea

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

compare and contrast DNA proofreading with MMR:

A

DNA proofreading can chew back on one base at a time, whereas MMR can recognize micro-satellite regions and correct INDELS (numerous base pairs)

both utilize exonuclease activity
DNA proofreading doesn’t use endonucleases? (DNA polymerase is what chews back on it)

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

explain how the NER pathway is multirole:

A

can remove both UV induced DNA damage (cyclobutane pyrimidine dimers, CPD) and bulky adducts cross linked to DNA

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

what is the nucleotide excision repair (NER) pathway:

A
  1. recognition (Centrin2)
  2. recruitment of TFIIH complex - a basal transcription factor)
  3. excision of damaged DNA strand (XPD, XPB helicases)
  4. incision of DNA strand (XPF, XPG incisionases)
  5. DNA synthesis and ligation (with replication machinery)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

what are the 2 forms of NER repair:

A
  1. transcription coupled repair (TCR)
  2. global genomic repair (GGR)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

what is transcription coupled repair (TCR)?

A

-targets transcriptionally ACTIVE genes
-“fast” repair
-RNA polymerase II works in conjunction with the XP proteins to remove UV damage
-uses CSA and CSB (transcription factors?)

the TFIIH complex is carried with the RNA polymerase II, TFIIH system is “built into” RNA polymerase

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

what is global genomic repair (GGR)?

A

-uses the NER pathway (not built-in)
-slow repair of UNTRANSCRIBED regions
-removal of DNA damage can take days

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

why is the double strand break repair process important?

A

A double-strand break repair model refers to the various models of pathways that cells undertake to repair double strand-breaks (DSB). DSB repair is an important cellular process, as the accumulation of unrepaired DSB could lead to chromosomal rearrangements, tumorigenesis or even cell death.

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

what are the 2 modes of double strand break repair?

A
  1. non-homologous end joining (NHEJ)
  2. homologous recombination (HR)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

what are some causes of double strand breaks (DSB)?

A

ionizing radiation
fork collapse
oxidative damage
drug induced damage

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

what are the 2 main signaling kinases and DSB damage sensors?

A

ATM
ATR

**MRN complex interacts with ATM and ATR directly and indirectly. it is the primary sensing complex for DSB. regulates cell cycle and initiates the repair of DSB.

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

what cells do NHEJ target?

A

cells at any stage of the cell cycle

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

what cells do HR (homologous recombination) target?

A

replicating or G2 cells

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

describe the NHEJ process:

A
  1. end recognition of the DSB by Ku heterodimers
  2. additional proteins recruited (DNA protein kinase K)
  3. processing of DNA ends, limited repair synthesis, and ligation
  4. end product = repaired DNA that has suffered a deletion of nucleotides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

what is the downside of NHEJ?

A

DNA can be lost

think fast, half hazard, and nonspecific!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

describe the homologous recombination process?

A

-occurs during S and G2 phase
-slower process than NHEJ (think slow and regulated)
-for sister chromatids

  1. MRN complex detects DSB
  2. exonuclease degrades 5’ ends
  3. strand invasion at branch point
  4. DNA synthesis and migration of branch point
  5. continued branch migration with pairing of newly synthesized DNA with the synthesis of the top strand
  6. DNA ligation
  7. DSB is accurately repaired
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

what would happen if there was a mutation in the DSB repair proteins?

A

MRN complex proteins: MRE11, Rad50, NBS1

ATM, ATR

mutation in these genes would have an negative impact on:
-sensing for DSBs
-cell cycle regulation (checkpoint repairs)
-transcription of DSB genes
-initiating repair of the DSB genes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

how are BRCA1 and BRCA2 related to the HR pathway?

A

BRCA1: ubiquitin ligase, works directly in HR repair, tumor suppressor, transcription factor

BRCA2: promotes HR repair and recombination, activates the S phase checkpoint

both promote HR repair and ensure that there are not any double stranded breaks present prior to DNA replication (S phase)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

what are the phases of the eukaryotic cell cycle?

A

G0: resting phase when cell has left the cell cycle and has stopped dividing

G1: cells increase in size, G1 checkpoint ensures that everything is ready for DNA synthesis, ensures there’s no DNA damage

S: DNA replication occurs

G2: gap between synthesis and mitosis, cells continue to grow, G2 checkpoint ensures everything is ready for mitosis

M: CELL GROWTH STOPS and cellular energy is focused on division into x2 daughter cells; metaphase to anaphase transition checkpoint (making sure spindle fibers are correctly attached)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

phases of meiosis:

A

meiosis I and II

prophase: chromosomes condense, nuclear membrane disintegrates, find sisters and tetrads form (crossing over)

metaphase: migrate to metaphase plate

anaphase: tetrads split, chromosomes pulled to either pole by spindle fibers

telophase: cleavage furrow forms, cytokinesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

difference between anaphase I and anaphase II?

A

anaphase I - 2 TETRADS line up in the middle, and whole chromosomes are pulled apart

anaphase II - a chromosome lines up in the middle, the axis divides it into 2 SISTER CHROMATIDS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

mitosis phases:

A

prophase
metaphase
anaphase (sisters are pulled apart, like meiosis II)
telophase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

differences in mitosis and meiosis:

A

mitosis (cellular division): cell division (2n–>2n)

meiosis (reductional division): reproductive, gamete producing
(2n –>n)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

what are the 2 types of cell death?

A

necrosis and apoptosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

morphological features of apoptosis:

A

-PROGRAMMED cell death
-can be intrinsic (stress) or extrinsic (induced by an external cell, i.e. macrophage)
-cell loses structure and the membrane blebs
-blebs are phagocytosed (cleaned up)
-NON-INFLAMMATORY
-controlled
-energy required

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

morphological features of necrosis:

A

-cell death due to injury or disease
-NOT PROGRAMMED
-external (e.g. infection) or internal triggers (e.g. ischemia)
-MESSY
-cell bursts and decomposes
-triggers INFLAMMATORY response
-passive/no energy needed
-debris is ultimately phagocytosed

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

what is common between the intrinsic and extrinsic apoptotic pathways?

A

both pathways eventually rely on caspase proteolytic cascades to achieve cell death

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

what is the differences between the intrinsic and extrinsic caspases?

A

intrinsic - caspase 9
extrinsic - caspase 8

BOTH have caspase 3 as the last step

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

describe the intrinsic apoptotic pathway:

A
  1. apoptotic stimulus disrupts the mitochondrial membranes (Bax and Bak poke holes)
  2. cytochrome C is released into the cytosol (also releases SMACS - apoptosis inhibitors)
  3. Cytochrome C binds to ATP and APAF-1 —> apoptosome
  4. procaspase 9 –> caspase 9
  5. caspase 3 activated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

describe the extrinsic pathway process for macrophage interaction:

A
  1. external cell initiates apoptosis via signaling proteins
  2. signaling proteins (i.e. TNF-alpha - inflammatory cytokine) bind to death receptor on membrane
  3. cytosolic part of death domain dives deep into the cell (the death domain)
  4. death inducing signaling complex - DISC (FADD and TRADD) change shape and binds numerous proteins to form signaling complex
  5. procaspase 8 –> caspase 8
  6. caspase 3 activated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

how is the extrinsic apoptotic pathway different for cytotoxic T cells:

A

T cell recognizes that the cell is expressing foreign antigens on its surface (MHC1)

Fas ligand (of T cell) binds the Fas receptor on the cell.

Changes the shape of the death domain signaling complex (DISC-FADD)

Same caspase pathway (caspase 8 –> caspase 3) as macrophage initiated.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

what is a mutual part of the macrophage initiated and T cell initiated apoptotic pathways:

A

-both cause the death domain to bind to other proteins to form the DISC
-both initiate the same caspase cascade (caspase 3)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

what is normal flora?

A

refers to the microorganisms including bacteria, fungi, and viruses that inhabit the human body or live on its surfaces

normal microbiota = normal flora = not harmful

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

why is the normal flora important?

A

-aid immune system in preventing infection
-key role in metabolism (GI)
-necessary for survival

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

what is symbiotics?

A

-live in harmony with their human host
-“mutualism”
-human body provides benefits necessary for bacteria’s survival and the bacteria offers benefits to the host
-e.g. symbiosis in the gut

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

what is an opportunist?

A

-potential pathogens that would not normally inhabit the body but can take advantage of an opportunity to
-in times of weakened immune responses or skin breaks

-cancer chemotherapy, anti-rejection drugs, catheter or central line insertions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

what is a pathobiont?

A

-typically a benign endogenous microorganism
-potentially pathogenic and may cause disease when overgrowth occurs

-e.g. C diff - usually present in GI tract and is usually kept in check by other bacteria. Growth spurt after antibiotics. Causes toxin release and inflammation, colitis, diarrhea, abd pain, fever. Has endospores that can survive the stomach.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

what is the role of human intestinal microbiota in health and disease?

A

GI tract bacteria keep one another in check
Prevents overgrowth
e.g. C diff infections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

what is the predominant normal flora of the skin?

A

staphylococcus epidermis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

what is the predominant normal flora of the nose?

A

staphylococcus epidermis, colonizing staph aureus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

what is the predominant normal flora of the oropharynx?

A

viridians group
S mitis
S mutans

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

what is the predominant normal flora of the mouth?

A

streptococcus mutans

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

what is the predominant normal flora of the colon?

A

bacteroides fragilis (anaerobe)
E coli (faculative)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

what is the predominant normal flora of the vagina?

A

lactobacillus, also colonized by group B strep (S agalactae) and E coli

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

what is tissue tropism?

A

pathogens and normal flora exhibit a PREFERENCE for specific tissues to live and grow

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

bacterial tropism is primarily driven by:

A

-tissue pH
-metabolic products
-immune factors
-oxygen levels
-other local tissue environmental factors

viral tropism is largely driven by host cell receptors that allow the virus to attach to the cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

what causes dysbiosis:

A

normal flora imbalance

causes:
-overgrowth
-too few bacteria
-inappropriate types

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

examples of dysbiosis in the gut:

A

small intestinal bacterial or fungal growth

-Klebsiella species (not normally present) found in the feces of IBS or gastroenteritis patients (indicating dysbiosis)

-alcohol induced dysbiosis can weaken the body’s defenses against Klebsiella pneumoniae (normally in mouth and skin), leading to PNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

example of dysbiosis in the vagina:

A

low pH maintained by Lactobaccillus

If Lactobacillus is decreased (i.e. Abx treatment), pH rises and potential pathogens can grow

yeast infection - Candida albacans overgrowth

BV - Lactobacilli replaced by Gardnerella vaginalis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

examples of dysbiosis in the oral cavity:

A

normal bacteria: diptheroids and corynebacterium (overgrowth is harmful)

Pseudomembranous pharyngitis: C diphteriae proliferation due to oropharyngeal dysbiosis

creates a pseudomembrane = thick, grayish adherant exudate formed by cell debris and inflammatory products

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

normal blood pressure:

A

normal systolic: 120
normal diastolic: 80

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

what are the 3 factors that affect blood pressure?

A
  1. cardiac output
  2. fluid volume in the artery
  3. degree of constriction by arterioles (resistance)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

what is blood pressure?

A

force of blood pressing against the artery walls

131
Q

systolic vs diastolic pressure

A

both in relation to AORTA PRESSURE:

systolic: maximal contraction of the heart, aortic valve is open and blood is pushed into the aorta

diastolic: minimum pressure in the aorta, aortic valve closed, no blood exits heart into aorta, blood moves away from aorta due to the lower downstream pressure

132
Q

normal heart rate:

A

60-100 bpm

133
Q

normal respiratory rate:

A

12-16 breaths per minute

134
Q

normal body temperature:

A

97.7-99.5
~98.6 F / 37 C

135
Q

pulse pressure equation:

A

PP = systolic - diastolic

136
Q

what is mean arterial pressure (MAP)?

A

-average pressure throughout one heartbeat
-“cardiac cycle”

MAP = 1/3 Ps + 2/3 Pd

Heart is in diastole 2/3 of the time. Therefore, factoring in the phase lengths.

137
Q

explain how a sphygmomanometer is used:

A

cuff pinches artery, release pressure:

blood first pushed through the artery, a whoosing sound is heard –> systolic (turbulent flow)

sound disappears, blood flows smoothly (laminar) down the pressure gradient because the artery is fully open –> diastole

138
Q

cardiac output equation:

A

CO=stroke volume x HR

139
Q

what is the effect of sympathetic activity on stroke volume, heart rate, and cardiac output?

A

increases HR and stroke volume, therefore increasing CO

CO=stroke volume x HR

140
Q

sympathetic activity ______respiratory rate.

A

increases

141
Q

what are some causes for increased sympathetic activity? how do they differ in their mechanisms?

A

exercise:
-stronger contractions of the heart
-increase SV and Ps
-increased heart rate
-increased CO
-CO physiologically and temporarily increased

stress:
-arterioles vasoconstrict, increased Pd
-heart needs to beat stronger to overcome resistance, increase SV, increase Ps
-increased HR
-chronically and pathologically increases

142
Q

respiratory buffer equation:

A

CO2 + H2O –> H2CO3–> HCO3- + H+

143
Q

effects of high and low respiratory rates:

A

high:
-more CO2 removed
-less H+
-pH increases, alkalosis

low:
-less CO2 removed
-increase in H+
-pH decreases, acidosis

144
Q

what are the 2 sources of internal body heat?

A
  1. metabolism (waste heat, non-shivering thermogenesis)
  2. muscle contractions (shivering thermogenesis, waste heat)
145
Q

factors affecting human body temperature:

A

-age
-gender
-time of day (lowest at night)
-time in menstrual cycle (higher from ovulation to menses)
-state of consciousness and emotions

146
Q

what are the different regions temperature can be taken? benefits/cons?

A

forehead: affected by environment, no body contact, quick

aural (ear): useful except during extremes

oral: 0.5 lower than rectal

axillary: too variable

rectal: most accurate for core body temperature, inconvenient

147
Q

body water is ______% of total body weight.

A

60

lower in obese, higher in lean people
higher in infants, lower in elderly

148
Q

what are the distributions of body water into compartments?

A

ICF: 66%
ECF: 33% (1/4 in plasma, 3/4 in interstitial fluid)

149
Q

hydration status effects on the body:

A

ICF swelling: HA
ICF shrinking: fatigue

ECF depletion: hypotension
ECF overload: edema

150
Q

effects of sodium on water weights:

A

water can freely diffuse between ICF and ECF. 33% kept in the ECF due to osmotic pressure created by sodium salts

151
Q

how does a low sodium diet decrease BP?

A

salt in the blood stream increases water pressure in the arteries, therefore increasing pressure.

want the opposite effect.

151
Q

how does a low sodium diet decrease BP?

A

salt in the blood stream increases water pressure in the arteries, therefore increasing pressure.

want the opposite effect.

152
Q

stages of linear growth:

A

-prenatal
-infancy
-childhood
-adolescence
-adulthood
-aging

153
Q

what is human linear growth?

A

period of time starting from the cleavage of the zygote (pre-natal) marked by the end of long bone growth (adolescence)

-long bone growth finished when estrogen is secreted, causing the epiphyses to fuse (growth plates)

154
Q

compare the relative linear growth velocity during each stage of
human life cycle:

A

fetal and infantile: highest growth rate, environment, nutrition, health and happiness

childhood: sharply decreases and levels off but contributes 40% to adult height, growth and thyroid hormones present, genes, environment

pubertal: females have a peak slightly earlier than males, due to hormones (sex and growth), 15% of adult height

155
Q

contrast hormonal control of neonatal growth to post-natal growth:

A

IGF 1 and 2 in pre-natal environment. pre-natal growth depends mostly on maternal size and uterine development. infancy is marked by the dependence of genetic factors. also thyroid hormones and infant environment and nutrition.

155
Q

contrast hormonal control of neonatal growth to post-natal growth:

A

hormones predominate in post-birth ???

neonatal depends on uterine environment

infants: thyroid hormones

childhood: thyroid and growth hormones

pubertal: growth and sex hormones

156
Q

Describe differences between genetic and environmental factors contributing to
aging

A

GENETIC:
-DNA repair defects - accumulation of mutations
-genetic abnormalities - abnormal cellular signaling

ENVIRONMENT:
-environmental insults - free radial damage
-reduced enzyme activity
-accumulation of damaged cellular proteins and organelles

157
Q

innate vs adaptive immune responses

A

innate: protection against infection that relies on a mechanism that existed before the infection, capable of a rapid response of microbes

adaptive: protection against infection that is mediated by lymphocytes and is stimulated by exposure to FOREIGN antigens. specificity for distinct molecules and long-tem immunity. AKA “acquired immunity”

158
Q

properties of the adaptive immune response:

A

-specificity
-diversity
-memory
-clonal expansion
-specialization
-contraction and homeostasis
-non-reactivity to self

159
Q

where do B lymphocytes mature?

A

bone marrow

160
Q

where do T cells mature?

A

thymus

161
Q

what are all lymphocytes derived from?

A

hematopoietic stem cells

162
Q

what are the 3 stages of lymphocytes:

A

1) naive cell - haven’t found antigen yet

2) activated/effector - encounters antigen, proliferates and carries out effector function

3) memory - immunological memory, quicker and more effective response with reinfection

163
Q

what is clonal selection?

A

every person has a vast number of unique clonally derived lymphocytes

“immune repertoire”

each clone is genetically distinct and created at random, each expresses ONE pre-determined antigen receptor

clonal selection explains how a single B or T cell that recognizes an antigen that enters the body is selected from the pre-existing cell pool of differing antigen specificities and then reproduced to generate a clonal cell population that eliminates the antigen.

164
Q

clonal selection occurs IF and ONLY IF _________.

A

a specific antigen enters the body and encounters a lymphocyte with that same specific receptor

165
Q

role of antigen presenting cells (APCs):

A

-capture antigens, process antigens, and present them to lymphocytes for more sophisticated immune functions

dendritic cells
macrophages
B lymphocytes

166
Q

innate vs. adaptive immunological memory

A

innate - very little/nonexistent
adaptive - memory T and B cells, enhanced secondary responses

167
Q

primary vs secondary immune responses

A

primary:
-first encounter with antigen
-induces production of plasma cells - antibodies
-LAG period before antibodies produced
-modest production

secondary:
-second encounter with antigen (B cells are no longer naive)
-faster response
-produces antibodies at a higher rate and maintains the higher levels

168
Q

what is humoral immunity?

A

adaptive defense mechanism mediated by B lymphocytes
antibodies produced
principal defense against EXTRACELLULAR microbes and toxins

169
Q

what is cell mediated immunity?

A

adaptive defense mediated by T lymphocytes

Helper T cells -secrete cytokines that recruit/activate phagocytes to kill
Cytotoxic T cells - directly kill

170
Q

list the phases of the immune response:

A
  1. recognition (antibodies for B cells, through APCs for T cells)
  2. activation phase (clonal expansion/differentiation/gain effector fx)
  3. antigen elimination phase
  4. contraction phase (return to homeostasis)
  5. memory phase
171
Q

describe the chronological process of hematopoiesis:

A

image

172
Q

what are the primary lymphoid organs?

A

bone marrow
thymus

173
Q

secondary lymphoid organs:

A

spleen
lymph nodes

174
Q

name some other secondary lymphoid tissue:

A

MALT - mucosa associated lymphoid tissue
GALT - gut associated lymphoid tissue

175
Q

the site for presentation and initiation of immune responses against BLOOD BORNE antigens?

A

spleen
therefore, a splenectomy would be associated with a predisposition for certain blood borne infections, especially of encapsulated bacteria (S pneumoniae)

also a reservoir for RBCs, lymphocytes, and monocytes

176
Q

describe the 3 different subdivisions of the spleen:

A

white pulp - immunologically active part
-periarteriolar lymphoid sheaths (PALS) - arteriolar supply of the spleen
-lymph follicles - dividing B lymphocytes

red pulp - hematologically active part
-most mass
-reserve storage of monocytes

marginal zone - in between the pulps
-contain the APCs (macrophages and dendritic cells)

177
Q

describe the role of MALT:

A

MALT = lymphoid aggregates that sit near the entrances of the digestive and respiratory tracts

MALT on the tonsils

  1. M (microfold) cells on surface detect pathogen
  2. activate tonsillar B cells
  3. proliferate in the germinal areas
  4. secrete IgA antibodies
178
Q

what are Peyer’s patches?

A

collection of lymphoid nodules found in the gut mucosal tissue

lymphoid nodules = small localized collection of lymphoid tissue located in the connective tissue beneath the epithelium

Peyer’s patches located in the ileum (SI)
-like MALT, contain M cells
-M cells detect antigen and transport into cell to the DCs
-DCs present antigen to lymphocytes
-T lymphocytes and Igx leave the efferent vessel - go to site of infection

179
Q

passive diffusion is dependent on:

A
  1. concentration gradient
  2. partition coefficient
  3. cell surface area
  4. pH differences for ionic compounds
180
Q

characteristics of active transport:

A

requires energy
moves drugs AGAINST electrochemical gradient
saturable (can saturate the channels)
selective (charge and size)
drugs can compete for transport

181
Q

factors that effect the transfer of drugs across a membrane:

A
  1. molecular size
  2. partition coefficient
    -(concentration of drug in lipid)/(concentration of drug in water)
    -p>1 lipophilic
    -p<1 hydrophilic
  3. ionic character
    -non-ionized can cross membranes (unless there’s an ion channel)
    -chemicals exist in an ionized form when exposed to an environment with the opposite pH
  4. effects of pH - Henderson Hasselbalch
182
Q

a _______intravenous diffusion rate, promotes the absorption of a drug.

A

rapid

183
Q

describe drug permeability of the CNS:

A

endothelial cells of CNS do not contain large pores (do not transmit ionized/large polar drugs)

therefore, lipid soluble drugs readily permeate the CNS membranes

184
Q

what are the 3 types of drug reservoirs:

A

plasma proteins (albumin and alpha-1 glycoprotein)
tissues (prolongs drug action)
fat (lipid soluble reservoir)

185
Q

what type of drugs does albumin bind?

A

acidic

186
Q

what type of drugs does alpha-1 glycoprotein serve as a carrier for?

A

basic drugs

187
Q

what are the 2 ways that drugs are eliminated in the body?

A

excretion - kidneys (unchanged drugs)
metabolism - liver (changed into metabolites)

188
Q

describe the function of the liver in drug metabolism?

A

kidneys are modest (can’t excrete lipophilic drugs as they can be reabsorbed/pass through the membrane)

the liver makes these drugs more hydrophilic so that they remain in the urine and are excreted. also terminates their pharmacological activity

189
Q

what enzymes catalyze phase 1 reactions in liver metabolism?

A

cytochrome P450 enzymes (family of CYP enzymes)

190
Q

what are the differences between phase 1 and phase 2 of liver metabolism?

A

phase 1 - hydrolysis, oxidations, reductions

phase 2 - glucoronidation, sulfation

191
Q

what are the 4 ways an drug can cross a membrane?

A
  1. passive diffusion
  2. active diffusion
  3. aqueous diffusion
  4. transporter
192
Q

list the 6 types of biological drug receptors:

A

water and ions (nonspecific, i.e. laxatives)
metal ions (chelating drugs, i.e. chelating poisonous ions)
enzymes
nucleic acids
membrane sterols
specific receptors (i.e. Ach receptors)

193
Q

what is affinity (drugs)?

A

tenacity for which a drug binds its receptor

194
Q

what is intrinsic activity (drugs)?

A

capacity of a drug to propagate a biological signal

195
Q

what is the structure activity relationship (SAR)?

A

affinity and intrinsic activity are closely related to the drug’s chemical structure

therefore, small modifications in the drug can drastically change its properties

196
Q

what are the 2 receptor domains?

A

binding domain - binds ligand
effector domain - propagates signal (direct or through second messengers)

197
Q

describe how steroid receptors enact a change:

A
  1. steroid drug passively crosses membrane and binds steroid receptor
  2. receptor activated and moves across nuclear membrane
  3. steroid binding induces conformational change in receptor, allowing it to bind response elements on DNA
  4. mRNA transcription and translation
  5. protein synthesis –> biological effect

**can also inhibit protein synthesis

198
Q

examples of enzyme linked receptors:

A

tyrosine kinase
adenlyl/guanlyl cyclases

199
Q

examples of enzyme linked receptors:

A

tyrosine kinase
adenlyl/guanlyl cyclases

**have ligand binding domain and intracellular catalytic domain

200
Q

explain receptor desensitization:

A

continued stimulation of cells with agonists generally results in desensitization
-down regulation of receptors on the membrane
e.g. repeated use of bronchodilators in asthma treatment

201
Q

explain supersensitivity (up-regulation) of receptors:

A

supersensitivity to agonists frequently follows a chronic reduction in receptor stimulation

with chronic inhibition, there is an upregulation of receptors

when inhibitor is removed, there are now more receptors available for drug binding

e.g. long term use of B adrenergic drugs such as propanolol. stop propranolol and will get hypersensitive B adrenergic response

202
Q

what causes myasthenia gravis?

A

autoimmune depletion of Nm receptors

(type II hypersensitivity - autoantibodies bind Ach receptors on skeletal muscle cells)

203
Q

what causes some forms of IDDM?

A

autoimmune depletion of insulin receptors
(type IV hypersensitivity)

204
Q

effect of propanolol on B adrenergic receptors?

A

propanolol = beta blocker
B adrenergic receptor agonists used in asthma treatment but can become desensitized with long term use

if patient receives long term administration of propanolol and then stops taking the drug, hypersensitivity to the B adrenergic asthma drugs may be seen

205
Q

types of bonds between drug and receptor (in order of decreasing strength)

A

covalent
ionic
H bonds
hydrophobic
van der waals

206
Q

what is the receptor occupancy theory?

A
  1. response occurs in receptor when drug is bound
  2. more receptors bound = greater response
  3. all receptors bound = maximal response
207
Q

what is the ED50 or EC50?

A

concentration or dose of drug that causes half of the maximal response (all receptors bound)

AKA the potency

208
Q

what does the dose-response relationship look like graphically?

A

image

208
Q

what does the dose-response relationship look like graphically?

A

image

209
Q

what are antagonists?

A

binds receptors without causing an effect but blocks the binding of endogenous agonists

therefore, have affinity but not intrinsic activity

210
Q

what are the three types of antagonism?

A

competitive
irreversible
uncompetitive

211
Q

what is physiological antagonism?

A

drug response mediated by the activation of a receptor is counteracted/opposed by the opposite response produced by another DIFFERENT receptor

e.g. epinephrine and anaphylaxis
histamine causes vasodilation, epinephrine is given to overcome hypotensive effect

212
Q

contrast graded and quantal responses

A

graded response: the more receptors bound, the greater the response (receptor occupancy theory)

quantal response: an all or nothing response. as dose is raised, population affected is raised (e.g. desired effect is absence of seizure, adverse affect is death)

213
Q

equation for therapeutic index:

A

TI = (LD50)/(ED50)

LD50 - median lethal effective dose that kills 50% of subjects
ED50 - median effective dose that protects 50% of subjects

therapeutic index = DRUG’S MARGIN OF SAFETY

214
Q

what kind of therapeutic index is desirable?

A

a high therapeutic index is good
means there’s a large difference between the lethal and effective doses (e.g. penicillin)

a low therapeutic index is bad
not much difference, very easy to overdose
e.g. warfarin and digoxin

215
Q

examples of decreased responses in dose-responses

A

tolerance - repeated exposures to a drug lead to diminished response (down reg of receptors)

tachyphylaxis - rapid loss of response upon repeated exposure to a drug (down reg of receptors)

resistance - diminished response to drugs that inhibit cell growth or death

216
Q

examples of increased dose relationship responses:

A

additive - two drugs given together are equal to the sum of their individual effects

synergism - response is greater than the sum of individual effects

potentiation - administration of a drug with little/no effect enhances the effect of another drug

217
Q

what is pharmokinetics?

A

study of the processes of absorption, distribution, and elimination of drugs as they relate to the time course of the drug’s presence in the body in one or more compartments

218
Q

what is the fundamental hypothesis in pharmacokinetics?

A

a relationship exists between the pharmacologic (toxic) effect of a drug and its concentration in the blood

219
Q

most assays do not distinguish _______from _______, therefore, the total drug concentration in the plasma includes both.

A

free drug, bound drug
the extent of binding and the fraction of free is relatively constant

220
Q

what is first order kinetics (drugs)?

A

the rate of decrease in drug concentration over time is directly proportional to the drug concentration

high drug concentration - faster elimination
as drug concentration decreases - slower elimination

221
Q

what are the 2 assumptions in pharmacokinetics?

A

-first order kinetics
-the body can be treated as a single compartment

222
Q

what is bioavailability (F)?

A

fraction of the unchanged drug that reaches systemic circulation

for IV drugs…F=1

223
Q

explain zeroth order kinetics:

A

the elimination of drug is constant and INDEPENDENT of concentration or the amount of drug

occurs when the system is rate limiting or a saturable process

e.g. alcohol dehydrogenase becomes saturated and then can only metabolize 9g/hr, regardless of concentration

**can’t use any of the pharm equations

224
Q

karyotyping involves freezing chromosomes during which phase of mitosis?

A

metaphase - most visible

225
Q

what cell type is ideal for karyotyping?

A

WBCs - enter mitosis easily when stimulated

226
Q

describe the karyotyping process for WBCs:

A
  1. WBCs from blood sample are cultured in the presence of growth factors
  2. arrested in metaphase using COLCHICINE
  3. added to a hypotonic solution to swell
  4. cells fixed with EtOH/acetic acid
  5. stained with Geimsa stain
227
Q

describe Geimsa staining (G banding) patterns:

A

dark bands
-heterochromatin
-stain binds A-T regions strongly

light bands
-euchromatin
-loosely packed chromatin
-stain does not bind G-C strongly
-gene rich regions

228
Q

3 centromere placements:

A

metacentric (p and q almost equal)
submetacentric (p arm is shorter)
acrocentric (p arm is very short)

229
Q

euploid

A

having the normal number of chromosomes

230
Q

polyploidy

A

presence of a complete extra SET of chromosomes

eg. triploidy (3N), tetraploidy (4N)

231
Q

possible mechanism of triploidy:

A

fertilization of one egg with two sperm
causes fetal loss in first 2 trimesters

232
Q

possible mechanism of tetraploidy

A

fusion of 2 diploid zygotes or mitotic failure
rare

233
Q

aneuploidy

A

missing or additional INDIVIDUAL chromosomes

234
Q

monosomy

A

only have one copy of a chromosome pair

nearly always leads to death
critical loss of genes

235
Q

what is turner syndrome?

A

genetic female missing an X chromosome

45 XO or 45X

236
Q

trisomy

A

presence of an extra chromosome
more compatible with survival than monosomy (no critical loss)

237
Q

what are the 3 trisomies:

A

trisomy 13 - Patau (lucky 13/ st. patrick)

trisomy 18 - edwards (stephanie edwards has wanted to become a doctor since she was 18)

trisomy 21 - down syndrome

238
Q

what is klinefelter syndrome?

A

47, XXY
genetic male due to Y presence but has an extra X

calvin klein, a man who’s into fashion

239
Q

clinical features of down syndrome:

A

depressed nasal bridge, upslanting palpebral fissures, epicanthal folds

congenital defects in 40%

moderate to mild ID

240
Q

what other disease is down syndrome linked to?

A

alzheimer’s

aberrant gene expression of amyloid precursor protein (APP) on chromosome 21

241
Q

3 causes of down syndrome:

A

nondisjunction during meiosis I in the female (90%)
nondisjunction during meiosis I or II in male (10%)

OR

robertsonian translocation involving chromosomes 21 and 14 (4%)

242
Q

what is the karyotype for a robertsonian translocation in a person with down syndrome?

A

45, XX/XY, t(14;21)

long arm of chromosome 21 translocated to the long arm of chromosome 14
short p arms lost

result: loss of one chromosome

243
Q

result of nondisjunction in meiosis I:

A

trisomy
trisomy
monosomy
monosomy

244
Q

result of nondisjunction in meiosis II:

A

euploid
euploid
trisomy
monosomy

245
Q

what is a 1st trimester US looking for (down syndrome)?

A

nuchal translucency (NT)
elevated NT indicates high risk for DS

246
Q

2nd trimester down syndrome US looks for…

A

major anatomic abnormalities

cleft lip, polydactyly, abdominal wall or cardiac irregularities

247
Q

what two serum biomarkers are elevated in maternal labs (down syndrome)?

A

B-hcg
inhibin A

248
Q

what is a translocation?

A

rearrangements between segments of NON-HOMOLOGOUS chromosomes

can be balanced or unbalanced
can be reciprocal, nonreciprocal, or robertsonian

249
Q

what is a robertsonian translocation?

A

results in the fusion of long arms of acocentric chromosomes
the 2 p arms are lost (usually no clinical significance)

250
Q

what are the acocentric chromosomes?

A

13, 14, 15, 21, 22

251
Q

3 types of deletions

A

terminal deletion (end of chromosome)
interstitial deletion (within chromosome)
microdeletion (very small, need FISH to see)

252
Q

cri-du-chat

A

crIIII-FIIIIve

deletion of the SHORT arm of chromosome 5

46, XX/XY, 5p-

253
Q

symptoms of cri du chat

A

high pitched CRI
microcephaly, ID, associated with cardiac abnormalities

254
Q

williams syndrome cause:

A

deletion of long arm 7

255
Q

symptoms of williams syndrome

A

“WL always had extreme friendliness with strangers”
ID, distinctive facial features, increased sensitivity to vitamin D, high calcium levels, cardiac abnormalities sometimes

256
Q

pericentric vs paracentric inversions

A

chromosome inversions

para - do not include centromere
pericentric - involve the centromere

257
Q

2 most common ring syndromes:

A

14, 21

258
Q

how does a ring chromosome form:

A

deletions on either end of the chromosome create “sticky” ends
attach to each other
ring is typically lost, leading to monosomy

259
Q

isochromosome cause:

A

chromosomes divide along axis that is perpendicular to the normal

lethal loss of genes

260
Q

cause of Edward’s syndrome

A

trisomy 18
isochromosome 18q
produces an extra long arm of chromosome 18

261
Q

results of unequal cross over?

A

variable number tandem repeat (VNTR) polymorphisms, deletions, duplications

262
Q

types of microscopy

A

light microscope
-uses light (photons)
-cells or tissues on slides
-stained with something to give color/contrast

electron microscopy -uses electrons:

  1. transmission electron microscopy (TEM)
    2D
    VERY THIN sections, stained with metal for contrast
  2. scanning electron microscope
    sample coated in metal
    electrons produce a 3D image
  3. freeze fracture (a type of TEM)
263
Q

light microscopy - cytospin vs blood smear

A

cytospin - cells centrifuged into a circular area

blood smear - blood is dragged along to smear it on the slide

264
Q

2 locations of goblet cells:

A

digestive or respiratory systems

265
Q

what are the 2 dyes commonly used - what colors do they stain

A

eoisin
-acidic/eosinophilic
-stains basic features PINK
-stains PROTEINS/CYTOPLASM

hematoxylin
-basic/basophilic
-stains acidic structures BLUE
-stains NUCLEI

266
Q

what is freeze fracture

A

membranes are “fractured” of split in between 2 layers of the lipid bilayer
type of TEM but appears to be 3D
ALWAYS shows membrane structure

267
Q

composition of whole blood:

A

erythrocytes (44%)
hematocrit = volume % of RBCs in blood

plasma (55%)
water, proteins, other solutes

buffy coat (<1%)
platelets, leukocytes

268
Q

types of monocyte derivatives:

A

langerhan’s - skin
kupffer - liver
dust - lungs
microglia - brain
osteoclasts - bone

269
Q

what are the charged AAs?

A

glutamic acid (E)
aspartic acid (D)
arginine (R)
lysine (K)
histidine (H)

270
Q

which AA has buffering capacity?

A

histidine
pka=6.2

271
Q

aspartate effector function

A

excitatory nt

272
Q

glycine effector function

A

inhibitory nt, heme precursor

273
Q

arginine effector function

A

urea and nitric oxide precursor

274
Q

glutamate effector function

A

excitatory nt, precursor of GABA (inhibitory)

275
Q

histidine effector function

A

precursor of histamine (inflammatory)

276
Q

tryptophan effector function

A

precursor of serotonin, melatonon

277
Q

tyrosine effector function

A

hormones and catecholamine precursor (epinephrine, norepinephrine, dopamine, thyroxine)

278
Q

heat shock protein (HSP) function

A

helps with protein folding
needs ATP
chaperone “cage” with 2 stacked rings
protein enters hydrophobic cage
folded and is released

279
Q

describe the process of amyloid formation in alzheimer’s:

A
  1. transmembrane portion of amyloid precursor protein is cut
  2. spontenous aggregation of the amyloid pieces (AB)
  3. forms insoluble fibrils of B pleated sheets

Beta sheets are Bad.

280
Q

describe how prions (PrP) lead to disease:

A
  1. infectious PrP has beta sheets
  2. PrP interacts with noninfectious PrP (alpha helix)
  3. healthy PrP converts into infectious (beta sheet)
  4. goes on and does the same (exponential infection)

healthy - alpha
unhealthy - beta

Beta sheets are Bad.

281
Q

2 possibilities for buffers:

A

weak acid with conjugate base
weak base with conjugate acid

281
Q

2 possibilities for buffers:

A

weak acid with conjugate base
weak base with conjugate acid

282
Q

what are the 2 protein degradative pathways for damaged or unneeded proteins:

A

ATP dependent ubiquitin proteasome system
-cytosol
-endogenous proteins
-damaged proteins
-MHC1

ATP independent lysosomes
-extracellular proteins/phagocytosis
-plasma proteins
-organelle recycling
-MHC2

283
Q

stomach acid is secreted by _________. it’s function is to__________.

A

parietal cells
denature proteins, making them more susceptible to proteases

Acid - pArietal

284
Q

______secretes pepsinogen in the stomach which is converted to active form (pepsin) by _______.

A

chief cells, HCl (or automatically)

285
Q

role of pepsin

A

creates peptides and a few free AAs (breaking down the protein)

286
Q

where does zymogen activity take place (protein degradation)?

A

zymogen enzymes secreted by the pancreas break down oligopeptides into amino acids in the small intestine

287
Q

what enzymes are secreted by the small intestine (protein degradation)?

A

di and tri peptidases
amino peptidases

288
Q

3 types of AA branched chain metabolism:

A
  1. transamination
  2. oxidative decarboxylation
  3. dehydrogenations
289
Q

what causes methylmalonic acidemia?

A

defects in the methylmalonyl mutase gene
defects in genes that are responsible for conversion of cobalamin (vit B12)

290
Q

splicing process:

A

mediated by snRNPs in the splicesome

  1. 5’ ss donor binds to branch point A
  2. intron lariat is formed
  3. 3’ ss acceptor site broken
  4. lariat discarded, exons ligated together
291
Q

describe how RNA processing is cotranscriptional:

A

RNA processing can add to transcriptional stability
therefore, want want processing to occur as transcription is taking place, before splicing occurs. splicing would just remove these modifications prior to translation.

e.g. inserting intronless genes into intronless versions of intron dependent genes

would want to insert the introns into the 5’ UTR
why?
-don’t want it to be expressed (just there for stability)
-want it down at 5’ end rather than 3’ because RNA is processed from 5’ to 3’ and you want STABILIZATION at the beginning

overall, if these modifications aren’t added during transcription, they’re just gonna be spliced out after the fact and then there would be no point for having them anyways.

292
Q

describe the RNA interference (RNAi) silencing process:

A
  1. DICER recognizes dsRNA (i.e. viruses)
  2. breaks the dsRNA into pieces (small interfering RNAs)
  3. 1 siRNA enters the RISC complex
  4. associates with the argonaute protein
  5. finds this small sequence in host and degrades it
  6. RNA virus genome degraded and infection stops

** can introduce in a lab setting to knock out genes, therapeutic

293
Q

what does alternative splicing in the untranslated regions (UTRs) lead to?

A

does not alter protein structure (not involved with exons)
however, may alter mRNA levels and protein levels

remember, UTR modifications can alter stability

e.g. AU/AT rich regions are unstable, splice them out and will get more stability and higher protein levels

294
Q

.

A
295
Q

loss of function vs gain of function:

A

loss of function - inactive or less active protein
(e.g. thalassemias: anemia due to inefficient erythropoiesis)

gain of function - overly active protein
(e.g. thrombophilia/blood clots: too much prothrombin gene)

296
Q

what are the types of mutations?

A

nonsense - immature stop codon
missense - point change, one AA changed for another
frameshift - addition or deletion changes reading frame
synonymous - mutation does not change AA
conservative - changes AA but new AA has similar biochemical properties as the original

297
Q

what polymerase is involved in bacterial clonal replication?

A

DNA polymerase III

298
Q

what are transposons?

A

allow for drug resistance genes to be transferred from plasmid to host DNA

they flank the sequence of interest and insert it into host bacterial DNA

299
Q

what are high frequency recombinations (bacteria)?

A

plasmid incorporates itself into the donor DNA
then DNA is transferred to receiver
receiver gets some plasmid DNA along with host DNA

300
Q

what does protein A (virulence factor) do?

A

binds IgG and prevents an immune response
e.g. staph aureus

301
Q

role of IgA protease (virulence factor):

A

binds and degrades IgA antibodies
prevents IgA from recognizing pathogens in mucous membranes

302
Q

role of M protein (virulence factor):

A

prevents opsonization/phagocytosis

has epitopes - confuses antibodies and causes the host cells to react to self-antigens that look similar to M protein - S pyogenes - rheumatic fever

303
Q

penetrance vs expressivity

A

expressivity - dimming lights (present but at varying extents)

penetrance (on/off switch) - how many individuals in a population with a certain genotype express the phenotype

304
Q

what is mosaicsim?

A

a mutation that occurs AFTER fertilization
therefore, not all cells are affected
the earlier the mutation, the more cells affected
cascade effect

305
Q

what is epistasis? what law does it contradict?

A

one gene effects the expression of another gene
e.g. baldness gene masks hair color gene (2 different genes)

disobeys law of dominance

306
Q

allelic heterogeneity vs locus heterogeneity

A

allelic - different mutations on the same locus (same gene) lead to a similar phenotype

locus - variants on different loci (different genes) lead to a similar phenotype

307
Q

what are the modifiable residues on N terminal histone sequences?

A

K - lysine
R -arginine
S -serine
T -threonine

308
Q

explain histone acetylation:

A

histone acetyl transferase acetylates the amino acid residue
amino acid loses its charge
loosens the interaction with DNA

309
Q

histone patterns:

A

acetylated - gene activation
non acetylated - gene inactivation

310
Q

what DNA base is methylated?

A

cytosine
via DNA methyl transferases

therefore, large number of cytosines - likely to be a silenced gene. in the circumstance that a C is switched for T in the UTR region for example, would most likely increase gene expression.

311
Q

DNA methylation _______ transcription.

A

silences
methylations are read by MBD (methyl binding domain)
DNA methylation DOES NOT effect base pairing

312
Q

cystine 5’ demethylation/hydroxylation ________ transcription.

A

increases

313
Q

what is the earliest signs of cell injury?

A

cloudy swelling
swelling of the organelles
rough ER becomes dilated - lose ribosomes

314
Q

what is hydropic degeneration?

A

continued swelling of the organelles in response to injury
vacuoles appear in the cytoplasm
loss of basophilia

315
Q

what is fatty change (steatosis)?

A

accumulation of triglycerides in the cytoplasm
common in the liver (fatty liver)

causes: toxins, hypoxia, alcohol

316
Q

what are fibrotic changes?

A

irreversible changes
fibrotic bands visualized
cells die - necrosis
nodular appearance on the liver

317
Q

atrophy/hypertrophy

A

decrease and increase in cell SIZE
cell count does NOT change

318
Q

hyperplasia

A

increase in the number of cells

319
Q

what happens to the thymus during adulthood?

A

active at a young age, involutes before adulthood via APOPTOSIS

controlled cell death

320
Q

what is an xanthoma?

A

intracellular accumulation of cholesterol within macrophages
clusters of foamy cells (lipid laden macrophages found in sub-epithelial connective tissue or skin and tendons
gross appearance - small plaques to large masses