• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/256

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

256 Cards in this Set

  • Front
  • Back
What are the different ways in which genes can regulate patterning along an axis? (2)
Varied expression of genes along the axis

Concentration gradient expression of single gene along an axis
In human development, what happens at:
Week 1?
Week 2?
Week 3?
Weeks 4-8?
Weeks 9-birth?
Week 1: Fertilization, implantation

Week 2: Bilaminar embryonic disc

Week 3: Trilaminar (mesoderm) embryonic disc

Weeks 4-8: Tissues, organs, body form (patterning)

Weeks 9-birth: Fetal period (growth)
What is a homeotic gene?
Determines the fate or identity of each body segment
How do HOX genes work?
Encode a DNA-binding homeodomain

Act as transcription factors

Can be expressed at different times, in different places = specific functions!
What is spatial colinearity? Temporal?
Spatial:
The order of genes maps an axis in the developing embryo

Temporal:
The order of genes reflects their temporal expression during development
What is the "posterior dominance" of HOX genes?
When one or more HOX genes are expressed in a given segment, the HOX gene whose expression pattern is the most posterior defines the phenotype of that segment.

Ex. from lecture:
lack of b4 → becomes atlas
What is synpolydactyl?

What is its inheritance pattern? What gene is involved?
Webbing of digits with extra digits

Autosomal dominant, incomplete penetrance

HOX D13
What gene regulates notochord signalling for the neural plate to form?
Sonic Hedgehog
What does it mean that the notochord is an organizing center?
Sonic Hedgehog affects development of adjacent areas (sclerotome/somites, dermomyotome)
What is the cascade of signalling for hedgehog genes?
Inctivates Ptc (which is inhibitory))
→ Disinhibition (activation) of GLI1/2/3 (+/-)
What does a mutation in sonic hedgehog cause?
No induction of signaling

Holoprosencephaly
- Small eyes
- Microcephaly
What does a Ptc mutation cause?
Pathway always on (doesn't inhibit like it's supposed to)

Nevoid basal cell carcinoma syndrome (NBCCS), aka Gorlin syndrome
- Large head
- Wide eyes
- Retardation

**Extreme septation - can see calcified falx cerebri
What does a GLI mutation cause?
Greig cephalopolysyndactyl syndrome
- Pathway abnormality in extremities - too many fingers/toes
What is the gene dosage effect?
Heterozygotes typically have 50% enzyme activity relative to homozygous normal.

ie. one copy of the gene works, one doesn't
What is the purpose of activator with GM2 → GM3 + GalNac conversion?
Creates interface between enzyme and substrate (lipid/water) - micelle!
Why is it hard to diagnose the AB variant of HEX <i>in vitro</i>?
Enzymes will cleave GM2 in the presence of detergent, which is used <i>in vitro</i> instead of activator.
&rarr; looks like normal
In what population is Tay-Sachs prevalent?
Ashkenazi Jews
Which mucupolysaccharidosis is NOT autosomal recessive?
Hunter (type II) - X-linked recessive
What is responsible for the variations in severity of mucupolysacchiridoses?
Allelic heterogeneity
What is responsible for similar phenotypes of mucopolysaccharidosis but different enzyme mutations?
Locus heterogeneity
If two cell types with MPS did NOT cross-correct each other, what type of mutation did they have?
Allelic mutations

(same enzyme deficiency, different phenotypes)
What is the pathology of I-cell disease?
UDP-GlcNac transferase deficiency = mannose not phosphorylated = enzymes not transported to lysozome
What are the 3 lysozomal storage diseases?
1. Tay-Sachs
2. Mucopolysaccharidoses
3. I-cell disease
What are the precursors to propionyl CoA?
VOMITS

Valine
Odd-chain fatty acids
Methionine
Isoleucine
Threonine
Side chain of cholesterol
What is the difference between late onset and early onset multiple carboxylase deficiency?
Early onset: holocarboxylase synthetase deficiency (can't make holocarboxylase)

Late onset: biotinidase deficiency (can't recycle biotin)

&rarr; treat with &uarr; biotin in diet
What are the four carboxylase enzymes?
Pyruvate
AcCoA
Propionyl CoA
&beta;-methylcrotonyl
What is haploinsufficiency?
One copy of the gene is NOT enough

(sort of opposite of gene dosage)
What genotype causes xanthoma and xanthelasma in familial hypercholesterolemia?
Homozygous dominant
What are the steps of collagen synthesis?
1. Synthesized in RER (ribosomes)
2. Lots of modification of aas
3. Associate to form triple helix, then secreted
4. N-propeptide and C-propeptide and cleaved
5. Associate to form fibrils
Is it better to have reduced amounts of normal collagen or normal amounts of abnormal collagen?
Reduced, but normal
What is the mechanism for development of secondary sexual characteristics in boys?
LH secreted from pituitary
Binds Leydig cells
Leydig cells make testosterone
Why aren't girls affected by LH receptor constitutive activity?
Puberty in girls depends on other hormones as well as LH
What is the structure of a chromosome?

What are the 3 types?
Centromere
Long arm, short arm

Metacentric (equal)
Submetacentric
Acrocentric
What are the parts of acrocentric chromosomes? What is unique about them?
Stalk
Satellite

Both are very redundant, so mutations in either don't do much.
What is the only factor that predisposes to chromosomal abnormality?
Maternal age
What are the results of first division meiotic non-disjunction?
Trisomic
Monosomic
What are the results of second division meiotic non-disjunction?
Trisomic
Monosomic
Euploid
What is a fragile site? How does it appear?
Satellites at the end of the long arm of chromosome X

Looks like a gap at Xq27.3
How many repeats of CGG in FMR1 are required before individuals show the phenotype?
>200
When do trinucleotide repeats expand in FMR1?
Premutation (50-200): meiosis in oogenesis

Mutation (>200): meiosis in oogenesis, mitosis
What is anticipation?
Non-penetrance?
Anticipation - appearance of a genetic trait at an earlier age or with greater severity in successive generations

Non-penetrance - females have 2 X chromosomes, so one is randomly inactivated in cells; if it's the mutated one, they'll be fine
Why are daughters of transmitting males of Fragile X normal?
Transmitting males have premutation, but it only expands in oogenesis, so their daughters will also have premutation.
What does a longer repeat length in Friedriech's Ataxia mean?
Earlier age of onset
What are the two diseases affecting androgen receptors?
Complete Androgen Insensitivity Syndrome (CAIS) - loss of function
SBMA - gain of function
What are the principles of teratogens? (4)
1. Specific
2. Dose-effect relationship
3. Timing! (stage of development)
4. Genotype of mother/fetus
What is the result (generally) of teratogenic exposure at:
&bull; First 2 weeks after conception
&bull; 2-8 weeks
&bull; > 8 weeks
&bull; All-or-none
&bull; Organogenesis defects
&bull; Growth defects (but malformation is unlikely)
What are the two common maternal illnesses that can lead to teratogenic effects?
1. DM
&bull; 3x increase in malformations (related to poor glucose control)
&bull; Caudal regression syndrome (underdevelopment of lower extremities)...

2. PKU
&bull; Exposure of fetus to high levels of phenylalanine
&bull; Mental retardation, microcephaly, heart defects
What is the effect of ionizing radiation at 0-2 weeks, 2-5 weeks, >5 weeks?
&bull; 0-2 weeks: all-or-none
&bull; 2-5 weeks: CNS damage
&bull; >5 weeks: growth retardation
What is the effect of thalidomide on fetuses?
Phocomelia
What is Fetal Warfarin Embryopathy?

What enzyme is likely to be involved?

What disease does it present similar to?
When exposed b/w weeks 6-9:
&bull; Calcification of of cartilagenous epiphyses
&bull; Shortening of distal phalanges
&bull; CNS, cardiac abnormalities

Arylsulfatase E

X-linked recessive Chondrodysplasia Punctata
What is the difference between major and minor anomalies?
Major - compromise survival
Minor - no functional significance, usually cosmetic
What are the 3 types of anomalies?
1. Malformation: poor formation of tissue
2. Deformation: unusual forces on tissue
3. Disruption: breakdown of normal tissue
What are the 3 flavors of malformation?
1. Incomplete: common
2. Redundant: uncommon (polysyndactyl)
3. Aberrant: rare (mediastinal thyroid)
What is the major cause of congenital malformations?
Idiopathic (unknown)
What are the types of deformations?
1. Extrinsic mechanical
2. Intrinsic
&bull; Malformational (ex. spina bifida leads to club foot)
&bull; Functional (ex. neurological or muscular damage)
Which anomalies can be corrected spontaneously or by posture?
Deformations
Which anomalies will occur during organogenesis? Which will occur during the fetal period?
Malformations

Deformations, disruptions
What is a sequence?
Collection of anomalies that occur together
What is the Robin sequence?
Malformation (mandibular hypoplasia)

OR

Deformation (mandibular constraint)

leads to failure of tongue descent

leads to cleft palate
------
Malformation: Treacher-collins
Deformation: Pierre-Robin

*recall that deformation can see spontaneous correction - better prognosis
What is the significance of minor anomalies?
Often associated with major anomalies

Can aid in diagnosis (i.e. sequences)
What are the goals of gene therapy?
1. Alter/supplement function of mutated gene
2. Directly alter/repair mutated gene
3. Provide a gene that adds missing function of or regulates another gene
What determines success of gene therapy?
1. Delivery to appropriate cell
2. Proper expression
What are important factors to consider when creating a gene therapy?
1. Short/long-term
2. Tissue specificity
3. Regulation of expression
4. Quantity of cells to hit
5. Ex vivo or in vivo?
What is pseudo-typing?
Changing the viral envelope protein to change tropism
What are the 5 primary types of receptors?
1. Intracellular (e.g. steroid)
2. Trasmembrane, intrinsic enzyme activity (e.g. EGFR)
3. Trasmembrane, auxiliary enzyme (e.g. cytokine R)
4. Ligand- or voltage-gated ion channel (e.g. ionotropic glutamate)
5. GPCR (&beta;-adrenergic)
What is positive and negative allosterism?
Positive allosterism - enhance agonist-mediated response but do not independently activate receptor

Negative allosterism
&bull; Agonist-dependent: reduce agonist-mediated signalling
&bull; Agonist-independent: like inverse agonist
What is the therapeutic index?
Toxic ED<sub>50</sub>/Good EC<sub>50</sub>
What are the properties of competitive antagonists? Non-competitive antagonists?
Competitive:
&bull; Bind irreversibly to same site as agonist
&bull; &darr; potency
&bull; Efficacy unchanged
&bull; Effect depends on [agonist] and [antagonist]

Non-competitive:
&bull; Bind irreversibly to same site as agonist OR to allosteric site
&bull; Potency unchanged
&bull; &darr; efficacy
How are agonist-dependent non-competitive antagonists described?
Inhibition-response relationship: measure response while varying concentration

Look for IC<sub>50</sub>
How are agonist-independent non-competitive antagonists (~inverse agonists) described?
Schild analysis

&bull; Plot [agonist] vs. response
&bull; Dose ratio = &uarr; [agonist] required to give same response with certain [inverse agonist]
What are spare receptors?
Maximum physiologic response can be achieved without 100% binding of receptors.

Commonly observed with receptors that do not mediate the DIRECT effect
ie. GPCR, but not voltage-gated ion channel
What happens if there are spare receptors and you give a low concentration of non-competitive antagonist?
Looks like competitive antagonist.

Keep increasing [ ] and THEN Effect<sub>max</sub> will drop.
What are inverse agonists?
Drugs that reduce activity of <b>constitutively active</b> receptor

Competitive/Non-competitive
What are the properties and types (6) of intracellular receptors?
Lipophilic ligands
Receptors are transcription factors

1. Thyroid hormone receptor-like
2. Retinoid X receptor-like
3. Estrogen receptor-like
4. Nerve growth facter 1B-like
5. Steroidogenic factor-like
6. Germ cell nuclear factor-like

E.g. tamoxifen (SERM)
What are the properties and types (6) of enzyme-linked receptors?
Ligands - extracellular proteins
Slow!

1. Receptor tyrosine kinases
&bull; Growth factors often ligands
2. Tyrosine kinase-associated receptors
&bull; Cytokines
&bull; Dimerization required
3. Receptor like tyrosine phosphatases
&bull; Unknown ligand
4. Receptor serine/threonine kinases
&bull; TGF-&beta;, BMP, activin
5. Receptor guanyl kinases (cGMP)
&bull; Natriuretic peptides
6. Histidine kinase-associated receptors
&bull; Bacterial chemotaxis
What are some ligands for receptor tyrosine kinases (RTKs)?

How are receptors activated?
Insulin, EGF, IGF, PDGF, VEGF...

1. Ligand binds
2. Receptor dimerizes
3. Kinase phosphorylates tyrosines
4. Downstream signalling
What receptors must dimerize to signal?
RTKs
Tyrosine kinase-associated receptors
What are the properties and types (3) of ligand/voltage-gated ion channels?
Receptors have semipermeable channels
Ligands are small molecule NTs

1. Cys loop receptors
&bull; Pentameric
&bull; GABA<sub>A</sub>, GABA<sub>B</sub>, glycine
&bull; Nicotinic AChR, 5-HT<sub>3</sub> (serotonin)

2. Glutamate receptors
&bull; Tetrameric
&bull; AMPA, NMDA, kainate

3. ATP receptors
&bull; Trimeric
&bull; P2X
How does memantine work?
Uncompetitive antagonist - binds in the ion pore, so requires it to be active first.

Thought to prevent some excitotoxicity in neurons from constitutively open NMDA receptors.
What are some channelopathies?
1. Cystic fibrosis - CFTR transporter
2. Long QT - KCNQ1, other K

Also epilepsy, migraine, hyperekplexia
What are the properties and types (3) of the GPCRs?
Much slower signalling than ion channels

1. Family A (Rhodopsin-like)
&bull; Rhodopsin
&bull; Biogenic amine receptors
&bull; Olfactory receptors

2. Family B (secretin-like)
&bull; GI peptides
&bull; Corticotropin releasing hormone (CRH) receptor

3. Family C (metabotropic)
&bull; mGluRs
&bull; GABA<sub>B</sub>
&bull; Calcium-sensing Rs
Give an example of tachyphylaxis.
&beta;-adrenergic receptor
- &beta;-arrestin binds, prevents association with Gs

= Loss of receptor function (receptor-dependent)
What's the different between pharmacodynamics and pharmacokinetics?
Pharmacodynamics - what drug does to the body

Pharmacokinetics - what body does to the drug
What are dialyzing membranes?
Most capillaries, renal glomerules

Fenestrated!
What are the transporters of primary active transport? What are the properties?
ATP-binding cassette (ABC) transporters

Unidirectional efflux
Saturable
Can be inhibited by metabolic inhibitors
What is MDR?
Multidrug resistance - p-glycoprotein, modulates drug permeability
1. Prevents GI absorption
2. Role in BBB
3. Excretion in bile, urine

Can be overexpressed in cancers
How is secondary active transport different from primary?
Doesn't use ATP
Couples transport of one solute going uphill with another going downhill.

Ex.
SLC5A - Na-dep Gluc symporter
SLC8 - Na/Ca exchanger
What type of transport is targeted by SSRIs?
SLC6A4 - secondary active transport
What is vectorial transport?
Transferring solutes/drugs from one side of cell membrane to the other in polarized cells (e.g. endothelial, epithelial)

Often polarized expression of transporters! (e.g. SLC5A brings in glucose against [], SLC2A2 takes it downhill on other side)
What are the two types of endocytosis?
Fluid-phase - like pinocytosis
Receptor-mediated - clathrin-coated pits
What are some proteins involved in intercellular junctions? Which are involved in paracellular transport?
Claudins
Occludins
Zo proteins
JAMs (junctional adhesion molecules)

Claudin-5 - BBB, <1nm pore
Claudin-2 - ~5nm pore in other tissues for paracellular transport
What is the Hendersen-Hasselbach equation?
log([base]/[acid]) = pH - pKa
What are the parenteral and non-parenteral routes of administration?
Parenteral:
&bull; IV
&bull; IM
&bull; SubQ

Non-Parenteral:
&bull; Oral
&bull; Rectal
&bull; Inhalation
&bull; Topical
How does food affect drug absorption? Give 2 examples.
Food increases gastric emptying time (longer gastric residence)

1. Didanosine
Acid-labile: take without food to decrease residence time

2. Nitrofurantoin
Delayed absorption prolongs dissolution time = &uarr; bioavailability (but &darr; Cmax) so take with food
What is the major metabolizing enzyme in the gut?
CYP3A4
How does grapefruit juice affect drug absorption?
1. Inhibits CYP3A4 (irreversible)
2. Inhibits OATP (organic anion-transporting polypeptide)
-Reversible
How do we know that grapefruit juice only affects GI drug metabolism?
Felodipine study: IV + grapefruit juice = no change, but PO + GFJ = &uarr; F
How much first-pass metabolism is present with rectal dosage forms?
~50% (some bypasses liver)
What determines initial distribution of drugs? Final distribution?
Initial: blood flow - brain, heart, liver, kidneys

Final: affinity of drug for tissue
&bull; Lipid - all organs, adipose
&bull; Ionic - plasma, interstitial compartments
What 2 protein subfamilies of ABC transporters make up the biochemical barrier of the BBB?
Pgp (MDR/ABCB1)
Multidrug resistant associated protein (MRP1, ABCC1)
Why can intrathecal injections be used to treat brain infections?
There is no good brain-CSF barrier (no tight junctions)
Why is albumin important in distribution?
Drugs bound to albumin are pharmacologically inactive and can affect transport to tissues.

Must adjust for albumin level
How can redistribution be dangerous (ie. thiopental, propofol)?
Gets stored in fat cells, released when drug is no longer given - can OD.
Why are lipid drugs hard to excrete renally?
They are reabsorbed after being filtered in the glomerulus
What is the major mechanism of excretion of ionized drugs (especially anions)?
Tubular secretion (proximal tubule)
What are the transporters in renal secretion?
Pgp
MRPs
OATs
Solute carrier transporters:

1. Weak bases (histamine)
- Procaine
- Quinine

2. Weak acids (uric acid)
- Penecillin
- Probenecid

ex. give probenecid - &darr; secretion of penicillin!
Where does reabsorption occur? What happens to ionic drugs?
Distal tubule

Ion trapping - ionic drugs excreted in urine
How can renal excretion of a weak base like amphetamine be increased?
Acidify the urine with arginine-HCl! Makes the charged form more prevalent (ion trapping)

Works for weak acids too - alkalinize urine!
How do drugs enter the bile?
Lipid diffusion
Small molecules through fenestrae
Transporters
What is the enterohepatic cycle? What is it's effect on elimination?
Glucuronides formed in liver
Enters bile (MW >500, esp.)
Glucuronides can be cleaved in colon, some drug reabsorbed

Delays drug excretion, prolongs drug action

E.x. birth control + anbitiotic - disrupts cycling, lower plasma levels, unexpected pregnancy!
What is cometabolism?
Enzymes that metabolize endogenous agents also metabolize xenobiotics
What are the types of drug metabolism?
Phase I: functional group
&bull; Hydrolysis
&bull; Oxidation
&bull; Reduction

Phase II: Conjugation to make more polar
&bull; Sulfation
&bull; Acetylation
&bull; Glucuronidation
&bull; Glutathione-idation
&bull; Methylation
What does caffeine do at low doses? High doses?

*boards question
Adenosine antagonist

Inhibits phosphodiesterase = &uarr; cAMP
What is one of the reasons alcoholics have many metabolic disorders?
NADH excess! (from dehydrogenase activity)
Why is ingesting foods with tyramine (cheese) with MAOIs a bad idea?
Tyramine releases NE which is inactivated by MAO. If inhibited, hypertensive crisis.
Which CYP enzyme has many polymorphisms? What is the distribution?
CYP2D6

Trimodal: poor, extensive, ultraextensive metabolizers
How should you adjust the dose of codeine in an Ethiopian patient with CYP2D6 ultraextensive metabolism?
DECREASE dose - metabolism of codeine is to activate it to morphine!
What are the two polymorphisms involved in ethanol metabolism?
Alcohol dehydrogenase overactivity

Aldehyde dehydrogenase underactivity
Fast and slow acetylators
Just know they exist.
Glutathione null genotypes
Mu
- Linked with cancers

Theta
- Adverse effects with cancer chemotherapeutics
What is the mechanism responsible for the increased toxic effects of acetaminophen in the presence of alcohol?
EtOH uses and INDUCES CYP2E1, which is responsible for the toxic metabolites of acetaminophen
What can happen to a patient on oral contraceptives who also takes St. John's Wort?
SJW induces CYP3A4 = can get preggo!

Also induces CYP2E1
What are the results of non-instantaneous drug distribution?
1. Termination - may be fast or slow, depending on when effect is terminated (rapid distribution phase or slow elimination phase)

2. Bolus, infusion only appropriate for drugs with high margin of safety
(initial volume < Vd)

3. Accumulation during continuous administration (effect of infusion duration and distribution)
What is the mechanism of inactivation of effect of IV anesthetics?
Redistribution to lean and fat tissues.
Why is thiopental typically a short-acting drug? How does increasing its dose make it a long-acting drug?
Falls below min effective conc (MEC) during rapid distribution.

If you increase dose, it will hit slow elimination phase and take a long time to fall below MEC
How does drug distribution change with redistribution of cardiac output (ie. hemorrhage)?
Ex. in hemorrhage, decreased blood flow to muscles, GI, increased to brain

Different distribution = different effects
What is hysteresis with pharmacodynamics?
There is a lag time between plasma concentrations and pharmacologic effect.

&rarr; delay in onset and offset

Use biophase model to adjust and collapse hysteresis loop
Which of the &beta;-lactams can you give orally?
Penicilin V (1)
Oxacillin (2)
Amoxicillin (3)

Cephalexin (1st)
Cefadroxil (1st)
Cefaclor (2nd)
Clavulonic acid cannot inhibit WHICH &beta;-lactamase? Why?
Type B - is a Zn enzyme that does not form a penicilloyl enzyme complex.
Why is vancomycin useful against staphylococcal enterocolitis or GI superinfection?
Not absorbed orally!
Describe the steps of bacterial protein synthesis.
1. Initiation
&bull; mRNA binds 30S + F3
&bull; fMet-tRNA binds + F1, F2
&bull; 50S binds, F1,2,3 released

2. Elongation
&bull; aminoacyl-tRNA + elongation factors binds
&bull; Peptide bond formed
&bull; Translocation

3. Termination
How are penicillins and aminoglycosides synergistic?
Penicillins degrade cell walls, allowing aminoglycosides to enter
= bactericidal!
What are the two fluroquinolones NOT excreted renally?
Moxifloxacin
Trovafloxacin
Which drug inhibits mammalian CYP450 most?
Fluconazole
Itraconazole
Ketoconazole
Flu < Itra < Keto
What does Nifurtimox treat?

Piperazine?
Trypanosoma cruzi

Ascaris lumbricoides
What is damaged non-dividing tissue replaced with?
Scar tissue (collagen)
How do stem cells hold on to tritiated-thymidine?
Stem cells retain the maternal strand
What determines the fate of stem cells?
Tissue microenvironment
What is the main function of:
1) TGF
2) VEGF
3) HGF
4) EGF
5) FGF
6) Prolactin
1) TGF - epithelial cells, liver regen
2) VEGF - angiogenesis
3) HGF - liver growth and function
4) EGF - epithelium proliferation
5) FGF - mitogen for fibroblasts and epithelium
6) Prolactin - proliferation/differentiation of breast epithelium
What is a metaplasia?
Dysplasia?
Neoplasia?
Metaplasia - change in differentiation from one type to another (reversible)

Dysplasia - abnormal hyperplasia, propensity for malignancy (sometimes reversible)

Neoplasia - uncontrolled growth and cellular autonomy (irreversible)
What are some examples of metaplasias?
1. Squamous metaplasia: glandular to squamous
- pseudostratified ciliated columnar to squamous (in respiratory tract)

2. Glandular metaplasia:
Squamous to glandular
(e.g. Barrett's esophagus - stratified squamous of esophagus to columnar intestinal)
What are the cellular responses to stress?
1. Ignore - quiescence
2. Adapt
3. Divide and differentiate
4. Die (necrosis, apoptosis)
What is coagulative necrosis?
Trigger: Ischemia, toxins

Scarring and removal (time-dependent)
What is liquefactive necrosis?
Form of coagulative necrosis

Necrotic area rapidly liquefied (extensive lysis)

BRAIN vascular injury &rarr; cells digest dead tissue but it's not scarred, refills with cysts and fluid
What is caseous necrosis?
Associated with caseating granulomas

Center of lesions undergoes liquefaction

Think TUBERCULOSIS
Caseous = cheese
What is fat necrosis?
Often fatty tissue after trauma; often tissue near pancreas

Pancreatic damage &rarr; acinar cells leak enzymes &rarr; digest fat (saponification) to FAs which chelate metal ions (esp Ca2+)

Can become hypocalcemic! (tetany, death)
What is tumor necrosis?
1. Poor blood supply
2. Bad angiotic factor production
3. Apoptotic factor produced by dying tumor cells
4. Therapy
What factors indicate irreversible cell injury?
Mitochondrial densities
Blebbing
Spilling of lysosomal enzymes
Rupture of plasma membrane
What are the main causes of necrosis?
1. ATP depletion
2. Loss of Ca2+ homeostasis (influx &rarr; activates degrading enzymes)
3. Free radicals (and DNA/protein damage)
4. Loss of membrane permeability
What are clinically important proteins released during necrosis?
Cardiac:
cTNI - 4-6hr of MI
Lactic dehydrogenase (LD): 1 = cardiac, 2 = skel muscle
If LD1 > LD2 = cardiac necrosis

Liver: ALT/AST

Pancrease: Amylase
What is a major difference morphologically between apoptosis and necrosis?
Necrosis - swelling and explosion

Apoptosis - shrinking, controlled; no inflammation!
What are the two pathways of apoptosis?
1. Intrinsic:
&bull; p53-triggered &rarr; Bax/Bak
&bull; Cytochrome c release from mitochondria
&bull; Activation of caspases (9, then 3,6,7)

2. Extrinsic
&bull; Triggered by pro-apoptotic receptors: Fas, TNF
&bull; Fas-assocated death domain (FADD)
&bull; Caspases 8, 10 &rarr; 3,6,7
What is fatty liver?
Too many FAs produced by liver - can't cope - puts them into droplets (accumulates)

Alcoholic
Obesity - metabolic syndrome
DM

Can see Mallory bodies

REVERSIBLE!
What are the 6 ways FA concentration in the liver can increase?
1. &uarr; FA uptake
2. &uarr; FA synthesis
3. &uarr; triglyceride synthesis

4. &darr; FA oxidation
5. &darr; Apoprotein synthesis
6. &darr; Lipoprotein secretion
What is the unfolded protein response (UPR)?
Response for misfolded proteins in the ER (unlike ubiquitination in cytoplasm)

Detector: BiP, recognizes mutant proteins and binds sensors

Sensors: IRE1, ATF6, PERK - signal

Effectors - UPR
&bull; Fast feedback: shut down ribosomal translation
&bull; Slow feedback: kinases, transcription factors

Effector: ERAD
&bull; If protein can't be refolded correctly, ERAD sends them to proteosome
&bull; May also get rid of foreign agents in ER

Effector: Apoptosis
&bull; Follows prolonged UPR response
What is lipofuscin?
Seen during aging process

Undigested cellular material accumulating in endstage liposomes
What are two other pigmented substances that can deposited in cells?
Melanin

Hemosiderin
&bull; Bruising
&bull; Poor vascular perfusion (nutmeg liver)
&bull; Hemochromatosis (mutation) - body takes in too much iron = heme deposition
What is the result of precipitated uric acid?
Gout, hyperuremia
What are exogenous accumulations?
Anthracosis - Carbon particles in airway, lung (ie. smoker, coal miner)
&bull; Anthracotic pigment
&bull; Can lead to cell death, fibrosis, cancer

Tattooing: increased risk of HBV, HCV, HPV
What are the characteristics of calcification?
1. Dystrophic
&bull; Calcification in dead and disease tissue (due to net negative charge in dead cells)
&bull; Normal serum levels
&bull; Psammoma bodies - tombs for cancer cells
ex. atherosclerosis

2. Metastatic
&bull; Calcification in normal tissues with hypercalcemia
&bull; Prefers net negative charge: lung, kidney, stomach
What is pathological ossification?
Myositis ossificans

Metaplasia following trauma
- Deposition of cartilage or bone on skeletal muscle
What is the Hayflick limit?
Point at which cells stop growing and senesce.

Eventually will either die or become malignant.
What are telomeres?
Caps on the end of all DNAs.

3-10kbp

Preserves DNA upstream from damage in replication

Shorten with age - RNA primer on lagging strand gets degraded = shorter telomere
&rarr; seen as biologic clock
What cells' telomeres do not shorten?
Stem cells
Reproductive cells (ie. spermatogonia)

Controlled by telomerase enzyme

Can see increase in telomerase in cancer
What is Werner syndrome?
Defective DNA helicase
= defective DNA repair
= faster aging!
What are the methods of immune tolerance (ie. specific inhibition of the immune response to self)?
Central:
&bull; Clonal deletion - in thymus of T/B-cells that are self-reactive
&bull; Clonal anergy - T-cell that is not co-stimulated (by B7) is turned off = anergic

Peripheral:
&bull; Clonal anergy
&bull; Sustained activation of T-cells = apoptosis
&bull; Suppression (Th2) - IL-4, IL-10, <b>TGF-&beta;</b>
What are the pathologic mechanisms of autoimmunity (4)?
1. Immunologic -
&bull; Upregulation of B7
&bull; Defective Fas-FasL apoptosis
&bull; &darr; Th2 suppression activity
&bull; Polyclonal B-cell activation
&bull; Cross-reactive antigens
&bull; Cryptic, sequestrated antigens

2. Genetic - prevalence of certain diseases in certain HLA haplotypes (incl. complement def.)

3. Viral - mitogens (B-cell activation), neoantigens (like haptens), T-cell suppression

4. Hormonal - female predilection, pregnancy
What is the likely effect of testosterone on autoimmune disease?
Inhibits! (mice experiments...)
Graves' Disease
&bull; Pathophysiology?
&bull; Symptoms
1. Endocrinopathy
&bull; Hyperthyroid - thyroid stimulating Igs (TSIs) bind TSH receptor &rarr; increase cAMP &rarr; make T3, T4
&bull; Anti-TPO, anti-thyroglobulin sometimes present as well

2. Ophthalmopathy
&bull; Edema, wide eyes

3. Dermopathy
&bull; Non-specific edematous thickening

Increased metabolism = sweating, tachycardia, etc.

*Loss of Th2 suppressor activity
Hashimoto's Disease
1. Hypothyroid

2. Hurthle cells
&bull; Degenerated thyroid cells

3. Autoantibodies
&bull; anti-TPO
&bull; anti-thyroglobulin
&bull; anti-colloid
&bull; anti-TSH receptor (block)

Slowed metabolism = fatigue

*Loss of Th2 suppressor activity
Myasthenia Gravis
1. Anti-nAChR autoantibody

2. Associated with thymic disorders

3. Fatigue with descending paralysis/weakness

Edrophonium test - give anticholinesterase = improvement!
Pernicious Anemia
1. Decreased B12 absorption
&bull; Bone marrow defective maturation of DNA of RBCs = megaloblastic anemia
&bull; Spinal cord - myelin degeneration

2. Autoantibodies
&bull; sIgA against parietal cells = achlorhydria (&darr; HCl in stomach)
&bull; Against intrinsic factor (IF) = blocks binding of IF-B12 = can't absorb B12
OR
binding Ab = binds IF-B12 complex = not absorbed

**B12 required for DNA synthesis
Megaloblast - large nucleus (RBCs in bone marrow)
What are the transitions on the way to fibrinoid necrosis in an artery?
Inflammation
Exudation
Dissolution
Necrotizing
&rarr; Results in fibrin-like material = fibrinoid

The result of autoantibody or immune-complex trapping in blood vessel wall
Systemic Lupus Erythematosus
1. Skin rash, joint pain!! (see below)

2. Autoantibodies - ANA!
&bull; anti-dsDNA
&bull; anti-ssDNA
&bull; anti-histone
&bull; Extractable Nuclear Antigens (ENAs): anti-Sm, anti-nRNP, anti-SSa, anti-SSb

3. Symptoms
&bull; Butterfly rash; atrophy of epidermis
&bull; Arthritis
&bull; Proteinuria
&bull; Raynaud's
&bull; Pleuritis, pericarditis, endocarditis
&bull; Anemia, leukopenia, thrombocytopenia
&bull; Vasculature: onion skin appearance

4. Kidney
&bull; Various forms of lupus glomerulonephritis
&bull; Diffuse proliferative - BAD prognosis
&bull; Membranous nephritis - better prognosis

Remissions, relapses
Late stage: disfigured face like a wolf = lupus
What is unique about anti-Smith antibody?

Which other antibody is specific for SLE?

How are they detected?
Titer does not fluctuate with progress/state of disease

Specific for SLE, found in 20-30% of patients. Speckled IF

anti-dsDNA also specific for SLE. Peripheral IF. Crithidia luciliae test
What is the crithidia luciliae test?
Protozoan, has kinetoplast rich in dsDNA

Used to test for anti-dsDNA Ab
What are the patterns of immunofluorescence for nuclear antigens?
Homogeneous - DNA, histones, nRNP (not specific)

Peripheral - dsDNA, soluble NP (specific for SLE)

Speckled - ENA, partially specific for SLE; hallmark of mixed CTDs

Nucleolar - RNA - scleroderma, polymyositis-dermatositis
Which has a better prognosis: diffuse proliferative (nephritic state) or membranous nephritis (nephrotic state)?
Membranous nephritis
What is the diagnostic feature of progressive systemic sclerosis?
Scl-70 (anti-DNA topoisomerase I)
What is the pathophysiology of PSS, and what are some of the symptoms?
Lymphokines stimulate collagen production systemically (delayed-type hypersensitivity)


S<sub>x</sub>
&bull; Loss of elasticity (mouth, skin)
&bull; Vasospasm, vasculitis
&bull; Honeycomb lung
&bull; Systemic HTN (&darr; RBF)
&bull; &darr; GI absorption, peristalsis
What is CREST syndrome?
Calcinosis
Raynaud's syndrom
Esophageal dysfunction
Sclerodactyl (hardening of digits)
Telengiectasia (capillary dilation)
What is Sjorgen's Syndrome
a) Pathophysiology?
b) Symptoms?
a) SSa, SSb autoAb (ENA)

b) Painful, but can't cry
&bull; Infiltrates in secretory glands
What is polymyositis-dermatositis
Patho and Sx?
a) Anti-myoglobin, anti-tRNA synthetase autoAb

b) Inflammatory/degenerative disease of muscle/skin
What is characteristic of Mixed Connective Tissue Disease (MCTD)?
Often female with multitude of symptoms

Anti-ribonucleoproteins: +
Anti-DNA, Anti-Sm: - (if positive, then SLE)

Speckled immunofluorescence
What is Polyarteritis Nodosa?
a) Pathophysiology?
b) Symptoms?
Males!
a) P-ANCA
Perinuclear-AntiNeutrophil Cytoplasmic Antibody

Anti-MPO (found in PMNs) in microscopic form

b) Nodosa = nodules = aneurysms of large arteries

Fibrinoid necrosis, immune complexes in all 3 layers of artery
What is karyorrhexis? When do you see it?
Fragmentation of nuclear material.

Specific to lupus
What is the pathophysiology of Raynaud's phenomonon?
Cryogenic Igs precipitate in cold
= cyanosis in fingers
+ vasospasm (endothelin)
Where do immune complexes typically deposit in nephritic (diffuse proliferative) and nephrotic (membranous nephritis)?
Nephritic = subendothelial

Nephrotic = subepithelial
How does discoid lupus differ from SLE?
Localized form in skin
What are some drugs that cause lupus?
NAT slow acetylators:

Procaineamide
Hydralazine
Diphenylhydantoin
What is the triad of symptoms in Wagener's Granulomatosis?
Nasopharyngeal granuloma (nasal septum destruction)

Vasculitis (microscopic)

Necrotizing glomerulonephritis
What is the cause of Wagener's?
C-ANCA
Cytoplasmic-Anti-Neutrophil Cytoplasmic Antibody

anti-PR3 (proteinase 3) in PMNs
How is Wagener's differentiated from Polyarteritis Nodosa, immunologically?
Wagener's: C-ANCA
PAN: Both C- and P-ANCA
What is the pathophysiology of Amyloidosis?
Stach-like deposits in various organs

MECHANICAL disturbances, NOT inflammatory
What does the starch-like deposit of amyloidosis contain?
1. Fibrillary proteins (eg. AA, AL, ATTR, A&beta;<sub>2</sub>m, etc)
2. Serum amyloid-P component (SAP)
3. Proteoglycans
4. Sulfated GAGs
What are the classifications of amyloidosis?
1. Systemic
&bull; Primary - myeloma
&bull; Secondary - chronic infections, hemodialysis
&bull; Hereditary - familial mediterranean fever

2. Localized
&bull; Endocrine
&bull; Senile
&bull; Atrial
&bull; Cutaneous
What is the reason for precipitation of fibrils (amyloid)?

Where do AA and AL precipitate?
Limited proteolytic cleavage

AA - liver
AL - plasma cells
What is the difference between sago and lardaceous spleen?
Sago - amyloid around arterioles

Lardaceous - amyloid in sinusoids (looks like fat = lard)
What does congo red stain for?
Amyloid! Causes birefringence due to &beta;-pleated sheet of amyloid protein
What is the length of fibrils to be considered amyloid?
8-10 nm
What are the characteristics of benign vs. malignant tumors? Include nomenclature.
Benign:
&bull; Resemble tissue of origin
&bull; &darr; mitosis
&bull; Expansive with encapsulation

"cell of origin" + "-oma"
&bull; Adenoma = glandular epithelium
&bull; Papilloma = squamous epithelium

Malignant
&bull; May/may not resemble tissue of origin
&bull; &uarr; mitosis
&bull; Infiltrative, invasive

"cell of origin" + "-sarcoma"
&bull; Rhabdomyosarcoma = striated muscle
&bull; Leiomyosarcoma = smooth muscle

For epithelium: "cell of origin" + "-carcinoma"
&bull; Adenocarcinoma
What are the exceptions to the nomenclature rules?
Melanoma = malignant (benign = nevus)

Lymphoma = malignant

Teratoma = benign (totipotent stem cells)
What is differentiation?
Extent to which parenchyma of tumor resembles normal cells

Well-differentiated = typically benign
What is anaplasia? What are some characteristics?
Lack of differentiation

Pleomorphism = multiple cell types, sizes, shapes

Hyperchromatic
&uarr; nuclear:cytoplasmic ratio
Abnormal mitosis
Loss of polarity
What is dysplasia?
Disordered growth - loss of architectural orientation
What is "carcinoma in situ"?
Dysplasia affects entire epithelium but confined to basement membrane
What is desmoplasia?
Extensive collagen produced in stroma (like in breast adenocarcinoma)
What affects tumor growth? How is it related to differentiation?
&bull; Doubling time
&bull; Fraction of tumor cells in replicative pool
&bull; Rate of apoptosis

Growth &alpha; 1/differentiation
(well-diff. = slow growing)
What are the steps for invasion of tumor cells?
1. Loosening of intracellular junctions (E-cadherins)

2. Degradation of ECM - MMPs (proteolytic)

3. Changes in attachment of tumor cells to ECM proteins

4. Locomotion and migration - ratcheting mechanism
What is metastasis? What are the 3 pathways?
Spread of malignant tumor clels to distant site

1. Direct seeding of body cavities/surfaces (peitoneal, pericardial, pleural, etc.)

2. Lymphatic

3. Hematogenous - affects liver, lungs especially (thin vessels)
What is tumor grading?
Degree of differentiation of tumor cells.

I &rarr; IV (well-diff. &rarr; not well)
What is tumor staging?
Degree of spread and size

UICC:
T = primary tumor
N = regional lymph nodes
M = metastasis
What is the monoclonal origin of tumors?
Derived from a <u>single cell</u>!
What are the types of oncogenes? Give an example of how each can lead to cancer.
1. Extracellular growth factors
&bull; PDGF overexpression

2. Growth Factor Receptors
&bull; Erb2 (her2/neu) expression
&bull; EGFR - 80% of squamous cell carcinoma of lung

3. Signal transducing proteins
&bull; RAS: codes for p21 GTP-binding proteins, turned off by GTP-ase activating protein (GAP)
&rarr; mutant p21 always on, resistant to GAP

4. Non-receptor tyrosine kinase
&bull; c-ABL - chromosomal translocation in CML

5. Nuclear transcription factors

&bull; C-myc: translocated adjacent to Ig heavy chain = dominant clone of B-cells (Burkitt's Lymphoma) due to oversupply of c-myc transcription factor
&bull; N-myc: neuroblastoma

6. Cyclins and CDKs
&bull; Overexpression of Cyclin D
&bull; Amplification of CDK4
&bull; In charge of G1&rarr;S transition
What is a tumor suppressor gene (TSG)? What is the inheritance?
Genes that code for proteins that STOP cell proliferation (anti-tumor)

Each cell MUST have both alleles knocked out. But if organism if heterozygous, there is a much higher chance to lose second allele in an individual cell.
&rarr; Susceptibility is dominant inheritance
Retinoblastoma
Malignant tumor from immature retinal neurons

Leukocoria - appearance of a white pupil (white mass = tumor)

Susceptibility is autosomal dominant (see TSG flashcard)
&bull; Homozygous normal requires 2 mutations (one in each Rb allele)
What is the normal function of Rb?
Prevents transition to S phase

Sequesters E2F

Phosphorylation (*by Cyclin D/CDK4) releases E2F...S phase!
What are the possible mutations in the G1/S checkpoint?
1. Rb
2. Cyclin D
3. CDK4
4. P16/Ink4a - CDK inhibitor
What is Li-Fraumeni syndrome?
1 defective p53 gene
What does p53 do?
Senses DNA damage

Upregulates p21 (CDK inhibitor), GADD45 (DNA fixer), bax (apoptosis)

Activates transcription of mir34 (miRNA) - inhibits growth promoting genes (myc, CDK4), anti-apoptosis genes (BCL-2)
What are BRCA1 and BRCA2?
TSGs in breast, ovarian cancer

Transcriptional gene regulation, DNA repair
What is adenomatous polyposis coli (APC)?
TSG

Seen in Familial Adenomatous Polyposis - defective APC allele - metric fuckton of polyps

Promotes degradation of &beta;-catenin, which can activate genes involved in proliferation (c-myc, Cyclin D, etc.)
What is WT-1?
Wilm's Tumor (TSG)

WT-1 gene essential for normal urogenital development

Can get renal tumors
What is NF-1?
Neurofibromatosis type 1 (TSG)

Neurofibromin - same as GAP, inactivates RAS-GTP
What is Von Hippel Lindau (VHL)?
TSG

Ubiquitin ligase - regulates hypoxia response genes in angiogenesis
What is Smad4?
TSG

90% of pancreatic adenocarcinomas, many colon adenocarcinomas too

Mediates TGF-&beta; suppression of epithelial cell growth
What genes regulate apoptosis?
Bcl-2 - anti-apoptotic &rarr; overexpression in many B-cell lymphomas

Bax - "death gene"
Ratio of bax + bad/Bcl-2 + Bcl-xL = determines apoptosis
What is Hereditary NonPolyposis colon cancer (HNPCC)?
Inherited predisposition to colon cancer

Defects in DNA mismatch repair genes (MSH2, MLH1, PMS1, PMS2)

Usually heterozygous - second hit occurs in colonic epithelium
What is xeroderma pigmentosum?
Autosomal recessive

Mutation in one of several nucleotide excision repair (NER) genes

Extreme photosensitivity, SUPER HIGH risk of skin cancer
What is ataxia telengiectasia?
Hypersensitivity to ionizing radiation

Cerebellar degeneration
Oculocutaneous telengiectasia
Immunological abnormalities

Predispose to cancer
What % of cancers are environmentally induced?
85-90%
What is initiation in carcinogenesis?

Is it sufficient to produce tumorigenesis?
Irreversible DNA damage in a critical target gene (oncogene, TSG, etc.)

NO
What is promotion in carcinogenesis?

Is it reversible?
Process by which initiated cell is selected for and amplified into benign tumor/preneoplastic condition

Reversible initially
What is progression in carcinogenesis?
Benign &rarr; invasive potential (carcinoma in situ)

Then, subpopulation of these cells selected for ability to metastasize
What is the electrophilic theory of carcinogenesis?
Direct-acting carcinogens - already electrophilic - bind DNA, RNA, proteins

Indirect-acting carcinogens must be metabolically activated into electrophilic species
What is a mutagen?
An agent that can permanently alter genetic constitution of the cell

Most carcinogens are mutagens, and vice versa
What is the Ames test?
Test for chemical carcinogens

Uses S. typhimurium that CANNOT produce histidine (can't grow on normal agar)

If agent causes bacteria to make histidine (mutated) = mutagen!
What is an example of a direct-acting chemical carcinogen?
Nitrogen mustard
Found in chlorambucil, melphalan (anti-cancer drugs)
What are some examples of indirect-acting chemical carcinogens?
Polycyclic aromatic hydrocarbons
&bull; Incomplete combustion of organic material
&bull; Benzo(a)pyrene

Aflatoxins
&bull; B1 - one of the most potent liver carcinogens
&bull; Aspergillus flavus - more common in Africa, Asia
&bull; Produces p53 point mutation ("footprint") in HCC - not seen in HCC in USA

Nitrosamines
&bull; Implicated in GI cancers
&bull; Nitrates (food) + acid = nitrosamines
What are some promoters in human cancers?
Cigarettes
UV
Hormones
Viral infection
High fat diet
What are the events that occur in colonic mucosa that result in adenocarcinoma?
1. APC gene mutation
2. K-Ras point mutation
3. p53 mutation
SMAD mutation

Telomerase overexpression, hypermethylation
What 3 cancers are caused by UV exposure?
Melanoma
Basal cell carcinoma
Squamous cell carcinoma
What cancers are associated with asbestos exposure?
Malignant mesothelioma
Lung cancer
What cancers does HPV cause?

How does it do this?
Papilloma (benign squamous cell cancer)

Cervical carcinomas (often with high-risk HPV - 16, 18)

E6 expression - binds and degrades p53
E7 - binds Rb (displaces E2F)
What cancers are associated with EBV?
(E. Thomas) Hodgkin disease
Burkitt's Lymphoma
Very Nasopharyngeal carcinoma
How does HBV cause cancer?
Mutation-favored environment from hyperplasia from chronic liver injury

X protein - binds p53
What cancer is linked to H. Pylori infection?
Gastric lymphomas of MALT ("Maltomas")

Gastric adenocarcinomas
What are paraneoplastic syndromes?
Effects not attributable to invasion or metastases (not local effects):
&bull; Fever
&bull; Weight loss (cachexia)
&bull; HyperCa (parathyroid hormone-related protein secreted by tumor)
&bull; Hypercoagulable state
&bull; Neurologic symptoms
What is the difference between edema and inflammatory edema?
Transudate (low in protein)

vs.

Exudate (high in cells)
What is Virchow's triangle?
Factors promoting thrombosis:
Endothelial injury
Abnormal blood flow
Hypercoagulable state
What is a paradoxical embolus?
One that starts in the venous system and gets systemic (often through septal defect or PDA)
Why can't a group A patient get a group O organ?
Organ anti-A will react with patient's A antigen (like graft-vs-host)
What is Neonatal Alloimmune Thrombocytopenia?
Mother's IgG antibodies against fetal platelets

Human Platelet Antigen (HPA)
1a = most common (on GPIIIa)
What is Hemolytic Disease of the Newborn?
Maternal IgG against fetal RBCs

Antigens:
&bull; A, B - mild
&bull; Kell, Duffy, Kidd, ... = severe alleles in each one
&bull; D in Rh system = most severe

Rh(D-) mom with Rh(D+) baby
&bull; 15% chance of anti-D in mom each time

Treat with anti-D IVIG
Does ABO incompatibility matter in newborns receiving heart transplants?
NO!

Too early to produce antibodies

But...may require careful monitoring, immunosuppression

Don't really start making anti-A/B antibodies even after they grow up
What are some strategies to deal with ABO incompatible organ transplants?
1. Donor exchanges
2. A<sub>2</sub> subgroup = milder
3. Japan method:
&bull; Remove ABO antibody
&bull; Immunosuppression
&bull; Inhibit antibody production
&bull; Monitor