• 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

1/670

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;

670 Cards in this Set

  • Front
  • Back
What does etiology stand for and what are the two major classes?
study of the cause of disease

Intrinsic (genetic)
Aquired
What is hypertrophy and what is a classic example of a cell that often undergoes hypertrophy?
increase in cell size

example: cardiac myocyte
What is hyperplasia and what are the two main types?
increase in cell number

Physiologic hyperplasia - normal hormonal (at puberty) and compensatory (due to damage)

Pathological Hyperplasia - abnormal (still responds to regulatory mechanism)
In what organ do hyperplasia and hypertrophy happen without being considered pathologic?
Gravid uterus
What is atrophy and what are some of the common causes?
decrease in cell size

decreased workload, denervation, decreased blood supply, aging, inadequate nutrition, loss of endocrine stim, pressure
What is the main mechanism of atrophy and how does this work in the cell?
imbalance b/w protein synthesis and degradation (proteolysis)

- proteolysis can be caused by lysosomes or the ubiquitin-proteasome pathway
What is a typical morphologic finding in a cell undergoing atrophy?
autophagic vacuoles (contain degraded cell components)
What is metaplasia, by what mechanisms does it occur, and what is the classic example?
change is cell type due to the reprogramming of tissue stem cells

example: Barrett's esophagus
Which factors affect the programming of cells undergoing metaplasia and what can persistent metaplasia lead to?
cytokines, growth factors, extracellular matrix components

persistent exposure to these factors may cause malignant transformation of metaplastic epithelium
What cellular characteristics may be seen in REVERSIBLE cell injury?
loss of microvilli, cell swelling, blebs, swelling of the mitochondria and ER, clumping of chromatin, autophagic vacuoles
What additional cellular characteristics may be seen in cells once they undergo IRREVERSIBLE cell injury?
in addition to Loss of microvilli, cell swelling, blebs, swelling of the mitochondria and ER, clumping of chromatin, autophagic vacuoles . . . .

loss of ribosomes from ER, kysosome rupture, myelin figures, nuclear condensation/fragmentation, amorphous densities in swollen mito
Which characteristic of cell injury is especially indicative of an IRREVERSIBLE process?
rupture of the lysosomes
How does the end result of necrosis differ from that of apoptosis?
necrosis: enzymatic digestion and leakage of cellular contents

apoptosis: phagocytosis of apoptotic cells and fragments
Is apoptosis a reversible process?
no
What are the four major abnormalities that can lead to cell damage?
decrease in ATP
membrane damage
increase in intracellular calcium
reactive oxygen species
How does ATP depletion cause cell death?
- decrease in Na/K pump = cell/organelle swelling
- increase in anaerobic glycolysis = decrease in pH (via lactic acid) causing clumping of chromatin
- detachment of ribosomes = decreased protein synthesis and lipid deposition
What causative agents can lead to mitochondrial damage?
increased cytosolic calcium, oxidative stress, lipid perioxidation
Which compound is associated with mitochondrial death?
cytochrome C (causes mitochondrial permeability transition - MTP)
What enzymes are activated due to increased cytosolic calcium and what is each associated with?
- ATPase --> decreased ATP
- Phospholipase --> decreased phospholipids = memb damage
- Protease --> disruption of membrane and cytoskeleton proteins
- Endonuclease --> nucleus chromatin damage
How is oxidative stress defined?
imbalance b/w free radical-generating and radical scavenging systems in the cell
What effects do reactive oxygen species have on the cell?
membrane lipid peroxidation (attack the unsat. FA in cell memb), DNA fragmentation (nuclear and mito), protein damage (oxidation of a.a. residues, formation of S-S bonds b/w proteins, protein fragmentation, enhance protein degradation)
What defines a free radical?
molecule with single unpaired electron in outer orbit which is highly reactice and propagates to create more free radicals in new compounds
What can cause the formation of free radicals?
absorbant radiant energy (UV, X-ray), drug and xenophobic metabolism, normal metabolic processes (energy metab., inflammation, intracellular oxidates), catalyzation by transition metals, nitric oxide
How do free radicals cause damage to lipids and proteins, specifically?
lipids - attack unsaturated fatty acids in the cell memb

proteins - oxidation of amino acid residues, formation of disulfide bones, protein fragmentation/degradation
By what mechanisms can membrane damage occur?
- mitochondrial dysfunction
- decreased phospholipids
- cytoskeletal damage
- ROS
- products of lipid breakdown
How do a decrease in oxygen and increase in calcium lead to membrane damage, respectively?
Decrease in oxygen --> mitochondrial dysfunction --> decrease in phospholipid deacylation/synthesis --> phospholipid loss

Increase in calcium --> phospholipase activation --> increase in phospholipid degradation --> phospholipid loss and formation of lipid breakdown products (cause damage by inserting themselves in the cell memb and changing the permeability)

increased calcium also leads to membrane damage by activating proteases which cause cytoskeletal damage
When does cell injury become irreversible?
inability to reverse mitochondrial dysfunction; profound disturbances in memb fcn (esp. lysosomal memb damage causing an influx of calcium or leakage of cell contents)
What is meant by the time lag of irreversible injury?
irreversible injury occurs before light microscopic changes appear (ultrastructural changes occur after cell death)
Which ultrastructural changes are seen in REVERSIBLE cell injury?
- distortion of microvilli
- blebs on memb
- loosening of intracellular adhesions
- myelin figures
- mito swelling /rarefaction/ amorphous densities
- dilation of ER
- nuclear changes (less granular/fibrillar)
What are the microscopic features of necrosis?
- eosinophilic cytoplasm (loss of cytoplasmic RNA/denatured proteins bind to eosin)
- homogenous, glassy cytoplasm
- vacuolated cytoplasm
- calcification of cells
What three nuclear changes are seen in necrosis?
- Karolysis = decreased number of basophilia
- Pyknosis = decreased nuclear size but increased nuclear basophilia
- Karyorrhexis = nuclear fragmentation
What type of necrosis is seen after ischemic injury (e.g. MI) to the cell and what classifies its morphology?
coagulative necrosis - cell outlines are preserved
What type of necrosis is seen with bacterial infection and ischemic injury to the brain?
liquefactive necrosis
What are the three historic forms of necrosis and what characterizes each?
1. Caseous (cheese-like) necrosis = same as coagulative but w/ loss of cell outlines and thick white appearance

2. Fat Necrosis = enzymes digest (saponify) fat

3. Gangraeous Necrosis = ischemia of limbs (wet = with bacterial involvement; dry = without)
What is the classic example of caseous necrosis?
tuberculosis
What disease often leads to fat necrosis of surrounding structures?
pancreatitis (release lipases that digest fats causing saponification
What is tested for when determining whether necrosis has occurred in a patient?
increased leakage of enzymes (e.g. CK, LDH)
What is the key difference in hypoxia and ischemia concerning cell injury?
Hypoxia: damage caused by reduced oxygen availability

Ischemia: damage caused by reduced blood flow leading to decreased aerobic AND decreased anaerobic metabolism (also, toxic metabolites accumulate since blood cant carry them away)
How does reperfusion of tissue after ischemia cause damage?
- reoxygenation means an influx of oxygen which causes an increase in ROS
- inflammation caused by the immune system in blood damages cells
- complement in blood binds the IgM deposited during ischemia and leads to further cell injury and inflammation
What are the two types of chemical injury and what are some examples associated with each?
1. Direct Actors (no metabolism needed)
- examples: mercuric chloride in GI/kidney, lead in blood/GI/nervous system, antibiotics, chemotherapy drugs

2. Indirect Actors (metabolites do the damage)
- example: CCl4 which generates free radicals in the liver
What can a substance that causes necrosis at high doses lead to when given at low doses?
apoptosis (programmed cell death)
What are the general features of apoptosis?
- plasma memb remains intact
- dead cell is rapidly cleared
- can be physiologic (part of normal process) or pathologic
What are some examples of physiologic apoptosis?
- embryogenesis
- hormone-dependent involution
- cell deletion in dividing populations
- eliminatino of self-targeting lymphoctes
- death caused by cytotoxic T-lymphocytes (including elimination of tumors, virus-infected cells, and transplants, so long as not part of pathologic condition)
What are some examples of pathologic apoptosis?
- self-destruction of injured/virus-infected cells (as part of the disease process)
- pathologic atrophy following duct obstruction
- tumor cell death
- cell death due to mito permeability changes
What morphologic characteristics are seen in apoptosis?
- cells shrink
- chromatin condenses (nucleus may fragment)
- cytoplasmic blebs (leading to apoptotic bodies)
What happens to apoptotic bodies?
they express a receptor that causes them to be digested by phagocytes
What is the distinguishing feature of DNA on electrophoresis between a cell that underwent apoptosis vs. necrosis?
Apoptosis: laddering of DNA due to othe effect of endonucleases

Necrosis: general dissolution of DNA creating a homogenous band
What are caspases and which caspase functions as an endonuclease?
Cystein-aspartic acid proteases; Capase 3 is an endonuclease
What types of injury can cause apoptosis at low doses?
radiation, toxins, free radicals - cause DNA damage; apoptosis is initiated through p53
What are the two pathways of apoptosis and how do they differ?
1. Extrinsic: death receptors on cell surface are activated (FAS, TNFalpha) which activate initiator capases (8-10), and subsequent executioner caspases (3)

2. Intrinsic: signals that stop apoptosis (Bcl-2/Bcl-x) are silenced and signals that promote apoptosis (Bax, Bak, Bim) begin; involves increased mito permeability which leads to the release of cytochrome C which activates initator caspase
What molecule is associated with the bypass pathway of apoptosis, what secretes it, and how does it cause apoptosis?
Granzyme B; secreted by cytotoxix T lymphocytes through pores (perforin) in the cell membrane

- granzyme B directly activates executioner caspases
Is the Bcl-2 family of genes pro-apoptotic or anti-apoptotic?
both, depending on the circulating form

Pro = Bax, Bak, Bim
Anti = Bcl-2, Bcl-x
What are the subcellular responses seen in cell injury?
lysosomal catabolism, induction of sER, mitochondrial changes, cytoskeletal abnormalities
What are the two types of lysosomal catabolism and what defines each?
1. Heterophagy: catabolism of extracellular material via phagocytosis or pinocytosis

2. Autophagy: catabolism of intracellular material via autophagic vacuoles
What are the possible end-results of heterophagy and what class of disease is caused by a failure of these mechanisms?
phagocytosed materials form a residual body which either be exocytosed or become a lipofuscin pigment granule

failure of these systems leads to lysosomal storage disease
When cells are exposed to toxins (e.g. polycyclic aromatic hydrocarbons from cigarette smoke), what is induced?
formation of lots of sER as well as cytochrome p450 (molecule that metabolizes many toxins)
What damaging side-effects can occur due to the metabolism of certain compounds?
metabolite can be more toxic than original compund; ROS are often formed in the process
In what disease are megamitochondria seen?
alcoholic liver disease
What cytoskeletal components are affected by toxins and what toxins specifically affect each?
- thin filaments: affected by phalloidin of Amanita phalloides

- microtubules: affected by some anti-neoplastic agents

- intermediate filaments: affected by alcohol (causes formation of Mallory bodies = keratin intermediate filaments)
What are three major processes by which intracellular accumulations occur and what are examples for each?
1. Inadequate rate of metabolism of a normal substance (protein reabsorption droplets in renal tubules)

2. Defective Metabolism, packaging, transport or secretion of a normal substance (storage disease)

3. Accumulation of exogenous substances (silica, asbestos)
What specific mechanisms can lead to intracellular accumulations?
abnormal metabolism, defective protein folding/transport, lack of an enzyme, ingestion of indigestible materials
What causes fatty liver?
any defect in the pathway from free fatty acid uptake by the liver to lipid secretion; most common in excessive alcohol use
What pathologies can result from the accumulation of cholesterol and cholesterol esters?
- atherosclerosis (accumulation in tunica intima)

- Xanthomas (in hyperlipidemic state)

- Inflammation and necrosis

- cholesterolosis (in gall bladder)

- genetic disease (e.g. Niemann-Pick disease, type C)
What are three causes of excess protein accumulation?
1. reabsorption

2. excess secretion/production

3. defects in protein folding (leading to defects in transport/secretion and aggregation of abnormal protein)
What is the function of chaperones?
assist in proper protein folding (i.e. creating the correct tertiary structure)

example: heat shock proteins
What is the pathophysiology of cystic fibrosis?
defective intracellular transport

- a genetic mutation delays the dissociation of a chloride channel protein from its chaperone, causing abnormal folding of the channel
Defective secretion of which protein can lead to a severe form of emphysema?
alpha1-antitrypsin
What does an accumulation of unfolded proteins in the ER lead to?
increase in chaperone production, decrease in protein translation, and possibly caspase activation if other safety mechanisms fail
What is the pathophysiology of amyloidosis?
aggregation of abnormal proteins (beta-pleated sheets)
In what diseases can glycogen accumulation occur and how does it show up on light microscopy?
diabetes mellitus and glycogen storage diseases - appears in muscle as less defined lipid vacuoles
What exogenous pigment is found occupationally in WV and what is the associated disease?
coal dust - excessive inhalation leads to anthracosis (better known as coal worker's pneumoconiosis)
What is lipofuscin and what is its presence indicative of?
an endogenous pigment consisting of lipid/phospholipid protein

does not cause damage itself but is indicative of free radical injury and lipid peroxidation
What is hemosiderin and what results from excess and extreme accumulation of this substance?
endogenous pigment composed of iron oxide - excess hemosiderin leads to hemosiderosis, whereas extremem accumulation in the liver, heat, and pancreas cause hemochromatosis
What is the distinguishing feature on light microscopy between lipofuscin and hemosiderin?
lipofuscin accumulates perinuclear whereas hemosiderin is found throughout the cell
Abnormal accumulation of what pigment causes jaundice?
bilirubin
In pathology, what is referred to as hyaline change?
a homogenous, glassy, pink appearance within or around the cell
What is the cause of metastatic calcification and what pathologies does it result from?
hypercalcemia w/ tissue deposition

caused by increased PTH, bone destruction, vitamin D, related disorders, and renal failure
Accumulation of what substance often results from sarcoidosis?
vitanim D (precursor form activated macrophages inside the granulomas)
How does dystrophic calcification differ from the metastatic form and what are its causes and effects?
- does not correlate with increased blood calcium levels --> caused by prior necrosis

- formation of crystalline calcium phospahte initiated intracellularly by mitochondria in dead/dying cells and extracellularly by phospholipids of membrane-bound vesicles

- propagation of abnormal crystallization can cause organ dysfunction
What biochemical changes are associated with aging cells?
- reduced mitochondrial oxidative phosphorylation

- reduced synthesis of proteins, nucleic acids, and transcription factors

- reduced chromosomal repair

- decreased nutrient uptake
What structural changes are associated with aging cells?
- mitochondrial pleomorphism and vacuolation

- abnormally shaped nuclei

- decreased ER

- distorted golgi

- increased lipofuscin

- glycation end-products and cross-linked proteins

- abnormal protein folding
What is meant by replicative senescence, what causes it and what disease exemplifies this process?
a decrease in population doubling level with increased cellular age, caused by a decrease in telomere length

exemplified in Werner syndrome (caused by defective DNA repair)
What is a common feature regarding telomeres in cells that have undergone malignant transformation?
telomerase reactivation (telomerase is not found in normal somatic cells)
What results from a decrease in IGF-1 signaling?
a leaner person w/ increased lifespan (lifespan also thought to increase from a decreased caloric intake)
What metabolic events are associated with a decreased lifespan?
oxidative damage, DNA damage, declining proteasomic fcn (causes damaged proteins and organelles)
Who wrote the first work on immunity to disease and what disease was it concerned with
Edward Jenner - small pox
Who coined the term pathogen, and who developed the first vaccine?
pathogen: Robert Koch

vaccine: Louis Pasteur
Who discovered antibodies and antigens?
Emil von Behring
What are the two types of immune responses and what characterizes each?
1. Innate Immune Response: nonspecific, not altered by exposure, rapid response, dominated by phagocytic cells

2. Aquired/adaptive Immune Response: specific, memory following exposure, slow response, dominated by lymphocytes
What do the innate and acquired immune responses have in common?
they have common effector mechanisms for the destruction of pathogens
What are the anatomical barriers of the innate immune system?
- skin (mechanical barrier)

- mucous membranes (trap and remove invaders)
What are the physiological barriers of the innate immune system?
- temperature (fever inhibits bacterial growth)

- low pH (acidity not conducive to bacterial growth)

- chemical mediators (lysozyme, interferon, complement)
When a pathogen gets past the anatomical and physiological barriers of the innate immune system, what barrier it encounter last?
phagocytic cell barrier
What did Eli Metchnikoff discover?
phagocytosis by leukocytes; specifically macrophages (activated monocyte) and microphages (neutrophils or polymorphonuclear cells - PMN)
What is the acquired immune response solely dependent on?
lymphocytic cells and their products
Who discovered the purpose of lymphocytes?
Bruce Glick and his chickens
What are the two types of acquired immunity?
1. Humoral immunity (in the blood)

2. Cell-mediated immunity (in the cells)
Where do myeloid and lymphoid immune cells come from?
both are produced in the bone marrow
Which hematopoietic cell is the precursor to all others and what characterizes this cell?
Pluripotent hematopoietic stem cell - 1/100,000 cells in bone marrow

can be transported to reform the entire hematopoietic system without losing its ability to self-renew
Which cells are the immediate offspring of the pluripotent hematopoietic stem cells and what do those cells give rise to?
committed stem cells (limited renewal) - give rise to progenitor cells (no renewal) which then form the functional end cells
What does it mean, clinically, when you have marrow with a left shift?
there are more immature hematopoietic cells than found in the normal individual
What are bone marrow stromal cells also known as and what is their purpose?
adventitial reticular cells - secrete cytokines that sim blood cell production in response to the humoral environment
What are the cytokines called that are produced by bone marrow stromal cells and what function do these cytokines have?
Colony Stimulating Factors (CSF) - stimulate the production of colonuy-forming units in culture (CFU-C)

CSF are also known as interleukins
What happens to hematopoietic cells that fail to be stimulated by appropriate survival signals (cytokines, adhesion contacts, etc.)?
they die via apoptosis
From what precursor are the bone marrow stromal cells derived?
Mesenchymal stem cells (these are also responsible for the formation of osteogenic cells, adipocytes, and chondrogenic cells)
From what embryonic region do hematopoietic stem cells originate and where do they migrate to?
Splanchnopleura and AGM region --> migrate to the yolk sac and fetal liver
To what organs do the hematopoietic stem cells migrate after leaving the liver (in mice)?
thymus (to make T-lymphocytes) and spleen

from the spleen they migrate to the bone marrow (this occurs just before birth in mice - whereas in humans the migration is finisehed at the end of the 1st trimester)
Where do the hematopoietic stem cells of the bone marrow arise from?
NOT from the yolk sac! instead from the fetal liver
Removal of which part of the immune system has the most lethal impact on patients?
removal of the innate immune system (seen w/ radiation/chemotherapy)

removal of the aquired immune system still provides the initial protection, but the disease continues for a long time
What distinguishes the immediate innate immune response from the early induced innate immune response?
immediate (0-4 hours): macrophages and preformed chemical mediators respond
early induced (4-96 hours): inflammation recruitment and activation of effector cells
What is meant by the microbiological aspect of innate immunity?
the normal flora of the skin and gut provide a layer of protection against pathogens by competing for the same nutrient and space
Which is the most numerous leukocyte in circulation and what is its lifespan?
polymorphonuclear neutrophil (PMN), comprises 70% of leukocytes, have a lifespan of 12 hours in circulation (36 hour total lifespan)
In which disease is there a deficiency in neutrophils that occurs every 21 days?
cyclic neutropenia (result of autosomal dominantly inherited mutations in ELA2, the gene encoding neutrophil elastase)
What is the primary cytokine that stimulates proliferation and differentiation of neutrophils?
G-CSF (granulocyte colony stimulating factor); can be used therapeutically (Neupogen)
What characterizes colony stimulating factors and what are the different types?
small peptide formed by bone marrow stromal cells;
stimulates proliferation and differentiation of cells with the receptor to the specfic CSF;
M-CSF: monocytes
G-CSF: neutrophils
GM-CSF: monocytes and neutrophils
Stem cell factor: mixed myeloid cells
What is the immediate response when bacteria enter the body through a break in the skin?
tissue macrophages recognize structures on the cell surface of bacteria and secrete cytokines (vasoactive and chemotactic factors) which cause inflammation of the surrounding tissues
Which cells are the first to respond to cytokines released by macrophages after its exposure to a pathogen and what do these cells do once stimulated?
Neutrophils - they attach themselves to the surrounding endothelial cells (margination), then squeeze through the vessel wall (extravasation - made possible by dilation of capillaries in response to cytokines), they migrate to the site of infection and finally phagocytose the microorganism
What is the purpose of complement and how does it achieve this?
complement is involved in pathogen recognition mechanisms - in the presence of a pathogen, the complement is enzymatically cleaved; one part of the complement attaches to the pathogen while the other serves as a chemotatic factor which signals effector cells (phagocytes) that a pathogen is present
What are the cardinal physical signs of inflammation?
pain, redness, heat, swelling
What is the lifespan of a monocytes/macrophage, what are some of the different types of macrophages and where are they located?
lifespan = weeks
Alveolar macrophages (lung)
Kupffer cells (liver)
Histiocytes (CT)
Mesangial Cells (kidney)
Microglial cells (brain)
When a macrophage is stimulated by a pathogen, what does it release?
pro-inflammatory cytokines (interleukins, TNF-alpha); these are also released by dendritic cells
Where are natural killer cells produced, and what do their cell surface receptors respond to?
produced in the bone marrow, they respond especially to virus-infected cells; they are also called lymphokine-activated killer cells since they response to IL-2
What is the main purpose of eosinophils, basophils, and (to a certain extent) mast cells, and what complications are associated with them?
they fcn in the killing of parasites - however, the substances they release (e.g. histamine) can cause severe complications regarding allergies
Where are dendritic cells produced and what are some examples of the different types?
produced in the bone marrow
Langerhans cells (skin)
Interstitial Dendritic cells (organs: heart, liver, kidneys, etc.)
Interdigitating dendritic cells (follicles of Lymph nodes)
Circulating dendritic cells (blood)
What is the main purpose of dendritic cells, and especially that of the Langerhans cell?
recognition of foreign invaders - Langerhans cells engulf a foreing invaders, travel to a lymphoid organ and activate the aquired immune system (it is the connection b/w the innate and aquired immune systems)
What are Toll-like receptors and where are they found?
very primative and highly conserved receptors on innate immunity cells that recognize foreign materials; they are found on both the outside and inside of the cell
What makes IL-1, IL-6, and TNF-α especially important cytokines?
they induce acute-phase protein secretion (incl. complement) which is essential for the inflammatory phase of the innate immune response
In a patient with hypoalbuminemia, what must be taken into account when giving drugs?
The patients free drug concentration will be higher than in normal individual
What is the major organ of drug excretion?
kidney

but in the case of inhalational anesthetics it will be the lungs
What is meant by enteral administration of a drug?
administration via the GI: oral, sublingual, rectal
What is IT drug administration?
intrathecal: into the spine
What are four factors affecting the absorption from the GI tract?
acidity, gastric emptying time, motility, GI content
Which compound will be absorbed easier by the GI tract, a weak acid or a weak base?
a weak acid, since it will be neutral when protonated by the slightly acidic environment of the GI tract
How do the GI contents affect absorption?
content can either impede absorption or it can enhance it by stimulating acid secretion and blood flow
What factors can reduce the efficiency of absorption of orally ingested medications?
inactivation by the intestinal enzymes, transport of absorbed drugs back into the intestine, metabolism in the liver due to the first-pass effect
What are the advantages of sublingual and rectal administration of certain drugs, respectively?
sublingual: rich in blood supply, no first-pass effect

rectal: useful when drugs not tolerated orally, for heavily sedated patients, when site of action is rectal, to avoid injection, and to avoid first-pass effect
What are some of the advantages and disadvantages of parenteral routes of drug administration?
advantages: rapid effect, maximizes bioavailabilty

disadv: once injected, cannot get back or retard its absorption, overdose may be a problem
What are some special features of IM drug administration?
drug absorption is affected by blood flow, so vasodilation/constriction can change absorption; lateral diffusion can be changed (e.g. with hyaluronidase); a disadvantage is local tissue damage and pain due to alkaline formation
How does subcutaneous drug administration differ from IM?
generally slower absorption due to less lateral diffusion and lower blood flow
What form of drug administration will result in the most rapid increase in concentration of that drug in plasma, and which methods are slower?
IV>IM>SC>PO
What are topical routes of drug administration and what are some of the features of each?
Percutaneous: thick skin and lipid poor stratum corneum are barriers to penetration (highly lipid soluble drugs pass through epidermis most easily; Mucous membranes: includes local anesthetics for operative manipulations, and nasal sprays for common cold; Corneal application: for ophthalmologic drugs
What are some of the advantages to inhalational drug administration?
High surface area, rich blood supply, and thin epithelium allow for rapid absorption
What are the two major types of lymphocytes and what is their classification?
B-Lymphocytes (humoral immunity)
T-Lymphocytes (cell-mediated immunity)
What is the difference between primary/central and secondary/peripheral lymphoid tissue?
primary/central: bone marrow and thymus
secondary/peripheral: adenoid, tonsil, lymph node, appendix, spleen, NOT blood
Once B-lymphocytes in secondary lymphoid tissue have been activated and become plasma cells, where do they travel?
to bone marrow
When lymphoid cells are in peripheral tissue, what happens to their cell cycle?
they are stuck in G0, if their cognate antigen is presented within 2-7 days, they will become activated and proliferates; if not they will undergo apoptosis
What are the two types of cells that are created after a B-lymphocyte has undergone clonal expansion?
Effector cells (make antibodies)
Memory cells (reside in the body for a long time)
How does lymph flow through the lymph node?
from afferent vessels, through the cortex (packed w/ lymphocytes), into the paracortex and medulla (contain macrophages and plasma cells), ending at the hilus in an efferent lymphatic vessel
What is the difference between the primary and secondary follicles (germinal centers) in the lymph node?
primary: accumulation of lymphocytes
secondary: location of stimulated lymphocytes (follicle has a germinal center)
What are the characteristics of the post-capillary venule of a lymph node?
high cuboidal epithelium
contain cell adhesion molecules (vascular addressins/homing receptors)
Integrin on lymphocytes attach to post-capillary venules
How does the spleen differ from the lymph node?
does not have lymphatic input
recognizes antigens in the vascular space (NOT in the tissue space)
Where in the spleen are the lymphoid cells found?
in the white pulp (around the arterioles)
bulk of the lymphocytes are found in the periarterial lymphatic sheath (PALS) where they await antigens
Where is mucosal lymphoid tissue found and what specific cell type do they possess?
Oral cavity (tonsils)
brochial associated lymphoid tissue (BALT)
gut associated lymphoid tissue (GALT)
they have M cells which allow antigens to pass into them so they can be identified
Where are Peyer’s patches found?
in the small intestine (mainly jejunum), under the lamina propria; a layer of M cells lies directly above the Peyer's patch to import foreign antigens
What are the receptors that recognize antigen called on B cells vs. those on T cells?
B Cells: immunoglobulins
T Cells: T cell receptor (TCR)
What is the difference in function between T-helper cells and T-cytotoxic cells?
T Helper Cells: release cytokines in response to a foreign antigen that pull in T-cytotoxic cells and B lymphocytes
T Cytotoxic Cells: recognize and immediately destroy foreign antigen
What co-receptor is found on T-helper cells and T-cytotoxic cells, respectively?
T-Helper: CD4
T-cytotoxic: CD8
What is meant by the T-suppressor cell?
it is not a cell in itself, but rather a fcn of T-cytotoxic cells
What is the difference in the variable and constant regions of the antibody?
variable region: antigen-binding site
constant regoin: effector fucntion (signals celll that an antigen has been recognized)
How does antigen recognition differ between B cells and T cells?
B Cell: recognizes native antigen
T Cell: recognizes "chewed up" antigen
What two signals are needed for lymphocyte activation for B cell and T cells, respectively?
B Cell: antigen receptor binding and activation by a T-Helper cell
T Cell: recognition of a peptide of antigen that is expressed on the surface of another cell (antigen-presenting cell) and activation by the APC
What is the anamnestic response?
secondary immune response of the aquired immune system to a particular antigen (much greater response than primary immune response)
What is presented by MHC-I and MHC-II, respectively, and which T cell interacts with each?
MHC-I: present foreign protein made inside the cell (i.e. viral protein) which is recognized by T-cytotoxic cells
MHC-II: presents a phagocytosed peptide which is recognized by T-helper cells
What is the subset of MHC-I and on which cells is it expressed?
HLA-A, HLA-B, HLA-C
expressed on ALL nucleated cells (since all cell can become infected w/ a virus)
What is the subset of MHC-II and on which cells is it expressed?
HLA-DP, HLA-DQ, HLA-DR
expressed on macrophages, dendritic cells, and B cells (i.e. antigen-presenting cells)
What are the two different results that can occur when a free antibody binds to a pathogen?
Neutralization: antibodies prevent pathogen from infecting host cells
Opsonization: macrophages bind to the constant region of the antibody and phagocytose it along w/ pathogen
What is the specificity for an Ig with a valence of 10?
1 - every Ig, no matter what the valence, has a specificity of one
What does polyclonal antibodies refer to?
a polyclonal response of many monoclonal antibodies to an antigen with multiple epitopes
What is known, in general terms, about the genes that comprise the variable region of Ig?
they consist of a V gene, which codes for the bulk of the variable region, and a J gene, which codes for a small portion of the variable region; the J gene is found separate on the human genome from the V gene
What changes does germline DNA undergo while forming the light chain of Ig?
Germline DNA undergoes somatic recombination which removes the DNA b/w the V and J genes --> this piece of DNA is then transcribed into a primary RNA transcripts --> mRNA splicing removes the RNA b/w the V-J genes and the C gene --> the formed mature RNA transcript is then translated to form the actual polypeptide chain of the Ig
What is meant by the 12-23 rule regarding the recombination signal sequence?
Each V/J gene has a DNA sequence consisting of a heptamer (conserved) - 12/23 nucleotide spacer - nonamer (conserved)
recombination can only happen b/w a gene w/ a 12 nucleotide spacer and another with a 23 nucleotide spacer (i.e. 1 turn of the helix vs. 2 turns of the helix)
What are the enzymes in lymphocytes called that activate the recombination signal sequence and how do they function?
RAG1/RAG2 (recombinase activating genes)
two RAG enzymes attach themselves to the V and J regions and subsequently combine
all of the genetic material b/w the V and J regions forms a loop which is cleaved off by RAG
the loop (singal joint) is then degraded by endonucleases
How is the V-J coding joint formed by RAG?
RAG complex binds to and randomly cleaves recombination signal sequences to yield a DNA hairpin --> RAG-mediated cleavage of hairpin generates palindromin P-nucleotides --> the enzyme TdT adds N-nucleotides to the ends of the P-nucleotides --> strands are paired, unpaired nucleotides removed (by exonucleases) and the gaps are filled by DNA synthesis
What is the DNA polymerase called that only functions in lymphocytes and what is characteristic of it?
TdT (terminal deoxynucleotidal transferase) - polymerases nucleotides without template
What are the three hypervariable regions of Ig comprised of, respectively?
2 of the regions are coded for by V genes and 1 is generated randomly by TdT
What portion of the rearrangements of an Ig gene are abortive and why?
~50% - occurs b/c when TdT adds nucleotides it causes a frameshift in 2/3 of the Ig genes, generating a stop codon in about half of all sequences
What enhances transcription of an Ig gene?
rearrangement if the Ig gene brings the enhancer closer to the promoter, resulting in enhanced transcription
How does the diversity region of the heavy chain gene get incorporated into its variable region?
by the 12-23 rule (RAG1-2) where J and D recombine first, followed by recombination of of V with the new J-D region
What is the key feature that distinguishes the heavy chain variable region from the light chain variable region?
the heavy chain variable region has V, J, and D segments, wheras the light chain variable region only has V and J segments
How is it possible that different isotypes possess the same idiotype?
After the VJD region has recombined to determine idiotype, the CH genes on chromosome 14 can recombine to form any isotype
Which is the first Ig isotype expressed and which is subsequently expressed?
IgM is first, then IgD (can be expressed in the bone marrow or shortly after leacing the bone marrow, but it is expressed PRIOR to antigen binding)
What characterizes IgD as opposed to other immunoglobulins?
it is never found in secretion, but only in its membrane-bound form (function is unknown)
Which two Ig isotypes can be expressed at the same?
IgM and IgD (only possibility) b/x they are expressed on the same strand of RNA
What keeps the Ig attached to the cell surface of a B cell?
IgAlpha and IgBeta
What happens if a B cell undergoes an abortive rearrangement?
it either recombines until a proper protein has been made (it senses the presence of the protein in the cytoplasm) or it dies by apoptosis
What is the difference between immunoglobulin and antibody?
Immuniglobulins are expressed on B cells but they do not necessarily recognize any available antigen
Antibodies are secreted from plasma cells (activated B cells) and they will respond to the antigen that activated the original B cell
Which of the globulins is immunoglobulin?
Gamma-globulin
Which parts of the immunoglobulin have an identical structure and where are the disulfide bonds located?
each light chain is identical and each heavy chain is identical
2 disulfide bonds are found b/w the heavy chains, and 1 b/w each heavy chain and light chains
Which end of the immunoglobulin is the variable region and what parts of the immunoglobulin does it comprise?
the amino end is variable and it comprises the ends of both the light and heavy chains
What are the 6 domains of immunoglobulin?
Variable: VH, VL
Constant: CL, CH1, CH2, CH3
What gives the immunoglobulins their strength and stability in a noxious environment?
they are mainly composed of beta-pleated sheets
Where on the immunoglobulin is the antigen-binding site found and what is the part of the antigen called that binds here?
between the VH and VL domains on each arm
this is where the epitope binds
What is the valence of IgG and what does this mean?
Valence = 2, this means that there are 2 binding sites on the immunoglobulin
What aspect of the immunoglobulin allows it the flexibility to bind multiple antigens at variable angles?
the hinge regions (located b/w CH1 and CH2)
What different parts does immunoglobulin divide into after proteolytic cleavage by papain and pepsin, respectively?
Papain: 2 Fab (antibody binding fragment) and Fc (crystallizable fragment)
Pepsin: F(ab)2 (both antibody binding regions still attached together) and pFc (chewed up crystallizable fragment)
What are the 5 different heavy chain immunoglobulin isotypes (classes) and on what chromosome are they all coded?
IgG, IgM, IgD, IgA, IgE, all coded on chromosome 14
What are the 2 light chain immunoglobulin isotypes (classes) and on what chromosome is each coded?
Kappa on chromosome 2
Lambda on chromosome 22
What does the concept of allelic exclusion refer to regarding heavy and light chains?
Heavy Chain: only one chromosome is used to create heavy chains of immunoglobulin (the other is silent)
Light Chain: either kappa or lamba is expressed on onlyy one chromosome (the other is silent)
What are allotypes in regard to the heavy chain of immunoglobulin?
differences in the variable region of Ig within the individual
What does idiotype refer to?
differences in the sequence of the heavy chain b/w individuals
What is the most common immunoglobulin found in serum and what are some of its characteristics?
IgG - it has 4 subclasses (IgG1-4) and a valance of 2; its MW is 160,000
What is the only immunoglobulin expressed on the cell surface in bone marrow and what are some of its characteristics?
IgM (first Ig expressed on cell memb) - 5-10% of total serum Ig, MW 180,000
How does IgM differ when found on the surface of lymphocytes vs. secreted IgM?
it appears on the surface of lymphocytes as a monomer (MW 180,000) but is a pentamer in its secreted form ((MW 9000,000 with a valence of 10); the monomers are polymerized by J chain (which is made by B cells) and the pentamer cannot cross the placenta because of its large size
Which is the primary Ig made in the lymphoid tissues and what are some of its characteristics?
IgA: 10-15% of serum Ig, predominant Ig in secretions, polymerized by J chain to form a dimer (MW 600,000 with a valence of 4); has 2 subclasses (IgA1-2)
What happens to dimeric IgA after it is secreted, as it moves from the basolateral surface to the lumen?
it binds to a poly-Ig receptor as it tracels through the epithelial cell; part of this receptor remians attached to the IgA as it exits the cell and is known as the "secretory piece"
Which Ig is responsible for symptoms of allergy, hives, and asthma, and what are some of its characteristics?
IgE - has very low serum concentrations; normally binds to receptors on mast cells causing degranulation and histamine release
How does the FcεR1 receptor on mast cells differ from almost all other Fc receptors?
it can bind IgE before the IgE has come in contact with its specific epitope; all other Fc receptors can only bind Ig that has already contacted its epitope
What kinds of forces are involved with antigen-antibody interactions and how do these relate to antibody affinity?
non-covalent forces: electrostatic forces, H-bonds, van der Waals forces, Hydrophobic forces
Anitbody affinity depends on the precise fit and forces elicited when the antigen comes in contact with the antibody (this can range from high to low affinity)
What are hypervariable regions and how many exist on each variable Ig domain?
regions of variable Ig domains that express a large amount of variability (also known as complementarity-determining regions); they comprise the actual antigen-binding site; there are three of these variable regions per variable domain (HV1-3)
What is the difference between a linear and a discontinuous epitope?
linear: antibody binds a linear, continous sequence of amino acids on the antigen
Discontinuous: antibody binds amino acids from different parts of the antigen
How are monoclonal antibodies created and what is their purpose?
B cells from a mouse are immunized with a specific antigen --> these cells are fused with myeloma cells to make them immortal --> they are grown in drug-containing medium so that only hybrid cells survive --> antigen-specific hybridomas (B cell + myeloma cell) are selected and used to clone massive amounts of antigen-specidic antibodies
What are the components of a T cell receptor (TCR)?
Variable region with antigen-binding site
constant region
transmembrane region
cytoplasmic tail
the receptor is composed of an alpha and beta chain (broke down in Valpha/Calpha/Vbeta/Cbeta)
together they have a similar structure to the Fab component of the B cell Ig
What post-translational modification is found on the TCR and what bond is found in the hinge region between the constant and transmembrane regions?
post-translational modification: hlycosylation of all variable/constant regions
bond: a disulfide bond connects the two segments of the hinge region
In regard to the genes used to produce the α and β chains, how do they compare to those used to make the light and heavy chains of the Ig molecule?
alpha: similar to the light chain, but found on chrom 14
beta: similar to heavy chain but found on chrom 7
Which segment of the TCR undergoes recombination first?
beta first (D-J, then V-DJ); then alpha (V-J)
When comparing Ig to TCR, what are some of the noticeable differences seen?
the beta chain only has 2 D segments (vs. 25 for heavy chain)
beta chain is often read in all three reading frames (vs. rarely for heavy chain)
alpha/beta junctional diversity is much greater than that of the heavy/light chain
What are the two classes of TCRs and what differentiates them?
alpha beta TCR and gamma delta TCR
there is much greater variability in the alpha beta TCR
gamma delta TCR are more commonly used in mucosal tissues and they appear to be used in innate immunity as well
Where on the genome is the δ-chain locus found and what is important about this location?
on chrom 14 b/w the V and J genes of the alpha chain locus
after the first rearrangement of the alpha chain. the delta chain locus is excised
What is the difference between the exocytic and endocytic pathways of antigen presentation using the major histocompatability complex (MHC)?
exocytic: peptides from endogenous proteins (and sometimes exogenous proteins) are presented by MHC-I to cytotoxic T cells
Endocytic: peptides from exogenous proteins are endocytosed and presented by MHC-II to helper T cells
When a pathogen is taken up in an endocytic vesicle of a macrophage, what causes its degradation?
a combination of low pH and acid activated proteases
When CD4+ T cells respond to a peptide presented on an MHC-II protein, what results?
either activation to kill the intravesicular bacteria/parasites; or activation of B cells to secrete Ig to eliminate extracellular bacteria/toxins
What is the importance of the highly polymorphic nature of MHC proteins?
their variability ensures that they can bind to many different types of foreign peptides as well as a wide variety of T cells
What is the key morphologic difference between the MHC-I and MHC-II?
MHC-I is anchored by one transmembrane tail, whereas MHC-II is anchored by two
How do MHC-I and MHC-II expression differ among cells and tissues?
MHC-I: found mostly on T cells, B cells, macrophages (and other APCs) and neutrophils
MHC-II: found primarily on B cells, macrophages (and other APCs) and epithelial cells of the thymus, but also on activated T cells
Which cells do not express MHCs?
RBCs (i.e. non-nucleated cells)
What are the major structural characteristics of MHC-I?
composed of one single heavy chain (alpha chain) with three domains (alpha 1-3), one signle light chain (beta 2- microglobulin - Beta 2 m); only alpha 3 is inserted into the cell membrane; the peptide binding cleft is formed by the alpha helical regions of the alpha 1 and alpha 2 domains with their underlying beta sheaths
What are the structural differences of MHC-II in comparison to MHC-I?
MHC-II consists of a two-chain heterodimer (alpha and beta chains), both of which are inserted into the cell memb (via alpha 2 and beta 2) ; the peptide binding cleft is made by the alpha 1 and beta 1 domains and presents peptides that are 13-17 residues long (vs. 8-9 for MHC-I)
By what process does MHC-I antigen processing and presentation occur?
intracellular antigen is degraded by a proteosome and enters the ER --> MHC-I in the ER binds the antigen --> it is transported through the Golgi and eventually presented on the EC surface
How does MHC-II antigen processing and presentation differ from that of MHC-I?
the foreign antigen enters the cell through an endocytic vesicle --> this antigen is processed in a phagolysosome --> MHC-II travels from the ER through the Golgi and eventually meets up with this antigen containing vesicle --> once they fuse, MHC-II binds the antigen and travels to the EC surface
What is the importance of the concept that MHC-II picks up antigen from a phagolysosome and not in the ER?
it prevents MHC-II from binding MHC-I antigen
In which MHC pathway does the proteasome play an integral part and what is its function?
MHC-I; it contains about 28 catalytic sites which cleave unfolded protein as it passes through the proteasome; the cleaved peptides are then transported into the ER where they bind MHC-I
What structure is utilized to transport the antigen product of the proteasome from the cytosol into the ER and what are its components?
TAP (transporter associated with antigen processing) - consists of 2 ATP-binding cassette (ABC) domains and 2 hydrophobic transmembrane domains; the 4 domains assemble into a heterodimer with the ABCs on the cytostolic side and the hydrophobic tails in the ER
How is the TAP transporter able to transport a wide variety of antigens in the ER?
both its domains (TAP1-2) are coded for in two different alleles which allows for some selective variation
What is MHC-I heavy chain bound to until β2-microglobulin binds it, and which chaperone proteins bind subsequently?
the MHC-I heavy chain is bound to calnexin; when beta2m binds, calnexin releases and the chaperones calreticulin and Erp57 take its place; tapasin binds the complex as well on the side of the beta2m domain and creates connections b/w MHC-I and TAP
When does MHC-I folding complete and what happens afterwards?
folding is complete when TAP delivers a peptide that binds MHC-I; when folding has copmleted, the MHC-I is released from TAP and exported to the cell surface
What activates the proteases found in early endosomes and what is their purpose?
as early endosomes become late endosomes. the pH drops significantly, activating the endosomal proteasomes; these proteasomes degrade foreign antigen which is presented to MHC-II once the endosome fuses with an MHC-II vesicle
How is MHC-II prevented from binding antigen in the ER and how is this process reversed when MHC-II leaves the trans-Golgi in its own vesicle?
the invariant chain binds to the cleft outside of MHC-II, blocking the antigen-binding cleft; once in its own cesicle, invarient chain is cleaved, but it leaves behind CLIP fragments in its antigen-binding cleft
How is the CLIP fragment removed from the antigen-binding cleft of MHC-II?
when the MHC-II vesicle fuses with an endosome, HLA-DM is activated and induces opening of the MHC-II antigen-binding cleft, this releases the CLIP fragment and allows antigen to bind instead
In what form can the invariant chain binding MHC-II be found, and where can they become dissociated from MHC-II?
invariant chain can form a trimer, binding three MHC-II compounds at once; they can become dissociated from MHC-II in the MIIC (MHC-II compartment)
Why does the MHC-I antigen-binding cleft bind residues that are almost exclusively 8-10 amino acids long?
because the cleft has strong anchoring sites on each end (or on the floor of the cleft) that determine residue interactions' since there are only two anchoring sits, antigen length is restricted to 8-10 residues long
What is the main difference between the MHC-II cleft anchoring sites and those of MHC-I?
they are anchored throughout the entire length of the cleft, allowing for greater differences in antigen binding length; MHC-II binds to a certain core sequence deep inside of its cleft
How are MHC-I and MHC-II genes organized on the HLA complex on chromosome 6?
they are located on different parts of the chrom, but the components that code for TAP and the proteasome (involved in MHC-I antigen binding) are found in between the genes that code for different MHC-II compinds
What is the meant by the fact that HLA expression is co-dominant?
Offspring will inherit either one of the HLA alleles from each of the parents, and both acquired alleles are expressed equally in the child
What accounts for the large diversity in MHC expression?
Polymorphism (individuals are heterozygous for HLA genes – i.e. they have two different copies)
Polygeny (duplication of the HLA genes – i.e. there is HLA-A, HLA-B, HLA-C, HLA-DP, etc.)
Where on the MHC molecules is the allelic variation found?
Almost entirely on the antigen-binding cleft – on both sides of the cleft for MHC-I, but mainly on the β chain for MHC-II
Which HLA isotypes are the most highly polymorphic?
The classical isotypes: HLA-A, HLA-B, HLA-C and HLA-DP, HLA-DQ, HLA-DR
Who discovered the function of the thymus, and who gave the first description of B and T cells?
Thymus: Good and Miller
B/T cells: Raff
What did Dennis Osmond discover in ’72?
that B lymphocytes differentiate from stem cells EXCLUSIVELY in the bone marrow
When hematopoietic stem cells differentiate into early pro-B cells, what detectable change occurs?
D-J rearrangment occurs in the heavy chain genes
When early pro-B cells differentiate into late pro-B cells, what change happens?
V-DJ rearrangment occurs
The presence of which cell type is absolutely essential for B cell differentiation?
bone marrow stromal cells
What relationship between the stromal cells and pro-B cells is required for differentiation to take place?
the pro-B cells (and stem cells) must be directly attached to the layer of bone marrow stromal cells
What are the different types of adhesion contacts between bone marrow stromal cells and pro-B cells and why are they important?
VCAM-1/VLA-4
Fibronectin/FN-receptor
CD44-CD44
ckit-ligand (SCF)/ckit
the adhesions are important b/c they allow survival and proliferative signals to be sent from the stromal cells to the pro-B cells
the ckit connections are especially important
Which free flowing cytokines, secreted by bone marrow stromal cells, cause tremendous proliferation of pro-B cells?
IL-7
IGF-1 in response to GH (IGF-1 is also required for pro-B cells to release from the stromal cells)
When VDJ is fully arranged and pro-B cells become large pre-B cells, what is the pre-B cell receptor composed of and what is the purpose of this receptor?
a fully formed heavy chain composed of the mu isotype (IgM) and two associated molecules which comprise the light chain (i.e. lamba5 and VpreB) - the presence of this surface receptors stops rearrangment by RAG1-2 and signals to the cell that everything is going ok (otherwise apoptosis follows)
What occurs after the μ chain is expressed on the large pre-B cell and the cell differentiates into a small pre-B cell?
RAG1-2 are reactivated (slightly) and V-J rearrangement occurs on the light chain genes; this is followed expression of the mu isotype in both the cytoplasm and cell surface
when VJ rearrangment is completed, the cell becomes an immature B cell with its IgM expressed on the cell surface
Which cytokine is required for pre-B cells to differentiate into immature B cells and where is this cytokine produced?
IL-4 secreted from T cells (IL-4 is a necessary survival signal)
What has to occur for immature B cells to differentiate into mature B cells?
IgD has to be expressed alongside IgM (the cells is now ready to respond to antigen)
What is the consequence of the fact that TdT expression is strong during heavy chain rearrangement but weak during light chain rearrangement?
heavy chain variable regions have large N regions (lots of N-nucleotide addition) whereas light chain variable regions have an almost non-existent N region
How does the concept of allelic exclusion apply to heavy chain rearrangement in the late pro-B cell?
after both chromosoms have undergone D-J rearrangment, V-DJ rearrangement occurs in only one chromosome
if this rearrangement is productive, the other chromosome will not rearrange
however, if it is not productive, the second chromosome will undergo rearrangement and if this fails as well, the cell dies by apoptosis
What is the sequence by which light chain rearrangement occurs in pre-B cells?
Rearrangement of the κ gene on the first chromosome is usually step 1
if this fails, rearrangement on the second chromosome will occur
if this fails as well, the process will repeat itself but this time with the λ gene
if failure occurs twice here, the cell will die by apoptosis
How is it possible for one κ or λ gene to undergo multiple rearrangements and why is this not possible on the heavy chain?
If, after the first V-J rearrangement, there are still V and J genes remaining (i.e. they were not lost in the signal joint), RAG1-2 can cause additional rearrangements until no more available Vs and Js remain; this is not possible on the heavy chains, because a second rearrangement would get rid of the D region
In the first several hours after formation of the immature B cell, what happens when this B cell encounters a multivalent self-antigen vs. a soluble self-antigen?
Multivalent: the B cell is destroyed by apoptosis
Soluble: the B cell becomes anergic (it can’t generate a response to antigen)
What can happen when an immature B cell encounters a self-antigen while RAG1-2 are still active?
Light chain rearrangement can continue and cause the B cell to express a different idiotype (very rare event)
What two functional regions are found in each lobe of the thymus and what is found within each of these functional regions?
cortex: immune thymocytes and cortical epithelial cells
Medulla: mature thympcytes, medullary epithelial cells, macrophages, dendritic cells
What happens to the vast majority of thymocytes in the thymus?
they die by apoptosis
On what T cells are the surface molecules CD3, CD4, and CD8 found, respectively?
CD3: all T cells
CD4: T helper cells
CD8: cytotoxic T cells
What is the region called where thymocytes first enter the thymus and what are they called at this point?
subcapsular region - they are called double-negative thymocytes because they have neither CD4 nor CD8 (nor CD3)
When double-negative thymocytes travel deeper into the cortex, what happens to them?
they become double positive thymocytes (they gain CD4 and CD8 - as well as CD3) mediated by the cortical epithelial cells
What is the most critical transition for the thymocytes and what happens at this transition?
cortico-medullary junction: dendritic cells mediate negative selection creating mature CD4+ or CD8+ thymocytes by destroying self-reactive thymocytes
What genetic rearrangements occur, and in what order, as the thymocytes goes from double-negative to double-positive?
first, there is D-J rearrangement in the beta chain creating CD25+, CD44low double-negative thymocytes --> these thymocytes then undergo V-DJ rearrangement and produce beta chain protein --> at this point CD4/CD8 induction occurs and transcription of un-rearranged alpha chain begins (called pTalpha) --> eventually V-J rearrangment takes place on the alpha chain, and surface expression of alpha:beta:CD3 complex is seen
What is the purpose of positive selection?
it allows thymocytes the chance to express a functional T cell receptor before undergoing negative selection in which self-reactive thymocytes are destroyed
What is meant by self MHC restriction?
a T lymphocyte will only respond to a peptide antigen on the same MHC type encountered during its selection process in the thymus (i.e. it will not react with any other native/foreign MHC molecule)
At which point does the thymocyte become either CD4 or CD8 single positive and what determines its fate?
during its interaction with the cortical epithelial cells
if the thymocyte comes in contact with an MHC-I molecule, it will continue to express CD8 but not CD4 (becoming a cytotoxic T cell)
if it comes in contact with an MHC-II molecule, it will continue to express CD4 but not CD8 (becoming a T Helper cell)
What must be true of a thymocyte for it to pass through positive and negative selection without dying by apoptosis?
positive and negative selection must have different specificity and avidity (binding strength)
also during positive selection, the self-antigen on the MHC must be recognized poorly by the thymocyte and deliver a partial signal (if it is not recognized well, the thymocute will die by apoptosis)
As T cells enter the lymph node, where do they go?
They interact with dendritic cells in an attempt to recognize their specific antigen
if this does not occur, they will move back into circulation and out of the lymph node
however, if they do encounter their antigen, they will stay in place, proliferate, and differentiate into effector T cells
Which cell-surface adhesion molecules are important for initiating the interactions of lymphocytes with APCs?
(T-cell:APC) CD2:LFA-3
LFA-1:ICAM-1
LFA-1:ICAM-1
ICAM-3:DC-SIGN
Once LFA-1 on the T cell binds with ICAM-1 on the APC, what does it allow for?
It brings the TCR and MHC-II closer together so that they can recognize each other; if recognition occurs, a signal will be sent by the T cell that induces a conformational change in LFA-1, increasing its affinity for ICAM-1 and prolonging cell-cell contact
When the TCR makes contact with an MHC molecule, how do they align?
The TCR will lie across diagonally with its α and β chain CDR3 loops contacting the center of the peptide
the CDR1 and CDR2 of the α chain contact the MHC helices at the amino terminus whereas the CDR1 and CDR2 of the β chain contact the MHC helices at the carboxyl terminus
What two events must occur for normal T-cell recognition of peptides to occur?
The T cell must be both antigen-specific and self-MHC restricted (i.e. it has to fit together with both the antigen and the MHC molecule)
How does T-cell recognition occur in the case of alloreactivity?
It involves direct recognition of a foreign MHC-peptide complex
this can involve peptide-dominant or MHC-dominant binding (i.e. the T cell binds strongly to either the foreign peptide or to the foreign MHC molecule)
How is a superantigen able to activate large numbers of T cells?
After binding the MHC-II molecule, the superantigen is able to activate TCRs on the basis of their Vβ region, not their hypervariable regions
since Vβ regions are much less specific than hypervariable regions, the superantigen can activate many T cells that share a similar structure in this region
What are some classic examples of superantigens?
Staphylococcal enterotoxins, toxic shock syndrome toxin (TSST), MMTV-7 (mammary tumor virus)
Where do the CD4 and CD8 receptors of T cells bind the MHC molecule?
CD4: binds MHC-II β2 domain
CD8: binds MHC-I α3 domain
What parts make up the T cell receptor complex?
Antigen-recognition proteins (i.e. the α and β chain); Invariant signaling proteins (i.e. CD3 complex: ε, γ, δ, and ζ chains)
How is activation of the CD3 complex achieved?
CD45 is a phosphatase which dephosphorylates Fyn and Lck, taking away their inhibition
clustering and activation of the TCR/co-receptors activates Fyn and Lck, allowing them to phosphorylate tyrosine residues on the CD3ε and ζ chain ITAMs; this allows ZAP-70 to bind and become phosphorylated
ZAP-70 then goes on to phosphorylate other molecules which in effect alters gene expression in the nucleus
What two signals are required to activate a naïve T cell and what happens when only one of these signals is present?
TCR/co-receptor binding to the MHC/peptide complex AND interaction between the T-cell CD28 with B7 on a dendritic cell (for example); if only the co-stimulators bind (CD28/B7) there is no effect, but if the TCR/co-receptor binds with the MHC/peptide complex without the co-stimulators, anergy of the T cell results (i.e. the T cell will no longer respond to antigen)
What happens after the initial cross-linking of CD28 with B7 in T cell activation?
Expression of CTLA-4 is induced; CTLA-4 binds B7 more avidly than CD28 did and delivers inhibitory signals to the activated T cell
What is the distribution of dendritic cells, macrophages, and B cell in lymph nodes, respectively?
Dendritic cells: cortex T-cell areas (not in follicles); Macrophages: throughout cortex and follicles; B cells: mainly in follicles
How do immature dendritic cells differ from their mature form?
They are located in peripheral tissues and are very phagocytic; once they have taken up foreign antigen, they migrate to the lymphoid tissues, lose their phagocytic function, and become much more potent antigen-presenting cells
How does the response of macrophages differ when taking up a nonbacterial protein vs. bacteria?
In the case of a nonbacterial protein, the macrophage will not express the B7 molecule and so any T cell that interacts with it will become anergic; however, when the macrophage takes up bacteria, it will express B7 and T cells will become activated – in certain cases, a macrophage can take up a nonbacterial protein and bacteria at the same time, in which case T cells will become activated in response to the nonbacterial protein as well
On what cells are high-affinity IL-2 receptors found and what is the response from the cell when this receptor becomes activated?
Only found on activated T cells – these activated T cells secrete high amounts of IL-2 and activate their own receptor in an autocrine fashion; activation of the IL-2 receptor leads to T-cell proliferation
After proliferation of naïve T cells, what type of cells do they differentiate into?
immature effector T cell (TH0); this cell differentiaties into either a TH1 or a TH2 cell
What is the difference between the TH1 and TH2 cells?
TH1: activates macrophages, induces B cells to produce opsonizing antibody
TH2: activates B cells to make neutralizing antibody, has various effects on macrophages
By what mechanisms can CD4 T cells cause activation of CD8 T cells?
if the CD4 T cell is an effector T cell (i.e. it has already encountered its antigen), it will activate the APC which in turn will broduce B7 which can co-stimulate the naive CD* T cell
alternatively, if a naive CD$ T cell (TH1) encounters its MHC/peptide complex and is co-stimulated by B7, it will begin to secrete IL-2, which in turn can activate a naive CD8 T cell without being co-stimulated by B7
What is the process by which a CD8 T cell (cytotoxic T cell) causes destruction of a cell?
the T cell makes nonspecific adhesions to its target cell --> if recognition occurs, redistribution of the cytoskeleton and cytoplasm of the T cell takes place, leading to the release of lytic granules at the site of cell contact --> this leads to destruction of the target cell (e.g. by apoptosis)
What distinct set of effector molecules is produced by cytotoxic T cells, TH1, and TH2 cells, respectively?
cytotoxic: perforin, granzymes, granulysin, Fas ligand
TH1: IFN-gamma, GM-CSF, TNF-alpha, CD40L, Fas ligand
TH2: IL-4, IL-5, CD40L
What are the effects of the lytic granules perforin, granzymes, and granulysin, respectively?
perforin: polymerizes to form a pore in the target membrane
granzymes: serine proteases which activate apoptosis once in the cytoplasm of the target cell
granulysin: induces apoptosis
How do cytotoxic T cells prevent from killing cells that neighbor its target cell?
they have a very polar/focused mode of secretion
When a T H1 cell causes activation of a macrophage, what changes take place in the macrophage?
they greatly increase their microbicidal effectiveness and amplify the immune response by presenting a large number of new molecules on their surface, such as CD40, B7, TNF-alpha, MHC-I and MHC-II
What happens when a T H1 cell secretes IFN-γ and CD40, and what is the effect of that T H1 cell secreting Fas ligand or TNF-β?
IFN-gamma/CD40: activates macrophages to destroy engulfed bacteria
Fas ligand/TNF-beta: kills chronically infected cells, releasing bacteria, to be destroyed in fresh microphages
What results when an intracellular pathogen or its constituents cannot be totally eliminated from the macrophage?
formation of a granuloma, which has a central portion that contains a still infected multi-nucleated giant cell, surrounded by a mass of T cells
When B cells become activated after their Ig binds epitope, what sequence of events takes place, eventually resulting in changes of gene expression?
The B cell receptors (Ig) cluster and cross-link --> this allows receptor-associated kinases (e.g. Blk, Fyn, Lyn) to phosphorylate the ITAMs (immunoreceptor tyrosine-based activation motif) on IgAlpha and IgBeta --> the tyrosine kinase, Syk, then binds to the doubly-phosphortlated ITAMs and is activated --> activation of the tyrosine kinase sends signals to the nucleus where changes in gene expression take place
Where is CD45 found and what is its purpose?
it is a tyrosine phosphatase found on all leukocytes which augments the activation of receptor-associated kinases (Blk, Fyn, Lyn) upon activation of B cells by antigen
What are the B cell co-receptors and by what mechanism do they enhance B cell activation?
CD81, CD19, CD2 (CD21)
when the complement C3d binds to a pathogen, it can interact with CR2 and by means of the co-receptors enhance B cell activation
What function does CD22 serve?
it is a negative regulator of B cell activation
What are the two parts of lymphocyte activation and what signal is responsible for each?
Competence (lymphocyte activation: caused by a naive lymphocyte in G0 binding to its antigen
Progression (lymphocyte proliferation/differentiation): induced by signals from T-helper cells --> causes S-phase entry of a B lymphocyte in late G1
What is meant by the concept of linked recognition?
in order to have a TD (thymus dependent) antibody response, the B cells and their associated T-helper cells must respond to the same antigen
Once a B cell has bound its antigen, by what mechanism does the T-helper cell cause activation of the B cell?
after capping of Ig on the B cell (Ig converges and cross-links in one area of the B cell), the antigen is endocytosed and degraded --> MHC-II binds peptides from the degraded antigen and moves to the surface --> T-helper cells recognize the peptides on MHC-II and bind --> it then produces CD40L (which binds the CD40 on the B cell) and cytokines causing proliferation and differentiation of the B cell
What does the increase in ICAM-1 due to CD40L binding cause and what is its purpose?
ICAM-1 is an intergrin which binds to the environment causing the B cell to stay put in close proximity to where it was activated, thus making time for the antibody response to take place
In general terms, what happens after the B cell has been activated the second time?
it divides about 6-10 times and differentiates to become a mass producer of antibody
What do the terms hapten and carrier refer to and what is their experimental significance?
Hapten: an epitope of an antigen recognized by B cells
Carrier: an epitope of the same antigen recognized by T-helper cells - if an antigen is so small that the B cell can recognize it (i.e. the hapten is antigenic) but the T-helper cell cant find an epitope to bind to, the hapten is not immunogenic (since it cant elicit an immune response)
What happens in terms of differentiation of the B cell when CD40L binding occurs?
the B cell switches from making IgM to making a different Ig isotype
What is the first cytokine released by the T-helper cell in response to its activation and from what location of the T-helper cell is it released?
IL-4
all of the organelles of the T-helper cell undergo capping and the secretory apparatus of the cell aligns so that the cytokines are only excreted where the B and T cells connect
After IL-4 has been produced by the T-helper cell following its activation, what other cytokines are formed and what is the function of all of these cytokines?
Proliferation Cytokines: IL-2, IL-4, IL-5
Differentiation Cytokines: IL-2, IL-4, IL-5, IFN-gamma, TGF-beta
How do activated B cells and T-helper cells meet in the lymph node?
B cells leak out of the high endothelial venules (HEV) and meet up with T-Helper cells which have been activated by the same antigen because of their interaction with a dendritic/Langerhans cell
Once they meet and interact, they form a primary locus/follicle in the lymph node where proliferation/differentiation of B cells occurs
Once a primary lymph follicle has been created in the T-cell area, what happens next?
The primary follicle migrates to the B-dependent area where it becomes a secondary germinal center
What are the various parts of a secondary follicle/germinal center?
Dark Zone: lymphocyte proliferation
Light Zone: contains a large amount of follicular dendritic cells
Mantle Zone: contains mature lymphocytes (e.g. plasma cells)
When do germinal centers begin to appear, comparing primary vs. secondary exposure?
Primary: ~ 1 week
Secondary: 2-3 days
What three key events occur in the germinal center?
B cell proliferation
Isotype switching (mostly to IgG)
Affinity Maturation (affinity get stronger as the B cell matures)
How do follicular dendritic cells help to prolong an immune response?
They have Fc receptors and complement receptors (CR2) which can hold the immune complex (iccosomes = antibody + antigen) in place so that B cells can contine to get stimulated; once B cells lose stimulation by antigen, they die by apoptosis
What is the half-life of a B cell comparing those that successfully enter lymphoid follicles (i.e. they have encountered antigen) and those that fail to enter?
Fail to enter: ~ 3 days
Successfully enter: ~ 2 weeks
How do general capillaries and brain capillaries differ?
General Capillaries: discontinuous and relatively permeable to unbound drugs
Brain Capillaries: physically joined and creat a significant barrier for drugs
What are some common ways for drugs to travel through cell membranes and what are examples of drugs that use those mechanisms?
Endocytosis (e.g. absorption of complexes of iron and proteins - transferrin)
Facilitated Diffusion (e.g. glucose and Vit B12)
Active Transport (e.g. Levo-Dopa and Gabapentin)
Diffusion (most important mechanism - required non-ionized moiety that is lipophilic)
What forms of a drug can move by bulk flow to the interstitial space?
both the ionized and unionized forms (but NOT hte bound forms)
In what case is it possible for protein-bound drug to diffuse through the discontinuities of general capillaries?
during inflammation
By what mechanisms can drugs flow across the blood-brain barrier?
by diffusion if the drug is unionized
through endocytosis of the ionized/unionized forms
during infammation
What two factors does the drug ionization of a weak base or a weak acid depend on?
pKa (-log of H+ dissociation constant) and pH (H+ concentration of the biophase in which it exists)
What does a high pKa indicate for any given compound?
that it easily attracts a hydrogen ion
8. What are the normal pH values for blood, CSF, gastric fluid, intestinal fluid, glomerular filtrate, and urine?
blood: 7.4
CSF: 7.2
gastric fluid: 1-2
intestinal fluid: 5-6
glomerular filtrate: 7.4
urine: 6
At a low pH, in what form are drugs found that are either a weak base or a weak acid?
Weak Base: ionized
Weak Acid: unionized
What is the Henderson-Hasselbach equation?
pH = pKa + log (A-/HA) and pH = pKa + log (B/HB+) for a weak acid and weak base, respectively
At what point is 50% of a drug found in its ionized form?
when their pKa is equivalent to the pH of their environment
What is meant by the term ion trapping?
it describes the unequal division of ions b/w different compartments in the body (i.e. the ionized form gets trapped in one compartment because of the pH in that compartment) - for a weak acid, the ions get trapped in the compartment with the higher pH whereas with a weak base the ions will be trapped in the compartment with the lower pH (i.e. in gastric juice)
What is the partition coefficient a measurement of?
Lipophilicity (is more important to diffusion rate than non-ionized fraction when comparing thiopental and pentobarbital)
What structures can a drug bind before binding its active receptor (i.e. to remain in the drug reservoir)?
Soluble proteins in blood, protein in tissue, fat deposits - the sequestration of drugs in these reservoirs prolongs its action (keeps it from being degraded or excreted)
What factors can influence a drug’s binding capacity and what effects can this have on the patient?
Two drugs can compete for the same binding site
certain diseases can influence a drug's binding capacity (e.g. hypoalbuminemia)
if the drugs binding capacity is decreased, plasma levels will increase causing possible toxicity of the drug
What are some examples of binding proteins that will bind basic drugs?
albumin (also binds acidic compounds), α1-acid glycoprotein, lipoproteins
What other processes function to balance out a sudden increase in free drug levels when the drug has been displaced from its binding protein by a competitor?
increased metabolism and excretion
Which diseases reduce albumin binding?
hypoalbuminemia, hyperbilirubinemia, uremia
During which diseases is α1-acid glycoprotein increased and what effect does this have?
cancer, arthritis, Chron's disease
causes an increase in the binding of basic drugs and thus higher doses are needed to exert the same therapeutic effects
For what sites of infection are antibodies effective?
Extracellular sites (interstitial spaces, blood, lymph, epithelial cells), but NOT for intracellular spaces (cytoplasm, vesicles)
By what DIRECT mechanisms can pathogens cause damage and what are some examples of diseases that result from each mechanism?
exotoxin production (secretions form pathogen): boil, food posioning, cholera, tetanus
Endotoxin (toxin is intrinsic part of pathogen): gram-negative sepsis, meningitis, pneumonia
Direct cytopathic effect (directly kill the cell): lots of viral infections
By what INDIRECT mechanisms can pathogens cause damage and what are some examples of diseases that result from each mechanism?
Immune complexes: kidney damage
Anti-host antibody: rheumatic fever
Cell-mediated immunity: tuberculosis
By what mechanisms do macrophages destroy pathogens?
acidification, toxic oxygen derived products, toxic nitrogen oxides, antimicrobial peptides, enzymes (lysozymes, acid hydrolases), competitors (Vit B12 protein and lactoferrin for Fe)
Which T effector cells respond to parasites?
CD4 TH2 cells - they are important for switching to IgE production
What is the purpose of the CD4 regulatory cells?
They suppress T-cell response to prevent auto-immune disease
What is the primary antigen-presenting cell used in the acquired immune response?
dendritic cell (it is ubiquitous throughout the body)
What do TH17 cells do?
they activate fibroblasts and epithelial cells to bring in neutrophils
What is the difference between a toxin and a toxoid?
a toxoid is the inactive form of the toxin which can be used as a vaccine
Which Ig isotypes are best at activating the complement system?
IgM and IgG3
What is meant by “original antigenic sin”?
after an initial antigenic response, the immune system will only respond to previously presented epitopes during a second exposure, even though the organism causing the second infection may have different epitopes that those found on the original organism - this concept has been found to exist in response to influenza, malaria, and HIV
What concept of drug metabolism does flurazepam exemplify?
Drugs which are both active and have an active metabolite – also, the half-life of the drug and the metabolite must be taken into consideration (for flurazepam T1/2 = 2-3 hrs, and its metabolite desalkylflurazepam has a T1/2 of 30-100 hrs)
What is the end-result from a CYP450 drug metabolism?
Oxidation of the parent drug
In which case can Phase II metabolism occur without prior Phase I metabolism?
If the parent compound already contains a functional group that can be directly conjugated
What type of reaction is involved in Phase II metabolism?
Biosynthetic metabolism (i.e. an endogenous substrate conjugates with the Phase I metabolite)
What are the characteristics associated with Phase I metabolism?
Usually oxidation that inactivates the parent compound by unmasking a functional group which makes it more polar; if the metabolite is polar enough, the metabolite will be excreted, otherwise it will undergo Phase II metabolism – instead of oxidation, the parent compound can also undergo reduction or hydrolysis in Phase I metabolism
What are the various aspects of “first-pass” metabolism?
Liver metabolism before entering general circulation; Intestinal metabolism (chlorpromazine, cyclosporine); Gastric acid (PPIs: omeprazole,esomeprazole); Digestive enzymes (insulin)
What is the main organ of drug metabolism?
Liver (also lungs, kidney, intestine, placenta in decreasing order)
What is the usual end-result of drug metabolism by the body?
Usually less pharmacologically active, more polar, more water soluble, more readily excreted
What are drugs called that are activated by metabolism and what is an example?
Pro-drugs: codeine  morphine (also cyclophosphamide, carbamazepine, proton pump inhibitors)
What are polarized drugs more readily excreted?
Non-ionized drugs can diffuse across cell membranes to be reabsorbed in the blood stream; however, if it is ionized, this diffusion will not be possible and the drug will be excreted
What P450 isoform is responsible for the metabolism of ethanol and acetaminophen?
CYP2E1
Which drugs are considered inhibitors of CYP2A6 and CYP2B6?
Tranylcypramine, methoxysalen
Which drugs are considered inhibitors of CYP1A1 and CYP1A2?
Fluoxetine, cimetidine, ciprofloxacin
What effect will a low sulfate diet, along with acetaminophen intake have on the patient?
Acetaminophen will not be metabolized as rapidly, leading to toxic levels of acetaminophen which can cause liver failure
By what mechanism does irreversible enzyme inhibition take place and how can this process be resolved?
An inhibitor is metabolized to a form that irreversible binds to and inactivates the enzyme (e.g. erythromycin and 3A4); can only be resolved by synthesizing new enzyme
By what two mechanisms can competitive enzyme inhibition occur?
Two drugs bind to the same active site on an enzyme (e.g. macrolide antibiotic and statin on 3A4); One drug that functions as a substrate and another that inhibits the enzyme (codeine and quinidine on 2D6)
What enzymes are responsible for Phase II metabolism, where in the cell are they found, and what can complicate their functioning?
Transferases; can be microsomal or cytosolic – the transferases use endogenous substances (e.g. sulfate) for their reaction mechanisms, so a lack of these substances in diet can result in problems with drug metabolism
What is the most predominant P450 isoform, and what is the importance of its closely related isoforms?
CYP3A4 (accounts for 50% of all drug oxidations) – CYP3A5 has a similar specificity but is not expressed in all individuals (determined by genetics); CYP3A7 is also similar but is only expressed in the fetus
Which drugs, with narrow therapeutic indices, are metabolized by CYP2C9?
Phenytoin, warfarin (also metabolizes NSAIDs) – responsible for 20% of all drug oxidations
Which P450 isoform comprises only about 5% of all liver isoforms, but actually accounts for around 30% of all drug oxidations?
CYP2D6 (responsible for codeine  morphine metabolism)
Which drugs are considered inhibitors of CYP3A4?
Cimetidine, erythromycin, ketoconazole, grapefruit juice, HIV antivirals
Which drugs are considered inducers of CYP2D6?
Dexamethasone, rifampin
Which drugs are considered inducers of CYP2C9 and other 2Cs?
Phenobarbital, dexamethasone, rifampin
Which drugs are considered inducers of CYP2A6 and CYP2B6?
Phenobarbital, dexamethasone
Which drugs are considered inducers of CYP1A1 and CYP1A2?
Smoking, char-grilled meat, omeprazole
What clinical problems can arise from enzyme induction?
Therapeutic failure when active drug concentration is lowered by enhanced metabolism; Increased drug effects when the metabolite is the active form (i.e. with pro-drugs)
What factors are able to induce (i.e. increase) enzyme metabolism and by what mechanisms can this occur?
Prolonged exposure to certain drugs, air pollutants, cigarette smoke, ingested materials – can occur by increased synthesis of new enzyme (e.g. dexamethasone and rifampin induction of 3A4) or by decreased proteolytic degradation of enzyme (e.g. ethanol-mediated induction of 2E1)
Which drugs are considered inhibitors of CYP2C9?
Fluoxetine, fluconazole, zafirlukast,
Which drugs are considered inhibitors of CYP2D6?
Fluoxetine, cimetidine, paroxetine, quinidine!
Which drugs are considered inhibitors of CYP2C19?
Fluoxetine, cimetidine, PPIs (omeprazole)
Which drugs are considered inhibitors of CYP3A4?
Cimetidine, erythromycin, ketoconazole, grapefruit juice, HIV antivirals
Which drugs are considered inducers of CYP2D6?
Dexamethasone, rifampin
Which drugs are considered inducers of CYP2C9 and other 2Cs?
Phenobarbital, dexamethasone, rifampin
Which drugs are considered inducers of CYP2A6 and CYP2B6?
Phenobarbital, dexamethasone
Which drugs are considered inducers of CYP1A1 and CYP1A2?
Smoking, char-grilled meat, omeprazole
What clinical problems can arise from enzyme induction?
Therapeutic failure when active drug concentration is lowered by enhanced metabolism; Increased drug effects when the metabolite is the active form (i.e. with pro-drugs)
What factors are able to induce (i.e. increase) enzyme metabolism and by what mechanisms can this occur?
Prolonged exposure to certain drugs, air pollutants, cigarette smoke, ingested materials – can occur by increased synthesis of new enzyme (e.g. dexamethasone and rifampin induction of 3A4) or by decreased proteolytic degradation of enzyme (e.g. ethanol-mediated induction of 2E1)
Which drugs are considered inhibitors of CYP2C9?
Fluoxetine, fluconazole, zafirlukast,
Which drugs are considered inhibitors of CYP2D6?
Fluoxetine, cimetidine, paroxetine, quinidine!
Which drugs are considered inhibitors of CYP2C19?
Fluoxetine, cimetidine, PPIs (omeprazole)
How does age affect drug metabolism?
Maturation of full function occurs slowly after birth; Decline in drug metabolism occurs with aging, especially affecting the elderly (this affects mainly Phase I metabolism!)
What causes “alcohol flush”?
A mutant allele causing a deficiency in alcohol dehydrogenase – common in Asian populations
Which conjugating enzyme is commonly polymorphic and how does this influence those affected?
NAT (N-acetyltransferase): acetylates isoniazid, procainamide, caffeine; people with less active enzyme will be slow acetylators and take longer to metabolize the aforementioned drugs
What can a deficiency of CYP2C19 lead to?
An enhanced therapeutic effect of omeprazole (reducing ulcers), possibly leading to toxicity
Which P450 isoform is a good example of polymorphism occurring in populations?
CYP2D6 (5-10% of European Caucasians are poor metabolizers as are 1-2% of Southeast Asians)
During which phases of pregnancy does induction of metabolism occur?
Second and third trimesters
Which drugs are thought to be affected somewhat by gender differences?
Ethanol, propranolol, benzodiazepines, estrogens, salicylates
Which drugs are considered inducers of CYP3A4?
Glucocorticoids, rifampin, phenobarbital, St. John’s wort!, carbamazepine, phenytoin, pioglitazone
What types of disease will have a large effect on drug metabolism?
Liver disease: hepatitis, alcoholic liver, biliary cirrhosis, hepatocarcinoma, fatty liver, altered liver perfusion secondary to cardiac disease
Which Ig isotype is best at opsonization and sensitization for killing by NK cells?
IgG – whereas IgE is best at sensitization of mast cells
What is meant by somatic hypermutation regarding Ig exposure to antigen?
Actively proliferating B cells experience a large amount of point mutations in their hypervariable regions (1/103 bp vs. 1/108 for normal somatic genes)
What Ig isotype does IL-5 augment the production of?
IgA
When IL-4 is secreted from T-helper cells, which Ig isotypes will be produced by the B cells?
IgG1 and IgE (also inhibition of IgG2a and IgG3)
Which Ig isotype can easily diffuse into extravascular sites?
IgG
Why do IgM and IgA have trouble crossing the placenta?
IgM is very large and IgA forms a dimer – IgG is best at crossing the placenta
What Ig isotype is needed for protection of mucosal surfaces and what characteristic makes it function so well?
IgA (dimer): can easily transport across epithelium
What are the different phases of the primary acquired immune response?
Lag phase (~1 week); Log Phase; Plateau phase; Decline
What is found on the 5’ side flanking each Ig isotype coding domain, and what is the function of this sequence?
A switch site (all switch sites have the same sequence) – switch recombinase links together two switch sites and loops out the connecting piece of DNA; this is how B cells can change Ig isotype in response to CD40L binding to CD40
What is the difference in the quality of the immune response when comparing primary and anamnestic (secondary) response?
Primary: IgM; Secondary: IgG, IgA, IgE
Once B cells have differentiated into plasma cells, where do they travel to and what do they secrete?
Travel to the bone marrow where they secrete massive amounts of antibody (3,000-30,000/sec)
How do TI-2 antigens cause activation of B cell in contrast to TI-1 antigens?
They cause extensive cross-linking of antigen receptors (since TI2 antigens are highly polymeric structures – e.g. bacterial cell wall polysaccharides and polymeric proteins like flagellin);
whereas TI1 antigens use the CD14 receptor
What is the receptor on B cells that responds to LPS and what effect does this type of activation have at low concentrations?
CD14 (via LBP) – at low concentrations, this signal decreases the threshold for B cell activation by another antigen on the bacterial surface
What substance (in high concentrations), made by Gram-negative bacteria, functions as a polyclonal activator of all B cells?
Lipopolysaccharide (LPS) = example of a TI1 antigen
What is the key difference between T-dependent B cell activation and T-independent B cell activation?
T-dependent B cell activation is normally used when the antigen is a protein whereas T-independent B cell activation is in response to carbohydrates (constituents of bacterial cell walls)
How do plasma cells make the switch from secreting membrane Ig to secreting antibody?
By differential RNA processing of the primary transcript so that the hydrophobic amino acids that normally insert in the membrane are spliced out
What functions have plasma cells lost in comparison to B cells?
No surface Ig, no surface MHC-II, no growth, no somatic hypermutation, no isotype switching; only high-rate Ig production
What does somatic hypermutation of the hypervariable region of Ig lead to?
Affinity maturation of the antibody response – i.e. mutations in the antibody variable region will lead to an increase in the affinity of the antibody for the antigen (since those mutations with lower affinity will die by apoptosis and those with higher affinity will proliferate more vigorously)
Which enzyme, found in B cells, is responsible for creating the mutations that cause affinity maturation and by what mechanisms does it achieve this?
Activation-induced cytidine deaminase (AID) – AID is an RNA editing enzyme which changes cytosine to uracil; it can also change single-stranded DNA, but ends up changing cytosine to adenine, thus changing a CG pair to an AT pair; (note, AID is also involved in isotype switching)
Why are point mutations only seen in the hypervariable regions and not in any of the structural regions?
Mutations in the structural regions will lead to a defective Ig and thus apoptosis
How does the B cell response to TI activation differ from that of TD activation?
Differentiation is limited to only IgM secreting plasma cells; there is no somatic hypermutation and no affinity maturation of the response; also, no memory cells are produced
What is meant by Antibody Dependent Cellular Cytotoxicity (ADCC)?
Cells with Fc receptors (e.g. NK cells, macrophages) recognize antibody bound to antigen  cross-linking of Fc receptors activates the cell and the antigen is destroyed;
The same process occurs with opsonization where antibody-bound antigen is recognized by Fc receptors on macrophages, thus facilitating phagocytosis of that antigen
What must happen before the Fc receptor will bind antibody?
The antibody must bind antigen in order to induce a conformational change so that the Fc receptor can accept it – the one exception is FcεRI, which will bind antibody without antigen bound to it (mostly associated with mast cells and allergens)
What deficiency is found in patients with Hyper IgM Syndrome, what are the two causes, and what pathology is characteristic in these patients?
Isotype switching is not possible – in Type 1, T cells do not produce CD40L, whereas in Type 2 there is a deficiency of the enzyme AID; both types do not have primary follicles or germinal centers in their lymph nodes
During the first year or two of life, which Ig isotypes are found at low levels?
IgG and IgA – they do not reach mature levels for the first several years; this normal process used to be diagnosed as perinatal immunodeficiency
What is the primary cell transformed in follicular lymphoma?
The B-1 B cell (as opposed to the much more common B-2 B cells)
How does the B cell response to TI antigen differ in infants?
There is no response to TI2 antigen and a limited polysaccharide response
When happens after C1/C1q has bound the antigen-antibody complex?
The serine esterase C1r activates is neighbor, serine esterase C1s
After the initial C3bBb is bound to the pathogen surface, what enzyme cooperates with it and with what end-result?
Properdin (factor P) binds to and stabilizes C3bBb which continues to act as a C3 convertase causing multiple C3b to bind to the pathogen surface
What is the mechanism by which the alternative pathway results in the binding of C3b to the pathogen surface?
C3 is hydrolyzed forming iC3  the enzyme B binds to iC3  enzyme D cleaves B into Bb and Ba where Bb remains attached to iC3 and Ba floats away  another C3 binds to the newly formed iC3Bb and is cleaved into C3a and C3bBb bound to the pathogen surface
What does C4b2a cleave and what happens to its products?
It binds to and cleaves C3 into C3a and C3b  C3b subsequently binds to the microbial surface
What is the function of C2a?
C2a binds to the surface C4b forming C4b2a, the classical C3 convertase
Besides cleaving C4, what other effect does C1s have?
It cleaves C2 into C2a and C2b, where C2a is the larger component
What does C1s exert its effect on and what are the two possible outcomes?
C1s cleaves C4a off of C4, leaving behind the C4b part which now contains an exposed thioester bond  nucleophilic attack on the thioester bond by H2O creates soluble C4b, whereas nucleophilic attack by R-OH or R-NH2 results in C4b binding to the pathogen surface
What three pathways are associated with complement activation and what is the common end-result of all three?
Classical pathway: antibody-antigen binding; Lectin pathway: mannose-binding lectin binds to surface pathogen; Alternative pathway: pathogen surface creates local environment conducive to complement activation – all three pathways lead to complement activation and covalent binding of C3b to surface component of the pathogen
How does C1 binding differ when comparing its interaction with IgM vs. IgG?
C1 binds to a single antigen-bound IgM pentamer whereas it requires two or more antigen-bound IgG to bind C1q
What are the early acting components of the classical pathway and which component has the largest plasma concentration?
C1q,r,s; C2; C3; C4 – C3 has the largest plasma concentration
What are the ligands of the CR1 receptor, and what is the receptor’s function?
C3b, C4b; CR1 promotes C3b and C4b decay, stimulates phagocytosis, and is involved in erythrocyte transport of immune complexes
When a C5 convertase cleaves C5, what happens to the formed C5b fragment?
C5b joins with C6 and C7 and inserts itself in the bacterial cell membrane  C8 then joints followed by a large number of C9s which form a pore in the bacterial cell wall
How do the C3 convertases in the classical, lectin, and alternative pathways become C5 convertases?
By binding an additional C3b (thus becoming C4bC2aC3b for the classical and lectin, and C3bBbPC3b for the alternative)
How do erythrocytes aid in the destruction of certain immune complexes?
Antigen-antibody complexes form in circulation  C3b becomes activates and binds to the complex  C3b then binds to the CR1 receptor on erythrocytes --> in the spleen and liver the immune complex is handed over to phagocytic cells that destroy it
What is the mechanism by which bacteria are engulfed by neutrophils?
Antibody binds to the pathogen and activates complement --> C3b binds to pathogen --> engulfment of pathogens is now mediated by Fc receptors and complement binding
Which receptor is found on Kupffer cells and functions in bacterial opsonization?
CRIg – binds to C3b and iC3b
Where are the CR3 and CR4 receptors found, what is the ligand, and what is their function?
Found on macrophages, monocytes, PMNs; their ligand is iC3b and they stimulate phagocytosis
What receptor on the macrophage surface binds to C3b and what happens next?
CR1 binding causes opsonization of the pathogen
In the lectin pathway, which segment associated with the MBL/ficolins cleaves C2 and C4 and which part of these molecules is the active part?
MASP-2; cleaves C2 and C4 so that C2a and C4b can form C4b2a, which is the C3 convertase
What does the MBL/ficolins attach to on the bacterial cell surface?
Repeating carbohydrate molecules
What are C3a, C4a, and C5a known as and what is their function?
Anaphylatoxins – they act on blood vessels to increase vascular permeability allowing for extravasculation of antibodies and complement as well as leading to increased migration of macrophages , neutrophils, and lymphocytes
Where in the body is the bulk of complement created, and what is special about the biosynthesis of C1, C7, factor D and especially factor P?
90% is synthesized in the liver – C1: intestinal epithelium, macrophages, monocytes; C7: bone marrow granulocytes, dendritic cells; factor D: adipose tissue; factor P: dendritic cells ONLY
Deficiency of which complement is the most commonly occurring (though still rare)?
C1INH, MLB, C2
What is the function of CD59 and how does it accomplish this?
It prevents pore formation by inhibiting the membrane attack complex at the C8 to C9 stage
What molecule can inactivate C5 convertase and what subsequent cleavage occurs?
CR1 and H both have the capability to displace C3b from C5 convertase, changing the C5 into a C3 convertase; this is followed by factor I coming in to cleave the C3b on the C3 convertase, completely silencing the enzyme activity
What three molecules have the capacity to displace C2a from the C4b2a complexes and how do they function?
DAF, C4BP, CR1 – after displacing C2a, factor I is drawn in which is a protease that cleave C4b (into inactive C4c and C4d) and prevents it from rebinding C2a
What is the molecule that can control complement activation by covalently binding to C1r and C1s, and in what disease does the patient have reduced levels of this compound?
C1INH (C1 inhibitor) – reduced levels are found in people with hereditary angioneurotic edema (HANE); they will have bouts of skin, gut, and airway edema triggered by stress
What are the ligands of the CR1 receptor, and what is the receptor’s function?
C3b, C4b; CR1 promotes C3b and C4b decay, stimulates phagocytosis, and is involved in erythrocyte transport of immune complexes
When a C5 convertase cleaves C5, what happens to the formed C5b fragment?
C5b joins with C6 and C7 and inserts itself in the bacterial cell membrane  C8 then joints followed by a large number of C9s which form a pore in the bacterial cell wall
How do the C3 convertases in the classical, lectin, and alternative pathways become C5 convertases?
By binding an additional C3b (thus becoming C4bC2aC3b for the classical and lectin, and C3bBbPC3b for the alternative)
How do erythrocytes aid in the destruction of certain immune complexes?
Antigen-antibody complexes form in circulation  C3b becomes activates and binds to the complex  C3b then binds to the CR1 receptor on erythrocytes --> in the spleen and liver the immune complex is handed over to phagocytic cells that destroy it
What is the mechanism by which bacteria are engulfed by neutrophils?
Antibody binds to the pathogen and activates complement --> C3b binds to pathogen --> engulfment of pathogens is now mediated by Fc receptors and complement binding
Which receptor is found on Kupffer cells and functions in bacterial opsonization?
CRIg – binds to C3b and iC3b
Where are the CR3 and CR4 receptors found, what is the ligand, and what is their function?
Found on macrophages, monocytes, PMNs; their ligand is iC3b and they stimulate phagocytosis
What receptor on the macrophage surface binds to C3b and what happens next?
CR1 binding causes opsonization of the pathogen
In the lectin pathway, which segment associated with the MBL/ficolins cleaves C2 and C4 and which part of these molecules is the active part?
MASP-2; cleaves C2 and C4 so that C2a and C4b can form C4b2a, which is the C3 convertase
What does the MBL/ficolins attach to on the bacterial cell surface?
Repeating carbohydrate molecules
What types of physiological interactions are seen in ADRs and what are examples of them?
Absorption: opioid analgesics --> altered gastric motility and drug absorption; Protein binding: tolbutamide lowers blood sugar, but in combination with dicumarol (which displaces tolbutamide from albumin) can cause hypoglycemia; Renal excretion: probenicid and penicillin; Inhibition of metabolism: acetaminophen in glutathione-depleted chronic alcoholics; Induction of metabolism: barbiturates; Interactions at the site of action: opioids and CNS depressants
How does the categorization of NSAIDs differ between 1st & 2nd trimesters and the 3rd trimester?
During 1st & 2nd it is a Category B drug whereas during the 3rd trimester it becomes Category D due to its effects on the premature closing of the ductus arteriosus
What distinguishes Category D from Category X drugs?
Both have positive evidence of risk of fetal harm, but with Category D drugs the benefits to the pregnant mother may outweigh the risks to the fetus (e.g. lithium, phenytoin), whereas Category X drugs should never be used in pregnant women (e.g. statins, isotretinoin, thalidomide)
What defines pregnancy Category C drugs?
There are no adequate, well-controlled studies in pregnant women and either animal studies have shown adverse effects or none have been conducted
How are pregnancy Category A and Category B drugs defined, respectively?
Category A: safe; Category B: either no harmful effects in animals but no human studies done OR adverse effects in animals but none found in well-controlled human studies (acetaminophen, penicillin, oxycodone)
What is the most dangerous time for the fetus to be exposed to drugs?
18-55 days after conception; earlier exposure usually causes death; later exposure produces minor malformations/retardation/functional deficits
What changes in pregnant women alter drug levels?
Increased plasma volume, GFR, CO (decreases drug levels); Increased body fat (increased Vd for fat-soluble drugs); Decreased albumin levels (increase free drug/increased renal excretion); Delayed gastric emptying (increases absorption in stomach/delays absorption from intestine); Increased gastric pH (decreased absorption of acid-requiring drugs); Estrogen/progesterone (modulate hepatic enzyme activity)
What is the difference between Type A and Type B ADR?
Type A: extension of the primary effect or an additional known pharmacological effects;
Type B: unusual or unexpected reactions (idiosyncratic) or immune-mediated reactions
What results when a patient is given IV heparin and antihistamine?
The acid and base will form an inactive salt (another example of a chemical interaction is tetracycline chelation by metal containing antacids like Maalox)
What percent of hospitalizations is due to ADRs and what percent of these is preventable?
3-5% (300,000); inpatients have ~30% chance of an ADR – 85% of ADRs are preventable
Why are opioids used in high doses during pregnancy considered Category D?
If used close to term, they can cause addiction by the fetus and depress respiration
How does infection/inflammation influence glomerular filtration?
Increases due to increased pore size (may allow protein-bound drugs to pass through)
What are colesevalam and activated charcoal examples of?
Drugs that are used to keep substances from being absorbed in the intestine – colesevalam for cholesterol and activated charcoal for a variety of toxins
What are some examples of drugs that are excreted through anion and cation transporters, respectively?
Anion transport: furosemide, penicillin G (must be given along with probenecid to block the transporter in order to offset excessive transport); Cation transport: cimetidine, neostigmine, morphine
How does active tubular secretion come about and for which drugs is this an important method of excretion?
Through the use of cation and anion transporters – important in drugs that are highly protein-bound and don’t filter through the glomerulus
What happens to aspirin when the urine is alkalinized?
Since aspirin is a weak acid, it will become ionized in alkalinized urine, making it more readily excreted (important in aspirin overdose)
How does acidification and alkalinization of the urine affect drugs that are weak bases and weak acids, respectively, in regard to tubular reabsorption?
Acidification: promotes reabsorption of weak acids and decreases reabsorption of weak bases; Alkalinization: promotes reabsorption of weak bases and decreases reabsorption of weak acids
What drug characteristics allow for greater back diffusion (i.e. tubular reabsorption)?
Non-ionized, lipophilic drugs
What are the principle organs of excretion?
Kidney(!), intestine, biliary system, lungs – also sweat, saliva, milk, hair
What drug characteristics lower their glomerular filtration?
Large size, charge (especially anions), globular shape , reduced protein-binding
What are the three processes that determine the concentration of substances in the urine?
Glomerular filtration, active secretion, tubular reabsorption
Why is the half-life of ezetimibe substantially longer that expected?
After being glucuronidated and excreted by the liver, it is hydrolyzed by the intestinal flora back to its original form and subsequently reabsorbed in the intestine to once again exert its effects
What are some examples of drugs that can be found in breast milk?
Caffeine, nicotine, ethanol, aspirin, barbiturates, morphine, ACE inhibitors
How does the pH of milk differ from that of plasma and how does it affect drug excretion?
Milk pH: 6.5 vs. plasma pH: 7.4; weak bases will be more ionized in milk and get trapped
What is the general rule of thumb regarding drug safety in pregnant women?
If the drug is safe to use in a neonate, it is safe to be taken by the lactating woman
What is the clinical significance of drugs being excreted through sweat?
They can sometimes cause dermatitis or other skin reactions
What happens to drugs secreted in saliva?
They are swallowed and thus reabsorbed
What determines whether a drug can be excreted via the lungs?
Whether it is volatile (e.g. N2O, alcohol, acetone, garlic)
By what mechanisms does biliary excretion occur and what is an example of a drug that uses this system?
Anion and cation transporters – cardiac glycosides are excreted this way
What is meant by enterohepatic recycling and what drugs use this system?
Drugs that are excreted by the liver into the bile are often reabsorbed in the intestine and thus not excreted from the body – examples are Vit. D3, Vit. B12, folic acid, estrogen
How does the dose-response curve shift when a nonequilibrium competitive antagonist is added to a particular agonist?
The curve shifts to the right AND the maximum response is decreased (because the total available receptors is reduced) – these effects cause the slope of the curve to be decreased
What mechanism is responsible for a cell’s basal level of functioning?
Without the presence of agonists or antagonists, a cell receptor is in equilibrium between its active and inactive form; the time it spends in the active form is responsible for its basal functioning
What effects do inverse agonists have, how do they achieve this effect, and what are some examples of receptors for which inverse agonists have been described?
They reduce basal functioning of the cell (i.e. they have negative efficacy) by binding to the inactive form of the receptor with higher affinity – found for 5-HT, opioid, β-adrenoceptor, benzodiazepine
How does the dose-response curve of functional antagonism compare to that of noncompetitive antagonism?
The are identical (nonspecific, not surmountable)
What are the characteristics of functional antagonism?
Two agonists that initiate signaling via independent and opposite cellular pathways will cause functional antagonism (e.g. simultaneous sympathetic and parasympathetic innervation)
How does the dose-response curve shift when a noncompetitive antagonist is added to a particular agonist?
The curve shifts to the right AND the maximum response is decreased; in contrast to competitive antagonism the effects are NOT specific and will affect many agonists (because it doesn’t function at the level of the receptor, but rather on a particular step in a signaling pathway)
W hat are the hallmarks of nonequilibrium competitive antagonism?
Specific, irreversible, competitive, not surmountable (i.e. it reduces maximum response)
What are the two types of antagonisms and what are examples of each?
Chemical: protamine/heparin or dimercaprol chelation; Pharmacological: most common type, many examples possible
What are the hallmarks of equilibrium competitive antagonism?
Specific (for a particular agonist), reversible, competitive, surmountable (i.e. it has no effect on maximum response)
How does the dose-response curve shift when an equilibrium competitive antagonist is added to a particular agonist?
A parallel shift to the right (i.e. a greater dose of agonist is required to get the same level of response, but the shape of the curve does not change)
To what form of the receptor does a (neutral) antagonist bind?
Both the active (R*) and inactive (R) forms – neutral simply means that is has an efficacy of 0 and does not change basal functioning of the cell
What results when you add an inverse agonist to an agonist?
The inverse agonist will reduce the response generated by the agonist
What calculation is used to calculate half-life?
[t1/2 = 0.693/ke] where ke is the elimination rate constant calculated by [rate of elimination/amount in body] or by looking at the slope of the plasma concentration vs. time log curve
When using the 2 compartment model of pharmacokinetics, what two phases can be seen on the drug plasma concentration vs. time graph?
Distribution phase and elimination phase
What is the relationship between clearance and half-life?
It is an inverse relationship (i.e. if you increase the half-life of a drug you will decrease its clearance)
What is the term that describes the volume of plasma from which drug is removed per unit time and how is it calculated?
Clearance – [Renal clearance = total amount in urine/AUC] and [systemic clearance = dose/UAC] or [systemic clearance = Vd x ke]
How does ascites affect volume of distribution?
It increases the total extracellular fluid volume
How is it possible for certain drugs to have a very large volume of distribution (e.g. amiodarone)?
If the drug gets selectively taken up by a tissue or binds to a binding protein, it can appear to have a much greater volume of distribution when compared to their plasma volume
What is meant by volume of distribution and how is it calculated?
Volume of fluid in which the drug appears to be dissolved – [Vd = total amount of drug absorbed/drug in plasma at t=0]
What does the pharmacokinetic parameter AUC (area under the curve) mean?
It is calculated from a drug plasma concentration vs. time curve; it gives an indication of drug exposure, clearance, bioavailability, etc.
What physiological factors affect half-life?
CYP inhibition/induction, renal/hepatic impairment
How many half-lives does it take to eliminate >95% of dose?
Between 4 and 5
What factors determine the absorption rate constant, Ka?
Chemical and physical properties of the drug formulation; the presence of food (ingesting food along with a drug delays the action of that drug because it delays the amount of time it takes for the drug to get to the intestine and it dilutes the drug)
What is a loading dose and how is it calculated?
Amount of drug given to a patient to reach steady state level faster than it would normally take (i.e. ~5 half-lives) – it is calculated as [LD = Css x Vd]
How long does it take to achieve steady state on multiple drug dosing and how is the concentration at steady state (Css) calculated?
It takes ~5 half-lives to achieve steady state – Css is calculated by either [Css = (F x DoseRate)/Clearance] or [Css = (F x Dose)/(Clearance x T)]
How does the zero-order elimination graph differ from first-order elimination and how does zero-order elimination occur?
In zero-order elimination a constant amount is eliminated per unit time, whereas in first-order elimination a constant fraction is eliminated per unit time; zero-order elimination most commonly occurs because of saturation of their elimination process (i.e. the drug’s degrading enzyme is constantly saturated) – examples of zero-order drugs are ethanol and phenytoin
What factors affect bioavailability and how does bioavailability affect Cmax and Tmax
Pre-systemic degradation, export transporters in the small intestine, drug formulation – bioavailability directly influences Cmax, but has no effect on Tmax
What is meant by bioavailability and how is it calculated?
It is the fraction of orally administered drug which is absorbed into the plasma – [F = AUCoral/AUCiv]
How does a higher value of Ka affect Cmax and Tmax?
If increases Cmax and decreases Tmax (more absorbed in a less time)
What are examples, concerning the 2 compartment model of pharmacokinetics, of rapid and slowly excreting compartments?
Rapid compartment: Brain, muscles, maybe visceral; Slow: fat, skeleton, maybe visceral
What factors affect Cmax and Tmax, respectively?
Cmax: Ke, Ka, F; Tmax: Ke, Ka
How does oral administration of a drug compare, pharmacokinetically, to IV administration?
Oral administration has an added absorption phase, which limits the maximum possible plasma concentration of the drug (Cmax) occurring a given time (Tmax)
By what mechanisms do various extended release drug formulas function?
Multiple layer tablets (zolpidem); Microparticle pH-dependent release (carvedilol); Dual polymer matrix (oxycontin); Osmotic extended release system (concerta)
What compound is responsible for the release of IκB-α inhibitor protein from NF-κB, and what two effects can NF-κB have in the nucleus?
IKK phosphorylates IκB-α inhibitor protein which is subsequently degraded; free NF-κB moves to the nucleus where it either binds with HAT to perform histone acetylation, or functions alone to increase transcription of κB elements
Where is TNF-α secreted and what are its effects?
Secreted by monocytes, macrophages; can kill tumor cells, induce production of pro-inflammatory cytokines, stimulate WBC adhesion/activation, cause fever, stimulate bone reabsorption/cartilage degradation
What are the effects of the leukotrienes LTC4, D4, E4, and LTB4?
LTC4, D4, E4: bronchoconstriction, increased vascular permeability; LTB4: chemoattraction, adhesion/activation of leukocytes, pain sensitization
What is the function of PGE1, PGE2, PGF2a, PGI2 (prostacyclin), TXA2?
PGE1: maintenance of patent ductus arteriosus; PGE2: vasodilation, gastric cytoprotection, pain sensitization, fever, uterine contraction/initiation of labor; PGF2α: vasoconstriction, uveoscleral drainage, contraction of uterine/GI/bronchial smooth muscle; PGI2 (prostacyclin): vasodilation, gastric cytoprotection, pain sensitization, fever, uterine relaxation (!), prevents platelet aggregation, limits action of TXA2; TXA2: stimulation of platelet aggregation
What characterizes eicosanoids?
Short half-life, released at site of action
What is the difference between the COX-1 and COX-2 isoforms?
COX-1: constitutively expressed, “housekeeping” functions, responsible for acute prostaglandin production following inflammatory stimuli, gastric cytoprotection, platelet aggregation; COX-2: inducible by inflammatory stimuli, responsible for sustained acute prostaglandin production following inflammatory stimuli
By what mechanism does synthesis of eicosanoids (prostaglandins/leukotrienes) occur?
Phospholipids are metabolized to arachidonic acid by PLA2 (generates PAF as a side-product) --> Arachidonic acid is then metabolized by either cyclooxygenase (COX-1, COX-2) to prostaglandins OR by lipoxygenase to leukotrienes
What is NF-κB and what is it involved in?
A family of transcription factors involved in inflammation, immunity, and many other processes
What stimuli cause NF-κB activation?
Antigen binding to T cell receptors; Pro-inflammatory cytokines (TNF-α, IL-1); Oxidative stress; Co-stimulatory activation signals; Bacterial LPS
What form of NF-κB is normally found in the cell and how is this maintained?
Inactive form is found in the cytoplasm; it is bound to IκB-α inhibitor protein; once the inhibitor protein releases NF-κB, it becomes activated and moves into the nucleus
What is the general therapeutic use of glucocorticoids and what are some examples of commonly used glucocorticoid drugs?
Used as immunosuppressants and anti-inflammatory drugs – examples include dexamethasone, methylprednisolone, prednisone, prednisolone; most glucocorticoids end on –sone or –lone
How can glucocorticoid use lead to an adrenal crisis and what are the symptoms associated with this?
Abrupt discontinuation of glucocorticoids or a stressful event can cause a deficiency in plasma glucocorticoid – this can lead to weakness, fainting, hypotension, cardiovascular collapse, and death
By what mechanism do high-dose, long-term adverse effects of glucocorticoid use occur?
Suppression of the hypothalamic-pituitary axis (HPA) which causes inhibition of corticotropin-releasing factor (CRF) in the hypothalamus, and inhibition of ACTH in the pituitary
What are the short-term adverse effects of PO or IV glucocorticoid use?
CNS stimulation, appetite stimulation, nausea/vomiting, slightly increased risk of infection
What are the chronic hematological effects of glucocorticoids?
Decrease in primary antibody response; decrease in production of established antibody
What are the acute hematological effects of glucocorticoids?
Increase in circulating neutrophils; decrease in circulating lymphocytes/monocytes/basophils/ eosinophils (they become sequestered in lymphoid tissues)
What are the molecular effects generated by glucocorticoids?
After passing through the cell membrane and binding to their cytosolic receptors, they inhibit histone acetylation/increase histone deacetylation (in order to decrease transcription of inflammatory genes), and bind to DNA promotors (in order to cause transcription of anti-inflammatory genes, or cause repression of certain genes leading to adverse side-effects)
What are the two receptors for TNF-α and how do they differ in cell-surface vs. soluble forms?
p55/p75; cell surface p55/p75 initiates inflammatory responses; soluble p55 initiates inflammatory responses; soluble p75 antagonizes inflammatory responses – overall effect is initiation of inflammatory response
What are the two most important biological oxidants that mediate the inflammatory response?
Superoxide (made by NADPH oxidase/detoxified by SOD) and nitric oxide (made by NOS/detoxified by oxyHb) – in inflammatory states NAPDH oxidase and iNOS are increased
What is special about the IL-1 surface receptors?
They have an endogenous receptor antagonist (IL-1ra) that competes for binding to IL-1 receptors, thus balancing the inflammatory response
What pathological problems are associated with high-dose, long-term use of glucocorticoids?
Increased risk of infection; masked signs of infection; osteoporosis; decreased bone growth in children; hypokalemia, sodium and fluid retention, hypertension (all due to mineralocorticoid activity); hyperglycermia, diabetes (since glucocorticoids decrease glucose utilization and increase gluconeogenesis); impaired wound healing; wasting of skeletal muscles; glaucoma, cataracts; redistribution of fat to face and back; dermatological changes (acne, skin atrophy, striae, growth of fine hair)
Glucocorticoids interact with what P450 isozyme?
They are inducers of CYP3A4
What molecule causes vasodilation in response to H1 receptor mediated activation?
Endothelial derived relaxing factor: NO
Where are H4 receptors located and what are they associated with?
Found in the GI tract and the CNS; they are associated with eosinophils and neutrophils causing shape changes, chemotaxis, and upregulation of adhesion molecules
What is the H3 receptor mediated response?
A decrease in Ca++ influx into the neuronal cells, modulating neurotransmitter release (the H3 receptor is presynaptic)
What changes are seen in response to activation of the H2 receptor and what are the associated physiological effects?
Gastric acid secretion: increased gastric acidity/irritation; Vasodilation of vascular smooth muscle: decreased blood pressure leading to increased rate and contractility of the heart (also due, in a minor part, to direct effects on the heart)
How does activation of the H1 receptor cause pruritus/cough reflex, nasal/bronchiole mucus, and wakefulness, respectively?
Pruritus/cough: activates selected sensory nerves; Nasal/bronchiole mucus: increased glandular secretions; Wakefulness: modulation of CNS functions
What effect does activation the H1 receptor have on post-capillary venules?
Contraction of the endothelium, creating gaps between cells which leads to edema
What are the different effects due to the activation of the H1 receptor regarding vascular endothelium and non-vascular smooth muscle?
Vascular endothelium: indirectly causes vasodilation; Non-vascular smooth muscle: directly causes vasoconstriction
What is meant by the statement that histamine is an autocoid?
It is a substance produced as a local hormone within the tissue in which it acts
What do histamine receptors have in common and how do they differ?
The four subtypes are all derived from the G protein-coupled superfamily, so they all function via intracellular signal transduction pathways; however, the end-result of receptor activation differs for each subtype
What are factors that trigger the release of histamine?
Noxious stimuli: cold/heat, bacterial toxins/insect bites, cellular injury; Pathophysiological reactions to an allergen; Therapeutic agents: morphine-like narcotics, curare-like neuromuscular blocking agents
What side-effects are associated with H1 receptor antagonist’s interaction with muscarinic and monoaminergic receptors, respectively?
Muscarinic: sedation (sometimes CNS excitation in children), dry mouth, blurred vision, tachycardia, urinary retention, suppression of Parkinsonian symptoms, reduction of nausea/vomiting/motion sickness/vestibular disturbances; Monoaminergic: anti-anxiety
Which Cimetidine drug interactions are particularly worrisome?
Interactions with Warfarin (small therapeutic index), Phenytoin (zero-order kinetics)
What drug interactions are associated with H2 receptor antagonists?
Ranitidine inhibits the glucuronidation of acetaminophen; Drugs that require an acidic environment for absorption (ketoconazole, itraconazole) will be less absorbed; Cimetidine inhibits CYP1A2/CYP2C19 as well as CYP2D6/CYP3A4 (weakly), altering their metabolism
What is the most important therapeutic action of H2 receptor antagonists and what are some examples of them?
They inhibit the secretion of gastric acid stimulated by histamine, gastrin, cholinometics, and vagal stimulation; Examples include Cimetidine (Tagamet), Ranitidine (Zantac), Famotidine (Pepcid), Nizatidine (Axid)
How does the duration of action differ when comparing first and second generation H1 receptor antagonists?
Second generation H1 receptor antagonists have a much longer duration of action (~24 hours vs. 4-8 hours for first generation drugs)
What are examples of second generation H1 receptor antagonist drugs and what symptoms can they treat?
Cetirizine (Zyrtec); Fexofenadine (Allegra); Loratadine (Claritin): appetite stimulant; Desloratadine (Clarinex): is the active metabolite of Loratadine
What are examples of first generation H1 receptor antagonist drugs and what symptoms can they treat?
Dimenhydrinate (Dramamine): motion sickness; Diphenhydramine (Benadryl): motion sickness, insomnia; Hydroxyzine (Vistaril, Atarax): anxiety, psychosis-induced tension; Meclizine (Antivert): vertigo, motion sickness; Promethazine (Phenergan): nausea, vomiting
\What are examples of drugs that inhibit release of mast cell granules, and what are some functional antagonists of the products released from mast cells?
Release inhibitors: cromolyn sodium, nedocromil; Functional antagonists: isoproterenol, theophylline, epinephrine
What is the difference between first generation and second generation H1 receptor antagonists?
First generation: not very selective for histamine receptor, enter CNS; Second generation: more selective for histamine receptor (meaning fewer side-effects associated with sedation/anti-emetic/anti-cholinergic), less lipophilic (do not enter CNS as readily)
What happens initially at the onset of inflammation to the vasculature?
Vasoconstriction of arterioles (seconds) followed by vasodilation of arterioles and subsequently the capillaries (due to histamine and NO)
How do selectins bind their ligands and what is an example of a disease in which this binding is deficient?
They bind using low affinity calcium-dependent binding; Leukocyte Adhesion Deficiency-2 (LAD-2) is an example of deficient selectin binding in humans
What are the three selectins used in adhesion and where is each found?
E-selectin (aka CD62E, ELAM-1): found on activated endothelial cells; P-selectin (aka CD62P, GMP140, PADGEM): found in platelet alpha granules and endothelial Weibel-Palade bodies;
L-selectin (aka CD62L, LAM-1): found on leukocytes
How is margination defined?
Movement from the central lumen to the luminal periphery
What substances are responsible for angiogenesis seen in inflammation?
VEGF, histamine, substance P
How do leukocytes mediate endothelial injury?
Via toxic oxygen species and proteolytic enzymes during margination and activation
Which substances are responsible for the endothelial gaps in venules (as part of the immediate transient response)?
Histamine, bradykinin, leukotrienes, substance P
What are some examples of agents that cause inflammation?
Microbial infections, physical agents, chemicals, necrotic tissue, immunological reactions
What is the difference between exudate and transudate?
Exudate (e.g. pus): high protein, specific gravity > 1.02, cellular debris; Transudate: low protein, specific gravity < 1.02, ultrafiltrate of blood
What are the cardinal signs of inflammation?
Heat (calor), redness (rubor), edema (tumor), pain (dolor), loss of functio (function laesa)
What is meant by diapedesis and what is responsible for this process?
Migration of leukocytes through the endothelium (also known as transmigration); stimulated by chemokines (e.g. PECAM-1, CD31) in venules – this process is aided by collegenases which help cells move through the basement membrane
What molecules are involved in the process of opsonization?
IgG Fc:FcR (activates complement cascade); C3b:CR1-3 (or via alternative pathway); Collectins:C1q
What responses are seen due to leukocyte activation and what signals are responsible for this activation
Production of arachidonic acid metabolites; degranulation and secretion of lysosomal enzymes; activation of oxidative burst; secretion of cytokines; modulation of leukocyte adhesion molecules – these functional responses are due to increased cytosolic calcium, and activation of protein kinase C & phospholipase A2
By what mechanism does chemotaxis occur?
Increased cytosolic calcium and activation of GTPases/kinases causes polymerization of actin within the cell --> actin and myosin generate contractions in pseudopodia in response to actin-regulating proteins (filamin, gelsolin, profilin, calmodulin) --> these contractions pull the cell closer to the source of the chemotactic agent
What chemical agents are responsible for chemotaxis?
Exogenous causes: bacterial products; Endogenous causes: C5a, Leukotriene B4, IL-8
What is the difference between LAD type 1 and type 2?
Type 1: defect in biosynthesis of β2 chains; Type 2: absence of Sialyl LewisX (ligand for E-selectin) – both deficiencies present with recurrent bacterial infections
Which molecules are the first to arrive in normal transmigration, and how does this differ in viral infections and hypersensitivity reactions, respectively?
Normally: neutrophils (6-24 hours); Viral infections: lymphocytes; hypersensitivity reactions: eosinophils – also, in pseudomonas infection neutrophils are delayed (2-4 days)
What is the purpose of integrins, what are the two classes of integrins, and what is the function of each class?
Promote cell-cell or cell-matrix interactions; Beta1-containing integrins (e.g. CD49a, CD29, VLA) are found on leukocytes for attachment to the endothelium; β2-containing integrins (e.g. CD11a-c, CD18, LFA-1) are responsible for leukocyte adhesion to other cells
What compounds are responsible for the redistribution and induction of adhesion molecules, respectively?
Redistribution: histamine, thrombin, PAF; Induction: IL-1, TNF
What are mucin-like glycoproteins composed of, what is their ligand, and where are these adhesion molecules found?
Composed of heparan sulfate; bind to leukocyte adhesion molecule CD44; found in the extracellular matrix and on cell surfaces
What is meant by diapedesis and what is responsible for this process?
Migration of leukocytes through the endothelium (also known as transmigration); stimulated by chemokines (e.g. PECAM-1, CD31) in venules – this process is aided by collegenases which help cells move through the basement membrane
What molecules are involved in the process of opsonization?
IgG Fc:FcR (activates complement cascade); C3b:CR1-3 (or via alternative pathway); Collectins:C1q
What responses are seen due to leukocyte activation and what signals are responsible for this activation
Production of arachidonic acid metabolites; degranulation and secretion of lysosomal enzymes; activation of oxidative burst; secretion of cytokines; modulation of leukocyte adhesion molecules – these functional responses are due to increased cytosolic calcium, and activation of protein kinase C & phospholipase A2
By what mechanism does chemotaxis occur?
Increased cytosolic calcium and activation of GTPases/kinases causes polymerization of actin within the cell --> actin and myosin generate contractions in pseudopodia in response to actin-regulating proteins (filamin, gelsolin, profilin, calmodulin) --> these contractions pull the cell closer to the source of the chemotactic agent
What chemical agents are responsible for chemotaxis?
Exogenous causes: bacterial products; Endogenous causes: C5a, Leukotriene B4, IL-8
What is the difference between LAD type 1 and type 2?
Type 1: defect in biosynthesis of β2 chains; Type 2: absence of Sialyl LewisX (ligand for E-selectin) – both deficiencies present with recurrent bacterial infections
Which molecules are the first to arrive in normal transmigration, and how does this differ in viral infections and hypersensitivity reactions, respectively?
Normally: neutrophils (6-24 hours); Viral infections: lymphocytes; hypersensitivity reactions: eosinophils – also, in pseudomonas infection neutrophils are delayed (2-4 days)
What is the purpose of integrins, what are the two classes of integrins, and what is the function of each class?
Promote cell-cell or cell-matrix interactions; Beta1-containing integrins (e.g. CD49a, CD29, VLA) are found on leukocytes for attachment to the endothelium; β2-containing integrins (e.g. CD11a-c, CD18, LFA-1) are responsible for leukocyte adhesion to other cells
What compounds are responsible for the redistribution and induction of adhesion molecules, respectively?
Redistribution: histamine, thrombin, PAF; Induction: IL-1, TNF
What are mucin-like glycoproteins composed of, what is their ligand, and where are these adhesion molecules found?
Composed of heparan sulfate; bind to leukocyte adhesion molecule CD44; found in the extracellular matrix and on cell surfaces
What is defective in chronic granulomatous disease?
Microbicidal activity
What are the two pathways of arachidonic acid metabolites and what are some of the metabolites that become activated as an end-result?
Cyclooxygenase: TXA2 (platelet aggregation/vasoconstriction), PGI2 (vasodilation/aggregation inhibition), PGD2 (vasodilation/edema potentiation), PGE2 (pain/fever); Lipoxygenase pathway: LTB4 (chemotaxis/aggregation of neutrophils), LTC4/LTD4/LTE4 (vasoconstriction/ bronchospasm/increased permeability), LXA4 (vasodilation) – TX means thromboxane, PG means prostaglandin, LT means leukotriene, LX means lipoxin
What effects does C5a have?
It causes release of inflammatory mediators via the lipoxygenase pathway of arachidonic acid metabolism, and causes increased adhesion/chemotaxis of leukocytes
What two plasma proteases function as chemical mediators and what are their characteristics?
Hageman factor: activated by collagen, basement membrane, or activated platelets (requires the co-factor HMWK); Kinin-kallikrein system: makes bradykinin which functions like histamine and causes pain; Thrombin: enhances leukocyte adhesion, generates fibrinopeptides which increase vascular permeability and is chemotactic for leukocytes, converts fibrinogen to fibrin clot
Besides transmitting pain signals, what other effects does substance P have?
Regulation of vessel tone and modulation of vascular permeability
Where is serotonin found and what are its effects?
It is found in platelet dense bodies and has effects similar to that of histamine – it is released in response to platelet aggregation
Where is histamine found and what is its release stimulated by?
Found in mast cell granules, basophils, platelets; Release stimulated by physical injury, immune reactions, anaphylatoxins, substance P, IL-1, IL-8 – histamine causes arteriolar dilation and increased vascular permeability
What effects do major basic protein and defensins have on pathogens, respectively?
Major basic protein: secreted from eosinophils for parasite toxicity; Defensins: form holes in microbial membrane
What pathology is seen in both Chediak-Higashi syndrome and Diabetes Mellitus?
Defective phagocytosis
What is neutropenia commonly caused by?
Chemotherapy in cancer treatment
What side-effects are associated with H1 receptor antagonist’s interaction with muscarinic and monoaminergic receptors, respectively?
Muscarinic: sedation (sometimes CNS excitation in children), dry mouth, blurred vision, tachycardia, urinary retention, suppression of Parkinsonian symptoms, reduction of nausea/vomiting/motion sickness/vestibular disturbances; Monoaminergic: anti-anxiety
Which Cimetidine drug interactions are particularly worrisome?
Interactions with Warfarin (small therapeutic index), Phenytoin (zero-order kinetics)
What drug interactions are associated with H2 receptor antagonists?
Ranitidine inhibits the glucuronidation of acetaminophen; Drugs that require an acidic environment for absorption (ketoconazole, itraconazole) will be less absorbed; Cimetidine inhibits CYP1A2/CYP2C19 as well as CYP2D6/CYP3A4 (weakly), altering their metabolism
What is the most important therapeutic action of H2 receptor antagonists and what are some examples of them?
They inhibit the secretion of gastric acid stimulated by histamine, gastrin, cholinometics, and vagal stimulation; Examples include Cimetidine (Tagamet), Ranitidine (Zantac), Famotidine (Pepcid), Nizatidine (Axid)
How does the duration of action differ when comparing first and second generation H1 receptor antagonists?
Second generation H1 receptor antagonists have a much longer duration of action (~24 hours vs. 4-8 hours for first generation drugs)
What are examples of second generation H1 receptor antagonist drugs and what symptoms can they treat?
Cetirizine (Zyrtec); Fexofenadine (Allegra); Loratadine (Claritin): appetite stimulant; Desloratadine (Clarinex): is the active metabolite of Loratadine
What are examples of first generation H1 receptor antagonist drugs and what symptoms can they treat?
Dimenhydrinate (Dramamine): motion sickness; Diphenhydramine (Benadryl): motion sickness, insomnia; Hydroxyzine (Vistaril, Atarax): anxiety, psychosis-induced tension; Meclizine (Antivert): vertigo, motion sickness; Promethazine (Phenergan): nausea, vomiting
\What are examples of drugs that inhibit release of mast cell granules, and what are some functional antagonists of the products released from mast cells?
Release inhibitors: cromolyn sodium, nedocromil; Functional antagonists: isoproterenol, theophylline, epinephrine
What is the difference between first generation and second generation H1 receptor antagonists?
First generation: not very selective for histamine receptor, enter CNS; Second generation: more selective for histamine receptor (meaning fewer side-effects associated with sedation/anti-emetic/anti-cholinergic), less lipophilic (do not enter CNS as readily)
What does release of platelet-activating factor result in?
Platelet aggregation/degranulation, bronchoconstriction; vasodilation/increased permeability; leukocyte adhesion/chemotaxis/degranulation/oxidative burst; stimulates synthesis of eicosanoids
What are some common causes of chronic inflammation?
Viral infections, persistent microbial infections, prolonged exposure to toxic agents (e.g. silica, lipids), autoimmune diseases (e.g. rheumatoid arthritis, DM-1)
Why are viral infections capable of causing chronic inflammation?
They require lymphocytes and monocytes to resolve the infection
How is tissue repair accomplished in chronic inflammation?
Via angiogenesis and fibrosis
What is a key feature of acute inflammation progressing to chronic inflammation?
Neutrophils are replaced by macrophages, lymphocytes, and plasma cells
How does the outcome of acute inflammation differ from that of chronic inflammation?
Acute inflammation causes limited injury with minimal tissue damage, damaged tissue is capable of regeneration, and histologic/functional normalcy can be restored – scarring or fibrosis can occur with acute inflammation
Which cytokines regulate lymphocyte function, innate immunity, and macrophage activation, respectively?
Lymphocyte function: IL-2 (stimulates), TGF-β (inhibits); Innate immunity: TNF, IL-1; Macrophage activation: IFN-γ, IL-12
What functions do the lipoxins have?
Endogenous regulators of leukotrienes; inhibit neutrophil chemotaxis/adhesion; stimulate monocytes adhesion
What substance specifically inhibits phospholipase A2 (PLA2)?
Glucocorticoids
How do aspirin and NSAIDS reduce inflammation?
They inhibit COX-1 and COX-2 (although the lipoxygenase pathway is not inhibited); overuse can predispose to gastric ulceration
What types of activation signals do macrophages respond to, and which substances induce the formation of giant cells?
Normal activation signals: IFN-γ, endotoxin, acute inflammatory mediators, fibronectin; Giant cells: IL-4, IFN-γ
How is the protective index calculated?
PI = ED50 for toxicity/ED50 for therapeutic effect – the greater the number, the safer the drug
What effects can a substance that causes potentiation of a drug have?
It can either shift the dose-response curve to the left, or increase the maximum response of the drug
What do efficacy and intrinsic activity mean and what determines them?
The maximum response that a given drug can produce – depends on the chemical structure of a drug
What determines the potency of a drug?
The log dose of a drug at its ED50 – this only works when comparing two drugs which produce similar effects
How does EC50 differ from ED50?
EC50 is only applied to in vitro settings where the concentration of a drug is fixed
What is meant by biological variability?
No two subjects have the same dose-response curve, meaning that the value of the ED50 is characteristic for each subject
What is the difference between a quantal dose-response curve and a graded dose-response curve?
In a quantal dose-response curve, every subject is given one dose to see whether or not there is an effect (“all-or-none” response); in a graded dose-response curve every subject is given a number of doses to see what the graded response is with each increasing dose – the quantal dose-response curve gives MEDIAN ED50, whereas the graded response curve gives MEAN ED50
What is meant by the ED50?
The median effective dose that produces a desirable effect in 50% of the population
What determines the safety of a certain drug and what measure is used to describe this?
The difference between the ED50 and LD50 – this calculated by the therapeutic index (LD50/ED50); the greater the therapeutic index, the safer the drug (TI >10 is desirable)
How does LD50 differ from ED50?
The LD50 is the dose for a particular drug that is lethal in 50% of the population
What does tachyphylaxis refer to, what does it depend on, and by what mechanisms can it occur?
A rapidly-occurring decrease in the response to a drug after repetitive administration – depends on the dose and interval of administration; can occur because a required intermediate is “used up”, receptors are internalized, or signal transduction becomes uncoupled
How do the values of α and e determine, respectively, whether a drug is an agonist or antagonist?
If α=1 the drug is a full agonist; if 0<α<1 the drug is a partial agonist; if α=0 the drug is an antagonist – if e>0 the drug is an agonist (there is no limit for e); if e=0 the drug is an antagonist
What parameters determine the ED50?
Intrinsic activity (α) or efficacy (e); and the dissociation constant (KA where [KA=k2/k1])
What concept determines the difference between intrinsic activity and efficacy?
The term intrinsic activity is used when the maximum response occurs when all receptors are occupied; Efficacy is used when the maximum response can occur when there are still spare receptors which are not occupied
What is the difference between an agonist and an antagonist?
Agonists resemble an endogenous substance which binds to a receptor and initiates a response (i.e. stimulation of the receptor); Antagonists also resemble an endogenous substance and bind to a receptor, but they do not trigger a response (rather, they block receptor responses to agonists)
What are the two types of interactions that a drug can have with a receptor?
Binding may alter the activity of a channel or an enzyme; Binding may initiate a biological response
What is the distinction between tolerance and tachyphylaxis?
Tolerance requires a physiological rearrangement in the body that occurs over a long period of time (i.e. days)
What is the concept called that states that the sum of two particular drugs is greater than their individual effect?
Synergism
In Type III hypersensitivity reactions, what is the immune reactant, the antigen, and the effector mechanism?
Immune reactant: IgG; Antigen: soluble antigen; Effector: complement, phagocytes
What are the spasmogens released from mast cells and what is their effect?
Histamine, PGD2, LTC4, LTD4 --> all cause bronchial smooth muscle contraction, mucosal edema, and mucus secretion
What happens to the armed mast cells on second exposure to bee venom?
They release spasmogens, activators, and chemoattractants – histamine peaks first, then leukotrienes appear (longer acting)
What happens to the IgE secreted by a plasma cell in response to bee venom?
The IgE binds to FcεR1 on a mast cell; this binding extends the half-life of the IgE from 2-3 days to >30 days (it inhibits signaling pathways that lead to apoptosis
What is the mechanism by which bee venom activates the immune system?
After entering the body, the venom is taken up by an APC and the peptide sequence presented --> a CD4 T Cell interacts with the APC and secretes IL-4 --> IL-4 activates a TH2 cell in combination with its interaction with the APC --> the TH2 cell secretes more IL-4 and activates a B cell --> the B cell differentiates into a plasma cell which secretes IgE
8.
What are some examples of Type I, II, III, and IV hypersensitivity reactions?
Type I: allergic rhinitis, asthma, systemic anaphylaxis; Type II: drug allergies, chronic urticaria; Type III: serum sickness, arthus reaction; Type IV: contact dermatitis, chronic asthma, chronic allergic rhinitis, contact dermatitis
In Type IV hypersensitivity reactions, what is the immune reactant, the antigen, and the effector mechanism?
Immune reactant #1: TH1; Antigen #1: soluble antigen; Effector #1: macrophage activation; Immune reactant #2: TH2; Antigen #2: soluble antigen; Effector #1: eosinophils activation; Immune reactant #3: CTL; Antigen #3: cell-associated antigen; Effector #3: cytotoxicity
What are the 4 types of hypersensitivity reactions according to Gell and Coombs?
Type I: IgE-mediated (2-30 min); Type II: Antibody-mediated (5-8 hours); Type III: Immune-complex (2-8 hours); Type IV: cell-mediated (24-72 hours)
In Type II hypersensitivity reactions, what is the immune reactant, the antigen, and the effector mechanism?
Immune reactant: IgG; Antigen #1: cell/matrix associated antigen; Effector #1: complement, FcR+ cells; Antigen #2: cell-surface receptor; Effector #2: antibody alerts signaling
In Type I hypersensitivity reactions, what is the immune reactant, the antigen, and the effector mechanism?
Immune reactant: IgE; Antigen: soluble antigen; Effector: mast-cell activation
What enzymes and toxic proteins are released from eosinophils and what are their effects?
Eosinophil peroxidase: toxic via catalyzing halogenation, triggers histamine release from mast cells; Eosinophil collagenase: remodels connective tissue matrix; Matrix metalloproteinase-9: matrix protein degradation; Major basic protein: toxic to parasites, triggers histamine release; Eosinophil cationic protein: toxic to parasites, neurotoxin
How does immune complex formation occur in Type III hypersensitivity reactions?
A constant presence of antigen leads to a massive production of antibody --> with enough antibody around, a soluble immune complex will form to which complement (C1qrs) can bind --> this activates the classical pathway of complement activation --> production of anaphylatoxins (C3a, C4a, C5a) leads to inflammation and tissue destruction
How can hemolytic disease of the newborn be prevented clinically?
The mother receives anti-Rh antibodies immediately after childbirth has occurred --> the anti-Rh antibodies bind to the FcγRIIB-1 receptor on naïve B cells --> binding activates immunoreceptor tyrosine-based inhibitor motifs (ITIMs) which prevent the formation of Rh antibodies; Anti-Rh antibody must be given with each pregnancy because memory B cells do not respond to it
In what scenario do problems with Rh factor cause hemolytic disease of the newborn?
If the mother is Rh- and the fetus Rh+, the fetus will expose the mother to Rh antibody, usually at birth; the mother will produce IgG antibodies in response, which, in subsequent pregnancies with an Rh+ fetus, cross the placenta and cause destruction of fetal RBCs (not via the complement system, but via macrophage phagocytosis)
By what pathway does a blood transfusion hypersensitivity reaction occur?
IgM antibody in the patient reacts with antigen on the transfused blood cells --> classical complement pathway is activated --> lysis of donor cells and agglutination; the patient will also produce large amounts of IgG in response to the blood antigen --> this leads to phagocytosis of blood cells by the liver and spleen --> circulatory shock and kidney necrosis
What type of hypersensitivity reactions are blood transfusions categorized as?
Type II – IgG
What route of action is needed to cause systemic anaphylaxis?
Intravenous (either directly or following massive absorption) – can be caused by drugs, serum, venom, peanuts
What are the activators released from mast cells and what is their effect?
Histamine: vasodilation, vascular permeability; Platelet activating factor (PAF): microthrombi; Tryptase: activates C3; Kininogenase: activates kinins --> vasodilation, edema
What aspect of anaphylaxis can be fatal?
Hypovolemic shock – due to vasodilation and edema
What are the chemoattractants released from mast cells and what is their effect?
IL-3, IL-5, CXCL8, CCL3, TNF-α --> attract neutrophils, eosinophils, basophils; LTB4, PAF --> attract basophils
How can Type III hypersensitivity reactions cause joint, kidney, and vascular disease?
Deposition of immune complexes in these areas activates the complement system which leads to localized tissue destruction
By what mechanism do Type IV hypersensitivity reactions occur?
A T-helper cell interacts with an APCs --> activation of the T-helper cell causes it to secrete cytokines (IL-3, TNF-α), chemokines (RANTES), and cytotoxins (TNF-β) --> they cause vasodilation leading to leaky vessels and swelling
What syndromes can result from Type IV hypersensitivity reactions and what antigens are associated with each?
Delayed-type hypersensitivity: due to proteins (insect venom, mycobacterial proteins); Contact hypersensitivity: due to haptens (poison ivy) or small metal ions (nickel, chromate); Gluten-sensitive enteropathy (Celiac disease): due to gliadin