Shared Flashcard Set

Details

SPM--Unit 1 Flaschards
N/A
614
Medical
Graduate
09/03/2010

Additional Medical Flashcards

 


 

Cards

Term
What is primary disease prevention?
Definition
removing risk factors b/4 there is disease (ex. "wear sunscreen")
Term
What is secondary disease prevention
Definition
early detection & treatment of asymptomatic disease (ex., remove & biopsy cancerous mole)
Term
What is tertiary disease prevention?
Definition
preventing a symptomatic disease from getting worse; normal disease treatment
Term
Normal respiratory rate
Definition
12-20 breaths/min
Term
normal heart rate
Definition
60-100 BPM
Term
Normal (healthy) blood pressure is less than: 
Definition
less than 120/80
Term
What is an underweight BMI?
Definition
<18.5
Term
What is a healthy BMI?
Definition
18.5-24.9
Term
What is an overweight BMI
Definition
25.0-29.9
Term
What is an stage 1 obese BMI?
Definition
30.0-39.9
Term
What is a stage 2 obese BMI?
Definition
>=40
Term
What does the mnemonic CUT the PY mean?
Definition
the 3 pyramidines are cytosine, uracil, & thymine
Term
What does the mnemonic PURe As Gold mean?
Definition
The 2 purines are arginine & guanine
Term
How do A, C, G, U, & T pair?
Definition

A pairs with T in DNA

A pairs with U in RNA

G pairs with C

 

 

Term
nucleotides are bound to one another by ________ bonds
Definition
phosphodiester
Term
What holds the 2 strands of DNA together?
Definition
hydrogen bonds between the base pairs
Term
Describe the 5 levels of DNA packaging
Definition

Level 1, the DNA double helix.

 

Level 2, DNA is wrapped around histone proteins, like “beads on a string”.

 

Level 3, DNA & histones are wrapped into a solenoid to form a chromatic fiber.

 

Level 4, solenoid loops attach to a non-histone scaffold.

 

Level 5, the scaffold is wrapped into a supercoil

Term
What is the start codon on mRNA in eukaryotes & what does it code for?
Definition

AUG codes for methionine

 

(on the DNA sense strand, it's ATG; on the anti-sense/template strand, it's TAC) 

Term
What are the 3 stop codons on mRNA?
Definition
UGA, UAG, UAA
Term
promoter sequence
Definition
Sequence upstream (toward the 5' end of the sense strand) of a gene that initiates & regulates transcription of the gene.
Term
splicing sequence
Definition
A sequence at the boundaries between an intron & an exon that says “splice here”; usually start with GU and end with AG.
Term

 

5' untranslated region

 

 

Definition

A DNA segment from the first transcribed base pair to the start codon; contains sequences that regulate mRNA’s stability or translation.

 

(the 5'untranslated region IS transcribed, just not translated.  Unlike the promoter, the 5'untranslated region is always contiguous with the translated region)

Term
3' untranslated region
Definition
A segment of DNA that begins at the stop codon and ends at the last transcribed base pair. Contains binding sites for factors that may affect mRNA’s stability. Also contains polyadenylation signal.
Term
Creating RNA from DNA is called
Definition
transcription
Term
Creating protein from RNA is called
Definition
translation
Term
An amino acid consists of a central carbon that is attached to what 4 things? 
Definition

a hydrogen (H)

an amino group (--NH3+)

a carboxyl group (--COOH, or --COO-)

and a side chain (varies)

Term
What are the 4 basic tissues of the body?
Definition
epithelia
connective tissue
muscle
nerve
Term
tight junctions (zonula occludens)
Definition
Junctions at the border of the apical (top) & basolateral (side & bottom) portions of epithelial cells. Membranes are fused by shared fibers, which forms cells into epithelial sheets.
Term
zonula adherens
Definition
A continuous belt of cadherin proteins just deep to tight junctions in epithelial cells. Very robust (like rings holding Coke cans in a 6 pack). Inside the cell, the cadherins are associated with intracellular proteins
Term
The zonula adherins are made of a continuous ring of _______ proteins.
Definition
cadherin
Term
spot desomosomes (maculae adherens)
Definition
"Spot welds" of cadherins between epithelial cells.
Term
Gap junctions between epithelial cells are made of many clustered ____________
Definition
hemichannels (aka connexons)
Term
What is a connexon?  What is it aka? 
Definition

A connexon is a a hexagonal arrangement of 6 connexin protein subunits. Many hemichannels make up a gap junction.

 

A connexon is aka a hemichannel

Term
A junction that sticks the basal domain of an epithelial cell to the basement membrane
Definition
hemidesmosome
Term
What kind of muscle cells look like large, elongated, multinucleated, striated fibers?
Definition
skeletal muscle
Term
What kind of muscle cells look like long, thin, grouped cells that seem to fuse into one another with little open space visible and no striations?
Definition
smooth muscle cells
Term
What kind of muscle cells look like long, thin irregular branched cells bound longitudinally by intercalated disks?
Definition
cardiac muscle
Term
Does a Scanning Electron Microscope or Transmission Electron Microscope allow you to look at 3D objects (such as molecules)?
Definition

Scanning Electron Microscope

 

(Scanning Electron Microscope scans the surface with a beam of electrons.  Transmission Electron Microscope transmits a thin beam of electrons through a very thin object) 

Term

Emits a beam of electrons through a very thin specimen slice, interacting with the specimen atoms as it passes through.

 

 

Scans a 3D surface with a beam of electrons. Allows you to look at 3D objects (such as molecules). 

 

Definition

 

Transmission Electron Microscope

 

 

Scanning Electron Microscope 

Term
Does the Scanning Electron Microscope (SEM) or Transmission Electron Microscope (TEM) have better (smaller) minimum resolution?
Definition
Transmission Electron Microscope (TEM) has better resolution than Scanning Electron Microscope (SEM)
Term
An extracellular layer that always lies at the interface of epithelial cells & connective tissues
Definition
basal lamina (basement membrane)
Term
A freeze fracture may break a lipid bilayer membrane into 2 layers. Describe the outer layer.
Definition

The E-face is the outer leaflet of the membrane (you’re looking from the inside of the cell out). The E-face has significantly fewer proteins than the P-face.

 

Remember:  E is for Exterior. E is for Empty of proteins.

 

(P is for Private layer.  P is for protein-filled)

Term
A freeze fracture may break a lipid bilayer membrane into 2 layers. Describe the inner layer.
Definition

The P-face is the inner leaflet of the membrane (you’re looking from the outside of the cell in). The P-face has significantly more proteins than the E-face.

 

Remember:  P is for Private (the inside).  P is also for Protein!

 

(as opposed to the E-face, which is the Exterior face and is relatively Empty of protein)

Term
The basal lamina is mostly made of __________ and the glycoprotein _________
Definition

collagen type IV

laminin 

Term

Do nerves cross the basal membrane?  Do blood vessels?  Do epithelial cells?

 

Definition

Nerves may cross, but blood vessels don't (so epithelial cells get receive nutrients and discard wastes by diffusion across the basement membrane).

 

Healthy epithelial cells do not cross (cancerous cells may).

Term
Mesothelium 
Definition

 

 

An epithelial membrane that forms the lining of several body cavities: the pleura (thoracic cavity), peritoneum (abdominal cavity including the mesentery) and pericardium (heart sac).

 

Term

Endothelium lines ____, _______, and ______.  What kind of cells is endothelial tissue made of?

 

 

Definition

 

blood vessels, lymph vessels, and heart chambers

 

simple squamous

 

 

Term
mitotic index
Definition

 

= (# of cells in a stage of mitosis) / (total # of cells) 

Term
For what 2 reasons are epithelial cells highly subject to cancer?
Definition

1. they have a high mitotic index (frequently divide)

 

2. they are exposed to many stresses

Term
papilloma
Definition

 

 

A slow-growing benign tumor that arises from an epithelial surface (ex., warts caused by Human Papilloma Virus, HPV, are a type of papilloma)

 

Term
Adenoma
Definition

 

bening tumor that arises from glandular epithelia

Term
carcinoma
Definition

 

a malignant neoplasm that arises from epithelial tissue

Term

adenocarcinoma

 

Definition
a malignant neoplasm that arises from glandular epithelia
Term

 

What is the apical domain of an epithelial cell?

 

 

Definition

the "top"

 

for an epithelial cell lining the lumen of a hollow organ, the apical domain faces into the lumen

 

for an epithelial cell lining the exterior of an organ, the apical domain faces out from the organ

 

epithelial cells have polarity, with apical, lateral, and basal domains

Term

 

basolateral domain of an epithelial cell 

 

 

Definition

the "bottom" & "sides" of an epithelial cell

 

for an epithelial cell lining the lumen of a hollow organ, the basolateral domain is facing away from the lumen (and toward other epithelial cells)

 

for an epithelial cell lining the exterior of an organ, the basolateral domain is facing in toward the organ (and toward other epithelial cells)

Term

What kind of epithelial cells line surfaces involved in passive diffusion of materials (e.g., air sacs of lungs, blood vessels, etc.)?

 

(that is simple, stratified, pseudostratified, squamous, cuboidal, columnar, transitional, ciliated, unciliated etc.) 

Definition
simple squamous
Term

Describe the shape and location of the nucleus of a simple cuboidal epithelial cell.  

 

 

Describe the shape and location of the nucleus of a simple columnar epithelial cell.  

 

Definition

simple cuboidal:  nucleus is usually round & located in center of cell

 

 

simple columnar:  nuclei are elongated (oval) & may be located at the base, center, or (occasionally) apex of the cell 

 

Term
What kind of epithelial cells line the small ducts and tubules that have excretory (outside the body), secretory (inside the body), or absorptive functions, (e.g. collecting tubules of kidney, small excretory ducts of salivary glands, small excretory ducts of pancreas). 
Definition
simple cuboidal epithelial cells
Term
Why are secretory cells simple cuboidal, not simple squamous epithelial cells?
Definition
They need space to hold the organelles necessary for secretion.
Term
Describe the nuclei of simple columnar epithalial cells.
Definition
Nuclei are elongated & may be located at the base, center, or (occasionally) apex of the cell
Term

What kind of epithelial cells primarily line the gut, and may have absorptive and/or secretory functions.

 

 

(that is simple, stratified, pseudostratified, squamous, cuboidal, columnar, transitional, ciliated, unciliated etc.) 

Definition
simple columnar epithelia
Term
Motile cell projections that beat in a wave-like manner, synchronized with adjacent cells.
Definition
Cilia
Term
Where are ciliated simple columnar epithelial cells found, and what do they do there?
Definition
in the fallopian tube (oviduct)
they move the egg (ovum) toward the uterus
Term
What are non-ciliated cells in ciliated epithelum called, and what is their purpose?
Definition
goblet cells
secrete mucous
Term
What is pseudostratified epitheilum?
Definition

epithelium in which differing heights of nuclei make cells seem stratified (not all cells have their apical domains at the surface), but actually all the cells sit on the basal lamina

 

 

Term
Simple columnar epithelia & pseudostratified columnar epithelia may have this organelle, but true stratified epithelia never has ________
Definition
cilia
Term
Where does pseudostratified ciliated columnar epithelia occur, and what is its purpose?
Definition

in the respiratory airways (pseudostratified ciliated columnar epithelia is aka respiratory epithelia)

 

ciliated cells move mucous (produced by goblet cells) & foreign particles trapped in the mucous up to throat.

Term

The cells of transitional epithelia may look like _______ eptithelial cells, or like _______ epithelial cells.  

 

Describe the surface of transitional epithelium.  

Definition

squamous/cuboidal

 

surface has a thick, densely-stained outline with a scalloped edge of "umbrella cells" (so called because they are round on the exterior edge & may cover 2 or more cells beneath them)

Term
Where is transitional stratified epithelum found?
Definition
found only in urinary outflow tract (that is, ureters, bladder, urethra, & gland ducts of the prostate)
Term
What is the advantage of transitional stratified epithelium being "transitional"?
Definition
ability of cells to stretch from round to flat makes transitional stratified epithelium stretchy
Term
Other than being stretchy, what 2 advantages make transitional stratified epithelium suitable to lining the urinary tract?
Definition

1. can withstand toxicity of urine (low metabolic activity in transitional epithelium makes it better able to withstand toxicity of urine)

 

2. waterproof-->can protect underlying tissues form toxicity of urine (unlike, say, cuboidal or columnar eptihelium, which is not typically waterproof)

Term

Name the parts in this single-cell reflex pathway

 

1. cell detects increased estrogen

2. estrogen/receptor complex enters cell, travels to…

3. cell nucleus

4. increased transcription & translation of proteins

5. cell grows

 

(options:  afferent pathway, effector, efferent pathway, integration center, receptor)

Definition

1. Receptor

2. Afferent pathway

3. Integration center

4. Efferent pathway

(mnemonic:  EFFERent-->EFFEctor)

5. Effector

Term
What is feed forward?
Definition
A response to an anticipated change (ex., heart rate may increase before you begin to exercise)
Term

 

gain (in terms of a feedback system)

 

 

Definition
a measure of the effectiveness of a feedback system
Term

About 2/3 of body water is ______ and 1/3 is ______

 

(location)

Definition

intracellular

 

extracellular

Term
Normal osmolarity of cellular or body fluids?
Definition
about 280-300 mOsmoles/L 
Term
If a cell membrane were suddenly made completely permeable to Na+ & K+, Na+ would flow ________ & K+ would flow __________
Definition

Na+ would flow into the cell

 

K+ would flow out of the cell

Term
A type of gradient-driven diffusion that requires transporter molecules
Definition
facilitated diffusion
Term

The Na+ K+ ATPase pump moves _________(# & ion) out of the cell and __________(# & ion) into the cell.

 

Is that active transport or facilitated diffusion?:

Definition

pumps 3 Na+ out of cell (up gradient)

pumps 2 K+ into cell (up gradient)

 

it's active transport--it requires energy from phosphorylation of ATP to make pump go

Term
What does a co-transporter pump do?
Definition
pumps 2 molecules in the same direction
Term
Is Na+ K+ ATPase a co-transporter (co-porter) or counter-transporter (anti-porter)
Definition
counter-transporter b/c it pumps Na+ (out) and K+ (in) in opposite directions
Term
How does glucose solution increase intracellular hydration faster than saline solution?
Definition

cells actively absorb glucose, and water passively follows after

 

(then the cell consumes or stores the glucose, leaving a higher concentration of water than before)

Term
What type of antibody crosses the placenta?
Definition
IgG
Term
What type of antibody is expressed in breast milk?
Definition
IgA
Term
what are the 2 generative (central, primary) lymphoid organs
Definition

bone marrow (where naive B cells arise & mature & where naive T cells arise)

 

thymus (where naive T cells mature)

Term
cytokines that work at very short range for cells near secretor cell
Definition

paracrine

 

(as opposed to autocrine cytokines, which act on the cell that synthesize it, either within the cell cytoplasm or excreted to work on surface molecules; 

 

endocrine cytokines, aka endocrine hormones, which are secreted directly into the bloodstream to work on distant cells;

 

exocrine cytokines, aka exocrine hormones, which are secreted into a duct) 

Term
cytokines that are directly secreted into the blood stream & work at long range
Definition

endocrine cytokines (aka endocrine hormones)

 

(as opposed to autocrine cytokines, which act on the cell that synthesize it, either within the cell cytoplasm or excreted to work on surface molecules; 

 

paracrine cytokines that work at very short range for cells near secretor cell;

 

and exocrine cytokines, aka exocrine hormones, which are secreted into a duct) 

Term
most numerous phagocyte in the body
Definition
neutrophils
Term
polymorphonuclear cells are aka ________
Definition
neutrophils
Term
Why are neutrophils also called polymorphonuclear cells (PMNs)
Definition
because they have a multi-lobed nucleus that looks like several nuclei
Term
Do macrophages or neutrophils give & receive more cytokines?
Definition
macrophages
Term
A phagocyte that lives in epithelia & most organs, and brings microbial antigens from the tissue to the T cells.
Definition
Dendritic Cell (DC)
Term
What causes lymph nodes to expand in size during infection?
Definition
large numbers of proliferating T & B cells
Term
Most numerous type of lymphocyte in blood
Definition
T cells
Term
An innate immune cell that wanders around the body lookin' fer somethin' tuh kill!
Definition
Natural Killer (NK) cells
Term
Natural Killer (NK) cells attack cells that do at least one of what 2 things?
Definition

1. don't express normal MHC class I surface molecules (invader cells or stressed self cells)

 

2. express a nonspecific activation signal

Term
What is the only lymphocyte that is part of the innate immune system?
Definition

Natural Killer (NK) cells

 

(note:  Neutrophils are White Blood Cells, and they are part of the innate immune system, but they are not lymphocytes)

Term
Are most of the bacteria on the skin gram negative or gram positive, and why?
Definition
gram positive b/c it's too dry for gram negative
Term

colonization

 

(microbiology)

Definition

 

establishment of a population of organisms (of one or more species) which can live and multiply

Term
What's the difference between colonizatin and infection? 
Definition

colonization is establishment of a population of organisms which can live and multiply 

 

infection occurs occurs when colonized bacteria cause a host reponse (such as inflammation) that leads to signs and symptoms of disease

 

So, the healthy gut is colonized by many beneficial flora, but is not infected. 

Term
Do prokaryotes have organelles?
Definition
Yes. Ribozomes are organelles. Prokaryotes don't have MEMBRANE-BOUND organelles.
Term
What kind of bacteria have a thick wall mostly made of peptidoglycan?
Definition
gram positive
Term
What kind of bacteria have a thin cell wall with very little of peptidoglycan between an inner cell membrane & outer membrane?
Definition
gram negative
Term
What colors are gram positive & gram negative bacteria on gram stain?
Definition

positive--purple or blue

negative--pink

 

(the bacterial world is upside down

positive bacteria are blue

negative bacteria have a rosy outlook)

Term
Do gram positive or gram negative bacteria have teichoic acids in their cell wall?
Definition

gram positive

 

 

The main function of teichoic acids is to provide rigidity to the cell-wall.  Lysis of Gram-positive bacteria and release of the teichoic acid into the bloodstream may cause fever, blood vessel dilation and possibly shock and subsequent death. Teichoic acid can also be used by bacteria to attach to mucosal membranes.

Term
What shape are bacilli?
Definition
rods
Term
a tiny, hairlike appendage found only on prokaryotes
Definition
pilus
Term
Fungus cell walls are mostly composed of
Definition

chitin

 

(A long-chain polysaccharide that also makes up the exosekeletons of arthropods) 

Term
Bacteria & fungi can both form long threads, but bacteria threads MOSTLY don't _______
Definition

branch

 

(one exception to the rule is Nocardia, which is a branching filamentous bacterium)

Term
hypha (pl. hyphae)
Definition

 

long, branching filamentous structure of fungus

Term
saprobic
Definition
eat dead things (usually by living on them); fungi are usually saprobic
Term
Describ the three forms of fungi:  yeast, filamentous, and dimorphic
Definition

yeast--a single-cell form of fungi

 

filamentous--a thread-like form of fungi

 

dimorphic--a form of fungi that can live in either yeast or filamentous forms

Term
trophozoite
Definition
a protozoan, esp. of certain parasitic species, in the feeding and growing stage in contrast with the reproductive and infective stages
Term
helminth
Definition
parasitic worm
Term
What are the 2 most common viral capsid shapes?
Definition

helical (a cylinder of coiled proteins)

 

icosahedral (having 20 faces of 1 or more proteins each; appears spherical)

 

 

 

other viral capsids have more complex shapes

Term
A protein, made by a mutated gene, that attacks self.
Definition
prion
Term
What are the 3 groups of helminths?
Definition

tapeworms

flukes

roundworms

Term
when a cell responds to stimulation by increasing in size
Definition

hypertrophy

 

 

(as opposed to hyperplasia, when a cell responds to stimulation by reproducing; hypertrophy may occur with hyperplasia)

Term
when a cell responds to stimulation by reproducing
Definition

hyperplasia

 

(Grk:  "beyond formation")

 

(as opposed to hypertrophy, when a cell responds to stimulation by increasing in size; hyperplasia & hypertrophy may occur together)

Term
Complete failure of a particular type of cell to reproduce. In fetal development it results in agenesis (where organ(s) never develop). In adults it results in lack of regeneration
Definition

aplasia

 

(Grk:  "no formation")

 

(as opposed to atrophy, which is decrease in cell size)

Term
when cells decrease in size, resulting in decrease in organ size
Definition

Atrophy

 

(such as muscle atrophy--disused muscles decrease in size because cells are decreasing in size, not b/c cell are failing to reproduce--aplasia--b/c muscle cells don't typically reproduce anyway)

Term
when a persistant irritant causes adult cells to be abnormally replaced by a different type of cells that are (usually) better adapted to the irritants (ex., Barrett’s esophagus)
Definition

metaplasia

 

(Grk:  "change in formation")

Term

heterolysis

 

Definition

apoptosis induced by enzymes from surrounding (usually inflammatory) cells 

 

(as opposed to autolysis, which is from the cell's own enzymes)

Term
genetically programmed cell death
Definition
apoptosis (note: doesn't have to be PRE-PROGRAMMED; the cell may have been re-programmed by a virus or external signal)
Term

necrosis

 

Definition
when an area of tissue in a living organism dies
Term
Compare cell size of apoptotic & necrotic cells.
Definition

Apoptotic cells shrink and eventually fragment (b/c, as part of the death program, the cell pumps out ions & water follows; this may be advantageous b/c smaller fragments are easier to phagocytize).

 

Necrotic cells swell and eventually lyse (b/c when the cell has low energy, ion transporters stop working, ions flow into the cell, and water follows).

Term
Compare the cell membranes of apoptotic & necrotic cells
Definition

apoptotic cell membranes remain intact (though the surface molecules may change)--they gradually shrink (by actively exporting ions & letting water follow) until the cell membrane touches itself and they break up into well-contained fragments that are easy to phagocytise

 

necrotic cell membranes are disrupted (the cells lose the ability to actively transport ions, swell up, and burst)

Term
Does apoptosis usually cause inflammation? Does necrosis?
Definition

apoptosis doesn't caused inflammation (b/c the cell contents are always contained by cell membrane until they're phagocytised)

 

necrosis usually does (b/c the cell swells up and bursts, releasing its contents into the extracellular fluid)

Term
Is apoptosis always pathological? Is necrosis always pathological?
Definition

apoptosis is usually "housekeeping" but may be pathological (ex., due to viruses re-programming the cell)

 

necrosis is always pathological

Term
What would you expect to see in the blood if a lot of liver cells were necrotic?
Definition
a lot of liver cell enzymes (different organs have different specific enzyme, and those enzymes are released when the cells die of necrosis)
Term

anthracosis

 

Definition

Accumulation of carbon in the lungs from inhaled smoke or coal dust

 

(may be a type of pneumoconiosus, or occupational lung disease)

Term
brown deposits that stain blue with Perl's staining
Definition
iron deposits
Term
refractive purple material
Definition
calcifications
Term
steatosis
Definition

abnormal deposits of fat within a cell due to one of a variety of impairments of normal movement of triglyceride fat throughout the cell 

 

most commonly associated with fatty liver (b/c the liver is the primary organ of lipid metabolism), but can occur in a variety of organs 

Term
A type of necrosis caused by ischemia without other significant types of injury; usually seen in heart or kidney. Tissue architecture is fine for several days after cell death, leading to a "ghostly" appearance under light microscopy.
Definition

coagulative necrosis

 

(note: ischemia leads to coagulative necrosis in most parts of the body, but in the brain it causes liquefactive necrosis b/c there's little structural framework in neural tissue)

Term
Macroscopically , coagulative necrosis will usually appear as:
Definition

a pale wedge (triangle) in the heart or kidney with the apex at the occluded vessel and the base at the periphery of the organ (the wedge corresponds to the area served by the occluded vessel)

 

Term
a special term for coagulative necrosis in hands or feet
Definition
gangrene (coagulative necrosis with some liquefaction due to infection is "wet gangrene")
Term
A mix of coagulative & liquifactive necrosis with a white, cheesy macroscopic appearance.
Definition

caseous necrosis

 

(the white cheesy appearance is caused by a leftover mass of protein from dead cells when, unlike in coagulative necrosis, cell architecture is obliterated)

 

(Frequently, caseous necrosis is seen in the lungs of people with TB infection, but it can also be caused in other organs and by other types of infection)

 

Term
TB causes ______ necrosis in the lungs.
Definition
Caseous
Term
white spots larger than a cell, containing no nuclei or cell divisions in adipose tissue indictate: 
Definition

adipose necrosis (fat necrosis)

 

 

(unlike coagulative necrosis, for instance, fat necrosis is not caused by a specific type of injury, though it's frequently associated with trauma.  The fact that adipose tissue has a distinctive appearance and structure b/4 death means that it has a distinctive structure & appearance after death)

 

 

Term
Areas of adipose tissue with no nuclei showing represent
Definition
fat necrosis
Term
Ischemia in brain usually causes ________ necrosis
Definition

liquefactive (ex., in preemies)

 

(in other tissues, ischemia causes coagulative necrosis, in which the cell dies and leaves its architecture behind, but the brain has little cell structure to leave behind and turns into mush, which turns into empty cavities)

Term
A type of necrosis characterized by fibrin-like proteinaceous materian within arterial walls
Definition
fibroid necrosis
Term
cephalad
Definition

toward the head (cephalad = "head" + "to")

 

antonym:  caudad

 

 

(sim. to rostral & caudal)

Term
rostral
Definition
toward the head
Term
caudal
Definition
toward the tail (butt)
Term
transverse plane
Definition

a plane that is perpendicular to the body's axis

 

(you typically look at a human transverse plane up from the feet, so left is right & right is left)

Term
sagittal plane
Definition
a plane that divides the body into right & left (parallel to the sagittal suture of the skull)
Term
coronal plane
Definition
a plane that divides the body into front & back (parallel to the coronal suture of the skull); aka frontal plane
Term
oblique plane
Definition

 

a diagonal or slanted plane

Term
longitudinal plane
Definition
any plane perpendicular to the transverse plane (could be coronal, sagittal, or oblique relative to those planes)
Term
flexion (of a joint)
Definition
a movement that decreases the angle of a joint
Term
extension (of a joint)
Definition
a movement that increases the angle of a joint
Term

With the arm hanging at the side, a rotation of the forearm that moves the palm from an anterior-facing position to a posterior-facing position

 

 

Definition

pronation

 

(if the arm were flexed so that the palm started out facing up, pronation would turn it so that it faced down)

 

(mnemonic:  a SUPlicant has their palm up for begging, which is the SUPine position; the Prone position is the opposite)

Term
The rotation of the forearm so that the palm faces anteriorly or up
Definition

supination

 

(mnemonic:  a SUPlicant has their palm turned SUPine for begging)

Term
Pronation of the sole of the foot is:
Definition
turning the sole of the foot laterally (letting the ankles sage medially)
Term
Supination of the sole of the foot is:
Definition
turning the sole of the foot medially (letting the ankles sag laterally)
Term
plantarflexion
Definition
Flexion of the entire foot inferiorly, as if pressing an automobile pedal.
Term
first-pass metabolism
Definition
drugs absorbed from the intestines go directly into hepatic portal circulation & are metabolized by the liver before they enter blood stream.
Term
A drug has a high first-pass metabolism. This would most affect what route of administration?
Definition
oral b/c things drugs absorbed from the intestines go directly into hepatic portal circulation & are metabolized by the liver before they enter blood stream.
Term
How are sublingual drugs absorbed?
Definition
Drug diffuses into the capillary network & enters systemic circulation directly
Term
How would you speed up or slow down absorption of an Intramuscular (IM) injection?
Definition

speed up by suspending in aqueous solution (since the extracellular fluid is aqueous, the drug can easily move from the aqueous solution to the extracellular fluid)

 

slow down by suspending in "depot solution" of non-aqueous liquid (such as polyethylene glycol), which forms a barrier between the drug and the aqueous extracellular fluid

Term
What is the difference between transdermal & topical drug application?
Definition
Topical--drug applied to skin (or eye) for local effect
Transdermal--systemic effect via skin absorption, usually via a patch
Term
A drug is not appropriate for oral consumption because it has a high first-pass metabolism. Is it appropriate for rectal application? Why or why not?
Definition
Could be. 50% of blood from rectal area bypasses hepatic portal circulation, so less first-pass metabolism than oral
Term

Explain the following mnemonic: 

 

Acid Accepts Electrons

Base Picks up a Proton

 

Definition

 

Acid Accepts Electrons (sometimes by donating a proton)

AH --> A- + H+

(note: A- is technically the conjugate base but strong acids typically exist in the conjugate base form so the conjugate base will sometimes be labeled with the acid's name.  For example COOH is carboxylic acid.  COO- is carboxylic acid's conjugate base, but COO- is sometimes labelled carboxylic acid.  The gods invented this system just to mess with your head) 

 

Base Picks up a Proton

 

B + H+ --> BH+

(the note above also applies) 

 

 

Term
Write the general equation for acid conjugation
Definition

HA <--> H+ + A-

 

 

mnemonic: Acid Accepts Electrons (sometimes by donating a proton)

Base Picks uP a Proton

Term
Write the general equation for base conjugation
Definition

B + H+ <--> BH+

 

Mnemonic:  Acid Accepts Electrons (sometimes by donating a proton)

Base Picks uP a Proton 

Term
Acidic drugs are absorbed better in a(n) _________ environment. Bases are absorbed better in a(n) _________ environment.
Definition

Acidic, basic

 

Because acids are better absorbed in their uncharged, acidic form than in their charged, conjugate base form, and bases are better absorbed in their uncharged, basic form than in their uncharged, conjugate acid form 

 

AH <--> A- + H+

B + H+ <--> BH+

Term
You give a person a dose of drug & test their maximum plasma concentration. What is the apparent volume of distribution (Vd)?
Definition
Vd = D/C
(b/c D = Vd * C)
(or work it out by units)
Term

For a base 10 log,

 

log(x) = y

 

x =

Definition

x = 10^y

 

 

So, log(100) = 2 

 

 

Term
Is the protonated or non-protonated form of an acidic drug more absorbable?
Definition

Protonated

 

AH <--> A- + H+ 

the non-ionized form is the protonated form (AH, not A-), and is more absorbable by non-polar cell membranes:

 

Mnemonic: 

Acid Accepts Electrons (sometimes by donating a proton)

Base Picks uP a Proton 

Term
Is the protonated or non-protonated form of a basic drug more absorbable?
Definition

non-protonated

B + H+ <-->BH+

 

Base Picks up a Proton, so the non-ionized (absorbable) form is non-protonated (B, not BH+)

 

Term
What equation can you use to determine what portion of an acidic or basic drug will be in a more absorbable form, given the pH of the environment and the pK of the acid or base?
Definition

[non]/[pro] = 10^(pH – pK)

 

where non = nonprotonated form (A- for acids, B for bases)

and pro = protonated form (AH for acids, BH+ for bases)

 

AH <--> A- + H+

B + H+ <--> BH+

(Acid Accepts Electrons, sometimes by donating a proton

Base Picks uP a Proton)

Term
What is the AUC of a drug?
Definition
The Area Under the Curve (of concentration over time)--tells you how much has entered the measured body compartment over a given amount of time.
Term
When is a person in the "fed" state (as opposed to fasting or starving)?
Definition
within 2-4 hours of eating a meal
Term
When is the "fasting" state
Definition
begins 2-4 hours after eating a meal
ends 3-5 days after eating a meal
Term
When does the "starved" state begin?
Definition
3-5 days after last meal
Term
Hormone that tells the liver to break down glycogen into glucose
Definition
glucagon
Term
What is the first step in glucose respiration in both the aerobic & anaerobic pathways?
Definition

glycolysis

 

(1 glucose in, 2 pyruvates out, 2 ATP are made)

Term
Glycolysis breaks glucose down into 2 _________, producing _______ ATP
Definition
pyruvate
2
Term
After glycolysis, the 2 pyruvates are transformed into acetyl CoA, enter theTCA cycle & enter the Electron Transport & Oxidative chains. How many more ATP does this produce?
Definition

34

 

(17 per pyruvate; when added to the 2 ATP of glycolysis this gives 36 ATP for aerobic respiration of 1 molecule of glucose)

Term
After glycolysis, pyruvate is fermented in anaerobic respiration. How many ATP does this produce?
Definition
0 ATP
Term
Fermentation produces 0 ATP. So what is its purpose?
Definition
To regenerate NAD+ from NADH to feed back into glycolysis
Term
How many ATP per glucose does aerobic respiration produce (total)?
Definition
36
Term
How many ATP per glucose does anaerobic respiration produce (total)?
Definition
2
Term
Cori cycle
Definition

The circular metabolic pathway in which

 

muscles convert glucose to lactate via anaerobic respiration

 

and liver converts lactate to glucose (via gluconeogensis), which then returns to the muscles 

Term
The _____ is an organ that will use glucose as its energy source except in extreme necessity, while the _______ can only ever use glucose.
Definition
brain
RBCs
Term
What is the major advantage of glycogen being a branched polymer?
Definition
branched makes it more soluble
it can be accessed faster b/c more branch ends to access
Term
Glycogen is primarily created and stored in the ______, but is also created & stored in the _______ for their own use.
Definition
liver
muscles
Term
When plenty of glycogen has been created, what kind of fat is glucose turned into?
Definition
triglycerides
Term
What must happen to fats in order for them to be transported in the bloodstream
Definition
they must be packaged inside of water-soluble proteins (chylomicrons: VLDL, IDL, LDL, HDL)
Term
3 fatty acids on a glycerol backbone
Definition
triglyceride
Term
What 3 things can the body do with amino acids?
Definition

1. use for energy (feed into TCA/citric acId/Krebs cycle)

 

2. build into proteins

 

3. transform into other amino acids or other substances (ex., histamine from histidine, nitric oxide from arginine, neurotransmitters, etc.)

Term
What is the body's goal during the fasting state?
Definition
to keep the blood glucose levels even & everything running smoothly until the next meal
Term
Glycogen has a (charged) phosphate attached that must be removed so glucose can leave cell. The ____ has an enzyme to remove the phosphate, but the ______ don't.
Definition
liver
muscles
Term
Glycogen has a (charged) phosphate attached that must be removed so glucose can leave cell. The ____ has an enzyme to remove the phosphate, but the ______ don't.
Definition
liver
muscles
Term
If you are well-fed during the day, roughly what portion of your glycogen do you use up overnight?
Definition
2/3
Term
Primary source of glucose during fasting
Definition
glycogen in liver
Term
The process by which glucose is synthesized from triglyceride or protein in the liver.
Definition
gluconeogenesis
Term
Low ________ (hormone) & high _________ (hormone) signal _______ (enzymes) in adipose tissue to break down triglycerides into fatty acids & glycerol, which are transported into the bloodstream.
Definition

insulin

glucagon

lipases

Term
How and where are fatty acids used for energy in the body?
Definition

Beta-oxidation:  Fatty acids-->acetyl CoA

 

can feed acetyl CoA into TCA /citric acid/Krebs cycle & electron transport

 

this can occur in many cells (ex., muscle cells)

(as opposed to the glycerol backbone, that can only be used for gluconeogenesis in the liver)

 

Term
How and where is glycerol used for energy in the body?
Definition

glycerol is used for gluconeogenesis by the liver only

 

(as opposed to fatty acids, that can be beta-oxidized into acetyl CoA and fed into the TCA/citric acid/Krebs cycle in many cells in the body) 

Term
Why do diabetics with poor control have "fruity" smelling breath?
Definition

During fasting or carbohydrate starvation, oxaloacetate is depleted in liver because it is used for gluconeogenesis. This impedes entry of acetyl-CoA into Krebs cycle. Acetyl-CoA then is converted in liver mitochondria to ketone bodies, which are exported to the rest of the body to use for energy.

 

One of the ketones produced is acetone, which has a fruity smell.


Term
When does the brain use ketone bodies for energy? When does it use fatty acids?
Definition
The brain only uses ketone bodies for energy in extreme need
the brain cannot ever use fatty acids
Term
What does the liver do with fatty acids during fasting & starvation?
Definition
transforms them into ketone bodies
Term
The preferred amino acid for gluconeogenesis.
Definition
alanine
Term
Breaking down amino acids during gluconeogenesis releases _______, which are transformed into toxic ________, which must be peed out as ________
Definition
amino groups (NH4+)
ammonia (NH3)
urea
Term
Ammonia (NH3) is mostly transformed into urea in the _____, but also in the________
Definition
liver
kidneys
Term
mobile DNA sequences that an move between plasmids, or between plasmids & the chromosome
Definition
transposon
Term
Prokaryote DNA organization doesn't use ________ proteins.
Definition
histone
Term
What are polycistronic genes? Are eukaryotes or prokaryotes more likely to have polycicstronic genes?
Definition
several genes controlled by one promoter
prokaryotes (in eukaryotes, one promoter usually controls one gene)
Term
Prokaryotes are mostly __________ (haploid/diploid). Eukaryotes are mostly _________
Definition
haploid (n)
diploid (2n)
Term
Ori
Definition
prokaryotic origin of replication (aka OriC)
Term
Do prokaryotes have one or many sites of origin of DNA replication? What about eukaryotes?
Definition
prokaryotes have 1 (called an Ori or OriC)
eukaryotes have several
Term
Prokaryote replication begins when a pile of _____ molecules bind to one of the 9-nuclotide repeats in the Ori (the site where DNA replication begins); the DNA melts and opens up
Definition
DnaA
Term
After DnaA opens the prokaryotic DNA at its Ori, a helicase called _____ opens the strands further, allowing elongation enzymes to attach and begin making the new DNA strand
Definition
DnaB
Term
Name the 2 prokaryotic forms of DNA replication. Which is semi-conservative?
Definition
theta replication
rolling circle
both are
Term
What kind of DNA replication begins when an initiator protein nicks one strand & frees a 3’ end?
Definition
rolling circle replication
Term
a type of prokaryotic nucleic acid replication good for rapidly making lots of copies of circular DNA or RNA
Definition
Rolling circle replication
Term
a prokaryotic enzyme that nicks one strand & releases some of twisting, allowing DNA replication enzymes that are essential to bacterial DNA replications
Definition
topoisomerase
Term
All new DNA is build in the ________ direction
Definition
5'-->3' (because polymerases can only attach new nucleotides to the 3' side)
Term
prokaryotic factors that cause RNA polymerase to recognize promoter & start transcribing
Definition
sigma factors
Term
Unlike eukaryotic RNA polymerase, prokaryotic RNA polymerase doesn't need a _________
Definition
primer
Term
bacteria "sex" (exchange of information via a sex pilus) is called _____
Definition
conjugation
Term
When a cell gains new genetic information from a lysogenic virus
Definition
transduction
Term
A virus that can't enter a lysogenic/temperate phase
Definition
lytic
Term
a bunch of genes in one operon (all turned on/off at the same time) that all contribute to pathogenicity of bacterium
Definition
pathogenicity island
Term
Antibiotics that attack the synthesis of the 30S ribosome subunit in prokaryotes
Definition
aminocglycosides
Term
Antibiotics that attack the synthesis of the 50S ribosome subunit in prokaryotes
Definition
Macrolides
Term
Prokaryotic ribosomes are made of a _______ subunit and a ________ subunit
Definition
30S
50S
Term
Tropoismerases, which nick one strand & releases some of twisting during DNA recpliaciotn (are essential to bacterial DNA replication) are a target of _____________ antibiotics
Definition
quniolone
Term
a functioning unit of genomic material containing a cluster of genes under the control of a single regulatory signal or promoter
Definition
operon
Term
The Operator of the LAC operon has a Repressor that turns the gene off unless chemicals turn the Repressor off. This is an example of _________ gene control.
Definition
positive (because the chemicals turn the gene ON)
Term
What 3 groups determine the charge of an amino acid
Definition
the amino group,
the carboxyl group,
and/or the side chain
Term
In a peptide chain, the peptide bonds use up the ____ & _____ groups, so only the _____, _________, & _______ can be ionized
Definition
amino & carboxyl
side chain, N-terminal amino, & C-terminal carboxyl group
Term
What are the 3 basic amino acids (B + H+ --> BH+; usually seen in conjugate acid form in body)
Definition
histidine
arginine
lysine
Term
Explain this mnemonic
HAL runs on BASIC
Definition
Histadine, Arginine, & Lysine are basic amino acids
Term
What are the 2 acidic amino acids (AH --> A- + H+; usually seen in conjugate base form in body).
Definition
glutamate (glutamic acid)
aspartate (aspartic acid)
Term
What 2 amino acids have pKas in the basic range, but their side chains act like acids
Definition
cysteine, tyrosine
Term
What is the ionized form of the amino group in amino acids?
Definition
NH2 + H+ --> NH3+
+1
Term
What is the ionized form of the carboxylic acid group in amino acids? What is its charge?
Definition
COOH --> COO- + H+
-1
Term
The stronger the acidic nature of a molecule, the ______ its pKa
Definition
lower
(like the more acidic the environment, the lower its pH)
Term
The stronger the basic nature of a molecule, the ______ its pKa
Definition
higher
(like the more basic the environment, the higher the pH)
Term
At pH = pKa, ___ of an acidic or basic substance is dissociated.
Definition
50%
Term
If pH is < pKa, more of a basic substance would be in its ______ and more of an acidic substance would be in its _________ form. (ionized/non-ionized)
Definition
base would be ionized
acid would be non-ionized
if pH < pKa, the environment is acidic (Acid Accept Electrons, sometimes by donating a proton & Base Picks Up Protons)& forces a proton on the substance. So the substances would both be more in their protonated form
so a base (B + H+ -->BH+) would be more ionized
an acid (AH --> A- + H+) would be more non-ionized
Term
For amino acids with 2 ionizable groups, what are those groups
Definition
NH2 (NH3+) & COOH (COO-)
Term
What amino acids have 3 ionizable groups
Definition
the bases: histidine, arginine, lysine
the acids: glutamate, aspartate
the weird ones: cysteine, & tyrosine (act like acids even tho they're not)
Term
the point at which the overall charge of the amino acid is neutral
Definition
isoelectric point (pI)
Term
What is an amino acid's pI?
Definition
isoelectric point: the point at which the overall charge of the amino acid is neutral
Term
For amino acids with 2 ionizable side groups, what is the pI?
Definition
pI = average(pKa1, pKa2)
Term
For amino acids with 3 ionizable side groups, what is the pI?
Definition
The pI is the average of the two pKas that are CLOSEST IN VALUE. (Note: pI is average the two pKa’s that are on either side of the neutral form; It just so happens that the 2 pKa's that are on either side of the neutral form happen to always be closest together in amino acids).
Term
Which amino acid is used by the body as a blood buffer?
Definition
Histidine.
Although histidine's side chain is basic, its pI of 7.65 is closest to blood pH of 7.4
Term
Primary structure of a protein
Definition
linear sequence of amino acids
Term
Protein's secondary structure
Definition
folding of primary structure into recurring structures (ex., beta-sheets, alhpa-helix) or non-recurring structures
Term
Beta-sheets & alpha-helices are examples of a protein's ______ structure
Definition
secondary
Term
A secondary protein structure that occurs due to hydrogen bonding occurs between the oxygen on carbonyl & the hydrogen on amide at every 4th carbon
Definition
alpha helix
Term
What 3 amino acids have side chains that are too bulky for alpha-helices?
Definition
phenylalanine, tyrosine, tryptophan
Term
What 5 amino acids are inappropriate for alpha-helices because their side-chains are charged?
Definition
the bases: histidine, lysine, arginine
the acids: aspartate, glutamate
Term
What 5 amino acids are inappropriate for alpha-helices because their side-chains are charged?
Definition
the bases: histidine, lysine, arginine
the acids: aspartate, glutamate
Term
Why are histidine, lysine, arginine, aspartate, and glutamate non appropriate for alpha-helices?
Definition
because their side-chains are charged
Term
Why are phenylalanine, tyrosine, and tryptophan not appropriate for alpha-helices?
Definition
because their side chains are too bulky
Term
What's the difference between an Ori & an OriC
Definition

An Ori is an origin of replication for DNA

 

each prokaryotic circular genome has one Ori, called an OriC

Term
Eukaryotes: an enzyme that unwinds DNA at the ORI, creating a replication bubble with 2 replication forks. Part of the multi-protein complex that attaches to the Ori.
Definition
helicase
Term
Eukaryotes: a protein that stabilizes the opening of the double helix
Definition
single-strand binding protein
Term
Eukaryotes: a protein that relieves the tension in front of the replication fork caused by unwinding the DNA double helix
Definition
DNA topoisomerase I
Term
Eukaryotes: a protein catalyzes synthesis of RNA primers (a primase)
Definition
DNA polymerase alpha
Term
Eukaryotes: How many DNA polymerase alphas does the leading strand need? How many does the lagging strand need?
Definition
The leading strand only needs one RNA primer on the 5’ end to start replication The lagging strand needs one RNA primer at the 5’ end to start each Okazaki fragment
Term
What does this mnemonic mean?
Alpha starts the fragment
Definition
In Eukaryotes, DNA Polymerase alpha creates the RNA primer that starts each fragment of replicated DNA
Term
A protein that catalyses formation of phosphodiester bonds between the 3’ end of the last nucleotide of a DNA chain & the 5’ end of a free nucleotide triphosphate
Definition
DNA polymerase delta
Term
What does this mnemonic mean? Delta forms Diester Bonds
Definition

In eukaryotes, DNA Polymerase delta forms diester bonds between the 3' end of the last nucleotide in the chain & the 5' end of a free nucleotide.

 

(DNA polymerase delta is the main eukaryotic DNA polymerase.  DNA polymerase epsilon 

Term
What eukaryotic protein has 3’-->5’ exonuclease activity & what does that mean?
Definition
DNA Polymerase Delta
In Eukaryotic DNA replication, DNA Polymerase Delta has a proofreeding mechanism--if one of the nucleotides was matched wrong (say, an A matched to a G), then DNA polymerase delta can go back one nucleotide & cut the mismatched nucleotide out
Term
Eukaryotic Is DNA replication at the Ori uni-directional or bi-directional?
Definition
Bi-directional (the bubble opens up on both directions)
so the leading strand on one side of the Ori is the lagging strand on the other side of the Ori, and v.v.
Term
Nucleotids can only be added to the _____ side of the polynucleotide chain
Definition
3'
Term
Eukarotic After replication ends, ________ are removed from the 5’ end of each newly synthesized DNA fragment (one from 5’ of leading strand daughter; one from 5’ end of each Okazaki fragment)
Definition
RNA primers
(Remember: RNA primers were synthesized by DNA Polymerase alpha & used to start each new fragment of replicated DNA)
Term
Eukaryotic: protein that joins the Ozaki fragments
Definition
DNA ligase
Term
Eukaryotic: protein that cuts & reseals the 2 newly synthesized DNA molecules so they can separate from each other
Definition
Topoisomerase II
Term
Eukryotic: What do Topoisomerase I & Topoisomerase II do?
Definition
DNA Topoisomerase I relieves tension ahead of the replication fork that is caused by unwinding the helix
DNA Topoisomerase II cuts & reseals the 2 newly synthesized DNA molecules so they can separate from each other
Term
Eukaryotic: What do DNA Polymerase alpha & DNA Polymerase delta do?
Definition
DNA polymerase alpha catalyzes synthesis of RNA primers (a primase)
DNA polymerase delta catalyses formation of phosphodiester bonds between the last nucleotide of a DNA chain & a free nucleotide triphosphate (Has a proofreading mechanism)
Term
What is the DNA primase for Eukaryotes?
Definition
DNA Polymerase Alpha
Term
Eukaryotic: What does telomerase do?
Definition
The lagging daughter strand can’t get all the way to the 3’ end of the lagging parent strand, so telomerase replaces the last bit with DNA formed form some RNA it carries around to act as a primer (template)
Term
Eukaryotic: RNA is primarily transcribed in the _______ and processed in the _______
Definition
nucleolus
nucleus
Term
RNA is copied from the ____ strand of DNA, so it has the same genetic sequence as the______ strand
Definition
anti-sense
sense
Term
When creating new RNA strands, ____________ act like DNA polymerases in that they catalyze phosphodiester bonds between nucleotides in a 5’-->3’ direction
Definition
RNA polymerases
Term
Unlike DNA polymerases, RNA polymerases don't need a:
Definition
primer to start the strand
(so there's no RNA equivalent of DNA polymerase alpha)
Term
Unlike DNA polymerases, RNA polymerases have no ability to:
Definition
proofread (not 3'-->5' exonuclease activity)
Term
first nucleotide in a gene to be transcribed (downstream of & near core promoter)
Definition
transcription start site
Term
The _____ _______ region “points out” the transcription start site for RNA polymerase
Definition
core promoter
Term
TNA polymerase is attracted to the _____ _____in the core promoter region
Definition
TATA box (5'-TATAAA-3', or similar)
Term
loosely-packed chromatin seen as dark splotches around the edge of the nucleus
Definition
euchromatin
Term
tightly packed chromatin seen as big empty spaces between the dark edge of the nucleus and the nucleolus
Definition
heterochromatin
Term
Eukaryotic: ________ are often (though not always) being transcribed while ______ are not freqently transcribed
Definition
euchromatin
heterochromatin
Term
What kind of cell secretes the extracellular matrix & collagen in most tissues
Definition
fibroblasts
Term
What's the difference between a fibroblast and a fibrocyte.
Definition
A fibroblast secretes the extracellular matrix and collagen.
A fibrocyte is a mature, fully differentiated, much less active version of a fibroblast
Term
What 4 cell types are msot common in connective tissue?
Definition
Fibroblasts
Macrophages
Mast Cells
Plasma cells
Term
What 3 fiber types are most common in connective tissue?
Definition
Collagen
Reticular fibers
Elastic fibers
Term
most abundant protein in body; provides tensile strength
Definition
collagen
Term
Although reticular fibers & collagen fibers have different names...
Definition
reticular fibers are actually made of a type of collagen
Term
Collagen fibers are made of packed, cross-linked fibrils, which are made of aggregates of...
Definition
triple-helices of alpha-1 & alpha-2 peptide chains
Term
A fine net-like protein that stains black in silver stain
Definition
reticular fibers
Term
Stretchy fibers or lamellae (sheets) that add resiliency to connective tissue
Definition
elastic fibers
Term
Marfans syndrom is characterized by problems with _______ fibers
Definition
elastic
Term
Elastic fibers are important in lungs, skin, & bladder, but they are most important in maintaining resilience of _______________
Definition
blood vessels
Term
Why is aortic disection a problem in people with Marfan's syndrome?
Definition
because Marfan's syndrome is caused by problems with elastin fibers, and elastin fibers are responsible for the resilience of blood vessels
Term
The most common glycosaminoglycan in loose connective tissue
Definition
hyaluronic acid
Term
Long, unbranched polysaccharide chains of repeating disaccharide units (uronic acid sugar + amino sugar) that are not flexible enough to glob up, so they stay expanded & form a flexible gel
Definition

glycosaminoglycan (GAG)

 

GAGs form an important component of connective tissues. GAG chains may be covalently linked to a protein to form proteoglycans. Some examples of glycosaminoglycan uses in nature include heparin as an anticoagulant, hyaluronan as a component in the synovial fluid lubricant in body joints, and chondroitins, which can be found in connective tissues, cartilage, and tendons.

Term
any protein with one or more glycosaminoglycan molecules attached
Definition
proteoglycan (aka glycoproteins)
Term
_________ are typically glycosaminoglycans covalently linked via a variety of protein molecules to a glycosaminoglycan (hyaluronan) backbone
Definition
proteoglycans (aka glycoproteins)
Term
What cell secretes the precursors of collagen fibers, elastin fibers, Glycosaminoglycans (GAGs), and proteoglycans such as fibrillin, laminin, & fibronectin
Definition
fibroblasts
Term
A multi-adhesive glycoprotein that is a major component of basement membranes & helps bind cells to membranes
Definition

laminin

 

(LAMINin is the major protein in the LAMINa)

Term
A multi-adhesive glycoprotein that elps control deposition & orientation of collagen in ECM & helps control cell binding to ECM
Definition
Fibronectin
Term
cell-to-cell adhesion proteins
Definition
cadherins
Term
cell-to-ECM adhesion molecules
Definition
integrins
Term
A type of tissue that consists of many loosely-arranged elastin fibers & collagen bundles with a few fibroblasts (may also contain mast cells, macrophages, & B cells)
Definition
loose connective tissue
Term
What kind of connective tissue looks like marble swirls of fiber with a few dark dots of cell nuclei?
Definition

loose connective tissue

 

(If the tissue is spread out on a glass slide, it will appear more like a loose net of fibers)

 

Compare to dense regular connective tissue, which looks like many parallel straight or wavy fibers, or dense irregular connective tissue, which looks like wood grain (neither as swirly & marble-like as loose connective tissue, nor as organized as dense regular connective tissue)

Term
What type of tissue consists of coarse, thick intertwined bundles of collagen fibers with sparse fibrobalsts (may also contain mast cells, macrophages, & B cells)?
Definition
dense connective tissue
Term
What type of connective tissue has a lot of fibers that look like woodgrain, and a few dark fibroblast nuclei?
Definition
dense irregular connective tissue (neither as swirly & marble-like as loose connective tissue, nor as organized as dense regular connective tissue)
Term
What type of tissue consists of regularly oriented parallel bundles of collagen fibers separated by linear rows of fiborcytes (appear as thin dark lines of fibrocyte nuclei)?
Definition

dense regular connective tissue

 

(compare to loose connective tissue, which looks swirly, like marble, and dense irregular connective tissue which looks like wood grain)

Term
What kind of connective tissue is found in tendons & ligaments?
Definition
dense regular connective tissue
Term
What kind of conective tissue is found in the dermis & the submucosa of digestive tube?
Definition
dense irregular connective tissue
Term
conception to 8 weeks post-conception; time during which basic body plan & organ systems develop
Definition
embryo
Term
solid ball of up to 16 cells
Definition
morula
Term
empty sphere of up to 128 cells
Definition
blastula (or blastocyst)
Term
Process by which blastula invaginates, becomes a tube-within-tube, and then cells migrate between the tubes to form 3 layers.
Definition
Gastrulation
Term
From the outside in, the 3 layers of the gastrula are
Definition
ectoderm, mesoderm, endoderm
Term
Process by which the notochord signals cells above it to begin formation of Central Nervous System (CNS)
Definition
neurulation
Term
What is embryonic folding?
Definition
the embryo forms most of its features by twisting & folding simpler features (ex., a sheet of cells folded in half to become a tube)
Term
the process by which the ectoderm, endoderm, and mesoderm develop into the internal organs of the organism.
Definition
organogensis
Term
Carnegie staging
Definition
A set of 23 stages used to describe the first 9 weeks of pregnancy based on internal & external physical characteristics of the embryo
Term
By stage 23 of Carnegie staging, all essential internal organ systems are:
Definition
present but incapable of sustaining independent life
Term
Time from 9 weeks post-conception (11 weeks post-period) to birth, during which organs develop & attain function; lots of growth!
Definition
fetal period
Term
Parturition
Definition
childbirth
Term
self-renewing progenitor cell
Definition
stem cell
Term
Embryonic stem cells are from a _______ and are ___potent
Definition
blastocyst (blastula)
pleuripotent (can produce any cell in body; not totipotent, which can also form placenta & extraembryonic membranes)
Term
pleuripotent stem cell
Definition
can produce or mature into any cell in body; not totipotent, which can also form placenta & extraembryonic membranes)
Term
totipotent stem cell
Definition
can generate the entire conceptus (body + placenta + extraembryonic membranes)
Term
For clinical purposes, gestational age is counted from:
Definition
the first day of the last period (day 1 of the menstrual cycle)
Term
For experimental purposes gestational age is counted from:
Definition
the day of conception (roughly day 14 of the menstrual cycle)
Term
An scientist who is studying gestation says that the embryo is 6 weeks old. An obstetricion would say that the gestational age is:
Definition
~8 weeks
Clinical gestational age = experimental gestation age + ~2 weeks
(b/c experimental counts from conception & clinical counts from first day of last period)
Term
A cytokine that acts on cells touching the signaling cell
Definition
juxtacrine
Term
Where are the receptors for all growth factors?
Definition
all growth factors are proteins, so their receptors are on cell surfaces
Term
Where are the receptors for steroids, retinoic acid (vitamin A), & other small, lipophilic molecules?
Definition
inside the cell (often inside the nucleus)
Term
Lipopolysaccharide (LPS), the most potent endotoxin, is only produced:
Definition
in the walls of gram-negative bacteria
(endotoxin: a toxin that is part of the organism rather than secreted by the organism)
Term
An inflammatory molecule produced in the cell walls of gram negative bacteria
Definition
Lipopolysaccharides (LPS)
Term
________ bacteria have an outer membrane
Definition
gram negative
Term
provides rigidity to cell wall; only found in gram-positive bacteria
Definition
teichoic acid
Term
What are the 4 steps of gram staining?
Definition
1. add crystal violet
2. add gram iodine (a mordant)
3. add alcohol or acetone (decolorizer)
4. add safranin red (to counter-stain gram negative bacteria)
Term
What color are gram positive bacteria after each of the 4 steps in gram staining?
Definition
1. crystal violet--purple/blue
2. gram iodine mordant--purple/blue
3. alcohol or aceteone decolorizer--purple/blue
4. safranin red counter-stain--purple/blue
Term
What color are gram negative bacteria after each of the 4 steps in gram staining?
Definition
1. crystal violet--purple/blue
2. gram iodine mordant--purple/blue
3. alcohol or aceteone decolorizer--clear
4. safranin red counter-stain--pink
Term
Is staphylococci gram positive or gram negative?
Definition
gram positive
Term
Is e. coli gram positive or gram negative?
Definition
gran negative
Term
Explain the following mnemonic:
The microbe world is opposite world:
Blue is positive!
Rosy-hued is negative
Definition
Gram positive bacteria stain purple/blue
gram negative bacteria stain pink
Term
Crystal violet attaches to
Definition
peptidoglycan
(gam negative bacteria also have peptidoglycan, but less and shielded from crystal violet by other molecules, including an outer membrane)
Term
Is mycoplasm gram positive or gram negative?
Definition
Neither. It has no cell wall.
Term
What color does mycobacteria turn if gram stained?
Definition
Stays clear. Mycobacteria have a lot of mycolic acid (waxy substance) in their cell wall & don't take up safranin red even though they're gram positive
Term
period during which bacteria adapt themselves to growth conditions & prepare to divide
Definition
lag phase
Term
period during which cells double, and double, and double…
Definition
growth phase
Term
period during which nutrients have begun to deplete, wastes have built up & rate of bacterial growth = rate of bacterial death
Definition
stationary growth phase
Term
period during which all nutrients have been used up or wastes have built up to toxic levels & all bacteria die
Definition
death phase
Term
dormant form of a bacterium with little water formed when environment is inhospitable
Definition
spore
Term
What are the only kinds of bacteria that can sporulate?
Definition
gram positive rods
Term
process during which spore coat is weakened (via heat, extremes of pH, etc.) & spore re-awakens
Definition
spore germination
Term
bacteria require or tolerate O2 at less than atmospheric partial pressure
Definition
microaerophiles
Term
bacteria that can live with or without O2
Definition
facultative anaerobes
Term
growth media that contain (extra) nutrients required to grow microbes
Definition
enriched media
Term
growth media contain or lack elements that preclude growth of all but a few kinds of microbes
Definition
selective
Term
growth media that react differently (ex., change colors) depending on which kinds of microbes are grown on them
Definition
differential media
Term
growth media contains blood that has been lysed by heating, releasing intracellular nutrients
Definition
chocolate agar
Term
MacConkey agar is selective because biles salts & crystal violet dye inhibit the growth of
Definition
gram-positive bacteria
Term
MacConkey agar is differential because
Definition
It contains a pH indicator that turns from reddish-orange to hot pink in the presence of lactic acid (from lactose fermentation)
Term
A bacteria is grown on MacConkey agar. The growth medium around the colony turns hot pink. What can be said about that species of bacteria?
Definition
It does lactic acid fermentation
(lactic acid turn a pH indicator in MacConkey agar from reddish to HOT PINK)
Term
Only staphylococci & micrococaceae can grow in Mannitol Salts Agar because
Definition
it's so salty! (NaCl)
Term
A bacteria grows on Mannitol Salts agar and produces yellow colonies with yellow zones. It is not a micrococcae. What is it?
Definition
coagulase-positive staphylococci (ex., Staph Aureus)
(only staphylococci & micrococcae can grow on Mannitol salts; coagulase positive staphylococcae produce yellow colonies with yellow zones)
Term
A bacteria grows on Mannitol Salts agar and produces small pink or red colonies with no color change in medium. It is not a micrococcae. What is it?
Definition
coagulase-positive staphylococci (ex., Staph Epidermidis)
(only staphylococci & micrococcae can grow on Mannitol salts; coagulase-negative staphylococcae produce small pink or red colonies with no zones)
Term
A bacterial cell colony grown on Eosin Methylene Blue agar has a dark blue-green center. What do you know about those cells?
Definition
They can do lactic acid fermentation (colonies that do lactic acid fermentation have low pH, which causes dye to flow in)
Term
A bacterial cell colony grown on Eosin Methylene Blue agar has a green sheen. What can you say about the colony?
Definition
It is e. coli
(e. coli grown on eosin methylene blue agar has a green sheen)
Term
A bacterial colony is growing on Thayer-martin agar. What can you say about that colony?
Definition
Its genus is Neisseria
(Thayer-martin agar is a chocolate agar with antibiotics that prevent all bacteria but Neisseria from growing
Term
A bacteria is growing salmon-red colonies on Hektoen enteric agar. What can you say about the bacteria?
Definition
it's gram negative (Hektoen enteric agar prohibits growth of gram-positive bacteria)
it does lactic-acid fermentation (lactic-acid fermenting colonies turn salmon-red)
Term
A bacteria is growing blue-green colonies with black centers on Hektoen enteric agar. What can you say about the bacteria?
Definition
it's gram negative (gram-positive can't grow on Hektoen enteric agar)
it produces H2S (cells that produce H2S form black colonies, or blue-green colonies with black centers)
Term
Hektoen enteric agar is mostly used to identify ______ & ________.
Definition
salmonella & shigella
Term
B cells in peritoneal cavity & at mucosal sites product _____ antibodies against certain encapsulated bacteria
Definition
natural
(you don't need to have met the microbe to produce "natural" antibodies)
Term
proteins expressed by cells of the innate immune system; bind to Pathogen-Associated Molecular Patterns (PAMPs) on stressed cells or microbes
Definition
PRRs (Pattern Recognition Receptors)
Term
Pattern Recognition Receptors (PRRs) bind to _________ on microbes or stressed cells
Definition
Pathogen-Associated Molecular Patterns (PAMPs)
Term
A type of PRRs (Pattern Recognition Receptors) expressed by phagocytes (PMNs, macrophages, DCs)
Definition
TLR (toll-like receptors)
Term
TLRs are a class of PRR expressed on _________
Definition
phagocytes (such as macrophages, neutrophils, & dendritic cells)
(PRR = pattern recognition receptors)
Term
Which 3 TLRs bind to bacterial peptidoglycan, lipoprotein, lipotechoic acid?
Definition
TLR-1, TLR-2, & TLR-6
Term
What 3 things do TLR-1, TLR-2, & TLR-6 bind to?
Definition
peptidoglycans
lipoproteins
lipotechoic acid
Term
Which TLR binds to LPS (on gram-negative bacteria, mannans (fungal polysaccharides), and viral envelope proteins?
Definition
TLR-4
Term
What 3 things does TLR-4 bind to?
Definition
LPS (on gram-negative bacteria)
mannans (fungal polysaccharides)
viral envelope proteins?
Term
What 3 things does TLR-4 bind to?
Definition
LPS (on gram-negative bacteria)
mannans (fungal polysaccharides)
viral envelope proteins?
Term
What molecule does TLR-5 bind to?
Definition
bacterial flagellin
Term
Which TLR binds to bacterial flagellin?
Definition
TLR-5
Term
What do TLR-3, TLR-7, TLR-8, & TLR-9 bind to?
Definition
microbial nucleic acids (ex., single-standed RNA, unmethylated CpG dinucleotides)
Term
What 4 TLRs bind to microbial nucleic acids?
Definition
TLR-3, TLR-7, TLR-8, TLR-9
Term
When a phagocyte's ____ binds to a microbial component, the phagocyte expresses molecules to help infection.
Definition
TLR
(WHICH molecules depend on which TLR & which type of phagocyte)
Term
Normally (when no infection), 60-70% of leukocytes are _____
Definition
neutrophils (aka PMNs)
Term
Leukocytes that rapidly enter cells after infection, but do not divide and die soon (in hours or days).
Definition
Neutrophiles (PMNs)
Term
Leukocytes that enter tissue slowly & live longer than earlier-entering leukocytes
Definition
monocytes (macrophages after they enter tissue)
(ther earlier cells are neutrophils/PMNs)
Term
___, ___, & ____ peptides form the complement cascade are anaphylatoxins that trigger acute inflammation reaction.
Definition
C3a, C4a, C5a
Term
C3a, C4a, C5a cause ___________to degranulate & release histamines
Definition
mast cells
Term
___ is a complement protein that stimulates neutrophils to move toward infection, adhere to endothelial cells, & release respiratory burse of Reactive Oxygen Species (ROS). It also induces endothelial cells to express adhesion molecules to promote neutrophil binding.
Definition
C5a
Term
C5a is a complement protein that stimulates ________ to move toward infection, adhere to endothelial cells, & release respiratory burse of Reactive Oxygen Species (ROS). It also induces _____ cells to express adhesion molecules to promote _______ binding.
Definition
neutrophils
endothelial
neutrophil
Term
The opsonin produced by the alternative complement pathway.
Definition
C3b
Term
The opsonin produced by the classical and lectin complement pathways.
Definition

C4b

 

(Opsonin:  a molecule that coats an invader and enhances phagocytosis)

Term
The 2 opsonins produced by the complement pathways
Definition
C3b C4b
Term
What is the C5b-C9 complex called & what does it do?
Definition
MAC (Membrane Attack Complex)
The MAC is an open pore in the microbial membrane that allows free movement of water & ions, which causes bacteria to swell & lyse.
Term
What is the C5b-C9 complex called & what does it do?
Definition
MAC (Membrane Attack Complex)
The MAC is an open pore in the microbial membrane that allows free movement of water & ions, which causes bacteria to swell & lyse.
Term
Complement receptor on phagocytes (neutrophils & macrophages) that binds to C3b or C4b.
Definition
CR1
Term
a TCR co-receptor that binds to MHCII
Definition
CD4
Term
A TCR coreceptor that binds the MHC I
Definition
CD8
Term
the 2 parts of the TCR complex that transduce signal
Definition
CD3, zeta chain
Term
the 2 parts of the TCR complex that become ITAMs when activated
Definition
CD3, zeta chain
(ITAM is an activation signal to immune cells. Unhealthy cells present ITAMs to NKs & cytotoxic T cells. But T cells & some other cells also produce ITAMs to self when activate
Term
the most important adhesion molecule on the T cell for antigen recognition
Definition
LFA-1
Term
chemokines from the innate immune system near sites of infection cause LFA-1 on T cells to
Definition
transform into a high-affinity state & cluster together
Term
The CD28 receptor on T cell binds costimulators APC, the most important type of which is
Definition
B7 proteins
Term
When PRRs (such as TLRs) on APCs see PAMPs, the APC expresses _____ proteins
Definition
B7
Term
What happens to an APC when the CD40L on an antigen-stimulated T cell binds to CD40 receptor on the APCs?
Definition
1. expresses more B7
2. secretes cytokines, such as IL-12
Term
When CD40L on an antigen-stimulated T cell binds to CD40 on an APC, the APC releases IL-12. What does IL-12 do to the T cell?
Definition
enhances T cell differentiation
Term
LFA-1 on a T cell binds to ________ on an APC
Definition
Intercellular Adhesion molecule-1 (ICAM-1)
Term
ICAM-1 on an APC binds to _______ on a T cell
Definition
LFA-1
Term
Injured ednothelial cells produce chemokines to attract T cells. What would some of these chemokines do to VLA-4?
Definition
cause it to enter a high-affinity state so that it binds better with VCAM-1 on the endothelial cell
Term
What does VLA-4 on T cells bind to?
Definition
VCAM-1 on endothelial cells
Term
The combination of TCR complex, MHC/peptide, & co-stimulators is called the _________ ___________
Definition
immunologic synapse
Term
What do CD3 & zeta chain do?
Definition
They help TCR transduce signal by becoming ITAMs (activation molecules) for the T cell when the TCR binds to a peptide/MHC complex
Term
What 3 membrane molecules do CD4+ Helper T cells produce after activation?
Definition
1. CD40L--binds to CD40 receptor on APC & causes it to express more IL-12 (stimulates T cell to differentiate) & 2. B7 (costimulator—-for TCR on other T cells)
3. IL-2R (IL-2 Receptor)--IL-2, which stimulates T cell growth
Term
After activation, what 2 cytokines do TH1 CD4+ Helper T cells make?
Definition
1.TFN-gamma--has many immune properties; activates macrophages
2. IL-2--stimulates growth of T cells
Term
Does activated TH1 or TH2 make IL-4 (which activated B cells)?
Definition
TH2
Term
Does activated TH1 or TH2 make IL-2 (which tells T cells to grow) & IFN-gamma (which has many actions, including activating macrophages)?
Definition
TH1 cells
Term
interleukin that tells T cells to grow
Definition
IL-2
Term
Cytokine that has many immune actions, including activating macrophages
Definition
IFN-gamma
Term
Immune system cytokines tend to be pleiotropic & redundant. What does that mean?
Definition
pleiotropic--one cytokine has multiple functions
redundant--multiple cytokines cover one function
Term
Naive T cells express a lot of ______ & a little bit of _____ immunoglobulin.
Definition
IgM
IgD
Term
Naive B & T cells are ONLY activated in:
Definition
peripheral lymphoid organs
Term
Why are T cells only activated in peripheral lymphoid organs? Why are B cells?
Definition
T cells need dendritic cells to activate & dendritic cells go to peripheral lymphoid organs when they find antigen
B cells need a high concentration of antigen to activate (also, need a survival signal from Follicular Dendritic Cells or T cells to survive)
Term
Only ________ express enough MHC, co-stimulatory molecules, & cytokines (such as IL-12) to activate a T cell.
Definition
dendritic cells
Term
Memory T cells rely on the cytokine _______ to help them survive & remind them to divide from time to time.
Definition
IL-7
Term
Which are easier to active: naive or memory cells?
Definition
memory are easier to activate (can be activated by plain antigen; doesn't need to be in lymph node)
Term
Which type of T cell mediates immunity to parasites & allergic reactions?
Definition
TH2
Term
Which type of T cell is responsible for alternative macrophage activation?
Definition
TH2
Term
TH-17 produces the cytokine ____, which mediates inflammation & is a powerful recruiter of neutrophils & monocytes.
Definition
IL-17
Term
Which type of T cell stimulates phagocyte-mediated ingestion (opsonization) and killing of microbes; activates B cells & macrophages?
Definition
TH1
Term
What are the 3 types of helper T cells?
Definition
TH1, TH2, TH17
Term
Which type of T cell makes IFN-gamma?
Definition
TH1
Term
IL-12 favors differentiation of naive CD4+ T cells into _______.
Definition
TH1
Term
Cytokines _____ & ______ favors differentiation of naive CD4+ T cells into TH1.
Definition
IL-12
IFN-gamma
Term
After dendritic cells & macrophages encounter a pamp, they produce cytokine ________, which activates the _______ pathway, which favors differentiation of naïve CD4+ T cells into ______ cells.
Definition
IL-12
STAT4
TH1
Term
NK cells that have encountered a PAMP & activated TH1 cells produce _________, which activates the ________ signaling pathway, which induces expression of a transcription factor called ___________.
Definition
IFN-gamma
STAT1
T-bet
Term
IFN-γ & CD40/CD40L reactions promote B cell isotype switch to:
Definition
IgG1 & IgG3
Term
After the Fab regions of IgG1 & IgG3 bind to a microbe, the Fc regions bind to ____ on phagocytes & tell it to eat the microbe
Definition

FcγRI

 

(FcγR's bind to IgG; FcεR's bind to IgE; Fcα/μR binds to IgA and IgM)

Term
When CD40L on T cells binds to CD40 on APCs (such as macrophages or B cells), this stimulates the T cell to produce more _____ ctyokine
Definition
IFN-gamma
Term
Cytokine ________ + reaction between _________ on T cells & ______ on B cells causes B cell to switch to IgG1 & IgG3
Definition
IFN-gamma
CD40L on T cells
CD40 on B cells
Term
Complement receptor on phagocytes that works with the classical complement pathway to induce phagocytosis
Definition
CR1
Term
_____ cytokine is the most potent activator of macrophages
Definition
IFN-gamma
Term
What are some things that activated macrophages produce or express more of?
Definition
Produces more NO (Nitric oxide)
Produces more ROS (Reactive Oxygen species)
Expresses more MHCII
Expresses more co-stimulatory molecules (B7)
Produces more cytokines (TNF- α, IL-1, IL-6)
Term
TH1 cells produce _______, which encourages naive T cells to turn into TH1 (positive feedback loop)
Definition
IFN-gamma
Term
What cytokine is the major autocrine/paracrine proliferation factor for T cells?
Definition
IL-2
Term
TH1 cells produce ____ & ____, which help activate naive CTLs & B cells.
Definition
IFN-gamma
IL-2
Term
IL-2 binds to _______ (receptors) on T cells
Definition
IL-2R (no kidding!)
Term
TH1 expresses ________, which converts IL-2R (IL-2 receptors) on other T cells from low to high affinity so they react better with IL-2
Definition
IL-2Ra chain
(IL-2Ra activates IL-2R)
Term
TH1 cells express IL-2Ra chain, which:
Definition
converts IL-2R (IL-2 receptors) on other T cells from low to high affinity so they react better with IL-2 (and proliferate more)
Term
What are two other names for Type IV hyper-sensitivity?
Definition
delayed-type hypersensitivity
cell-madiated hypesensitivity (b/c all of the other types are mediated by antibodies)
Term
You have to go back days later to get a TB skin test b/c it's a type ______ hypersensitivity reaction
Definition
IV (aka dalayed or cell-mediated hypersensitivity)
Term
Which type of hypersensitivity is particularly useful against intracellular parasites?
Definition
Type IV
Term
Type _____ hypersensitivity is due to tissue damage by macrophages releasing toxic products (like ROS), CTLs killing infected cells, & neutrophils actions
Definition
IV
Term
What is the role of CD4+ TH1 cells in Type IV hypersensitivity?
Definition
TH1 cells release IFN-gamma, which activates macrophages
Term
Compare the types of intracellular microbes eliminated by CD4+ TH1 cells and CD8+ cytotoxic T cells (CTLs)
Definition
CD4+ TH1 cells help macrophages kill cells that infect MACROPHAGES
CD8+ CTLs kill infected cells that are expressing the CTL's antigen
Term
_____ cells produce ____, which helps macrophages kill cells that they have phagocytosed, but can't kill on their own.
Definition
CD4+ TH1 cells
IFN-gamma
Term
When a TCR on a CD8+ CTL binds to a peptide/MHC I complex on an infected cell, the LFA-I adhesion receptor on the CTL binds to _______ on the infected cell
Definition
ICAM-1(Intercellular Adhesion Molecule-1)
(The is the same in CTLs as for other T cells)
Term
When a TCR on a CD8+ CTL binds to a peptide/MHC I complex on an infected cell, the ______ adhesion receptor on the CTL binds to InterCellular Adhesion Molecule 1 (ICAM-1)on the infected cell
Definition
LFA-1
Term
After a CD8+ CTL synapses with an infected cell, it releases perforins & granzymes. What do those 2 things do?
Definition
perforins--inserts into cell membrane & makes holes (like MAC)
granzymes--enter holes & activate capsase-3 in cytoplasm, causing apoptosis
Term
Granzymes enter holes created by perforin & & activate ______ in cytoplasm, causing apoptosis
Definition
capsase-3
(This is how CTLs kill infected cells)
Term
Minimum Inhibitory Concentration (MIC)
Definition
the weakest concentration of antibiotic that stops bacteria from growing (ex., 2%). Dose used in therapy should give body fluid drug concentration > MIC
Term
Minimum Bactericidal Concentration (MBC)
Definition
the weakest concentration of antibiotic that kills 99.9% of bacteria
Term
Extended-spectrum antibiotics work on:
Definition
nearly all gram-positive bacteria & a few gram-negative
Mnemonic: eXtended for + (cross, cross)
Term
infection following treatment of a previous infection; more likely if a broad-spectrum antibiotic is given
Definition
super-infection
Term
How do broad-spectrum antibiotics cause super-infections?
Definition
o Broad-spectrum antibiotics can kill normal bacterial flora, leaving the way for antibiotic-resistant microbes (such as candida albicans) to take over
Term
________ _______ (component of complex lipids in cell membranes) is turned into prostaglandin in the presence of COX-1 & COX-2 proteins
Definition
arachidonic acid
Term
Arachidonic acid(acomponent of complex lipids in cell membranes) is turned into prostaglandin in the presence of _____ & ______ proteins
Definition
COX-1 & COX-2
(cyclooxygenase-1 & cyclooxygenase-2)
Term
List the 4 NSAIDs (Non-steroidal Anti-Inflammatory Drugs)
Definition
1. aspirin
2. ibuprofin
3. acetominophin
4. indomethacin
Term
How is aspirin different from the other NSAIDs?
Definition
it IRREVERSIBLY inactivates COX-1 & COX-2, while the others (ibuprofin, acetominophin, indomethacin) irreversibly inactivate it.
Term
How is acetominophin different from the other NSAIDs?
Definition
it is not anti-inflammatory
(no one knows why)
Term
The 4 NSAIDs are aspriin, ibuprofen, acetominophen, & ______
Definition
Indomethacin
Term
NSAIDs block production of prostaglandins, which has what 3 desired effects on the body?
Definition
1. analgesic (blocks pain)
2. anti-pyretic
3. anti-inflammatory (except for acetominophen; no one knows why)
Term
Is cortisol's receptor inside the cell or outside?
Definition
Inside. Cortisol is in the steroid/thyroid family, which is cell membrane permeable
Term
Estradiol binds to a receptor protein in/on the _________.
Definition
the cytosol
Estradiol is in the thyroid/steroid family of signaling molecules, which are membrane permeable
Term
After testosterone binds to its receptor protein, it moves to:
Definition
inside the nucleus
Testosterone is in the steroid/thyroid family of signaling molecules, which are membrane permeable.
Term
After thyroxine binds to its receptor protein in the cytosol, the two move to the place of action &:
Definition
activate transcription of the target gene
Thyroxine is a member of the thyroid/steroid family of signaling molecuels, which are membrane permeable.
Term
Cortisol, estradiol, testosterone, & thyroxine are members of the membrane-permeable ______/________ family of signaling molecules.
Definition
steroid/thyroid
Term
2N + O2 --(__________)-->NO
Definition
Nitrous Oxide Synthase (NOS)
Term
For vasodilation, nerves terminating at _________ cells of the blood vessel stimulate _________ enzyme to produce NO inside the cell.
Definition
endothelial
NO
Term
NO rapidly diffuses out of the endothelial cell where NOS (Nitrous Oxide Synthase Produced it) & goes:
Definition
into neighboring smooth muscle cells
Term
Inside a smooth muscle cell, NO binds to & activate ____________________
Definition
guanylyl cyclase
Term
Inside a smooth muscle cell,
______ --(gyanylyl cyclase + NO)-->________
Definition
GTP (guanosine triphosphate)
cGMP (cyclic guanosine monophosophate)
Term
Inside a smooth muscle cell,
______ --(gyanylyl cyclase + NO)-->________
Definition
GTP (guanosine triphosphate)
cGMP (cyclic guanosine monophosophate)
Term
Inside a smooth muscle cell,
GTP --(_______________ + __)--> CGMP
Definition
guanylyl cyclase
NO
Term
No is the first messenger of smooth muscle relaxation. _________ is the second messenger.
Definition
cGMP
Term
Generic name for Viagra
Definition
sildenafil
Term
Normally, ___________ ends vasodilation by degrading cGMP, ending the signal for the smooth muscle to relax.
Definition
PhosphoDiEsterase (PDE5)
Term
Normally, phosphodiesterase (PDE5) ends vasodilation by degrading ____, ending the signal for the smooth muscle to relax.
Definition
cGMP
Term
How does sildenafil improve an erection?
Definition
Sildenafil interfers with the action of phosphodiesterase (PDE5) in degrading cGMP. This allows cGMP to continue to cause smooth muscles of the blood vessels to relax, continuing vasodilation.
Term
Nitroglycerin increases synthesis of ______, which allows blood vessels to the heart to vasodilate & send more blood to the heart.
Definition
NO
(no one knows how nitroglycerin does this)
Term

Nisoglycerin causes vaso____(dilation/constriction) by increasing synthesis of ________, and is used to treat angina. 

 

Definition

vasodilation

Nitric Oxide (NO)

Term
The cell normallly has a slightly _____ charge relative the the extracellular fluid.
Definition
negative
Term
What does the nictoinic acetylcholine receptor due when acetylcholine binds to it?
Definition
the receptor is a ligand-gated channel, which opens & allows cations to flood in. This decreases the cell's normally slightly negative charge relative to the extracellular fluid (membrane resting potential), which causes the membrane to depolarize & the muscle to contract.
Term
When acetylcholine binds to nicotinic acetylcholine receptor (a ligand-gated ion channel) _____ (cations/anions) flow into the cell and depolarize the membrane (causing muscle contraction).
Definition
cations
Term
What ultimately happens (what is the ultimate goal) when acetylcholine binds to a nictoinic acetylcholine receptor.
Definition
the muscle contracts
(acetylcholine is a ligand-gated ion channel that allows cations to flood into the cell, depolarizing it. This ultimately causes muscle contraction).
Term
The G-Protein Coupled Receptor (GPCR) passes through the membrane _____ times. Each trans-membrane region is a _______ _______
Definition
7
alpha helix
Term
G-Protein Coupled Receptor (GPCR) is associated with G protein, which has 3 subunits: _______ & ______ subunits are attached to the membrane by lipid tails & ______ subunit stays at the plasma membrane because it is associated with the other two subunits.
Definition

alpha, gamma

 

beta

Term
In the un-stimulated state of G protein, the alpha subunit holds a _____.
Definition
GDP
Term
In the un-stimulated state of G protein, the _____ subunit holds a GDP.
Definition

alpha

 

G proteins are made up of alpha (α), beta (β), and gamma (γ) subunits. α and γ are attached to the cell membrane by lipid tails the β subunit stays at the plasma membrane because it is chemically attracted to the other two subunits.  In the unstimulated state, the α holds a GDP.  When a signal molecule binds to the G-Protein Coupled Receptor (GPCR), it lets go of the GDP and picks up a GTP, which activates the α subunit.  (What happens next depends on what kind of G protein).  Eventually:

 

GTP—(alpha subunit of G protein + GTPaseActivating Proteins, GAPs)-->GDP + P

 

And the α subunit is inactivated.  (Alpha subunit has some GTPase activity on its own, but has more with GAPs)

Term
When a signal molecule binds to the G-Protein Coupled Receptor (GPCR), the alpha subunit of the G protein lets go of its ____ & picks up a ____, which activates the alpha subunit
Definition
GDP
GTP
Term
The alpha unit of a G protein is inactivated when:
Definition
GTP is phosphorylated into GDP
Term
GTP—(alpha subunit of G protein + ____)-->GDP + P
Definition
GTPase Activating Proteins (GAPs)
(Alpha subunit has some GTPase activity on its own, but has more with GAPs )
Term
What does the G protein Gs do?
Definition
activate adenylyl cyclase
ATP –(activated adenylyl cyclase)-->cAMP
Term
ATP –(activated ______ _______)-->cAMP
Definition
adenylyl cyclase
Term
What does the G protein Gi do?
Definition
activate adenylyl cyclase
ATP –(activated adenylyl cyclase)-->cAMP
Term
In the Protein Kinase A (PKA) pathway:
Gs activates adenylyl cyclase (Gi inhibits adenylyl cyclase)
ATP –(activated adenylyl cyclase)-->cAMP
What does cAMP do?
Definition
activates Protein Kinase A, (PKA) by turning off its 2 inhibitory regulatory subunits
Term
In the Protein Kinase A (PKA) pathway:
Gs activates adenylyl cyclase (Gi inhibits adenylyl cyclase)
ATP –(activated adenylyl cyclase)-->cAMP
What does cAMP do?
Definition
activates Protein Kinase A, (PKA) by turning off its 2 inhibitory regulatory subunits
Term
_______ (cGMP/cAMP) causes smooth muscles to relax. ______ (cGMP/cAMP) activates Protein Kinase A (PKA), allowing it to phosphorylate serines & thyrines on certain proteins & change the function of those proteins
Definition
cGMP
cAMP
(PKAAAAA is dependent on cAAAAAAMP)
Term
Protein Kinase A (PKA) is a tetramer. What do its 3 subunits do?
Definition
1 subunit catalyzes phosphorylation of specific serines & threonines on specific intracellular proteins (which proteins varies by cell type), altering the protein’s activity
The other 2 subunits inhibit the catalase unless turned off by cAMP.
Term
PKA catalyzes phosphorylation of specific ____ & ____ (amino acids) on specific intracellular proteins (which proteins varies by cell type), altering the protein’s activity
Definition

serine (side chain CH2OH)

 

threonine (side chain CHOHCH3)

 

Serine and threonine have similar polar uncharged side chains, consisting of carbons, hydrogens, and a hydroxyl group (-OH).  Serine/threonine-specific protein kinases, such as PKA, phosphorylate (add a phosphate group, PO4, to) the OH group of serine or threonine 

Term
cAMP activates Protein Kinase A (PKA). What quickly breaks down cAMP, inactivating PKA?
Definition
cAMP phosphodiesterase
Term
inositol phospholipid—( activated _____________)-->inositol 1,4,5-triphosphate (IP3) + diaglyerol (DAG)
Definition
phospholipase C
Term
inositol phospholipid—( activated phospholipase C)-->inositol 1,4,5-triphosphate (IP3) + diaglyerol (DAG)

What happens to IP3 & DAG?
Definition
DAG stays attached to the plasma membrane
IP3 floats off into the cytosol and opens opens Ca2+ ion channels in the endoplasmic reticulum, allowing Ca2+ flows out of ER storage into cytosol
Term
IP3 allows ______ ion to flow from ___________ to ____________
Definition
Ca2+
inside the endoplasmic reticulum
the cytosol
Term
Normally Ca2+ is kept at ______ (high/low) levels in the cytosol.
Definition
low (very low)
Term
What happens when a Receptor Tyrosine Kinase (RTK) binds to its ligand?
Definition
It gets together with another RTK & they will phosphorylate each other’s intracellular receptor tails. Then a whole bunch of proteins attach to the phosphorylated tails (including, in some pathways, Ras).
Term
The largest class of enzyme-coupled cell surface receptors, including receptors for insulin, insulin-like growth factors (IGFs), epidermal growth factor (EGF), Nerve Growth Factor (NGF), platelet-derived growth factors, macrophage-colony-stimulating factor, fibroblast growth factors, vascular endothelial growth factors, ephrins, etc.
Definition
Receptor Tyrosine Kinase (RTK)
Term
Ras stimulates ____________, which stimulates ___________, which stimulates _____________, which stimulates transcription factors & other proteins.
Definition
MAP Kinase Kinase Kinase
MAP Kinase Kinase
MAP Kinase
Term
Interleukin ____ favors T cell maturation as a TH2
Definition
IL-4
Term
IL-4 activates _____, which induces ________, which encourages a T cell to mature into a TH2 cells
Definition
STAT 6
GATA-3
Term
T cells that provide immunity to helminths, cause allergies, cause alternative macrophage activation, & produce neutralizing antibodies (IgG4).
Definition
TH2
Term
Neutralizing antibodies produced by TH2
Definition
IgG4
Term
TH2 produces cytokines ____ & ____, which favor B cell isotype switching to IgE
Definition
IL-4 & IL-13
Term
____ , produced by TH2 cells, encourages B cells to switch to IgG4 isotype.
Definition
IL-4
(without IL-13, IL-4 + IL-13 encourages IgE)
Term
_____, produced by TH2 cells, activates eosinophils & encourages them to degranulate against helminths.
Definition
IL-5
Term
What 3 cytokines, produced by TH2, encourage macrophages to undergo alternative maturation & become M2 cells?
Definition
IL-4, IL-10, IL-13
(IFN-gamma encourages M1)
Term
IL-4, IL-10, & IL-13 encourage macrophages to:
Definition
undergo alternative activation & become M2 cells
(IFN-gamma encourages M1)
Term
________ encourages macrophages to become M1 cells.
Definition
IFN-gamma
Term
_____ macrophages are primarily concerned with inflammation, and _____ with healing.
Definition
M1
M2
Term
____ cells produce _____, which encourages B cells to switch to IgG1 & IgG3 isotypes.
Definition
TH1
IFN-gamma
(IFN-γ also stimulates macrophages to phagocytize the opsonized microbe)
Term
IFN-gamma from TH1 cells encourages B cells to switch to IgG1 & IgG3 isotypes, and helps IgG1 & IgG3 work better by:
Definition
encouraging macrophages to phagocytize microbes opsonized with IgG1 & IgG3
Term
The Fc regions of IgG1 & IgG3 binds to ______ on neutrophils & macropahges
Definition
FcγRI
Term
The complex of Fc regions (from IgG1 & IgG3 antibodies) & FcγRI activates phagocytes to produce what kinds of chemicals in their lysosomes?
Definition
Reactive Oxygen Species (ROS), (large amounts)
NO
Proteolytic enzymes
Term
What often must happen before a phagocyt can phagocytose an encapsulated microbe (such as pneumococci)?
Definition
the microbe must be opsonized with antibody (otherwise, phagocytes have trouble grabbing on to microbe)
Term
FcγRs (gamma) bind to ____ antibodies
FcεRs (epsilon) bind to ______ antibodies
Definition
IgG
IgE
for an Fc receptor named FcxRI (or FcxRII, or FcxRIII), the x will be a Greek letter that starts with the same letter as the antibody whose Fc region the receptor binds to
Term
Fc binding sites on antibodies are inactive until
Definition
the Fab portion meets antigen
Term
Which antibody has the longest half-life, and why?
Definition
IgG, b/c neonatal FcR (FcRn), found in placenta, endothelium, & a few other cells, recycles it rather than digesting it in lysosome
Term
What class of antibodies neutralizes microbes & toxins, opsonizes antigens, activates classical complement pathway, and provides negative feedback to B cell activation?
Definition
IgG
Term
IgG provides ________ (negative/positive) feedback for B cell activation.
Definition

negative

 

(When the Fc portion of an IgG antibody interacts with the Fc receptor FcR gamma on the B cell, the B cell produces suppresive B cell factor.  This keeps the B cell from just going nuts and taking over the world after it's awakened.)

Term
Which type of antibodies provides negative feedback for B cell activation?
Definition
IgG
Term
Antibody responsible for mucosal immunity & neutralization of microbes & toxins
Definition
IgA
Term
Antibody responsible for mucosal immunity & neutralization of microbes & toxins
Definition
IgA
Term
Which antibody encourages mast cell degranulation (immediate hypersensitivity reactions)?
Definition
IgE
Term
a receptor on NK cells that binds to arrays of IgG antibodies attached to (opsonizing) a cell, causing the NK cell to degranulate & release proteins that kill opsonized targets
Definition
FcγRIII
Term
What happens when the FcγRIII binds to an array of IgG antibodies attached to a cell?
Definition
The Natural Killer cell degranulates, releasing proteins that kill opsonized targets
(FcγRIII is on Natural Killers cells)
Term
Describe Antibody-Dependent Cellular Cytotoxicity (ADCC)
Definition
FcγRIII on Natural Killer cells binds to an array of IgG antibodies attached to a cell, causing the NK cell to degranulate, releasing proteins that kill opsonized targets
Term
Where is FcγRIII & what does it do?
Definition
It’s on Natural Killer cells. FcγRIII on Natural Killer cells binds to an array of IgG antibodies attached to a cell, causing the NK cell to degranulate, releasing proteins that kill opsonized target cells
Term
A receptor on Natural Killer cells that binds to an array of IgG antibodies attached to a cell, causing the NK cell to degranulate, releasing proteins that kill opsonized target
Definition
FcγRIII
Term
Why are helminths tough for the body to kill?
Definition
Too large to be phagocytosed
Thick integument is resistant to many microbicidal substances produced by neutrophils & macrophages
Term
On what 2 types of cells is FcεRI expressed?
Definition
Eosinophils & mast cells
Term
How does IgE cause eosinophils to degranulate against helminths?
Definition
IgE binds to helminths. FcεRI on eosinophil binds to IgE & causes eosinophil to degranulate (This process probably also involves cytokines form nearby T cells)
Term
What happens when FcεRI on mast cells binds to IgE on helminths?
Definition
The mast cells secrete cytokines that call in more help (more white blood cells)
Term
Granules from ________ cells contain proteins that can kill helminths
Definition
Eosinophil
Term
Eosinophil reacts with IgE bound to a helminth via the ____ receptor
Definition
FcεRI
Term
Process by which mast cells are coated with IgE antibody specific for the antigen(s) to which the person is allergic
Definition
Sensitization
Term
Why don’t you have an allergic reaction the first time you meet an allergen to which you are predisposed toward allergy?
Definition
The first time you meet an allergic (for you) allergen, B cells produce IgE against it. Mast cells are coated with this IgE. When mast cells coated with enough of that type of IgE meet enough allergen, the mast cell degranulates.
Term
Where in the body will you find sensitized mast cells?
Definition
In connective tissue where you would find the allergen. Mast cells sensitized to food allergens are in connective tissue of the gut. Mast cells sensitized to respiratory allergens are in connective tissue of the respiratory system.
Term
What kind of hypersensitivity is caused by degranulation of sensitized mast cells?
Definition
Type I Hypersensitivity
Term
When exposed to a stressful event, what is the first physiological reaction of the body to the situation?
Definition
activation of the sympatho-adrenomedullary axis (SAM axis), aka sympathetic nerve system
Term
What 2 axes deal with stress?
Definition
sympatho-adrenomedullary axis (SAM axis), aka sympathetic nerve system

hypothalamic-pituitary-adrenal axis (HPA or HTPA axis)
Term
What kind of stress temporarily (for a few hours) ramps up the immune system by mobilizing leukocytes (neutrophils, macrophages, NK cells, B cells, T cells, etc.) to skin & other likely-to-be-wounded parts of body
Definition
acute stress (<2 hours)
Term
What is the role of glucocorticoids (cortisol) when an individual is not under stress?
Definition
• Preparing the body for future stress exposure by storing energy & promoting memory formation of previous stressors
Term
Chronic stress causes _____ (increased/decreased) release of cortisol in response to acute stress, _____ (increased/decreased) circadian variations, & _______ of the negative feedback loop for stress response.
Definition
increased
decreased
impairment
Term
What are the 5 top stressors for adult on the Holmes-Rahe scale?
Definition
1. death of spouse
2. divorce
3. marital separation
4. jail term
5. death of close family member
Term
If you add the numerical value of all of a person's stressors together, you receive a stress score that can tell you how likely they are to become ill:
Score of ___: High risk of illness.
Score of ___: Moderate risk of illness
Score ___: Slight risk of illness.
Definition
>=300
150-299
<=150
Term
Does chronic stress increase circulating CRH (from the hypothalamus) & ACTH (from the pituitary) levels?
Definition
No, it doesn't increase circulating CRH & ACTH
Term
The antigen for T-dependent immune response is
Definition
protein
Term
The antigen for T-independent immune response is:
Definition
polysaccharide, primarily
(can be any other non-protein, such as lipids & DNA)
Term
B cells are activated via T-dependent immune response in the
Definition
lymphoid follicles (in lymph nodes)
Term
B cells are activated via T-independent immune response in:
Definition
the marginal zones of the white pulp of the spleen
Term
What type of B cells only ever produce low-affinity IgM antibodies
Definition
marginal zone B cells (activated in marginal zone of spleen via T-independent immune response)
Term
PeriArteriolar Lymphoid Sheaths (PALS)
Definition
sheaths of T cells that live around spleen arterioles
Term
Marginal zone B cells can be activated by ONE polysaccharide (instead of needing a bunch of free-floating antigen or an array of antigen presented by a folllicular dendritic cell) b/c...
Definition
• Polysaccharide antigens tend to have long units of repeating epitopes. So one antigen can cross-link several B Cells Receptors (BCRs) on a marginal zone B cell, which activates the B cell
Term
The T-independent response is not well developed in
Definition
infants
Term
_____ turns a T-independent antigen into T-dependent antigen
Definition
conjugation (with a protein)
Term
What does this mnemonic mean?
Type TWO is against YOU
Definition
Type II Hypersensitivity is auto-immune response (and transplant/transfusion response)
Term
What type of hypersensitivity causes auto-immune response and transplant/transfusion response.
Definition
Type 2
(type TWO is against YOU)
Term
Type 2 Hypersensitivity (auto-immune & transfusion/transplant) is mediated by which antibodies?
Definition
IgG1 & IgG3
Term
Phagocytes can be activated by compliment protein ____ or ____ binding to receptor ____.
Definition
C3b
C4b
CR1
Term
In Type II Hypersensitivity, phagocytes can't phagocytyze the offending things because ___________________, so the phagocytes _______________________
Definition
self cells & tissues are too big to phagocytyse

phagocytes release enzymes & Reactive Oxygen Species (ROS) & cause tissue injury
Term
Autoimmune disease in which autoantibody reacts to acetylcholine receptors on muscle cells, causing paralysis
Definition
myasthenia gravis
Term
A type of hypersensitivity caused by reaction to incompletely cleared immune complexes (complexes of antigen/antibody/complement)
Definition
type 3
Term
hapten
Definition
a small chemical that can bind to antibody, but can’t elicit T cell-dependent immunity
Term
In conjugated vaccines, the B cell makes vaccines against the _____, but the response is T cell-dependent because the T cell responds to the ____
Definition
hapten (non-protein antigen)
carrier (protein chemically attached to the hapten)

so you get antibodies against a non-protein antigen, but you also get class switching, affinity maturation, & memory
Term
What type of vaccine is the tetanus vaccine?
Definition
toxoid
Term
DTaP (tetanus, diphtheria, pertussis) vaccine is a subunit vaccine that contains 2 _____ & a ______
Definition
toxoids (for tetanus & diptheria)
pertussis protein
Term
What chemical is the most common vaccine adjuvant?
Definition
alum
Term
antibodies from the same B cell lineage (come from one B cell), which all bind to the same epitope on the antigen; made in a lab or by cancerous B cells
Definition
monoclonal antibodies
Term
antibodies from different B cell lineages, which bind to different epitopes on the antigen; all normal immune responses are of this type
Definition
polyclonal
(monoclonal are only produced in labs or by cancerous B cells)
Term
What test is used to differentiate Staphylococcus aureus from staphylococcus epidermidis?
Definition
coagulase test
s. aureus is coagulase +
s. epidermidis is coagulase neg
Term
What test is used to differentiate beta-hemolytic strep (ex., S. pyogenes) from beta-hemolytic staph (ex., S. aureus)?
Definition
resistance to bacitracine
beta-hemolytic staph are resistant
beta-hemolytic strep are not
Term
Both botulinum toxin & tetanus toxin work by ___________, but:
Definition
blocking neurotransmitter release
but botulinum blocks release of acetylcholine (causes flaccid paralysis)
& tetanus blocks release of inhibitory neurotransmitters (causes prolonged, severe spasms)
Term
Golden colonies that look like an egg yolk.
Definition
Staph aureus
Term
Among human flora/pathogens, staphylococcus is uniquely resistant to ________
Definition
salt
(that's why it can live on skin)
Term
What bacterial genus do tetanus, botulism, & perfringens belong to?
Definition
clostridium
Term
angina 
Definition
heart pain due to insufficient blood flow to the heart
Term
opsonin
Definition

a molecule that coats an invader and enhances phagocytosis

Term
What is phosphorylation?
Definition
add a phosphate group, PO4, to a protein or other organic molecule
Term

What kind of junction forms epithelial cells into waterproof sheets?


What kind is a continous belt of cadherins (like coke can rings)?


What kind consists of "Spot welds" of cadherins between epithelial cells?


What kind of junction sticks the basal domain of an epithelial cell to the basement membrane?

Definition

tight junctions (zonula occludens)  

zonula adherens 
spot desomosomes (maculae adherens)

hemidesmosome 

Term

Explain the following mnemonic:

 

EFFErent-->EFFEctor

Definition

An efferent pathway is the pathway away from the integration center toward the effector (ex., from the brain to the muscles, or from the nucleus of a cell to a molecule on the cell surface)

 

the afferent pathway is the pathway from the receptor toward the integration center (ex., from the eyes to the brain, or from a receptor on the cell surface to the nucleus)

Term

Osmolarity 

Definition

the measure of solute concentration, defined as the number of osmoles (Osm) of solute per litre (L) of solution (osmol/L or Osm/L). 

 

1 osmole = 1 mole of osmotically active particles

1 mole NaCl = (1 osmole Na+)  +  (1 osmole Cl-) = 2 osmoles NaCl 

Term

osmole

Definition

1 osmole = 1 mole of osmotically active particles

1 mole NaCl = (1 osmole Na+)  +  (1 osmole Cl-) = 2 osmoles NaCl 

Term
What's the difference between wet and dry gangrene?
Definition

gangrene is coagulative necrosis of the limbs

 

coagulative necrosis with some liquefaction due to infection is "wet gangrene"

Term
ketone bodies
Definition

Three water-soluble compounds produced as by-products when fatty acids are broken down for energy in the liver and kidney.  

Supporting users have an ad free experience!