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

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/422

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

422 Cards in this Set

  • Front
  • Back
Chapter 3
Chapter 3
3 challenges of free living unicellular organisms
nutrition, occupancy, resistance
gram positive cell wall
thick, contains murein/peptidoglycan
components of murein
sugar and amino acid polymer
function of rigidity of murein coat
allows organism to survive in hypoosmotic media
lysozyme
murein hydrolytic enzyme found in human and animal tissues, causes bacteria to lyse in low osmotic pressure environments or to become spheroplasts in iso-osmotic media
gram stain procedure
1. stain with crystal violet 2. modify with potassium iodide 3. wash with alcohol, only gram positive will remain purple. 4. counterstain with safranin which causes gram negatives to become pink
acid fast stain procedure
1. stain with hot carbol fuchsin (red) 2. decolarize with acid alcohol, only acid fast remain red 3. counterstain with methylene blue
describe the structure of murein
alternating units of N-acetylmuramic acid and N-acetylglucosamine with crosslinking peptides resulting in a fence like structure
3 components of LPS
lipid A, core, and O antigen
Lipid A
anchors LPS in outer leaflet of gram negative membrane
components of LPS core
short series of sugars (ketodeoxyoctanoic acid and a heptose)
O antigen
long carbohydrate chain that covers bacteria and protects it from hydrophobic compounds
how are hydrophilllic compounds transported through gram negative cell wall
via porins
periplasm
space between the double plasma membranes in gram negative organisms
contents of periplasm
gel like solution containing murein and various enzymes to reduce large molecules to diestible size and protect against bacteriophages and antibiotics
effect of lipid A on body
also known as endotoxin, results in fever and mobilization of host defense, if present in high enough amounts can cause shock
explain the advantages of acid fast solution
relatively impervious to envionmental agents but growth is very slow because the waxy membrane makes for slow uptake of nutrients
antibiotic classes that block synthesis of murein
beta lactams (penicillin, cephlosporins, carbapenems)
describe the synthesis of murein
chain of sugars alternating between N-acetylglucosamine and N-acetylmuramic acid (which has a peptide chain attached to it). The units are transferred from cytoplasm to periplasm by a lipid carrier
MOA of vancomycin
prevention of the addition of disaccride (NAM-NAG) to growing chain of murein
inhibits regeneration of the lipid carrier for murein
bacitracin
final reaction of murein synthesis
transpeptidation to link the long chains of disacchrides (cross linking)
what forms the cross link in murein
the subterminal D-alanine of one chain joining with a lysine or diaminopimelic acid on the other chain
what antibiotics inhibit the cross linking reaction
penacillins and cephalosporins
how do antibiotics that inhibit murein synthesis kill bacteria
via lysis, caused by disruption in murein synthesis. This disruption results in overgrowth of the cytoplasm
autolysin and penicillin tolerance
bacteria tolerant to penicillin are deficient in an autolysin which cleaves murein,
facilitate the entry of most metabolites into bacteria
permeases
group translocation
phosphorylation linked transport used to bring in certain sugars such as glucose, resulting in the transported substance being chemicaly altered in the process
proton motive force
protons expelled from bacteria during oxidation of metabolites creates a gradient that can be utilized to bring in energetically unfavorable molecules such as lactose via symport
how do organisms within the body take up iron
excretion of chelating compounds called siderophores which bind free iron and can be specifically be taken up by the organism
nucleoid
irregular coiled DNA structure found in bacteria
DNA gyrase
introduces supercoils into circular DNA
topoisomerase 1
relaxes supercoils by introducing single strand nicks in the DNA
brief description of bacterial DNA replication
bidirectionally at a precice replicative origin that requires precice unwining and rotation of supercoiled nucleoid
metronidazole is active in what form
partially reduced
what organisms are metronidazole most effective in, why
anaerobes and amoebas, they partially reduce the molecule which is the active form
MOA of metronidazole
partially reduced form incorporates into DNA of bacteria and causes it to become unstable
nalidixic acid
inhibits DNA gyrase
MOA of fluoroquinolones
interfere with DNA gyrase and topoisomerase to cause dsDNA breaks
antibiotic that inhibits bacterial transcription at initiation step
rifampin
principal biosynthetic activity of rapidly growing bacteria
protein synthesis
RNA is made in bacteria at a rate proportional to _________________
number of RNA polymerase molecules
protein synthesis in bacteria occurs at rate proportional to _____________ ; what does this tell us
number of ribosomes, that regulation occurs with initiation of protein synthesis and not the actual speed of chain elongation
MOA of macrolides
block formation of peptide bonds by binding near.at the tRNA binding site on the 50s subunits, this prevents chain elongation
are macrolides bacteriostatic or bacteriocial, why?
bacteriostatic, eventually the effected portions of the ribosome are hydrolyzed and the remaining subunits are able to join another mRNA molecule
what type of patient would not respond well to macrolides, why
immunocomprimised patients cant clear bacteria effectively, meaning the macrolides would halt the growth but the immune system wouldn’t finish the job
bactericidal protein synthesis inhibitor antibiotic class
aminoglycosides (the -mycins)
where do aminoglycosides bind
to the smaller 30s unit
MOA of aminoglycosides
bind to 30s subunit and cause increased 30/50s interaction leading to accuulation of 70s particles, it also prevents elongation of peptide chains. The aminoglycoside molecules cannot be removed from the bacteria and result in death of the molecule

slimy outer coating made by certain bacteria

capsule

2 types of filaments that protrude through surface of bacteria
flagella and pili
another name for pili
fimbrae
explain the mechanics of the 2 forms of bacterial motion
chemotaxis involves bacteria being attracted or repelled by substances in ins environment. If bacteria are attracted the flagella will beat counterclockwise which results in straight line movement, if repelled they rotate clockwise which results in random tumbling, while often impersise there is a net movement toward attractants and away from repellants
MOA of imidazoles
block synthesis of ergosterol required for fungal cell wall integrity (-azole drugs)
MOA of echinocandins
block beta glucan synthesis, a component of fungal cell walls
lipopeptide antibiotics MOA
form channels in cell membranes of G+ bacteria
MOA of polyene: amphotericin B
bind to sterols in eukaryotic cell membranes leading to lysis
MOA of folate antagonists
sulfonamides inhibit dihydropteroate synthesis . Trimethoprim inhibits DHF reductase
antigenic variation
capability in some bacteria to change amino acid composition of pili to evade immune detection/response
phase variation
some organisms such as salmonella undergo rapid changes between expression/non expression of genes that code for flagella proteins
2 groups of nutritional requirements for bacteria
photo/chemosynhetic bacteria that use light/chemical energy to subsist on CO2 and organisms that require organic compounds
all pathogenic organisms have this nutritional requirement
require organic compounds to survive
strict aerobe
requires oxygen to grow
strict anaerobe
cannot grow in presense of oxygen
facultative anaerobe (include common example)
can grow with or without oxygen, example: E Coli
final electron acceptor for strict aerobes
molecular oxygen, this is respiration
final electron acceptor (give 2 examples) for anaerobes
pyruvate (to form lactate) and acetyl coA to form ethanol. This is fermentation
bacterial species that can grow on hundreds of organic compounds and are omnipresent in water/ soil
pseudomonas
2 bacterial species that can grow on many types of media
pseudomonas and e coli
minimal media
contains a solution of glucose and several other minerals
nutrient broth
rich in nutrients and complex proteins that are required for some nutritionally fastidious organisms to grow
nutritionally fastidious
organisms that can only grow in complex media such as the human body, often these are highly parasitic organisms with nutritional needs near that or exceeding those of humans
obligate intracellular parasites (3 examples)
organisms that cannot be cultured in artificial media, include chlamydia, treponema pallidum (syphilis) and mycobacterium leprae (leprosy)
generation time
time it takes for a bacteria to become two
total count
count of the number of bacteria in a sample, counts as particles without discrimination between living and dead
how do you determine the number of living bacteria
via a colony count, which is done by applying a diluted sample to a solid medium then multipling the numbner of colonies that form by the dilution factor to determine the CFU or colony forming units originally present
law of growth
Nt = No x e^kt (where Nt is number at time t , No is initial number at time 0, k is the growth constant)
stage of culture where growth stops
stationary phase
SOS response
response of some bacteria to stresses such as E coli activating DNA repair genes when exposed to UV radiation

sporulation

some bacterial species produce spores which are exceptionally resistant to damage but are metabolically inert

metabolic parsimony
bacteria tend not to make products that they cannot use at the time, this arises from the need for bacterial strains to be metabolicly efficient to survive
operon
multigenic segment of DNA found in prokaryotic cells that contain enzymes for a particular pathway, by stringing these genes together regulation can be controled by a single signal
explain the regulatory steps and mechanisms of the b-galactosidase operon
if lactose is only source of sugar, the bacteria requires b-galactosidase to convert it to glucose. The lactose allosterically inactivates the Lac repressor protein, releasing it from the operator sequence, allowing the RNA polymerase to progress with transcription of mRNA
explain regulation of enzyme synthesis by attenuation
using the example of leucine, a small bit of mRNA is produced from the beginning of the leucine operon, the RNA polymerase will reach the attenuator sequence, if leucine is present, the transcription stops as there is no need to synthesize leucine , if leucine is not present the operon is transcripted in full, so that the machinery to produce the needed amount
Chapter 5
Chapter 5
3 substances/methods for nonselectively killing microorganisms
heat, radiation, strong acids
how do sulfonamides exert their antibiotic activity
they compete with PAB (para aminobenzoic acid) which is a precursor to folic acid, without the ability to produce folate, bacteria cannot replicate
why are our cells unaffected by sulfonamides while bacteria are
we require preformed folic acid, sulfa drugs disrupt the production of folic acid not the utilization, fortunately bacteria cannot uptake and utilize the folic acid present in our bodies and must make it themselves, making them susceptable to sulfa drugs
why are bacteriocidal drugs preferable to bacteriostatic
organisms that remain alive can still harm host by producing toxins, or becoming drug resistant and resuming growth, however there are instances where bacteriostatic drugs are prefered
explain how trimethoprim differs from sulfa drugs
trimethoprim blocks the function of folic acid, by blocking DHFR, this is a required enzyme for humans as well as bacteria, but the concentration of TMP needed to block DHFR is substantially lower for bacteria/protozoa than humans (.005 mM vs 250 mM)
how do beta lactam antibiotics expand their spectrum
modification of B lactam ring and other parts to increase access to different bacteria, improve absorbtion, or counter bacterial resistance
steps of beta lactam antibiotics
1. associate with bacteria 2. (if G-) penetrate outer membrane and periplsm 3. interact with penacillin binding proteins on cytoplasmic membrane 4. activation of autolysin that breaks down cell wall murein

beta-lactamase

bacterial resistance enzymes that degrade penacillins, cephalosporins, and/or carbapenams

gram postive bacterial resistance to beta lactam drugs
usually involves production and release of beta lactamases into surrounding media to prevent drug from reaching bacteria, if the antibiotic does reach the gram positive organism, it produces massive amounts of beta lactamase that cannot be overcome with higher doses of drug
gram negative bacterial resistance to beta lactam drugs
secreted constitutivey into periplasmic space
can beta lactam resistance be overcome with higher concentrations of drug
usually not in G+ but it is possible with gram negative because the amount of beta lactamases produced is constant
other than beta lactamases, what are some other methods by which bacteria become resistant to beta lactam drugs
gram negative organisms undergo porin mutatios which lead to exclusion of beta lactams from periplasmic space, PBPs can undergo mutations themselves, bacteria can become tolerant or lack autolysin

MRSA

methaicillin resistant Staph aureus, resistant to all penicillins and cephalosporins due to acquisition of an altered PBP (penacillin binding protein)

IV antibiotic commonly used for resistant Gram positive infections
vancomycin
explain how strains of enterococcus became resistant to vancomycin
vancomycin targets the peptide chain of murein at its D-alanine -- D-alanine terminus, however VRE strains acquired a plasmid that altered the terminus to D-alanine -- D-lactate, which vancomyicin cannot act upon
why is VRSA worse than VRE
enterococci are considered less invasive and virulent than staph aureus
how does daptomycin work?
it is a lipopeptide antibiotic that is able to interfere with the gram positive cell membrane and form a destrictive ion channel
what organisms are most commonly resistant to daptomycin
VISA
how do quinalone antibiotics work in gram + /- organisms
act by inhibiting the topoisomerases DNA gyrase (+) and topo IV (-) resulting in strand breaks and loss of bacterial chromosomal integrity
2 methods of resistrance to quinolone antibiotics
mutation in genes coding for DNA gyrase and topo IV ; formation of quinolone efflux pumps in bacteria

aminoglycoside MOA

1. penetration of outer membrane of G- 2. associate with 2 stage active transport in cell membrane 3. bind to 30s ribosome to inhibit protein synthesis at or near initiation step

methods of bacterial resistance to aminoglycoside drugs
most commonly, acquired plasmids code for genes that add modifiers to the aminoglycosides such as adenyl or phosphate groups rendering them inert. Less commonly bacteria can inactivate the transport of aminoglycosides
major method of tetracycline resistance
reduction of intercellular concentration by utilization of efflux pumps which remove the drug from the cell
major method of macrolide resistance
modification of the 50s ribosomal subunit. This is via methylation of the 23s ribosomal RNA. The modification confers macrolide resistance

major method of resistance to oxazolidones

similar to macrolide resistance, in that mutations to the 23s RNA of 50S ribosomal subunit block access of drug to ribosome

MOA of polyene antibiotics
antifungal agents that target ergosterol in membranes of fungi
MOA of imidazoles
antifungal agents that target cyp P450 demethylase which is involved in sterol synthesis
MOA of echinocandins
inhibitors of B-glucan which is found in fungal cell walls
synergism
in combo therapy, 2 or more drugs have effects that augment each other
antagonism (drug therapy)
action of one drug reduces effectiveness of the other
indifference (drug therapy)
each drug works no better/worse than if it was administered alone
read table 5-1
then read it again, if you still don’t know it, read it a 3rd time
Chapter 8
Chapter 8
adhesin that is found directly on cell surface and not on protrusions
invasin
integrin
normal cellular receptor found on host cells that can be reconized by invading microbes
fibronectin
receptor for certain G+ bacteria that normally coats mucosal surfaces of epithelial cells
bacterial adhesins of G- bacteria (3)
pili, non fimbral surface proteins, capsules
bacterial adhesins of G+ bacteria (2)
surface proteins, capsules
fibronetin deficiency increases risk of
increased exposure to gram negative organisms
scarcity of this nutrient limits potential for growth of bacteria in body
free iron
bacterial adaptations to free iron scarcity in body (3)
secretion of high affinity iron chelators that can be taken up by the specific bacteria ; surface molecules that steal iron from IBPs of the host; bacterial hemolysis of red blood cells to acquire iron from hemoglobin
why does brucella abortus cause spontaneous abortion in cattle but not other animals
these animals have erythritol in their placentas, which B abortus has a high affinity for
most effective way to protect against the antimicrobial action of compliment
prevent its activation
how do gram neg/+ organisms mask against compliment activation
secretion of capsules to prevent compliment recognition of LPS (g-) or teichoic acids (g+)
sialic acid
sugar that inhibits compliment activation that is commonly incorporated into bacterial surface molecules

describe the strategy that meningococci employ to avoid compliment activation (hint: wolf in sheep's clothing)

become coated with circulating IgA which does NOT cause compliment activation. The IgA blocks access of other antibodies to the bacteria

how does HSV avoid detection from complement
it has an envelope that binds C3b inhibiting activation by alternative pathway
how does vaccinia virus avoid compliment activation
it secretes a C4bp inds C4b and limits the extent of complement activation
how does E coli hinder the success of the MAC
smooth strains of E coli are able to avoid destruction by the MAC due to prolonged O antigen portion of LPS which limits access of the MAC to the organism surface. Rough mutants with little/no O antigen are readilly killed by the MAC
O antigen and pathogenicity
more O antigen correlates with higher pathogenicity , smooth = virulent , rough = not
C5a peptidase
secreted by group A streptococci, eliminates C5a which is a neutrophil chemotactic
leukocidans
exotoxins secreted by some pathogenic bacteria that kill neutrophils and macrophages
NETS
neutrophil extracellular traps, many bacteria can secrete nucleases that can destroy nets and enhanse survival
protein A
found on surface of staphlococci, streptococci and other bacteria, bind to wrong end of IgG molecule and stop them from acting as opsonins
tuberculosis, psittascosis and legionnares disease survive within phagocytes how
they inhibit lysosome fusion wih phagosome they are contained in
how do listeriam shigellae, monocytogenes, and rickettsiae survive phagocytosis
they escape the phagosome and enter the cytoplasm where they are safe from lysosomal enzymes
listeriolysin
required for escape of bacteria from phagosome, secreted by l monocytogenes
how does the protozoa leishmania escape digestion in phagocytosis
it is resistant to lysosomal enzymes
how does legionella escape digestion in phagocytosis
inhibition of hexose shunt and oxygen consumption to reduce power of respiratory burst
syncytia and multinucleated giant cells
result from viruses which spread by inducing fusion with other uninfected cells
how can bacteria move through cytoplasm
through the induction of polymerization of actin in the cytoskeleton. This can push the bacteria along the edges where it can escape the membrane into a neighboring cell
superantigens
toxins secreted by certain streptococci that stimulate a nonspecific T cell response resulting in diversion of immune resources and a toxic cascade of cytokines. They are able to do this because unlike regular antigens, they form a bridge between the APC and the T cell that is not variant, where as the bridge between APC and T cells in normal antigen binding occurs in a highly specific region
what is the causative agent of sleeping sickness, how does it evade immune destruction
trypanosoma brucei is a protozoan that causes sleeping sickness, it is covered with a thick variable surface glycoprotein that undergoes antigenic changes throughout infection, the antigens presented to the body on the surface are constantly changing, meaning the immune system is constantly playing catchup
what is pilin and how do gonococci use them
pilin is a protein that makes up pili which attaches to potential host cells, gonococci have pilin that changes and allows for multiple reinfections, as the strand that causes one infection is likely to be antigenically different than one that causes a recurrent infection
antigenic drift/shift
occurs in viruses such as influenza at the population level. A drift occurs about every 2-3 years and a shift occurs every 10 years or so.
hemaglutinin, neuraminidase
responsible for antigenic shift/drift in influenza virus, hemagglutinin serves to bind to cell surface receptors, neuraminidase changes these receptors
staphytlokinase
cleaves host plasminogen into plasmin at bacterial cell surface of staphylococci, this derades IgG and C3b preventing opsonization
fabulation
occurs when an organism cleaves IgA and remains bound to the inactive fragment (Fab) this prevents binding of intact antibody molecules
Chapter 9
Chapter 9
3 methods of cell lysis during microbial infection
1. production of toxin that affects cell membrane integrity 2. organism multiplies within the cell causing lysis. 3. microorganism is targeted and lysed by NK cells or cytotoxic lymphocytes
general definition of toxin
proteins that alter the normal metabolism of host cells, with deleterious effects on host
how are bacterial toxins categorized
by site of action in host
where do exotoxins, type 3 cytotoxins and type 4-7 cytotoxins modulate their effects
intracellular targets
where do endotoxin, superantigens, membrane-damaging toxins and exoenzymes act
cell surface, exoenzymes modulate ECM
type 1 secretion system
autotransporter built into exotoxin
type 2 secretion system
transport apparatus within bacterial membrane
how do exotoxins reach their target
they are secreted by bacteria and circulate until they encounter receptors on surface of specific cells and become internalized into the target cell
are toxins required for bacterial growth
not directly but may be needed for survival and spread within host
when are toxins released by bacteria
some release them continuously, others only when bacteria are not growing, for example diptheria toxin is only made when the diptheria are iron starved, this toxin causes lysis of iron containing cells
A and B domains
protein configuration of many exotoxins, with the B domain "Binding" to the membrane" so the A domain can enter the cell and Attack
organization of cholera toxin
AB5
organization of anthrax toxin
2A + B (they interact at the host membrane)
edema factor
an adenylate cyclase seen in the anthrax toxin
lethal factor
a protease seen in an A domain of the anthrax toxin
ADP ribosyltransferases
seen in many bacterial toxins, these transfer ADP ribose from NAD to target proteins covalently
organization of diptheria toxin
AB
EF-2
elongation factor 2, target of diptheria toxin, this is required movement of ribosomes on mRNA
how does diptheria toxin work
it’s A domain a ADP ribosyltransferase that targets EF-2 , severely limiting a cells ability to produce proteins. This is because the A domain covalenty attaches an ADP-ribose to EF2, perminantly inactivating it and preventing progression of ribosomes along mRNA. A single A subunit of diptheria toxin is sufficient to kill a cell, due to its long activity
primary target of cholera toxin
small intestine epithelium
in 1 sentence how does cholera toxin affect cell
by irreversibley raising intracellular cAMP, resulting in mass movement of ions and water into intestinal lumen
how does cholera toxin raise cAMP in the cell
the A subunit is a ADP ribosyltransferase, it transfers the ADP-ribose to a G protein which is then activated, resulting in continual stimulation of Adenyl Cyclase and high intracellular cAMP levels
pertussus toxin and heat liable enterotoxin of E coli work similarly to what toxin
cholera toxin, namely they activate AC by ADP ribosylation of G proteins
blocks muscle contraction yielding flaccid paralysis
botulinum toxin
produces irreversable muscle contraction yielding spastic paralysis
tetanus toxin
most potent protein toxin to humans
botulinum toxin
anaerobic spore forming bacteria of genus clostridium produce these toxins
neurotoxins (botulinism toxin and tetanus toxin)
structure of tetanus and botulinism toxins
single proteins with an A and B domain
how does the effect of the A domain in Tetanus and botulinsim toxin differ
in tetanus, the toxin migrates up an axon into inhibitory neurons where they block release of inhibitory neurotransmitters (resulting in spastic paralysis. In botox the A domain migrates to the peripheral nerve ending, blocking release of stimulatory NT such as ACh (resulting in flacid paralysis)
what type of protein is the A domain of tetanus toxin/botulinism toxin
protease that cleaves proteins responsible for fusion of synaptic vesicles with plasma membrane
how are type 3 cytotoxins introduced to a host cell
by contact depentant mechanism where bacterium directly contacts host cell
injectisome
type 3 secretion system (T3SS) used by pathogenic bacteria to deliver type III cytotoxins to host cells in a contact dependant matter
salmonella, shigella, pseudomonas, cholera, and plague bacilli deliver toxins this way
T3SS
type IV to VII cytotoxins
we don’t know much about it, so Im not bothering to learn much about it.
endotoxin
another name for the LPS of the outer membrane of G neg bacteria
what is misleading about the name endotoxin
it is not internalized, and only toxic at high concentrations
what is a pattern recognition receptor
highly conserved factors seen in bacteria that are recognized by our immune system (Toll like receptors) these include endotoxin (from G neg bacteria) lipoteichoic acid (from G + bacteria) and lipoglycans from mycobacteria
components of endotoxin
lipid A, sugar core, O antigen
biologically active portion of endotoxin
lipid A….duh….A=Active (just like in A/B domains of exotoxins)
result of small/ large amounts of endotoxin
small is alarm reaction, large is shock an intravascular coagulation
alarm reaction results in
the increase in cytokines triggered by endotoxin binding to TLRs results in fever, vasodilation, inflammation, and increased antibody synthesis
fever induced by endotoxin is caused by _________ being released from __________
IL1 and TNFa being releced from mononuclear phagocytes
how does endotoxin activate compliment, what does this cause
endotoxin triggers the alternative pathway, resulting in MAC production, phagocyte chemotaxis and opsonisation. The compliment also causes anaphylatoxins which result in inflammatory response
effect of endotoxin on macrophages
increases everything they do. They make more lysozymal enzymes, eat more, and secrete more hydrolases
waterhouse friderichsen syndrome
adrenal insufficency due to infarction seen in DIC leads to death, seen in some cases of severe meningococcal infection
3 ways endotoxin contributes to pro-coagulative state
1. activates factor XII (intrinsic factor) 2. causes platelet degranulation. 3. causes neutrophils to release fibrin stabilizing proteins
lecithinase
lipase toxin produced by clostridium perfringenes which causes gas gangrene.
hemolysins
another name for membrane damaging toxins, due to their ability to be identified in laboratory by lysing RBCs on blood agar
example of homogeneous pore former
alpha toxin of staphylococcus aureus. Forms pores with same number of molecules (similar to pores formed by MAC)
heterogenous pore forming toxins (example and mechanism)
form membrane damaging pores of various sizes, prototype is streptolysin O that binds to cholesterol in cell membrane destroying RBCs
hyaluronidase and DNAse are examples of ___________ that act as ____________
exoenzymes that act as spreading factors, the hyaluronidase breaks down ground substance of CT and DNAse thins pus made viscous by dead WBCs
how do streptococci eliminate fibrin barriers
by releasing streptokinase which activates plasminogen to remove fibrin clots
toxoid
chemically inactivated toxin
Chapter 10
Chapter 10
Big 4 typical bacteria
gram positive/gram negative rod/cocci
2 most medically relevant genera of Gram positive cocci
streptococcus and staphylococcus
how do streptococci grow
in strings of spherical cells like pearls
how are streptococci subdivided
by weather they produce changes when grown on blood agar
beta hemolytic streptococci
destroy red blood cells, clear area around colony
alpha hemolytic streptococci
affect red blood cells partially, result in green area around colony
do streptococci carry out respiration
no, only fermentation meaning streptococci are STRICT ANAEROBES
oxygen tolerant anaerobe
although they do not respire they can grow in presense of air many pathogenic streptococci are oxygen tolerant
main pathogens in streptococcus genus are the _______ strains
beta hemolytic
most important b-hemolytic streptococci strains
streptococcus pyogenes aka group A strep
major alpha hemolytic streptococcus species
streptococcus pneumoniae
2 main pathogenic species of staphylococcus
staphylococcus aureus and S. saprophyticus
how to distinguish S. aureus from other staphylococci
with coagulase test, because only S. aureus produces it
most likely organism to cause pus in wounds
staphylococci
often produce serious infections in deep tissues such as osteomyelitis and endocarditis
staphylococci
most significant genus and species (2) of gram negative cocci
neissera gonorrhoeae which causes gonorrea and meningococcus ( N. meningititis) which causes meningitis/septicemia
toxin common to all gram negative organisms
endotoxin (lipopolysaccride)
what is the classification of the organism that causes diphtheria
gram positive rod, Corynebacteriu diphtheriae
in human environment most common gram positive rods are
sopre forming
2 categories of common gram positive rods found in humans
aerobic bacillius (B. anthracis which causes anthrax) strict anaerobes (belong to clostridium genus)
medically important agents of clostridium (4)
C botulinum, C tetani, C perfringens, C difficile
how do members of the genus clostridium cause disease
through powerful exotoxins
causes gas gangrene
C perfringens
associated with antibiotic use, causes pseudomembranous colitis
C difficile
gram positive rod that causes serious infections in immunocomprimised infants/adults
listeria monocytogenes
major clinical grouping of gram negative rods
enteric
enteric bacteria can be further divided by what characteristic
ability to ferment lactose
major lactose fermenter among enteric bacteria, major lactose non fermenter(s) among enteric bacteria
lactose (+) = e coli lactose (-) = shigella and salmonella
name several families of enteric bacteria that lie outside the family of enterobacteriaceae
vibrio, pseudomonas, campylobacter, helicobacter
name some genuses that are included in the fastidious and small G(-) rod family (6)
Haemophilius (pneumonia/meningitis) Brucella (brucellosis) , francisella (tularemia) Bartonella (cat scratch fever) Legionella (legionaires disease)
__________ are the most noteworthy strictly anaerobic gram negative rods
bacteroides
name some places that bacteriodes are found and some of their uses to us
they can break down some polysaccrides found in our bodies and serve as a stimulus for proper tissue development, usually these bacteria are harmless unless deposited in deep tissues
most common acid-fast bacteria
mycobacterium which include tubercle bacillus (M. Tuberculosis) and leprocy bacillus (M. leprae)
Ziehl-Neelsen technique
procedure for identifying acid fast bacteria
what color do acid fast bacteria stain
red, from the fuchsin stain that was not washed away by the acid, all other cells stain blue, which is the color of the counterstain
most common of the spirochete bacteria 2
treponema pallidum (causitive agent of syphilis) ; Borrelia (such as borrelia burgdorferi which causes lyme disease)
how do you culture chlamydiae
we cant, they are only known to grow intracellularly
most common cause of STD
infection with chlamydia trachomatis
describe the features of rickettsiae
these are small obligate intracellular parasitic bacteria similar to chlamydiae. They cause RMSF, endemic typhus, and usually require transmission from an arthropod vector (exception being Coxiella burnetti which causes Q fever and can be acquired by inhalation)
L form bacteri
mycoplasmas lack cell walls and resmble L form or laboratory formed bacteria
most common pathogenic mycoplasma
mycoplasma pneuoniae
Chapter 30
Chapter 30
emperic treatment
based on clinician knowledge of usual causative agents in certain clinical situations (ie: E coli and uncomplicated UTI)
disk diffusion method
test for microbal sensitivity that involves placing a disk soaked in antibiotic on an agar innoculated with a microbe. The distance which the microbe doesn’t grow from the disk gives an estimate of the MIC
does MIC indicate bacteriocidal concentration or bacteriostatic
bacteriostatic
MBC
minimal bactericidal conentration
proportion of administered dose of drug that reaches circulation
bioavailability
for drugs that kill bacteria slowly what do you want to maximize
time above the MIC
for drugs that kill bacteria quickly what do you want to maximize
AUIC or area under the inhibitory curve, this is achieved by creating a large C max
effect of pH on aminoglycosides, chloramephenicol, erythromyin, and tetracycline
decrease effect of (AG, B lactam, erythromycin) increase chloramphenicol and tetracycline
effect of pH, low redox potential and high divalent cation concentration on aminoglycides
all decrease
most common antibiotics for allergic responses, why
penacillins, cephlasporins and sulfonamides, penacillins cause allergies due to their ready protein binding which can elicit IgE response
some side effects of aminoglycosides
impaired renal functions and inner ear toxicity. Renal toxicity can result in toxic levels building up in blood, while inner ear toxicity presents as balance and hearing loss)
stevens johnson syndrome
serious skin allergic reaction leading to formation of bullae on the skin and inflammation of eyes/mucous membranes
in the clinical case for this chapter they discuss the effect of rifampin on the patient's warfarin therapy, give a brief explaination of this
the rifampin induced faster clearance of the warfarin which lead to hypercoaguability and resulted in thrombophlebitis for the patient
name 2 purposes for prophylactic antimicrobial therapy
prevent infection in individuals who have high risk/exposure to exogenous pathogens, prevent commensal organisms from spreading to sterile sites in body (ie post surgically)
Chapter 31
Chapter 31
fundamental difference between viruses and other infectious agents
viruses are obligate intracellular parasite that carry out replication entirely with host cell machinery.
structure that surrounds viral nucleic acid
capsid
nucleic acid + capsid
nucleocapsid
2 basic structural patterns for viruses
icosahedral and helical
some viruses have this, which surrounds the nucleocapsid
envelope
contents of viral envelope (in addition to nucleocapsid) include
virus specific proteins, lipids, and carbohydrates, sometimes a matrix protein
adsorption
attachment of virus to host cell surface
clathrin coated pits
aggregation sites for some viruses such as influenza on the plasma membrane,
these viruses cannot fuse with cellular membranes
nonenveloped viruses
uncoating
once the virus is adsorpted and releases its payload into the host cell, the capsid is removed to make the viral genome asessable to the cellular transcription/translation machinery
how do ss + RNA viruses such as picornaviruses (polio) and flaviviruses (west nile) produce proteins, reproduce their RNA
to produce protein, the + RNA genome simply acts as mRNA and is translated by cellular ribosomes into proteins. To replicate viral RNA, a virus encoded RNA dependant RNA polymerase produces a negative complimentary RNA molecule, in turn this acts as a template to make complimentary + RNA molecules for progeny
how do ss- RNA viruses such as influenza virus produce proteins, reproduce their RNA
negative sense RNA cannot act as mRNA. It must be converted to a complimentary positive sense RNA to produce proteins. Since our cells have no machinery for RNA dependent RNA polymerase, the virion must contain this. Once the + sense mRNA is created proteins can be replicated using ribosomes, and - sense RNA can be regenerated for progeny using the RNA dependent RNA polymerase
segmented genomes
some viruses have genomes that exist as multiple strands of RNA, often there are individual genes or groups of genes on these strands
how do ds RNA viruses produce proteins, reproduce their RNA
like ss -RNA viruses, they must come preloaded with a RNA dependent RNA polymerase. This polymerase takes the neg sense RNA strand and produces a single pos sense RNA strand from which the negative sense strand can be replicated, or ribosomes can use as mRNA to make viral proteins
describe the unique replication strategy of retroviruses
retroviruses like HIV are ss + sense RNA viruses however, they come preloaded with a RNA dependent DNA polymerase (reverse transcriptase) which created DNA from the original RNA molecule, this DNA is then incorporated into the host genome where it stays forever, and is transcribed using DNA dependent RNA polymerase
early vs late transcripts
in DNA viruses the first (early) transcripts contain regulatory proteins and proteins required for viral DNA replication, while late transcripts are structial proteins of virion
exons vs introns
exons contain the gene sequences, introns are junk sequences….just like in our DNA
how does the release of nonenveloped viruses differ from enveloped
nonenveloped released when cell lyses, enveloped released by budding
acute vs latent vs chronic viral infections
acute is the virus undergoes multiple replication rounds and there is death of host cell. Latent is seen in DNA/retroviruses, and chronic is seen is Hep C where viral particles are still shed following period of acute illness
immunoglobulin that contributes to defense at mucosal membranes such as respiratory and GI tract
IgA
iatrogenic inoculation
transmission of virus caused by diagonistic or thereputic procedures
horizontal/vertical sprea
horizontal spread is between members of susceptable population and vertical is between mother and fetus when virus is carried in germ cell line,placenta, or maternal birth canal
HSV rabies virus and VZV all spread how
through nerves using mechanisms involved in fast axonal transport
most important adaptive defense mechanism against viral infections is
cell mediated immunity
interellular defense mechanisms against viruses include
apoptosis and autophagy (which results in sequestration of virus particles)
early and late cytotoxic defenses against viruses
early is from NK cells while later its CTLs and virus specific antibody responses
viruses are delt with by T lymphocytes that express MHC1 or MHC2
MHC1
interferons
synthesized by host cells in response to viruses that induces the expression of proteins that INHIBIT the protein synthesis machinery to limit viruses ability to replicate
alpha interferon is produced by
leukocytes
beta interferon is produced by
fibroblasts and epithelial cells
gamma interferon is produced by
T cells activated by speciic anigens
Chapter 44
Chapter 44
primary treatment for HCV infection (2). These agents were derived emperically
interferon and ribavirin
pegylated interferon
longer acting interferon used in combonation with ribavarin to treat HCV
explain some of the mechanisms by which ribavirin exerts its antiviral effects
1. inhibition of DNA dependent DNA polymerases and some RNA dependent RNA polymerases 2. inhibition of inosine monophosphate dehydrogenase, lowering cellular GTP and restricting nucleic acid synthesis 3. prevention of capping of viral mRNA
limitations of ribavirin
toxicity and lack of specificity
gp120
viral glycoprotein found in the envelope of HIV that allows it to bind to surface of CD4 T cells
CCR5 and CXCR4
coreceptors for HIV that interact with the CD4 T cells after gp120 has bound CD4
how does maraviroc aid in the treatment of HIV
antagonizes the CCR5 receptor of HIV, limiting its ability to bind to T Cells
what is gp41
HIV transmembrane protein that changes conformation when gp120 binds CD4, this conformational change exposes the fusion peptide and allows for fusion of viral/cell membranes
R5 vs X4 tropic viruses
in patients recently infected with HIV, the virus more commonly binds CCR5 whereas patients with advanced disease have the HIV particles bind CXCR4
what is the MOA of Enfurvirtide
this drug treats HIV by targetin a specific domain of the HIV gp41 fusion peptide. This prevents the gp41 from undergoing conformation changes nesessary to fuse membranes with the host cell
what do amantadine and rimantadine treat, how do they do it
amantadine and rimantadine treat influenza A. They do this by blocking the M2 transmembrane protein in the influenza virus. This protein acts as a hydrogen chanel. Once influenza enters the cell these channels help to lower the pH inside the endosome, creating conformation changes which result in release of viral genome. These drugs block the M2 channel
prototype antiviral drug
acyclovir
what is the MOA of acyclovir and why is it effective against HSV
it is a DNA polymerase inhibitor specfic to herpes viruses. The reason it is so effective is that the HSV molecule has a thymidine kinase which phosphorylates acyclovir, this activates it. Herpes viruses phosphyorylate acyclovir much more effectively than other viruses, making it a drug of choice for treating these viruses
thymadine kinase
activates acyclovir by phosphorylating it. The thymadine kinase of HSV phosphorylates acyclovir at a much larger rate than those of our cells and other viruses
MOA of ganciclovir, what does it treat
ganciclovir treats CMV, it works the same way acyclovir does, except it is preferentially phosphorylated by CMV kinases
difference between acyclovir and valacyclovir
valacyclovir is a prodrug of acyclovir that is absorbed much better than acyclovir
difference between valganciclovir and ganciclovir
valganciclorir is a well absorbed prodrug of ganciclovir
what is penciclovir
acyclovir analog with a longer halflife
famciclovir
prodrug to penciclovir, it is a longer lasting more well absorbed version of acyclovir
most acyclovir resistant herpes strains have mutations in what
thymidine kinase
what are benefits and risks associated with foscarnet
it is a phosphonate which inhibits herpesvirus DNA polymerase directly, but it is nephrotoxic
NRTIs and NtRTIs
nucleotide/side reverse transcriptase inhibitors, these are the target of much research due to their potential to treat HIV
MOA of NRTIs
these drugs lack a 3' hydroxyl required for chain elongation, once these nucleotide analogs are incorperated into the growing chain by reverse transcriptase, chain elongation stops
NNRTIs
non-nucleotide reverse transcriptase inhibitors, these work by inhibiting a catalytic step in reverse transcription
raltegravir
prototype integrase inhibitor that targets HIV at the step of strand transfer it is refered to as a "INSTI or integrase strand transfer inhibitor"
HAART
highly active antiretroviral therapy, this is the standard for HIV treatment and involves using multiple drugs at once (at least 3)
name the class of drugs that prevent release of influenza A and B virus and how they work
neuraminididase inhibitors (oseltamivir/zanamivir). These inhibit cleavage of sialic acid, which is essential for influenza release
name the class of drugs that prevent assembly of HIV/HCV
protease inhibitors, these prevent cleavage of precursor peptides and prevent the formation of mature HIV proteins
name 2 classes of drugs that prevent entry of HIV into cell
HIV fusion inhibitor (enuvirtide) which binds to gp41 and CCR5 antagonist maraviroc which prevents binding of gp120 to CD4
name a class of drugs that prevents viral dissessembly in influenza
influenza M2 antagonist (amantidine and rimantidine) which block the hydrogen channel that induces conformation shift and release of influenza genetic material
thymidine kinase activated polymerase inhibitor effective against HSV, CMV
acyclovir, ganciclovir
name a drug that directly inhibits CMV polymerase
foscarnet (this is a phosphonate drug)
name 3 classes of drugs designed to target HIV replication
NRTI/NtRTI , NNRTI, INSTI
Chapter 46
Chapter 46
endemic mycoses
infections caused by pathogens that are restricted geographically
oppurtunistic mycoses
often seen in immunosuppressed patients
subcutaneous mycoses
involve skin, subQ tissue, and lymphatics
superficial and cutaneous mycoses
common fungal infections limited to skin/skin structures
sterols of fungal cell membrane
ergosterol
components of fungal cell wall
chitin, mannin, glucan
conditions that favor fungi that cause disease
35-37 C
yeast
unicellular form of fungi
mold
multicellular filamentous form of fungi
how do most yeasts divide
asexual budding
filaments that facilatate mold growth
hyphae
mass of hyphae is refered to as
mycelium
septate/nonseptate hyphae
septate are divided by cross walls running perpendicular to length while nonseptate exist as a multinucleated syncitium
dimorphism
ability of many fungi to exist as yeast and mold
with most dimorphic fungal pathogens which form is found in environment and which is found in tissues
mold in environment, yeast in tissues
conidia
asexual fungal reproductive structures
where are conidia found
at the tips of growing hyphae on the conidiophore, directly off the hyphae, or within the hyphae
how do fungi reproduce sexuallty
throuh development of spores
primary mechanisms that prevent establishment of fungal infections
once fungi spread to a tissue neutrophil phagocytosis/killing is usually sufficient
most important host defense against most fungi
cell mediated immunity, as evidenced by the high incidence of fungal disease in AIDS patients
Chapter 51
Chapter 51
parasites include mostly
helminths and protozoa
do protozoa multiply in human hosts, helminths?
yes, no.
common protozoan that many americans are infected with, but show no symptoms of disease
toxoplasma gondii
disease associated with parasites usually associated with what conditions
prolonged, repeated, unusually burdensome infection
zoonoses
parasitic infections caused by aents that infect animals such as birds,reptiles,mammals (particularly livestock)
dead end hosts
some animal parasites don’t require a development stage in humans and can infect us anyways. This makes us a dead end host for these parasites
swimmer's itch
caused by a blood fluke that alternated between birds and snails, when it attempts to infect us it fails to pass the skin and dies, causing a mild itch which resolves spontaneously
why do many protozoa not have an environmental stage of their life cycle
with only a cell membrane (no wall) to protect from environment, many cannot survive outside hosts
trophozoite
protozoan life stage that involves growth and replication within host by binary fission
cyst
protozoan life stage that is dormant and is more resilliant to environment, the cyst form allows for transfer between hosts
how do amebas move? What is their life cycle?
they extend pseudopods toward stimulus and stream cytoplasm in desired direction. They alternate between trophozoite and cyst form.
how do flagellates move, where do they usually infect
flagellates have flagella (duh). They are mostly intestinal and blood parasites. Glagellates that invade the GI form cysts to resist environment
ciliates
protozoa which move with cilia.
what are the apicomplexans/sporozoa and what diseases do they cause
these are protozoa which move in a gliding or tractor tread mechanism. Diseases caused by these include malaria and toxoplasmosis
helminths
multicellular worms that are considerably larger than protozoa and reporduce sexually
roundworms
circular in cross section, can grow up to 20cm, ascaris lumbricoides looks like an earthworm but they have no visible body segmentation
two types of flat worms
flukes and tapeworms
flukes
ex include schistosoma and fasciola ; these worms are short and nonsegmented
tapeworms
segmented worms tht can vary from a few mm to several meters in legnth, a long tapeworm is actually a chain of independent organisms (a colony)
vectors
living transmitters of disease, most vectors are arthropods
transmits malaria
anopheles mosquito
transmitts sleepin sickness
tetse flies
transmitts river blindness
black flies
transmitts chagas disease
reduviid kissing bugs
reserviors
sources of parasites in environment that do not participate directly in the transmission of parasites to humans
how does fecal oral and penetration of unbroken skin usually occur
ingestion of contaminated food and water or exposure to wastes in soil/water
how do arthropods transmit infection to humans
by biting you, like mosquitos, f**k mosquitos
how do plasmodia species evade immune response
antigenic variation, intracellular infection, and supression of cellular immune response
how do toxoplasma species evade immune respnse
by infecting intrecellularly
these organisms camoflouge from immune ditection
schistosomes
these organisms cleve antibodies and compliment
amebas and leishmania species
this surface factor is necessary for entry of malaria into RBC
duffy factor
what immune responses can often be the only manifestations of parasitic disease
chronic inflammation and hypersensitivity reactions due to leaking of parasitic antigens
leukocytes responsible for clearing parasitic infections
eosinophils
cytokine that triggers eosinophilia, immunoglobulin associated with parasitic infection
IL-5, IgE
3 major antiparasitc control strategies
1. produce drugs for prevention, 2. immunization 3. field control measures
example of sucessful chemopropholaxysis for malaria
administration of chloroquine before travel to malaria striken regions was sucessful in preventing infection, however resistant strains of p falciparum have emerged
how do schistosomes evade the immune response
they cover themselves with host antigens
how do trypanosomes evade host immune response
they alter surface antigens, preventing development of long term immunity
stage specific antigens and vaccines
to develop an effective vaccine against parasites, antigens from all parts of life cycle must be accounted for
Chapter 58
Chapter 58
true positive
when a test correctly predicts the presense of a pathogen
true negative
when a test correctly detects the absense of a pathogen
false negative
laboratory test is negative in presense of pathogen
false positive
laboratory test is positive in absense of a pathogen
sensitivity
likelihood that a test will be positive when the pathogen is present
specificity
likelihood that a test will be negative when the pathogen is not present
how to calculate sensitivity
true positives / (true positives + false negatives) x 100
how to calculate specificity
true negatives / (true negatives + false positives) x 100
positive predictive value
true positives / (true positives + false positives) x 100
negative predicitve value
true negatives / (true negatives + false negatives) x 100
why is it better to do an acid fast stain than a gram stain on a sputum sample
gram stain will just display normal microbiota of respiratory/oral tract but an acid fast stain can reveal if mycobacterium tuberculosis is present
what is a giemsa stain used for
systemic protozoal infections
what is an iodide stain used for
intestinal helminths
what is a silver stain used for
detecting systemic fungal pathogens
how do direct fuorescent antibody tests work
monoclonal antibodies, specific for a microbe of interest are conjugated to a flourescent compound. If the microbe is present the antibodies will attach to the microbe and flouresce, making them visible with a fluorescence microscope
why is the sepcificity of most DFA tests low, how can you raise it (see fig 58-2)
most of the antibodies that are used are polyclonal and bind multiple antigens, meaing there is cross rectivity and other microbes can be included. This raises the instance of false positive lowering specificity. To raise it monoclonal antibodies which bind a single, specific antigen, remove the risk of cross reactivity and reduce false positives
explain how a thayer martin chocolate agar is useful for detecting gonococci
the agar is a selective media, designed to inhibit normal microbiota of the genitourinary tract (with antibiotics) but not gonococci
describe the lysis centrifugation technique for blood culture
blood sample is placed in solution that lyses RBCs and spun. The dense material is removed and innoculated onto an agar based medium
in serological testing how is the amount of antibody in a sample quantified
by progressivly diluting the serum to the highest extent that antibodies can be detected
explain the basic steps in an ELISA serology protocol
1. patients serum is incubated with antigen in question on a solid support 2. the unbound antibodies are washed away. 3. antibodies to the antibodies which were in the patients serum are introduced. These are labeled with an enzyme. 4. the amount of activity from the bound enzymes is measured
explain the western blot procedure
antigenic molecules from a pathogen are seperated using electrophoresis. The separated antigens are then blotted onto a solid support and innoculated with patient serum. This allows to see what specific antigens are being bound with patients antibodies, and if there is cross reactivity.
how can physicians determine if a positive serology is due to past or present illness
they can take titers at two points in time (acute and convalescent titers) or can measure specific IgM antibodies. IgM antibodies appear in acute infection and fade over time
how do antigen detection tests work
they are serology in reverse. Specific antibodies are used to try to capture antigens from patient sample. The ELISA procedure can be done to detect antigens as well as antibodies
how does a competitive assay for antigens work
a sample is coincubated with a enzyme labeled antigen, which competes for binding to a fixed amount of antibody. If there is a large amount of unlabeled antigen in the patient's sample, it will outcompete the labeled and a color reaction will not occur, this is a positive finding
how does a DNA probe test work
a radiolabled short sequence of DNA is added to a sample. The sample is heated such that the double strand of DNA seperates, allowing the probe sequence to bind to the complimentary single strand. Upon cooling the amound of hybridized probe to non-hybridized probe can be quantified.
what is required for PCR
2 short primers, dNTPs, and heat stable DNA polymerase, in each heating/cooling cycle the sequence between the 2 primers is replicated
real time "qPCR"
PCR where flourescent labeled primers are amplified by increasing cycles of PCR, the amount of time it takes to reach the level of flourescent detection gives insight into how much DNA was present in the original sample, which is useful in detecting viruses in human fluid samples.
explain the rationale for microarray testing
bacteria have nonconserved sequences wedged between conserved sequences. If we do a PCR between these conserved sequences with labeled primers we will create DNA which has specific (non-conserved regions in it) that can be visualized with flourescense. From there a microarray with hundreds of known non-conserved sequences spanning many organisms can be tested. The sequence that hybridizes our labeled PCR product will reveal the identiy of the organism in the original sample