• 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

1/244

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;

244 Cards in this Set

  • Front
  • Back
What does "sarcina" mean?
bacteria that arrange in packets of 8 cells
Name one exception to the rule that bacteria do not contain sterols in their cell membranes.
mycoplasma pneumoniae
-it lacks a cell wall so sterols in the cell membrane are necessary to provide rigidity and support, also provide flexibility
What are the functions of the cell wall in bacteria?
-maintenance of cell shape
-prevent osmotic lysis
-environmental protection
-anchor for external structures like flagella, etc
What molecule is contained in virtually all cell walls?
peptidoglycan
-repeating disaccharide cross-linked by tetrapeptides
-backbone is alternating NAG/NAM which are then crosslinked by chains of 4 AAs
What are some ways that humans use the ability to target peptidoglycan to our advantage?
-peptidoglycan is important for bacterial cell support so we can interfere with the structure/synthesis of this molecule to target bacteria (known as cell wall inhibitors and only affect actively growing cells)
-we can use the following methods:
1)secretion of various antimicrobial compounds (taken from bacteria that naturally produce antimicrobial compounds)
2)secretion of lysozyme (found in egg whites and in granules of PMNs), tears, saliva, and mucus
What is the significance of lysozyme as an antimicrobial mechanism?
it is found in tears, saliva, and mucus and degrades the glycan backbone of peptidoglycan in bacterial cell walls
-it weakens the integrity of cell wall, even in bacteria that are not actively growing
-
What is the main difference between gram positive and gram negative cells (what differentiates them)?
Gram Positive=thick peptidoglycan layer and retain the primary stain, crystal violet

Gram Negative=thin PG layer, retain the counterstain, safranin
Describe the composition of the cell wall of gram negative bacteria.
-they contain 1-2 layers of peptidoglycan plus:
-outer membrane with LPS, trimeric porins and lipoprotein (external to the PG layer), it is a bilayer where inner layer is identical to cell membrane and outer is mostly LPS
-perisplasmic space
What is the "O" antigen?
the outer portion of LPS found in gram negative bacteria
-they are long linear repeating units of carbs
-serves as attachment sites and has been shown to inhibit phagocytosis
-this portion is highly variable and immunogenic
What is the structure of LPS?
1)O antigen on outer portion
2)core polysaccharide: connects lipid A to the O antigen
3)lipid A-embedded in the outer membrane has toxin activity, responsible for bacterial sepsis, heat stable and is a B cell mitogen, induces production of cytokines
What is the significance of LPS in bacterial septic shock?
-LPS directly activates the alternative complement pathway
-binds to receptors and causes release of cytokines leading to symptoms of septic shock
What is the function of lipoproteins in the outer membrane of gram negative cells?
the most abundant protein of gram-negative cells
-functions to stabilize the outer membrane and anchors the outer membrane to the PG layer
Describe the periplasmic space found in gram negative organisms.
-located btwn. the outer membrane and the cell membrane
-contains the PG layer, transport proteins, and a variety of hydrolytic enzymes
(some bacteria can concentrate antibiotic hydrolases which lends to resistance so important to do antibiotic susceptibility tests)
Describe the structure of gram positive bacteria cell walls.
-contain a thick PG layer along with one or both of the following:
-Wall teichoic acids anchored to PG
-Lipoteichoic acids anchored to cell membrane
-both provide elasticity and stability
What is the clinical relevance of wall teichoic acids and lipoteichoic acids found in gram positive bacteria?
they have 2 main functions that increase the virulence of gram-positive organisms:
-they function as adhesins (structures for adherence, the first step in the invasion process)
-they can initiate endotoxin-like activities when released
Describe the composition of acid fast cell walls.
they usually contain an intermediate thickness of PG along with the following:
-mycolic acid layer-covalently linked to PG, gives organism a waxy coat to resist dessication, resistance to antibiotics and inhibits phagocytosis
-tetrameric porins-allow passage of small hydrophilic molecules
What are the 3 major components of a flagellum?
1)filament-contains the H antigen
2)Hook-attaches filament to the cell surface, links to basal body
3)basal body-anchors flagellum in the cell wall and cell membrane
-they allow for motility
What are pili?
hairlike structures with adhesins at the tips
-they promote adherence to other bacteria or to host cells
-2 types are common pili (fimbriae, contains tips with adhesins usually lectins that bind to sugars) and sex pili
-can target these for antimicrobial therapy to inhibit bacterial colonization
What are sex pili (conjugation pili)?
-produced by some gram negative bacteria
-adherence btwn. 2 cells for exchange of DNA
What is the glycocalyx?
-polysaccharide layer outside of the cell wall
-source of K antigen
-commonly referred to as a capsule or slime layer
-can contribute to increased virulence, has many other functions as well including induces abscess formation
-glycocalyx of strep mutans allows for adherence to the tooth enamel
What is the Quellung reaction?
-a diagnostic test for Strep. pneumoniae
-used to detect the presence of a capsule in respiratory infections when S. pneumoniae is suspected
-test has anticapsular abs that cause the capsule to swell, aiding visualization
What are plasmids?
they encode for ancillary (non-essential) information
-includes genes that encode antibiotics, resistance, heave metal resistance, toxins, etc...
What ribosomal subunits do bacteria have?
30s
50s
=70s
-also, transcription and translation are coupled
-the ribosomes are the target of our protein synthesis inhibitors
What are some different types of inclusion bodies?
-all of the following function as storage depots
-volutin granules-polymers of
inorganic phosphate
-glycogen granules-polymers of alpha-D-glucose
-PHB granules-chains of beta-hydroxybutyric acid
What is an endospore?
-a resting stage that allows an organism to survive harsh environmental conditions
-conversion from a vegetative state to an endospore often triggered by nutrient depletion
-each endospore contains one chromosome, high concentrations of Ca bound to dipicolinic acid (gives increased resistance to heat), low amounts of essential proteins and ribosomes, and a keratin spore coat
-resistant to many chemicals and toxins (can persist for months to years in this state)
What are the 2 clinically relevant genera of bacteria that produce endospores?
1)bacillus
2)clostridium
-both gram positive
-the location and size of the spore varies for different species and aids identification
Does the optical density measure viable or non-viable cells?
both
-the more cells, the more absorption occurs or the light
What does a colony forming unit measure?
only VIABLE cells
-from diluting and culture and plating it, each colony is from one cell
Does the optical density measure viable or non-viable cells?
both
-the more cells, the more absorption occurs or the light
What does a colony forming unit measure?
only VIABLE cells
-from diluting and culture and plating it, each colony is from one cell
What is biomass?
-measures bacterial growth
-sample is washed, dried, and weighed, dry weight is taken
-measures all cells in culture (viable and non-viable)
Describe the lag phase of the bacterial growth curve.
-there is no cell division here
-increase in biomass from synthesis of macromolecules
-the length depends on form and availability of nutrients and the condition of the inoculum
What is biomass?
-measures bacterial growth
-sample is washed, dried, and weighed, dry weight is taken
-measures all cells in culture (viable and non-viable)
Describe the lag phase of the bacterial growth curve.
-there is no cell division here
-increase in biomass from synthesis of macromolecules
-the length depends on form and availability of nutrients and the condition of the inoculum
Describe the log phase of the bacterial growth curve.
-exponential growth occurs in this phase, rapid cell division
-slope reflects doubling/generation time (determined by the rate of cell division and varies with the microbe)
-pathogens often produce virulence factors at this time
-primary metabolites
Describe the late log phase of bacterial growth curve.
-transition to stationary
-secondary metabolites (antibiotics, pigments)
Describe the log phase of the bacterial growth curve.
-exponential growth occurs in this phase, rapid cell division
-slope reflects doubling/generation time (determined by the rate of cell division and varies with the microbe)
-pathogens often produce virulence factors at this time
-primary metabolites
Does the optical density measure viable or non-viable cells?
both
-the more cells, the more absorption occurs or the light
Describe the stationary phase of the bacterial growth curve.
-supplies of energy and nutrients exhausted
-resources are then renewed by cell death (release peptides and nucleic acids)
-zero population growth (no change in CFU/mL, OD, biomass)
What does a colony forming unit measure?
only VIABLE cells
-from diluting and culture and plating it, each colony is from one cell
What is biomass?
-measures bacterial growth
-sample is washed, dried, and weighed, dry weight is taken
-measures all cells in culture (viable and non-viable)
Describe the late log phase of bacterial growth curve.
-transition to stationary
-secondary metabolites (antibiotics, pigments)
Describe the death/decline phase of the bacterial growth curve.
-insufficient resources for growth
-exponential cell death (but still less than the growth rate in log phase)
-determined only by CFU/mL not OD or biomass
Describe the lag phase of the bacterial growth curve.
-there is no cell division here
-increase in biomass from synthesis of macromolecules
-the length depends on form and availability of nutrients and the condition of the inoculum
Describe the stationary phase of the bacterial growth curve.
-supplies of energy and nutrients exhausted
-resources are then renewed by cell death (release peptides and nucleic acids)
-zero population growth (no change in CFU/mL, OD, biomass)
Describe the death/decline phase of the bacterial growth curve.
-insufficient resources for growth
-exponential cell death (but still less than the growth rate in log phase)
-determined only by CFU/mL not OD or biomass
Describe the log phase of the bacterial growth curve.
-exponential growth occurs in this phase, rapid cell division
-slope reflects doubling/generation time (determined by the rate of cell division and varies with the microbe)
-pathogens often produce virulence factors at this time
-primary metabolites
Describe the late log phase of bacterial growth curve.
-transition to stationary
-secondary metabolites (antibiotics, pigments)
Describe the stationary phase of the bacterial growth curve.
-supplies of energy and nutrients exhausted
-resources are then renewed by cell death (release peptides and nucleic acids)
-zero population growth (no change in CFU/mL, OD, biomass)
Describe the death/decline phase of the bacterial growth curve.
-insufficient resources for growth
-exponential cell death (but still less than the growth rate in log phase)
-determined only by CFU/mL not OD or biomass
Describe the death/decline phase of the bacterial growth curve.
-insufficient resources for growth
-exponential cell death (but still less than the growth rate in log phase)
-determined only by CFU/mL not OD or biomass
Describe the stationary phase of the bacterial growth curve.
-supplies of energy and nutrients exhausted
-resources are then renewed by cell death (release peptides and nucleic acids)
-zero population growth (no change in CFU/mL, OD, biomass)
Describe the late log phase of bacterial growth curve.
-transition to stationary
-secondary metabolites (antibiotics, pigments)
Describe the log phase of the bacterial growth curve.
-exponential growth occurs in this phase, rapid cell division
-slope reflects doubling/generation time (determined by the rate of cell division and varies with the microbe)
-pathogens often produce virulence factors at this time
-primary metabolites
Describe the death/decline phase of the bacterial growth curve.
-insufficient resources for growth
-exponential cell death (but still less than the growth rate in log phase)
-determined only by CFU/mL not OD or biomass
Describe the lag phase of the bacterial growth curve.
-there is no cell division here
-increase in biomass from synthesis of macromolecules
-the length depends on form and availability of nutrients and the condition of the inoculum
Describe the stationary phase of the bacterial growth curve.
-supplies of energy and nutrients exhausted
-resources are then renewed by cell death (release peptides and nucleic acids)
-zero population growth (no change in CFU/mL, OD, biomass)
What is biomass?
-measures bacterial growth
-sample is washed, dried, and weighed, dry weight is taken
-measures all cells in culture (viable and non-viable)
Describe the late log phase of bacterial growth curve.
-transition to stationary
-secondary metabolites (antibiotics, pigments)
What does a colony forming unit measure?
only VIABLE cells
-from diluting and culture and plating it, each colony is from one cell
Describe the log phase of the bacterial growth curve.
-exponential growth occurs in this phase, rapid cell division
-slope reflects doubling/generation time (determined by the rate of cell division and varies with the microbe)
-pathogens often produce virulence factors at this time
-primary metabolites
Does the optical density measure viable or non-viable cells?
both
-the more cells, the more absorption occurs or the light
Describe the lag phase of the bacterial growth curve.
-there is no cell division here
-increase in biomass from synthesis of macromolecules
-the length depends on form and availability of nutrients and the condition of the inoculum
What is biomass?
-measures bacterial growth
-sample is washed, dried, and weighed, dry weight is taken
-measures all cells in culture (viable and non-viable)
What does a colony forming unit measure?
only VIABLE cells
-from diluting and culture and plating it, each colony is from one cell
Does the optical density measure viable or non-viable cells?
both
-the more cells, the more absorption occurs or the light
Define sterilization.
a physical or chemical process that destroys or removes all microbial life, including spores
Define disinfection.
a physical or chemical process that eliminates nearly all microbial life, but some forms may persist (ex. spores)
Define decontamination.
a physical or chemical process that reduces microbes to an acceptable level (not able to cause disease)
What is antigenic drift?
-the accumulation of mutations which alters protein product
-over time leads to variability in population and forms antigenically distinct strains
-can produce an unlimited number of strains in a spp.
-
Describe the example of antigenic drift relating to the influenza virus.
hemagluttinin (glycoprotein spike that aids in attachment to host) and neuraminidase (enzyme on spike, allows virus to release from host)
-these 2 molecules undergo drift over time and can change from year to year
-this is responsible for the yearly flu vaccine
Describe the example of influenza A and antigenic shift.
-reassortment of viral genome when cell (pig) is infected by 2 different strains of the flu A virus
-this can produce a new subtype and lead to possible pandemic b/c there is no immunity to the new flu A strain
-this happens quickly and is drastic
What is phase variation and what is it an example of?
-one type of antigenic switching
-it refers to the genetic reversible ability to turn off and turn on something
-usually happens to genes coding for surface antigens (capsule, pili, flagella, LPS), these genes are always part of genome
-can be regulated by site specific inversion of DNA sequence by recombinase or posttranscriptionally by stability of mRNA
Describe a clinical application of phase variation.
uropathogenic E. coli (UPEC)
-uses fimbriae to attach to host tissue during colonization of bladder
-during phase ON it produces fimbriae and attaches to uroepithel.
-during phase OFF it does not produce fimbriae and remains free floating in urine
What is gene conversion/cassette mechanism of antigenic switching?
-it is due to recombination within specific group of genes
-results in alteration of surface antigen characteristics but maintains biological function
-also known as "gene shuffling"
-a surface antigen expresses different antigenic structures
example: N. gonorrhoeae has 20 genes that code for pilin, majority are not expressed but shuffling and recombining chromosomal regions leads to multiple antigenically different pilin with different AA sequences
What is relapsing fever?
disease caused by Borrelia recurrentis (a spirochete)
-it expresses variant surface ags
-due to gene conversion/cassette mechanism
-body responds with a fever each time the organism "gene shuffles" and expresses a new variant of its own epitope on the surface ag
Describe the example of Plasmodium falciparum using the cassette mechanism.
it is a protozoa which causes malaria
-spread through the anopheles mosquito
-this organism generates variable surface glycoproteins (VSG) which is a detectable ag on surface of RBC-->hides from immune system
Describe the example of Trypanosoma brucei using gene conversion/cassette mechanism.
it is a protozoan that causes African sleeping sickness
-vector is the tsetse fly
-produces VSG which leads to it hiding from the immune system
What is homologous recombination?
-sequence similarity
-transformation, transduction, and conjugation are some types
-newly obtained DNA replaces homologous DNA on chromosome
example: "naked DNA" codes for a lipase gene which is taken up by a host cell from environment, new lipase ssDNA aligns with resident lipase and rec proteins carry out recombination
-the old chromosomal DNA is replaced by NEW homologous DNA
What is nonhomologous recombination?
-site directed recombination, NO rec proteins involved as in homologous
-results in addition to NOT replacement of chromosomal DNA
-can disrupt a gene, if insert into the promoter or coding sequene of gene (NULL mutation)
What are the 3 mobile genetic elements?
1)insertion sequence
2)composite transposon
3)pathogenicity island
-does NOT mean they replicate on their own, they don't!
-all undergo nonhomologous recombination
-they can transfer DNA within a cell from one position to another or transport DNA from one cell to another
Describe an insertion sequence.
-the simplest form of mobile genetic element
-made of 150-1500 bp of DNA
-requires transposase gene integration into replicon
-has inverted repeats at end of molecule, does not carry additional coding regions/genes
-inserts in specific sequence and creates direct repeats upon insertion
What is a transposon?
-has 2 insertion sequence sequences at either end of the molecule
-found in prok. and euk.
-can carry genes for antibiotic resistance, toxins,
-each gene=promoter + coding sequence
-can insert into and inactivate a gene (including chromosomes or plasmids) and if that gene was essential =cell death
What is a pathogenicity island?
-distinct genome segments of pathogens which encode virulence factors
-unusual clusters of genes (10-200 kb in length) inserts in tRNA and tRNA like genes
-has direct repeat DNA motifs at boundaries as other MGEs do, and insertion elements within sequence
-genes carried in PAI have a different %G+C than parent cDNA
-overtime unstable and is lost, these are not found on nonpathogenic forms of the same spp.
ex: can create different pathogens of same bacterial spp. as in Enterotoxic E. coli and uropathogenic E. coli
What are plasmids?
-small genetic elements which replicate independently of chromosome
-supercoiled CCC ds DNA which vary in size
-carry information that may not be essential but can confer a selective advantage such as antibiotic resistance, heavy metal and toxin resistance, virulence factors, etc.
-
What is an "R plasmid"?
-AKA representative plasmid
-carries 1 or more antibiotic resistance and contains resistance transfer factor genes that mediate the transfer of the plasmid from 1 cell to another
What are episomes?
plasmids that can integrate into the hosts chromosome
What is the exception to plasmid and chromosome structure?
borrelia burgdorferi-causative agent Lyme disease
-has linear and circular plasmids, linear chromosomal DNA
What are the plasmid encoded virulence factors that we discussed in class (3)?
1)tetanus neurotoxin-clostridium tetani
2)exfoliative toxin-staph aureus
3)enterotoxins-E. coli

TET, EET, SET
What is a bacteriophage?
-species specific and utilizes host cell machinery to complete replication of vDNA, assemble viral particles
-uses a lytic or lysogenic cycle to produce new virions
What are the 3 ways in which gentic exchange of DNA can occur among bacteria?
1)transformation
2)conjugation
3)transduction
What is transformation?
-acquisition of genetic information
-incorporation of exogenous/naked DNA into cytoplasm (plasmid) or chromosome of recipient cell by homologous recombination
-usually develops a log phase during lab cell culture b/c takes time to occur

-
What is conjugation?
mating or quasisexual exchange of genetic information
-one bacterium to another
-occurs with most if not all eubacteria, usually bacteria of same spp. or closely related
-requires contact btwn. 2 bacteria
-results in the ONE way transfer of DNA from donor to recipient
-genes that direct conjugation are carried on conjugative plasmids which may also carry antibiotic resistance (carries genes for transfer of plasmid, ability to make sex pilus, and initiate DNA synthesis)
-transfer of conjugative plasmid as linear ssDNA, rolling circle replication produces dsDNA plasmid in recipient
What is transduction?
transfer of bacterial DNA from one bacterium to another by a bacteriophage (virus)
-phage picks up fragments of bDNA and packages it into phage particles/virions
-the bDNA is then delivered to infected cell and incorporated into bacterial genome
-
What type of secretory system does conjugation use?
type IV
-carries out process of DNA transfer
-identified in gram pos and neg bacteria
Describe rolling circle replication.
mobilization begins with plasmid-endoded protein makes single-stranded site specific cleavage at oriT
-Nick initiates rolling circle replication
-the dislaced linear strand is then directed to the recipient
-transferred DNA recircularizes and replicates
Describe conjugative gram negative bacteria.
have a specialized sex pilus-conjugative pilus
-example: E. coli where pilus is produced by donor
Describe conjugative gram-positive bacteria.
no sex pilus
-adhesin molecules on surface of donor aid in contact establishment
Describe the structure of bacteriophages.
capsid-protein shell
tail with/without tail fibers
nucleic acid core (RNA or DNA)
Describe the lytic cycle of virulent phages.
-lytic cycle is key to generalized transduction
-during phage assembly of lytic cycle DNA packaging does not discriminate (bDNA vs. viral DNA)
-capsid filled bDNA (defective phage), does not contain vDNA
-the phage then attaches and injects bDNA, after injection generalized transduction completed and bDNA recombines with host chromosomal DNA
-the phages are host cell specific and therefore carry out homologous recomb.
Describe the lysogenic cycle of the temperate phage.
-this cycle is the key to specialized transduction
-the phage attaches, injects/pentrates, lysogeny then occurs in which there is integration of vDNA into bDNA/chromosome in site directed nonhomologous recombination-->latent prophage formed vDNA (lysogenic conversion of bacterial cell turns it into a lysogen)
-viral DNA replicates w/bacterial host DNA and it is passed to bacterial daughter cells
-until stress activates prophage and the lytic cycle occurs
Describe specialized transduction.
lysogenic cycle is key to this
-during phage assembly of stress-induced lytic cycle the prophage is incorrectly excised from bDNA which incorporates bDNA located next to integration site of vDNA
-defective phages are produced when the capsids are packed which includes vDNA and bDNA, lysis releases the phages and they infect new host cells -->specialized transduction occurs of the bDNA
-if the prophage carries a virulent gene then a pathogenic lysogen is produced
What is mutualism?
each member provides benefits for other
What is commensalism?
one member benefits and the other is not harmed
example: strep pneumo
neisseria meningitidis
-virulence in these 2 not normally turned on
What are some benefits of normal microbiota?
1)synthesize and excrete vit. K and B12 (enteric bacteria)
2)prevent colonization by pathogens (competition for resources)
3)antagonize pathogens (produce substances to inhibit or kill)
4)stimulate development of tissues
5)stim. prod. of cross-reactive abs (behave as ags, induce Ab-mediated immunity, ex:E. coli ab cross reacts with H.influenzae type B to prevent infection)
What gram staining group predominates skin colonization?
gram positive
-gram negative predominate mucous membranes, lower intestine, and colon
What is a sign?
any objective evidence of disease as noted by an observer (changes due to disease, observed by a clinician)
What is a symptom?
the subjective evidence of disease as sensed by the patient (changes due to disease, experienced by the patient)
What entity is both a sign and a symptom?
fever!
What microbe causes strep throat?
strep pyogenes
What microbe causes dental caries?
strep mutans
What are the 5 stages of disease?
incubation
prodromal
acute
decline
covalescent
Describe stage 1 of disease-Incubation (preclinical stage).
-the pathogen has entered the body through the "portal of entry"
-NO signs or sx
-innate immune system has NOT been activated
-the pathogen must be an infectious dose/inoculum to gain entry and inititate growth
-the person is NOT infectious under most circumstances in this stage
What does inoculum mean?
the number of microbes required for disease
Describe stage 2 of disease-prodromal (warning stage).
-appearance of signs and sx of an illness, pathogens generate early signs and sx such as rashes, headaches, etc (general malaise)
-activation of the innate immune system
-numbers have increased since incubation stage
-easily transmitted even before host realizes they have the pathogen
Describe stage 3 of disease-acute stage. (also called clinical illness)
-most severe during this period = ACME of the illness
-signs and sx characteristic for the disease
-balance btwn host immune system and pathogen virulence, either tipped to recovery or death, acquired immune system has been activated
-pathogen numbers are stationary and may increase or decrease
-communicable disease are easily transmitted in this stage
Describe stage 4a of a disease-decline stage.
-the illness is still apparent but the signs and sx dwindle
-immune system activity is reduced
-pathogen is cleared from the host
-the disease is contagious if the individual becomes a carrier (ex. S aureus in nasal passages)
Describe stage 4b of a disease (alternative decline stage in which the pathogen is not cleared from the host).
-the pathogen is not cleared and instead does one of the following:
1)becomes dormant-mycobacterium tb results in latent infection
2)undergoes antigenic variation-presents immune system with unknown ag such as borrelia recurrentis-results in relapse of infection
3)resists antimicrobial tx-staph aureus (MRSA)-results in recurrent infection
-the individual is communicable is they re-enter the prodromal stage
Describe stage 5 of a disease-convalescent.
-pt. is now returning to full health
-see end of signs and sx
-no activity by immune system toward pathogen
-pathogen cleared from host
-not communicable since NO presence of pathogen (unless that person becomes a carrier)
What are opportunistic pathogens?
organisms that can be members of pts. normal flora and do not produce disease in normal setting but can establish disease when introduced to an unprotected site (such as blood tissues, etc)
-pt.subject to trauma, w/defective immune system, or preexisting condition more susceptible

ex: E. coli, S. aureus, C. difficile
What are some of the most notable examples of opportunistic pathogens?
1)P. aeruginosa-skin of burn victim or lungs of CF pts.
-gram neg. bacillus, motile, obligate aerobe
2)nocardia asteroides-resp. infections in immunosuppressed or compromised ppl
-gram pos. obligate aerobe, weakly acid fast
What are virulent bacteria (strict pathogens)?
-always associated with disease
-signs and sx due to damage or loss of tissues and organs, also host inflammatory response
-ex: mycobacterium tb, N. gonorrhoeae
Describe how bacteria use tissue damaging metabolites to create pathogenesis.
-they use acid, gases, or other byproducts formed during growth to harm their host

ex: Clostridium perfringens-produces hydrogen gas which inflates and separates the tissues so the organism can move through the tissues
Describe how bacteria use invasins or spreading factors to create pathogenesis.
-can use enzymes/proteins which act locally to damage host cells or affect tissue matrices and intracellular spaces
-these facilitate growth and spread of the pathogen which can lead to a certain pathology
-ex: neuraminidase, collagenase, etc
What does hyaluronidase do?
used by bacteria, attacks the interstitial cement of CT and depolymerizes hyaluronic acid
What does neuraminidase do?
used by bacteria, degrades neuraminic acid which is the intercellular cement of epithelial cells of the intestinal mucosa
What do streptokinase and staphylokinase do?
used by bacteria
-they are fibrinolysins that prevent the clotting of blood
What are adhesins?
-they allow pathogens to bind to receptors on host cells and tissues (bacterial ligand + host cell receptor)
-most bind to carb moieties like glycoproteins
ex: vibrio cholerae and e.coli attachment to brush border cells of host villi in sm. intestine
Describe how strep. mutans adheres to the enamel of teeth.
produces a glycocalyx material called dextran (complex-branched chain of glucans) by secreting glucosyltransferase which binds the bacteria to enamel of teeth
-this is a type of adhesin
What is a biofilm and describe an example of the organism that can colonize IV catheters.
a biofilm is colonization of inert materials and organic materials (like catheters and native heart valves)
ex:staph. epidermidis forms a biofilm on IV catheter, this is continual source of infection
Describe an organism that uses fimbriae for attachment to host molecules.
uropathogenic strains of E. coli use P fimbriae that bind to epithelium of the kidneys
What are afimbrial adhesins and what are some examples of microbes that use these?
-they are proteins in the cell envelope of bacteria, have mediated binding specificity, used to adhere to hose organism
-use type V secretory system
examples: Staph. aureus and Strep. pyogenes-use fibronectin binding proteins that recognize the host cell glycoprotein (called MSCRAMMs and are a class of microbial surface components that bind fibronectin)
What is an exotoxin?
-they are proteins that are toxic to cells, usually by direct action
-they are secreted into the ECF or associated with bacterial cell surface
-have components which bind host cell receptors
-they are produced by both gram pos and neg. cells
What is an enterotoxin?
an exotoxin that produces GI sx
-example: s. aureus causes vomiting and diarrhea
What are functional exotoxins?
they are immunogenic and deadly at extremely low concentrations
What are toxoids?
denatured toxins that are used for immunization
-ex: diphtheria toxoid vaccine
tetanus toxoid vaccine (dTaP)
Describe the A-B exotoxins.
-have 2 domains called A and B
A is the enzymatic component which attacks and enters cell
B binds the toxin to the host cell receptor
-B binds, then A attacks
What is the mechanism of Corynebacterium diphtheriae?
lysogenic conversion of host cell, diptheria toxin stops protein synthesis of host cell -->leads to cell death
-uses AB exotoxin
Describe the mechanism of action of E. coli relating to exotoxins.
uses the AB exotoxin class
-plasmid that acts as an enterotoxin which stim. Adenylate cyclase and causes diarrhea
Describe the mechanism of Clostridium botulinum.
uses AB exotoxin system
-lysogenic conversion of host cell, the botulinum toxin prevents release of Ach at synapse, no mm.contraction-->flaccid paralysis
Decribe the mechanism of Clostridium tetani.
uses the AB exotoxin system
-plasmid that contains the tetanus toxin and prevents inhibitory NT release-->no mm. relaxation and spastic paralysis occurs
Describe the membrane active toxins (type of exotoxin).
they are not as well characterized as AB toxins, they attack host cell membrane and lyse or kill host cell
-examples:
1)proteases-elastase in cell membrane of P. aeruginosa (type II secretory system)
-metalloprotease of legionella pneumophila
2)lipases-C. perfringens-alpha toxin-lecitinase in cell membrane
Through what secretory system do shigella and salmonella enter host cells?
type III
What uses the type I secretory system in bacteria?
hemolysins
What are superantigens? Give examples.
-they activate 1 in 5 T cells in the absence of antigen
-cause release of massive amounts of cytokines which leads to a possible life-threatening response and T cell death
examples: S. aureus toxin TSST-1
S. pyogenes-pyrogenic exotoxin A
What are endotoxins?
-refers to the LPS of gram-negative bacteria
-heat stable, toxicity associated with lipid A
-immunogenicity associated with polysaccharide components
-elicits variety of inflammatory responses, activates complement (alternative pathway)
-leads to fever, hypotension, shock and possible death
-NO TOXOIDS TAKEN FROM THESE
Describe some examples in which common microbes invade normally sterile tissues.
1)shigella dysenteriae-resists lysozyme, acid and bile, colonizes GI tract
2)E.coli and Bacteroides fragilis-enter sterile peritoneal cavity by appendix rupture or endoscopic puncture
Describe an example of a bacteria that can circulate via blood or other means spreading from primary infection site.
-in general, bacterial toxins and other antigenic molecules which are transported by blood and lymph can cause cytotoxic effects at remote sites
example: S. aureus-scalded skin syndrome (SSS), exfoliative toxin from desquamation of upper epidermis which then goes to umbilicus or conjunctiva
Describe encapsulation and how bacteria can use this mechanism to evade the host immune response.
-the bacteria is covered with poorly antigenic polysaccharide which deters phagocytosis
-this protects it against degradation in phagolysosomes of macrophages
ex: Strep. pneumoniae
Describe how some bacteria can use the mechanism of intracellular growth to evade the host immune response.
-there is no inactivation of host cell so the microbe escapes immune detection
examples: Rickettsia-obligate intracell.
Mycobacterium:facultative intracellular organism
Describe an example of a bacterium that can reduce phagocytic cell function to evade the host cell immune response.
example: Strep. pyogenes-has streptolysins which are cytotoxins that kill phagocytic cells
What are examples of bacteria that can inactivate antibody to evade the host immune response.
1)S. pneumoniae-IgA degrading proteases
2)S. aureus-surface protein A which binds Fc portion IgG
What are examples of how some bacteria can inhibit complement to evade the host cell immune response?
1)O antigen of LPS protects cell from complement mediated lysis
2)antigenic variation
What are siderophores and what are their function?
-they chelate available iron and transport it into the cell
-scavenge from host iron-binding proteins (lactoferring and transferrin)
-the lack of iron in the host can INDUCE toxin production
examples:
1)corynebacterium diphtheriae
2)P. aeruginosa-exotoxin A
(both are AB toxins-stop protein synthesis)
What is a major contraindication for using bacteriostatic drugs?
should not be used on immunocompromised patients or for infections in privileged sites (in which immune cells cannot invade and clear infection)
-example: HIV pts, pregnant women
What are some examples of narrow spectrum antibiotics?
erythromycin, vancomycin, clindamycin
What are some examples of broad spectrum antibiotics?
amoxicillin, tetracycline, streptomycin, chloramphenicol
Describe the difference btwn. euk. and prok. ribosomes.
euk=80s, 60+40

prok.=70s, 30+50
What is the molecule found in yeast that is structurally very similar to cholesterol?
ergosterol
What is empirical therapy when talking about antimicrobials?
predicts the sensitivity of the organism to antimicrobials
What is rational therapy?
antimicrobials prescribed against specific known organism
Describe the B-lactam antibiotics.
they are cell wall synthesis inhibitors (and therefore considered bactericidal)
-penicillins, cephalosporins
-they are all structurally related by possessing a B-lactam ring
-they block the transpeptidation step in bacterial cell wall synthesis (no cross linkage of tetrapeptides)
-
What is the function of beta-lactamase?
enzyme that inactivates penicillin
Describe penicillin G.
-narrow spectrum, inactivated by B-lactamase
-often combined with probenecid to decrease tubular excretion rates
-sensitive to acid hydrolysis
-has a very short half-life
Describe Penicillin V.
-more stable to acids
-rarely used clinically (b/c resistance is common)
-poor bioavailabillity
-little toxicity and short half-life
Why were synthetic penicillins developed?
-increase stability to acids
-to be resistant to B-lactamase
-to expand the spectrum of activity
3 groups are:
1)penicillinase-resistant penicillins
2)extended spectrum penicillins
3)anti-pseudomonal pens
Describe penicillinase resistant penicillins.
more toxic than natural penicillins
-resistant to B-lactamase
-still narrow spectrum of activity, used for gram pos. and some staph infections
examples:flucloxacillin and dicloxacillin
Describe extended spectrum penicillins.
-spectrum of activity expanded to many gram neg, less effective on gram positives
-acid stable
-sensitive to B-lactamase
-examples: ampicillin, amoxicillin
Describe the anti-pseudomonal penicillins.
-spectrum of activity exapdned
-highly effective ongram neg. rods, less effective on gram pos.
-administered parenterally (not orally)
-sensitive to B-lactamase
-examples: piperacillin, carbenicillin
What are combination products and give an example.
a penicillin + a penicillin analog
-they expand the spectrum of activity
one example is clavulanic acid which is used in combo with amoxicillin (drug called augmentin)
-the clavulanic acid has a b-lactam ring that serves as a competitive inhibitor and slows breakdown of amoxicillin
Describe cephalosporins in general.
-they are bactericidal
-usually more resistant to B-lactamase than penicillins
-there are 4 generations
Describe 1st generation cephalosporins.
-fairly broad spectrum, work on gram pos. cocci except MRSA and enterococci, some gram neg. enterics.
-not often used as first drug of choice!
Describe 2nd generation cephalosporins.
-extended spectrum, more gram neg. but weaker against gram pos.
-
Describe 3rd generation cephalosporins.
-broad spectrum, used mainly for gram neg.
-increased resistance to b-lactamase
-some can cross BBB (can be used for meningitis)
Describe 4th generation cephalosporins.
-extended spectrum: P. aeruginosa and some gram pos. cocci (effective in meningitis cases)
-increased resistance to b-lactamase
-cross BBB, parenterally admin.
Describe bacitracin.
a cell wall synthesis inhibitor and therefore is bactericidal
-effective on gram pos. and gram neg. cocci
-used topically due to toxicity
-interferes with movement of PG precursors through the cell membrane and so precursors never even reach the cell wall, works upstream of penicillin effects
Describe vancomycin.
a cell wall synthesis inhibitor
-narrow spectrum, effective on gram pos. cocci, used mainly for methicillin resistant organisms and pts. allergic to B-lactam antibiotics
-has toxicity issues (ototoxicity, nephrotoxicity, red man syndrome from increased histamine release)
-binds to terminal peptides so can't cross link and build cell wall
Describe polymyxins.
-work by disrupting cell membranes, found in OTC antibiotic topicals
-effective against gram neg. bacteria
-act as cationic detergents and increase water uptake by cell, efflux of ions and cell will lyse
-toxic to host, used on localized, external infections
Describe daptomycin.
-disrupts bacterial cell membrane function
-irreversible binding to cell membrane results in rapid depolarization, efflux of K+
-inhibits RNA, DNA, and protein synthesis=bacterial cell death
-used for gram pos.
-inactivated by pulmonary surfactants and therefore not used for tx of pneumonia
-toxicity risk in elderly esp.
What are the 2 classes of 30s ribosome subunit inhibitors?
aminoglycosides and tetracyclines
What are the 50s ribosome subunit inhibitors?
chloramphenicol, macrolides, clindamycin, streptogramins, linezolid
Describe aminoglycosides.
-they are bactericidal
-broad spectrum-often used for enterics, used synergistically with b-lactams (ex: streptomycin, gentamicin)
-irreversible binding to 30s subunit, interference with initiation complex leads to misreading of mRNA and aberrant proteins-->premature release of mRNA from ribosomes leads to incomplete protein
Describe tetracyclines.
-they are bacteriostatic
-broad spectrum used mainly for intracellular bacteria, enterics, some protozoa and UTIs
-NOT given to children (can penetrate bone) or pregnant women
ex: doxycyline, etc.
-reversilby bind to 30s ribosomal subunit
-block binding of aminoacyl tRNA to the acceptor site on the mRNA ribosome complex
Describe chloramphenicol.
-is bacteriostatic
-broad spectrum antibiotic used only for certain infections (typhoid, meningococcal)
-it is toxic
-binds to 50s ribosomal subunit reversibly and inhibits peptidyl transferase and prevents elongation of peptide chain
-poor selective toxicity
Describe macrolides.
-are bacteriostatic broad spectrum that are readily used
-ex:erythromycin, azithromycin, etc.
-binds to 50s subunit reversibly
-block formation of the initiation complex and translocation step in protein elongation
Describe clindamycin.
-mechanism similar to macrolides (binds to 50s), bacteriostatic
-useful against some MRSA strains, anaerobes and some protozoa (malaria)
-used topically for tx of acne
-moderate toxicity issues
Describe streptogramins
-combo drug
-each drug binds distinct sites on 50s ribosome
-they are bacteriostatic individually, but bacteriostatic in combo
-used in tx of most gram pos. bacteria, resistant E. faecium, some MRSA
Describe linezolid.
-prevents formation of inititation complex
-can also bind "A" site on ribosome complex if formed
-effective against gram pos., but also used on vancomycin resistant organisms
-relatively safe drug
Describe rifamycins (rifampin).
-is bactericidal, effective against M. tuberculosis and some gram pos. cocci
-used in combo usually with isoniazid
-inhibits DNA dependent RNA polymerase
Describe fluoroquinolones.
-synthetic antibiotics initially used primarily for UTIs
-are bactericidal
-broad spectrum against many gram neg. aerobes
-inhibit DNA gyrase
ex:cipro, norfloxacin
-moderate tox. issues, should not be given to kids or preggers women
Describe metronidazole.
-is bactericidal
-narrow spectrum: used for anaerobes and protozoans
-reduction of metronidazole results in cytotoxic substances formed that disrupt DNA
Describe sulfonamides.
-they are analogs of PABA (one of precursor to make folic acid in bacteria)
-bacteriostatic, broad spectrum
-compete with PABA for dihydropteroate synthase which inhibits folic acid synthesis, bacteria need this for synthesis of nucleic acids and therefore can't make DNA
Describe trimethoprim.
-analog of the pteridine portion of dihydrofolic acid
-bacteriostatic
-usually used in combo with sulfa drugs (bactericidal in combo) -CALLED TMPSMZ
-inhibits the enzyme dihydrofolate reductase, inhibits folic acid synthesis =no DNA synthesis
Describe dapsone.
-mechanism identical to sulfa drugs
-bacteriostatic
-used in tx of leprosy and pneumocystis carinii pneumonia
-toxicity b/c blocks cytochroms p450 function, can result in toxic hepatitis
Describe isoniazid.
-analog of nicotinamide and pyridoxamine
-bactericidal against growing organisms
-used for tx of TB
-interferes with synthesis of mycolic acids
What 2 groups are antifungal agents divided into?
polyenes and azoles
Describe nystatin.
a polyene antifungal
-too toxic for systemic use, not absorbed from alimentary canal
-used for mucosal candidiasis and GI fungal infections
-interact with sterols in the cell membrane to form channels through which small molecules leak from inside of the fungal cell to the outside
Describe amphotericin.
-polyene antifungal
-can be used IV
-used to tx of systemic candidiasis and exotic mycoses
-similar mode of action to nystatin and can be very toxic to humans
Describe miconazole.
-antifungal azole
-interferes with cell membrane formation (blocks synthesis of ergosterol)
-dual action against fungi and some gram pos. cocci
-resistance rare
Describe fluconazole.
-wide spectrum of activity on yeasts and other fungi
-used to prevent Candida infection in HIV pts.
-minor toxicity
Describe intraconazole.
-similar to fluconazole, with a broader spectrum and increased toxicity
-inhibits the fungal cyt. P450 oxidase mediated synthesis of ergosterol (precursor to Vit. D)
-no penetration in CNS
Describe terbinafine.
an antifungal, allylamine drug that blocks ergosterol synthesis in fungal cell membrane
-used in dermatophyte infections (parasitic fungal infections)
-used as a topical and tablet, risk of hepatic toxicity with admin. as tablet
ex:lamisil
Describe echinocandins.
-used in systemic candidiasis and aspergillosis
-inhibits production of glucan in cell wall (targets cell wall directly)
-known as penicillin of antifungals
Describe how bacteria can become resistant through altering drug targets. (ex ribosome mutation)
the bacterium makes a protein product with an altered binding affinity for the drug
-often modifies the bacterial ribosome
-resistance to erythromycin and rifamycin occurred this way
What 3 organisms are becoming resistant to everything?
Enterococcus faecalis
mycobacterium TB
pseudomonas aeruginosa
What 2 organisms are indicated in otitis media?
turicella otitidis (bacillus) and alloiococcus otitis
-both are aerobes found in external auditory canal
What organisms are predominantly found in healthy sinuses?
healthy sinuses are sterile due to the mucociliary blanket
What was historically called doderlein's bacillus?
lactobacillus acidophilus (found in vagina)
What are the predominant bacteria of the vagina and what is their significance?
lactobacillus acidophilus and corynebacterium sp
-they metabolize glycogen to lactic acid or acetic acid
-the resulting low pH of the vagina prevents grwoth of most bacteria (including potentially pathogenic yeast candida)
What microbe dominates the oral microbiota from birth until the appearance of teeth?
streptococcus salivarius
-makes up 98% of total oral microbiota until appearance of teeth
What changes occur in the oral microbiota after the eruption of teeth?
-leads to colonization by S. mutans and S. sanguis
-require a nondesquamating/nonepithelial surface in order to colonize
-these organisms will persist as long as teeth enamel remains
-also creation of the gingival crevice area increases habitats for anaerobic spp such as prevotella, fusobacterium, veillonella)
What changes occur in the oral microbiota at puberty?
the complexity of the biota increases with time
-bacteroides and treponemal spirochetes last to colonize
What microbe usually plays a dominant role in causing abscesses of alveolar bone, lung, brain or extremities after gaining entrance via surgical wounds through the mouth?
prevotella melaninogenicus

-also these wounds can introduce oral strep (like strep. mutans) which adheres to damaged heart valves and initiates bacterial infective endocarditis
Describe the microbiota in the GI tract.
-differences in composition are influenced by age, diet, and cultural conditions
esophagus: contains only bacteria swallowed with saliva and food
stomach:few bacteria here, too acidic,mainly acid-tolerant lactobacilli (at least 1/2 pop. in US has helicobacter pylori-gastric ulcers)
small intestine:proximal has sparse gram pos. including lactobacilli and enterococcus faecalis, distal portion-more bacteria including coliforms and bacteroides
large intestine:similar to feces, coliforms group, enterococci, clostridia, and lactobacilli found
-predominant spp. are anaerobic bacteroides, anaerobic lactic acid bact. in genus bifidobacterium
-also significant numbers of anaerobic methanogenic (Archaea) bacteria in colon
Describe the ecological succession of the GI tract.
-at birth, entire tract is sterile until first feeding
breast fed infants: 90% intestinal bacteria are bifidobacteria (human milk contains Growth factor for this), low proportions of enterobacteriaceiae and enterococci
bottle fed: bifidobacteria are NOT predominant
cow's milk or solid food: bifidobacteria joined by enterics, bacteroides, enterococci, lactobacilli, and clostridia
What are the normally sterile areas of the body?
body fluids, tissues (bone, mm. , blood, CT), upper urinary tract including kidneys and bladder
What are coliforms?
associated with bacteriologically polluted water and consist of a group of related bacteria
total coliforms: (CEEK) citrobacter, Escherichia, enterobacter, klebsiella
What are enterics?
gram neg. bacilli
-enterobacteriaceae are C PESSKEY Strains of bacteria
-citrobacter, proteus, escherichia, salmonells, shigella, klebsiella, enterobacter, yersinia, serratia

other pathogenic enterics include: vibrio, campylobacter, helicobacter
What 2 methods of moist heat do not reliably kill endospores?
boiling and pasteurizing,
autoclaving does kill endospores
What type of UV light is used as a germicide?
UVC-short
doesn't penetrate the atmosphere, used to sterilizing surfaces, glass/plastic labware, and thin layers of liquid
-damages DNA and creates thymine dimers
-PPE for eyes and skin recommended
What is a high level disinfectant?
chemical sterilization
-kills all microorganisms but will not effectively kill large numbers of bacterial spores or prions
-used for heat sensitive critical and semi-critical equipment
-standard procedures not effective against prions
What is an intermediate level disinfectant?
kills mycobacteria, vegetative bacteria, most viruses and fungi, but not spores
-used on some semi-critical and non-critical equipment
What are the phenolic compounds?
ex: triclosan which is common in antibacterial soaps, also found in deodorant, toothpaste, etc

ex: listerine-4 phenolics +alcohol
-denature proteins and disrupt membranes, surface disinfectant, personal hygiene products, not effective against spores and non-env. viruses
Describe Quaternary ammonium compounds.
ex: benzalkonium chloride
-antiseptic towelettes and contact lens solutions
-they denature intracell. components
-disinfections of non-critical surfaces
-not effective against spores and viruses
What are some compounds you can use when you can't autoclave? (endoscopes, resp. equip., etc)
-aldehydes
-ethylene oxide (gas-explosive)-long sterilization times and post-steriliazation aeration required to remove toxic residues
-peroxides-peracetic acid=hydrogen peroxide+acetic acid-stronger biocide than H2O2 alone
What can be used to disinfect medical equipment surfaces?
alcohols
-hypochlorites
-iodine tinctures/iodophores (antiseptic iodophores not suitable for use a disinfectants)
What can be used for disinfection of non-critical surfaces?
phenolic compounds (as found in lysol)
-quaternary ammonium compounds
-hypochlorite-.1% chlorine can be used for high level disinfection with contact time of about 20 mins.
What are the 2 main functions of wall teichoic and lipoteichoic acids in gram positive bacteria that help to increase the virulence?
1)they function as adhesins (for invasion of host cells)
2)can initiate endotoxin like activities when released
What are the 2 common acid fast genera?
nocardia and mycobacterium
What 2 pigments does the acid fast stain process use?
carbol fuschin and methylene blue
What are the 2 clinically relevant genera that produce endospores?
clostridia and bacillus
-both gram positive
What is MacConkey agar?
it is differential and selective
contains peptone and lactose
-bile salts which inhibit Gram +
-crystal violet inhibits Gram +
neutral red is the pH indicator
What are the 3 basic categories of growth factors for bacteria?
1)purines and pyrimidines-nucleic acid synthesis
2)AAs-protein synthesis
3)vitamins-act as coenzymes
What are mesophiles?
most pathogenic bacteria are this type
-optimum temp for survival and growth is 30-37 C
-survive best in moderate temps
What are obligate Psychrophiles/cryophiles?
survive best in temps under 20 degrees celsius
-deep sea, cold marine environments, etc
What are facultative psychrophiles/psychrotrophs?
survive best in temps between 0-35 Celsius
-not found in extremely cold condition, can live in fridge
What is a microaerophile?
requires oxygen, but at reduced levels
What is an aerotolerant anaerobe?
does not require O2 and does not use O2 but has enzymes for ROS usually
What is a facultative anaerobe?
does not require O2 but will use it if present
What does an organism with a BSL-1 classification signify?
these are agents that are not known to consistently cause disease in healthy adults
What does BSL2 classification mean?
agents associated with human disease
-transmission by ingestion, percutaneous injury, or mucus membrane exposure
What does BSL3 classification of microbes mean?
indigenous or exotic agents associated with serious to fatal human disease with the potential for aerosol exposure
What does BSL4 classification of microbes mean?
dangerous/exotic agents with a high potential for causing life-threatening disease, potential aerosol transmission or the risk of infection is unknown, but related to agents with aerosol transmission
What are standard precautions?
-they protect both pts. and health care workers,
-prevent spread of infectious organisms from pt. to pt., environment to pt., health care worker or pt., pt. to health care worker, environment to health care worker