• 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

1/322

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;

322 Cards in this Set

  • Front
  • Back
1. What are topoisomerases?
These enzymes catalyze DNA replication and segregation by breaking, rotating, and religating DNA strands.

The enzymes that have been most successfully targeted by antibiotics are topoisomerases.
2. What are the two types of topoisomerases?
1. Type 1
-form and reseal single-stranded breaks in DNA to decrease positive supercoiling.

2. Type 2
-performs nuclease and ligase operations on both strands of DNA.
3. What are the similarities/differences between types 1 and 2 topoisomerases?
Both types of topoisomerases can remove excess DNA supercoils during DNA replication.

However, only type 2 topoisomerases can resolve intertwined copies of double-stranded DNA to permit segregation of the DNA to daughter cells.

Type 2 enzymes are both more complex and more versatile than type 1 topoisomerases, and the type 2 enzyme serves as a more frequent molecular target for chemotherapeutic agents.
4. What is the MOA of a type 2 topoisomerase?
1. The enzyme binds to a segment of DNA and forms covalent bonds w/phosphates form each strand, thereby nicking both strands.

2. The enzyme causes a second stretch of DNA from the same molecule to pass through the break, relieving supercoilding.

This passage of double-stranded DNA through a double stranded break is what permits separation of intertwined copies of DNA following replication, and thereby, segregation of DNA into progeny cells.
5. What are the two main bacterial type 2 topoisomerases?
1. DNA gyrase
2. Topoisomerase IV

DNA gyrase is particularly crucial for segregation in some bacteria, while topoisomerase IV is the critical enzyme in other bacteria.
6. What enzyme catalyzes transcription?

How is this different in prokaryotes/eukaryotes?
RNA polymerase.

In bacteria, one RNA polymerase synthesizes all the RNA in the cell. Furthermore, bacterial RNA polymerase is composed of only five subunits.

In contrast, eukaryotes express three different RNA polymerases, and each enzyme is considerably more complex in its subunit structure than the bacterial counterpart.
7. What are inhibitors of toposiomerases called?
Quinolones.

Quinolones are a major class of bactericidal antibiotics that act by inhibiting bacterial type II topoisomerases.
8. MOA of quinolones
Quinolones act by inhibiting one or both fo the two prokaryotic type 2 topoisomerases in sensitive bacteria, DNA gyrase and topoisomerase IV.

Quinolones primarily inhibit DNA gyrase in Gram-negative organisms, and they inhibit topoisomerase IV in Gram-positive organisms such as S. aureus.
9. Quinolone activity at low vs. high concentrations
At low concentrations, quinolones inhibit type II topoisomerases reversibly, and their action is bacteriostatic.

At higher concentrations, however, quinolone convert the topoisomerases into DNA-damaging agents by stimulating dissociation of the enzyme subunits form the broken DNA.

Doubly-nicked DNA cannot be replicated, and transcription cannot proceed through such breaks. Topoisomerase dissociation from the DNA and/or the bacterial response to the double stranded break lead ultimately to cell death.

Thus, at therapeutic doses, the quinolone antibiotics are bactericidal.
10. What are the names of the quinolones?
1. Ciprofloxacin
2. Gatifloxacin
3. Levofloxacin
4. Moxifloxacin
5. Norfloxacin
6. Ofloxacin
11. What are the quinolones used to treat?
They are widely used to treat common urogenital, respiratory, and GI infections caused by E. coli, Klebsiella pneumoniae, Campylobacter jejuni, Pseudomonas aeruginosa, Neisseria gonorrhoeae, and Enterobacter, Salmonella, and Shigella species.
12. Ciprofloxacin
MOA: Inhibits prokaryotic type 2 topoisomerases. At therapeutic doses, it has a bactericidal effect by causing double-stranded DNA breaks and cell death.

PURPOSE: Gram-negative infections

ADVERSE: *Cartilage damage, tendon rupture, peripheral neuropathy, increased ICP, seizure, severe hypersensitivity reaction, rash, GI disturbance

CONTRA: Concomitant tizanidine administration and hypersensitivity to quinolones

NOTES: Avoid coadministration of thioridazine due to increased risk of cardiotoxicity.
13. Gatofloxacin, Levofloxacin, Moxifloxacin, Norfloxacin, Ofloxacin
MOA: Inhibits prokaryotic type 2 topoisomerases. At therapeutic doses, it has a bactericidal effect by causing double-stranded DNA breaks and cell death.

PURPOSE: Gram-negative infections

ADVERSE: Cartilage damage, tendon rupture, peripheral neuropathy, increased ICP, seizure, severe hypersensitivity reaction, rash, GI disturbance

CONTRA: Hypersensitivity to quinolones

NOTES: Avoid coadministration of thioridazine due to increased risk of cardiotoxicity.
14. How do bacteria evolve resistance to the quinolones?
Bacteria evolve resistance through chromosomal mutations in the genes that encode type 2 topoisomerases, or through alterations in the expression of membrane porins and efflux pumps that determine drug levels inside the bacteria.
15. What are the inhibitors of transcription called?
Rifamycin derivatives, such as:

1. Rifabutin
2. Rifampin

These are semisynthetic derivatives of the naturally occurring antibiotic rifamycin B.
16. Rifampin
MOA: Form a stable complex w/bacterial DNA-dependent RNA polymerase, thereby inhibiting RNA synthesis. It targets the β-subunit of bacterial RNA polymerase.

PURPOSE: Prophylaxis of meningococcal disease and for mycobacterial infections, including tuberculosis

ADVERSE: Thrombocytopenia, hepatotoxicity, *saliva, tear, sweat and urine discoloration, influenza like illness, elevated LFTs, GI disturbance

CONTRA: Active Neisseria meningitidis infection
17. Rifampin NOTES
NOTES: Particularly effective against phagosome-dwelling mycobaceria b/c it is bactericidal for intracellular as well as extracellular bacteria.

Rifampin increases the in vitro activity of isoniazid

Displays high selectivity for bacteria, so it is generally well tolerated, and the incidence of adverse effects is low.

Rifampin is not used as a single angent b/c of rapid development of resistance

May reduce cyclosporine concentration and efficacy
18. Rifabutin
MOA: Form a stable complex w/bacterial DNA-dependent RNA polymerase, thereby inhibiting RNA synthesis. It targets the β-subunit of bacterial RNA polymerase.

PURPOSE: Mycobacterial infections, including tuberculosis

ADVERSE: Thrombocytopenia, hepatotoxicity, saliva, tear, sweat and urine discoloration, influenza like illness, elevated LFTs, GI disturbance

CONTRA: Active Neisseria meningitidis infection

NOTES: Avoid concurrent administration of clarithromycin w/rifabutin, b/c clarithromycin increases plasma concentration of rifabutin and rifabutin reduces plasma concentration of clarithromycin
19. What are three general consideration that apply to inhibitors of bacterial translation?
1. Translation inhibitors target either the 30S or 50S subunit of the bacterial ribosome.

2. In addition to their inhibitor effects on bacterial ribosomes, protein synthesis inhibitors can affect mammalian mitochondrial ribosomes, cytosolic ribosomes, or both.

3. Complete inhibition of protein synthesis is not sufficient to kill a bacterium.
20. What is the most common mechanism by which inhibitors of translation cause adverse effects?
Through inhibition of host ribosomes.
21. What are the three categories of drugs that target the 30S ribosomal subunit?
1. Aminoglycosides
2. Spectinomycin
3. Tetracyclines
22. What are aminoglycosides?

What are the names of the aminoglycosides?
Aminoglycosides are used mainly to treat infections caused by Gram-negative bacteria. These agents are charged molecules that are not orally bioavailable so they must be administered parenterally.

They include:
1. Streptomycin
2. Amikacin
3. Gentamicin
4. Kanamycin
5. Neomycin
6. Netilmicin
7. Paromomycin
8. Tobramycin
23. Aminoglycosides
MOA: Bind to 16S rRNA of the 30S subunit and elicit concentration-dependent effects on protein synthesis. Aminoglycosides are bactericidal due to induction of mRNA misreading; misread mRNA causes synthesis of aberrant proteins that insert into membrane, forming pores that eventually lead to cell death.

PURPOSE: Serious Gram-negative infections

ADVERSE: *Ototoxicity, acute renal failure, neuromuscular blockage, respiratory paralysis

CONTRA: Hypersensitivity to aminoglycosides

NOTES: Act synergistically w/β-lactam antibiotics
24. What are the 3 mechanisms by which bacteria can become resistant to aminoglycosides?
1. Plasmid-encoded production of a transferase enzyme or enzymes that inactivate aminoglycosides.

2. Impaired drug entry, possibly by alteration or elimination of porins or other proteins involved in drug transport.

3. Mutation of the drug target on the 30S ribosomal subunit.
25. What is the David model?
The David model frames the story of cell death in terms of the concentration dependent effects of aminoglycosides.

When drug first enters the cell, it is poorly transported across bacterial membranes. At these initial low concentrations, misreading occurs, leading to synthesis of aberrant proteins. Some of these proteins insert into membranes and cause the formation of membrane pores, which allow aminoglycosides to flood the cell and halt protein synthesis completely.

As a result, the damage to the membrane cannot be repaired, and leakage of ions and, later, larger molecules leads to cell death.
26. What is the single most important factor restricting aminoglycoside use?
Ototoxicity (manifesting as either auditory or vestibular damage).

The aminoglycosides are known to accumulate in the perilymph and endolymph of the inner ear and at high concentrations, to damage highly sensitive hair cells.

Aminoglycosides can also cause acute renal failure, apparently as a result of drug accumulation in proximal tubular cells.
27. Spectinomycin
MOA: Structural relative of the aminoglycosides that also binds to the 16S rRNA of the 30S ribosomal subunit. Spectinomycin permits formation of the 70S complex but inhibits translocation. It is not bactericidal but is bacteriostatic.

PURPOSE: Alternative therapy for gonorrheal infections

ADVERSE: Injection site pain, nausea, dizziness, insomnia

CONTRA: Hypersensitivity to spectinomycin
28. What are tetracyclines and what are their names?
Tetracyclines are broad-spectrum, bacteriostatic antibiotics that are used widely.

They are:
1. Chlortetracycline
2. Demeclocycline
3. Doxycycline
4. Methacycline
5. Minocycline
6. Oxytetracycline
7. Tetracycline
29. MOA of tetracyclines
Tetracyclines bind reversibly to the 16S rRNA of the 30S subunit and inhibit protein synthesis by blocking the binding of aminoacyl tRNA to the A site on the mRNA ribosome complex.

This action prevents the addition of further AAs to the nascent peptide.
30. What is the reason for the high selectivity of tetracyclines in bacteria?
The high selectivity derives from active accumulation of these drugs in bacteria but not in mammalian cells.

Tetracyclines enter Gram-negative bacteria by passive diffusion through porin protein in the outer membrane, followed by active transport across the inner cytoplasmic membrane.

In contrast, mammalian cells lack the active transport system found in susceptible bacteria.
31. Tetracyclines
MOA: Bind reversibly to the 16S rRNA of the 30S subunit and inhibit protein synthesis by blocking the binding of aminoacyl tRNA to the A site on the mRNA ribosome complex.

PURPOSE: Used to treat a variety fo infections, including Corynebacterium acnes, Haemophilus influenze, Vibrio cholerae, spirochetes, Mycoplasma pneumoniae, Chlamydia species, and rickettsial species. Also used for malaria prophylaxis (doxycycline).

ADVERSE: Bulging fontanelle, *discoloration and hypoplasia of teeth and temporary stunting of growth, hepatotoxicity, pseudotumor cerebri, *photosensitivity, rash, GI disturbance, vestibular disturbance (minocycline), candidal infection.

CONTRA: Last half of pregnancy, infancy, < 8 y/o; patients with sever renal impairment should not be treated w/any tetracyclines except for doxycycline.
32. How does tetracycline resistance occur?
Tetracycline resistance occurs by plasmid-encoded efflux pumps, production of proteins that interfere w/binding of tetracyclines to the ribosome, or enzymatic inactivation of tetracyclines.
33. Important pharmacokinetic feature of tetracyclines

What do tetracyclines have to do with acitretin?
Interaction of these drugs w/foods high in calcium.

B/c these products and medicines impair the absorption of tetracyclines, the tetracyclines are generally taken on an empty stomach.

Once they are in the circulation, however, the same interaction w/cations, can cause sequestration of the drugs in bone and teeth.

*Also, avoid coadministration with acitretin due to increased risk of elevated ICP
34. What are the two most problematic adverse effects of the tetracyclines?
Kidney toxicity and GI distress.

Nausea and vomiting are the most common reasons for premature discontinuation of a course of tetracycline.
35. What's so special about doxycycline?
Compared w/other tetracyclines, a lower fraction of doxycycline is eliminated via the kidney, making this drug safer for use in patients w/renal failure.

Also, doxycycline is excreted in the feces largely in an inactive form, so this agent has the added advantage of minimally altering intestinal flora.

Hence, doxycline use is associated w/a lower incidence of nausea, vomiting, and superinfection w/pathogenic organisms than the other tetracyclines, especially in an immunocompromised patient.
36. What are the glycylcyclines?
They are a new class of antibiotics.

It includes Tigecycline, which has a broad spectrum of activity and has been approved for IV administration in the treatment of serious skin and abdominal infections.
37. Tigecycline
MOA: Resembles that of the tetracyclines; Bind reversibly to the 16S rRNA of the 30S subunit and inhibit protein synthesis by blocking the binding of aminoacyl tRNA to the A site on the mRNA ribosome complex. It is bacteriostatic.

PURPOSE: Skin or subcutaneous infection, complicated abdominal infection

ADVERSE: GI disturbance

CONTRA: Hypersensitivity to tigecycine
38. What are the five categories of antibiotics that target the 50S ribosomal subunit?
1. Macrolides and ketolides
2. Chloramphenicol
3. Lincosamides
4. Streptogramins
5. Oxazolidinones
39. What are macrolides?

What is their MOA?
Macrolides are named for their large lactone rings. They are especially important in the treatment of pulmonary infections, including Legionnaire's disease.

They are bacteriostatic antibiotics that block the translocation step of protein synthesis by targeting the 23S rRNA of the 50S subunit. Macrolides bind to a specific segment of 23S rRNA and block the exit tunnel form which nascent peptides emerge.
40. What are the names of the macrolides and ketolides?
1. Azithromycin
2. Clarithromycin
3. Erythromycin
4. Telithromycin
41. What is erythromycin used to treat?
Erythromycin is used to treat a variety fo infections, notably those due to Corynebacterium acnes, Legionella pneumophila, Treponema pallidum (syphilis), Mycoplasma pneumoniae, and Chlamydia species
42. What is clarithromycin used to treat?
Clarithromycin has increased activity against H.. influenzae.
43. What is azithromycin used to treat?
Azithromycin has increased activity against H. influenzae and Moraxella catarrhalis.
44. Adverse effects and contraindications of macrolides and ketolides
ADVERSE: *Acute cholestatic hepatitis, *ototoxicity, fulminant hepatic necrosis (rare, telithromycin), and GI disturbance

CONTRA: Hepatic dysfunction
45. How is resistance accomplished by bacteria against macrolides?
Resistance can be onferred by chromosomal mutations leading to alteration of the 50S ribosomal binding site, production of methylases that alter the 50S binding site, or production of esterases that degrade macrolides.
46. What is the most frequent reason for discontinuing erythromycin?
GI intolerance, as the drug can directly stimulate gut motility and cause nausea, vomiting, diarrhea, and sometimes anorexia.

Azithromycin and clarithromycin are generally well tolerated, although these drugs can also cause liver impairment.
47. What are the pharmacological interactions with macrolide use?
Macrolides and ketolides inhibit hepatic metabolism of cyclosporine, carbamazepine, warfarin, and theophylline, and can lead to toxic levels of these drugs.

Macrolides eliminate certain species of intestinal flora that inactivate digoxin, thereby leading to greater oral absorption of digoxin in some patients.
48. Telithromycin
This is the ketolide that has a MOA similar to that of the macrolides, but w/a higher affinity for the 50S ribosomal subunit due to its ability to bind an additional site on 23S rRNA.

This higher affinity allows the use of telithromycin in treating infections due to certain bacterial strains that are resistant to macrolides.
49. Chloramphenicol
MOA: Binds to 23S rRNA and inhibits peptide bond formation, apparently by occupying a site that interferes w/proper positioning of the aminoacyl moiety of tRNA in the A site.

PURPOSE: Bacteriostatic broad-spectrum antibiotic that is effective against Gram+ and Gram- bacteria (especially anaerobes), and rickettsiae.

ADVERSE: *Hemolytic anemia in patients with low levels of G6PD, aplastic anemia, **gray baby syndrome

CONTRA: Hypersensitivity to chloramphenicol

NOTES: Most adverse effects are due to inhibition of mitochondrial function.
50. What are the organisms most highly susceptible to chloramphenicol treatment?
1. Haemophilus influenzae
2. Neisseria meningitidis
3. Some strains of Bacteriodes

This drug is still used occasionally in the treatment of typhoid fever, bacterial meningitis, and rickettsial diseases, but only when safer alternatives are not available.
51.How have microbes developed resistance to chloramphenicol?

Which two mechanisms are at work here...?
1. Low level resistance has emerged by the selection of mutants w/decreased permeability to the drug.

2. More clinically significant is the type of resistance that has arisen from the spread of specific plasmid-encoded acetyltransferases that inactivate the drug.
52. What is the gray baby syndrome?
Occurs when chloramphenicol is administered at high doses to newborn infants.

B/c newborns lack an effective glucuronic acid conjugation mechanism for the degradation and detox of chloramphenicol, the drug can accumulate to toxic levels and cause vomiting, hypothermia, gray color, respiratory distress, and metabolic acidosis.
53. What are the pharmacologic interactions with chloramphenicol?
Cloramphenicol antagonizes the bactericidal effects of penicillins and aminoglycosides

Like the macrolides, it also inhibits hepatic metabolism of warfarin, phenytoin, tolbutamide, and chlorpropamide, and thereby potentiates their effects.
54. Lincosamides: clindamycin
MOA: Clindamycin blocks peptide bond formation, apparently through interactions with both the A site (like chloramphenicol) and the P site.

PURPOSE: Bacterial infections due to anaerobic organisms

ADVERSE: **Psuedomembranous colitis, increased liver function tests, jaundice, GI disturbance, rash

CONTRA: Hypersensitivity to clindamycin

*NOTES: Clindamycin is associated w/overgrowth of C. difficile, which can result in pseudomembranous colitis.
55. What are the drugs that are mixed in streoptogramins?
1. Dalfopristin, a group A steptogramin

2. Quinupristin, a group B streptogramin
56. Streptogramins
MOA: Inhibit protein synthesis by binding to the peptidyl transferase center of bacterial 23S rRNA.

PURPOSE: Vancomycin-resistant enterococcus (VRE) infection, skin and subcutaneous infection caused by staphylococcal or streptococcal species.

ADVERSE: Injection site inflammation, GI disturbance, hyperbilirubinemia, arthralgia, myalgia, headache

NOTES: Should not be coadministered w/SSRIs, due to risk of serotonin syndrome. Coadministration with pimozide should be avoided due to increased risk fo cardiotoxicity
57. How are streptogramins unusual among the 50S antibiotics?
They are bactericidal against many, but not all, susceptible bacterial species.

Current hypothesis is that, unlike other 50S antibiotics, the streptogramins induce a conformational change in the ribosome that is reversible only after subunit dissociation.
58. Oxazolidinones: Linezolid
MOA: Linezolid appears to act at the 50S ribosomal subunit b/c mutations in 23S rRNA can confer drug resistance.

PURPOSE: Gram+ bacterial infections, especially VRE, MRSA, S. agalactiae, S. pneumoniae (including multi-drug resistant strains) and S. pyogenes, nosocomial pneumonia, complicated diabetic foot infections

ADVERSE: *Myelosuppression, peripheral neuropathy, optic neuropathy

CONTRA: Hypersensitivity to linezolid

NOTES: Available in both oral and IV formulation; precise mechanism of action remains uncertain.
59. What is the log cell kill model?
The log cell kill model states that the cell destruction caused by cancer chemotherapy is first-order; that is, that each does of chemo kills a constant fraction of cells.

If the tumor starts with 10^12 cells and 99.99% are killed, then 10⁸ malignant cells will remain. The next dose of chemo will then kill 99.99% of the remaining cells, and so on.
60. What is the composition of gram-positive bacteria?
In Gram-positive bacteria, the cell wall is composed of a thick layer of murein, through which nutrients, waste products, and antibiotics can diffuse. Lipoteichoic acids in the outer leaflet of the cytopasmic membrane intercalate thru the cell wall to the outer surface of Gram-positive bacteria.

The hydrophilic side chains of these molecules are involved in bacterial adherence, feeding, and evasion of the host immune system.

These bacteria appear purple.
61. What is the composition of gram-negative bacteria?
In gram-negative bacteria, the murein layer is thinner and is surrounded by a second, outer lipid bilayer membrane. Hydrophilic molecules cross this outer membrane through channels, which are formed by a cylindrical arrangement of porins.

Gram-negative bacterial also have LPS in the outer membrane, which is a major antigen for the immune response to Gram-negative organisms.

These bacteria appear pink.
62. What is the main difference between Gram-negative bacteria and mycobacteria?
Both mycobacteria and Gram-negative bacteria are enclosed by an inner membrane, a murein layer, and an outer membrane.

The main structural different is that in mycobacteria, the two leaflets of the outer membrane are asymmetric in size and composition; the inner leaflet of the outer membrane is composed of arabinogalactan and mycolic acids, whereas the outer leaflet is composed of extractable phospholipids.
63. What are the four inhibitors of murein monomer synthesis?
1. Fosfomycin
2. Fosmidomycin
3. Cycloserine
4. Bacitracin
64. Fosfomycin
MOA: Inhibits murein monomers. It is a PEP analogue that inhibits bacterial enol pyruvate transferase by covalent modification of the enzyme's active site. It enters the cells via G-6-P that are normally used by bacteria

Purpose: Gram-negative urinary tract infections: E. coli, Klebsiella, Serratia, Clostridia. ***It is excreted unchanged in the urine***

ADVERSE: Headache, diarrhea, nausea

CONTRA: Hypersensitivity
65. Therapeutic considerations for fosfomycin
1. Synergistic with beta-lactams, aminoglycosides, and fluoroquinolones
2. Decreased absorption when coadministered with antacids or motility agents such as metoclopramide
3. Less effective against Gram-positive bacteria b/c these bacteria generally lack selective glycerophosphate and G6P transporters.
66. Fosmidomycin
Fosmidomycin, another PEP analogue, acts by the same mechanism as fosfomycin, and resistance typically arises via mutations in glycerophosphate or G6P transports.

Fosmidomycin also has activity against malaria, but the drug has a different MOA and is not currently in clinical use for that organism.
67. Cycloserine
MOA: Cycloserine is a structural analogue of D-Ala; it irreversibly inhibits both the alanine racemase and the synthetase that joins together two D-Ala molecules.

PURPOSE: Multidrug resistant tuberculosis infection and M. avium complex

ADVERSE: Seizures, somnolence, peripheral neuropathy, psychosis

CONTRA: Epilepsy, depression, anxiety, psychosis, severe renal insufficiency, alcohol abuse
68. Cycloserine therapeutic considerations
1. Alcohol, isoniazid, and eithionamide potentiate cycloserine toxicity
2. Pyridoxine may prevent cycloserine-induced peripheral neuropathy
3. Cycloserine inhibits hepatic metabolism of phenytoin
69. Bacitracin
MOA: Peptide antibiotic that inhibits dephosphorylation by forming a complex with bactoprenol pyrophosphate, rendering this lipid carrier useless for further rounds of murein monomer translocation.

PURPOSE: Cutaneous and eye infections (topical), GI decontamination of C. difficile or VRE (oral)

ADVERSE: If systemic absorption occurs: nephrotoxicity, neurotoxicity, bone marrow suppression; With topical: contact dermatitis, blurred vision, red eye

CONTRA: Coadministration with nephrotoxic agents or neuromuscular blocking agents.

NOTE: Drugs that act as metal chelators could interfere with the activity of bacitracin.
70. What are the two inhibitors of murein polymer synthesis?

How do they work?
Vancomycin and teicoplanin are glycopeptides with bactericidal activity against Gram-positive rods and cocci.

These agents interrupt cell wall syntehsis by binding tightly to the D-Ala D-Ala terminus of the murein monomer unit, inhibiting transglycosidase and thereby blocking the addition of murein units.
71. Vancomycin and teicoplanin
MOA: Interrupts cell wall syntehsis by binding tightly to the D-Ala D-Ala terminus of the murein monomer unit, inhibiting transglycosidase and thereby blocking the addition of murein units.

PURPOSE: MRSA (IV), and C. difficile enterocolitis (oral)

ADVERSE: Neurtropenia, ototoxicity, nephrotoxicity, anaphylaxis, *Red man syndrome*

CONTRA: Solutions containing dextrose in patients with known corn allergy
72. Vancomycin and teicoplanin therapeutic considerations
1. Increased nephrotoxicity w/aminoglycosides
2. Red man syndrome can be avoided by slowing infusion rate or pre-administering antihistamines
3. Resistance occurs thru acquisition of DNA encoding enzymes that catalyze formation of D-Ala D-lactate
4. Teicoplanin is not used clinically in the US.
73. What is the "red man syndrome"?
Skin flushing or rash caused by fast infusion rates of vancomycin.

Other adverse effects of vancomycin are drug fever, hypersensitivity rash, and anaphylaxis.

These can be prevented by slowing the rate of infusion and by giving antihistamines prophylactically.
74. What are the four families of β-lactam agents?
1. Penicillins
2. Cephalosporins
3. Monobactams
4. Carbapenems
75. What are the two factors that determine a β-lactam's spectrum of action?
1. The degree to which it can penetrate the outer membrane and cell wall

2. Its ability to bind to specific transpeptidases once in the periplasmic space.
76. What are the broad spectrum β-lactams?
Hydrophilic agents such as ampicillin, amoxicillin, and especially, piperacillin, ticarcillin, carbenicillin, and mezolocillin tend to have broader spectrum of action.
77. What are the narrow spectrum β-lactams?
Hydrophobic agents such as oxacillin, cloxacillin, dicloxacillin, nafcillin, methicillin, and penicillin G tend to have narrow spectrum of action.
78. Penicillin G and V
MOA: β-lactams inhibit transpeptidase by forming a covalent dead-end acyle enzyme intermediate (Suicide substrate inhibition). Penicillins have a 5-membered accessory ring attached to the β-lactam ring.

PURPOSE: Penicllin-sensitive S. aureus and S. pyogenes, rogal anaerobes, N. meningitidis, Clostridia species; Syphilis, Yaws, Leptospirosis. Prophylaxis of rheumatic fever (penicillin V)

ADVERSE: Seizures, pseudomembranous enterocolitis, drug-induced eosinophilia, hemolytic anemia, acute interstitial nephritis, anaphylaxis, rash, fever, injection site reaction, Jarisch Herxheimer reaction when used to treat syphilis.

CONTRA: Hypersensitivity to penicillins
79. Penicillin G and V notes
1. Penicillin G is the IV preparation; Penicillin V is the oral preparation
2. Anticoagulant effects of warfarin may be potentiated by concomitant penicillin administration
3. IV penicillin G is preferred to oral penicillin V in hospital settings
4. *β-lactamase sensitive
80. What are the common uses for penicillin V?
Used mostly to treat mixed aerobic-anaerobic infections of the head and neck, such as dental abscesses.

Additionally, it is used to prevent recurrent rheumatic fever and recurrent streptococcal cellulitis in patients with lymphedema.
81. What are the five antistaphylococcal penicillins?
1. Oxacillin
2. Cloxacillin
3. Dicloxacillin
4. Nafcillin
5. Methicillin
82. Oxacillin, Cloxacillin, Dicloxacillin, Nafcillin, Methicillin
MOA: β-lactams inhibit transpeptidase by forming a covalent dead-end acyle enzyme intermediate (Suicide substrate inhibition). Penicillins have a 5-membered accessory ring attached to the β-lactam ring.

PURPOSE: Skin and soft-tissue infections or systemic infection with β-lactamase producing MRSA

ADVERSE: Diarrhea, nausea, vomiting, pseudomembranous colitis (cloxacilling, dicloxacillin), Hepatitis (oxacilling) interstitial nephritis, phlebitis (nafcillin)

CONTRA: Hypersensitivity

NOTES: *β-lactamase resistant! These agents are narrow spectrum.
83. What are the names of the four amino penicillins?
1. Ampicillin
2. Amoxicillin
3. Amoxicillin/clavulanic acid
4. Ampicillin/sulbactam

These agents have a positively charged amino group on the side chain which enhances diffusion through porin channels but does not confer resistance to β-lactamases.
84. What are the uses for IV ampicillin?
Invasive enterococcal infections and Listeria meningitis
85. What are the uses for oral amoxicillin?
Uncomplicated ear nose and throat infections, endocarditis prevention, dental surgery prophylaxis, and component of combination therapy for H pylori infection.
86. Adverse, contra, and notes for the amino penicillins
ADVERSE: Rash, anusea, vomiting, diarrhea

CONTRA: Hypersensitivity

NOTES:
1. Broad spectrum anti-bacteria activity
2. Ampicillin and amoxicillin are beta-lactamase sensitive as single agents; clavulanic acid and sulbactam are beta-lactamase inhibitors
3. Positively charged amino group on side chain enhances diffusion thru porin channels of Gram-negative bacteria
87. What are the two carboxy penicillins?
1. Carbenicillin
2. Ticarcillin

These agents are broad spectrum antibiotics that have a side chain carboxyl group that provides a negative charge which confers some resistance to some β-lactamases
88. What are the two ureido penicillins?
1. Piperacillin
2. Mezlocillin

These drugs have both positive and negative charges on their side chains and are generally more potent than the carboxy penicillins.

Their spectrum of action is similar to that of carboxy penicillins; in addition, they have activity against Klebsiella and enterococci
89. Carbenicillin, ticarcillin, piperacillin, mezlocillin
PURPOSE: Primarily used as treatment or prophylaxis against P. aeruginosa infection; Hospital acquired pneumonia due to resistant Gram-negative organisms

ADVERSE: Rash, nausea, vomiting, diarrhea

CONTRA: Hypersensitivity to penicillins

NOTES: Broad spectrum activity but primarily used against P. aeruginosa. Generally beta-lactamase sensitive except for carbenicillin and ticarcillin
90. What are the cephalosporins?
Cephalosporins differ structurally from penicillins by having a six-membered ring rather than a five membered ring attached to the β-lactam ring.
91. First generation cephalosporins:

Cefazolin & cephalexin
MOA: β-lactams inhibit transpeptidase by forming a dead-end acyl enzyme intermediate; relatively good Gram-positive coverage; sensitive to many β-lactamases

PURPOSE: Proteus mirabilis, E. coli, Klebsiella pneumoniae, Skin and soft tissue infections, surgical prophylaxis

ADVERSE: Psuedomembranous enterocolitis, leukopenia, thrombocytopenia, hepatotoxicity, nausea, vomiting, rash, diarrhea

CONTRA: Hypersensitivity to cephalosporins (rarely cross-react w/penicillins)
92. Second generation cephalosporins:

Cefuroxime, cefotetan, cefoxitin
MOA: β-lactams inhibit transpeptidase by forming a dead-end acyl enzyme intermediate; relatively broader Gram-negative coverage than first gen; more resistant to β-lactamases than first gen

PURPOSE: H. influenzae (cefuroxime), Enterobacter spp., Neisseria spp., P. mirabilis, E. coli, K. pneumoniae (cefotetan and cefoxitin)

ADVERSE: Psuedomembranous enterocolitis, leukopenia, thrombocytopenia, hepatotoxicity, nausea, vomiting, rash, diarrhea, **Cefotetan may produce disulfiram-like reaction w/alcohol ingestion and block synthesis of vitamin K-dependent coagulation factors

CONTRA: Hypersensitivity to cephalosporins (rarely cross-react w/penicillins)
93. Common uses for cefuroxime, cefotetan and cefoxitin?
Cefuroxime is primarily used in community acquired pneumonia

Cefotetan and cefoxitin are primarily used in intra-abdominal and pelvic infections.
94. Third generation cephalosporins:

Cefotaxime, ceftizoxime, ceftriaxone, cefoperazone, ceftazidime
MOA: β-lactams inhibit transpeptidase by forming a dead-end acyl enzyme intermediate; *Highest CNS penetration of the cephalosporins.

PURPOSE: N. gonorrhoeae, Borrelia burgdorferi, H. influenza, most Enterobacteriaceae (ceftriaxone), H. influenzae (cefotaxime), P. aeruginosa (ceftazidime).

ADVERSE: Psuedomembranous enterocolitis, leukopenia, thrombocytopenia, hepatotoxicity, nausea, vomiting, rash, diarrhea, **Ceftriaxone may cause cholestatic hepatitis, and cefoperazone may produce disulfiram-like reaction w/alcohol ingestion and block synthesis of vitamin K-dependent coagulation factors

CONTRA: Hypersensitivity to cephalosporins (rarely cross-react w/penicillins)

NOTE: Resistant to many β-lactamases; highly active against Enterobacteriaceae, but less active against Gram-positive organisms than are first gen cephalosporins
95. Fourth generation cephalosporin:

Cefepime
MOA: β-lactams inhibit transpeptidase by forming a dead-end acyl enzyme intermediate; *resistant to many beta-lactamases

PURPOSE: Enterobacteriaceae, Neisseria, H. influenzae, P. aeruginosa, Gram-positive organisms

ADVERSE: Psuedomembranous enterocolitis, leukopenia, thrombocytopenia, hepatotoxicity, nausea, vomiting, rash, diarrhea, **Cefepime may produce erythrocyte autoantibodies w/o significant hemolysis

CONTRA: Hypersensitivity to cephalosporins (rarely cross-react w/penicillins)
96. What are the inhibitors of polymer crosslinking?
1. Aztreonam
2. Imipenem/cilastatin
3. Meropenem
4. Ertapenem
97. Aztreonam
MOA: A monobactam that inhibits transpeptidase by forming a dead-end acyl enzyme intermediate

PURPOSE: Gram-negative bacteria; *used in penicillin allergic patients*

ADVERSE: Same as penicillins

NOTES: No Gram-positive coverage
98. Imipenem/cilastatin, Meropenem, Ertapenem
MOA: A carbapenem that inhibits transpeptidase by forming a dead-end acyl enzyme intermediate

PURPOSE: Gram-positive and Gram-negative bacteria except MRSA, VRE, and Legionella

ADVERSE: same as penicillins, except *High plasma levels of imipenem and meropenem may cause seizures
99. Carbapenem notes
NOTES:
1. Cilastin inhibits renal dehydropeptidase I, which would otherwise inactivate imipenem
2. Probenecid may increase meropenem levels
3. All three agents decrease valproate levels
4. Ertapenem can be give one-daily.
100. What are the three antimicrobial agents?
1. Ethambutol
2. Pyrazinamide
3. Isoniazid (INH)
101. Ethambutol
MOA: Inhibits the arabinosyl transferase that adds arabinose units to the growing arabinogalactan chain and thus decreases arabinogalactan synthesis.

PURPOSE: Mycobacteriostatic, and used in combo with other antimycobacterials, including rifampin and streptomycin

ADVERSE: Optic neuritis, blindness, peripheral neuropathy, neutropenia, thrombocytopenia, hyperuricemia, mania, nausea, vomiting

CONTRA: Known optic neuritis, patients unable to report visual changes such as young children, *Coadministration with antacids
102. Pyrazinamide
MOA: Pyrazinamide is a prodrug that must be converted to its active form pyrazinoic acid, which inhibits fatty acid synthestase 1 (FAS1) and thus inhibits mycolic acid synthesis.

PURPOSE: Mycobacterium species; used in combo w/rifampin and streptomycin

ADVERSE: Anemia, hepatotoxicity, arthralgias, hyperuricemia (usually asymptomatic)

CONTRA: Acute gout, severe hepatic dysfunction
103. Isoniazid and ethionamide
MOA: Targets the FAS2 complex and thus inhibits mycolic acid synthesis.

PURPOSE: Mycobacterium species, and is used in combo with other antimycobacterials

ADVERSE: Hepatitis, *neurotoxicity (paresthesias, peripheral neuropathy, ataxia), SLE, seizure, hematologic abnormalities

CONTRA: Acute liver disease

NOTES: Can inhibit or induce p450 enzymes and thus interacts w/other drugs such as rifampin, antiseizure meds, antifungals, and alcohol; *INH neurotoxicity can be prevented by pyridoxine supplementation
104. How does resistance to INH occur?
Resistance to INH results from an inactivating mutation in the mycobacterial enzyme catalase-peroxidase, which converts INH to ints antimycobacterial form. Mutations in the INHA gene, which is required for mycolic acid synthesis, also confer resistance to INH.
105. How does resistance to ethambutol occur?
Results from mutations in the arabinosyl transferase gene, some of which cause overexpression of the target enzyme
106. How does resistance to pyrazinamide occur?
Due to mutations in the pyrazinamidase gene, which result in the inability to convert the prodrug into its active form.
107. What are 7 congenital or development defects of the lung?
1. Agenesis or hypoplasia of both lungs, one lung, or single lobes
2. Tracheal and bronchial anomalies (atresia, stenosis, tracheoesophageal fistula)
3. Vascular anomalies
4. Congenital lobar overinflation (emphysema)
5. Foregut cysts
6. Congenital pulmonary airway malformation
7. Pulmonary sequestrations
108.What is pulmonary hypoplasia?
Pulmonary hypoplasia is the defective development of both lungs resulting in decreased weight, volume, and acini compared to the body weight and gestational age.

It is a common anomaly, seen in 10% of neonatal autopsies, and is most often secondary to space-occupying lesions in the uterus, oligohydraminos, or impaired fetal respiratory movements, as may occur in congenital diaphragmatic hernia, renal cystic disease, renal agenesis, PPROFM, and anencephaly.
109. What are foregut cysts?
Foregut cysts represent an abnormal detachment of primitive foregut and are most often located in the hilum or middle mediastinum.

Depending on the wall structure, these cysts are classified into bronchogenic (most common), esophageal, or enteric cysts.
110. What are bronchogenic cysts?
Bronchogenic cysts are rarely connected to the tracheobroncheal tree. It presents in children and young adults as an incidental finding or with symptoms related to mass effect or secondary infection.

Its size varies from 1-4 cm in diameter, but it may be larger in older pts.

*Microscopically, the cyst is lined by ciliated pseudostratified columnar epithelium w/squamous metaplasia occurring in areas of inflammation. The wall contains bronchial glands, cartilage, and smooth muscle.
111. What is congenital pulmonary airway malformation (CPAM)?
CPAM is a hamartomatous lesion of the lung, with an incidence of about 1 in 5,000 live births. It an be separated into five types based on clinical and pathologic features.

CPAM type 1 is the most common w/large cysts and good prognosis. CPAM type 2, with medium-sized cysts, often has a poor prognosis owing to its frequent association with other significant anomalies.
112. What is pulmonary sequestration?
Pulmonary sequestration refers to the presence of a discrete mass of lung tissue w/o any normal connection to the airway system. Blood supply to the sequestered area arises not from the pulmonary arteries but from the aorta or its branches
113. What are extralobar sequestrations?
Extralobar sequestrations are external to the lung and may be found anywhere in the thorax or mediastinum.

Found most commonly in infants as abnormal mass lesions, they may be associated with other congenital anomalies.
114. What about intralobar sequestrations?
Intralobar sequestrations are found within the lung substance and are usually associated with recurrent localized infection or bronchiectasis.

Although a small percentage of these are clearly congenital in origin, the vast majority are probably acquired lesions formed thru repeated episodes of pneumonia.
115. What does atelectasis mean?

What are the three types of acquired atelectasis?
Atelectasis refers either to incomplete expansion of the lungs or to the collapse of previously inflated lung, producing areas of relatively airless pulmonary parenchyma.

Acquired atelectasis, encountered principally in adults, may be divided into resorption (or obstruction), compression, and contraction atelectasis.
116. What is resorption atelectasis?
Resorption atelectasis is the consequence of complete obstruction of an airway, which in time leads to resorption of the oxygen trapped in the dependent alveoli, w/o impairment of blood flow thru the affected alveolar walls. Since lung volume is diminished, the mediastinum shifts toward the atelectatic lung.

Resorption atelectasis is caused principally by excessive secretions (e.g., mucous plugs) or exudates w/in smaller bronchi and is therefore most often found in bronchial asthma, chronic bronchitis, bronchiectasis, and postoperative states and with aspiration of foreign bodies.
117. What is compression atelectasis?
Compression atelectasis results whenever the pleural cavity is partially or completely filled by fluid exudate, tumor, blood or air (pneumothorax) or tension pneumothorax.

Compression atelectasis is most commonly encountered in pts with neoplastic effusions w/in the pleural cavities. Similarly, abnormal elevation of the diaphragm induces basal atelectasis.

*W/compressive atelectasis, the mediastinum shifts away from the affected lung.
118. What is contraction atelectasis?
Contraction atelectasis occurs when local or generalized fibrotic changes in the lung or pleura prevent full expansion.

B/c collapsed lung can be re-expanded, atelectasis is a reversible disorder *except that caused by contraction*
119. What are the cytokines that mediate acute lung injury?
Mediators include cytokines, oxidants, and growth factors, such as TNF, IL-1, IL-6, IL-10, and TGF-beta.

Lung injury may manifest as congestion, edema, surfactant disruption, and atelectasis, and these may progress to acute respiratory distress syndrome or acute interstitial pneumonia.
120. What can cause pulmonary edema?
Pulmonary edema can result from hemodynamic disturbances (hemodynamic or cardiogenic pulmonary edema) or from direct increases in capillary permeability, owing to microvascular injury.
121. What is the most common hemodynamic mechanism of pulmonary edema?
Increased hydrostatic pressure, as occurs in left-sided CHF.
122. What are the characteristics of hemodynamic pulmonary edema?
Whatever the clinincal setting, pulmonary congestion and edema are characterized by heavy, wet lungs. Fluid accumulates initially in the basal regions of the lower lobes b/c hydrostatic pressure is greater in these sites (dependent edema).

Histologically, the alveolar capillaries are engorged, and an intra-alveolar granular pink precipitate is seen. Alveolar microhemorrhages and hemosiderin-laden macrophages ("heart failure" cells) may be present.
123. What are the characteristics of the lungs in long-standing pulmonary congestion, such as those seen in mitral stenosis?
In long standing cases, hemosiderin-laden macrophages are abundant, and fibrosis and thickening of the alveolar walls cause the soggy lungs to become firm and brown (brown induration).

These changes not only impair respiratory function, but also predispose to infection.
124. What is ARDS?
ARDS is a clinical syndrome caused by diffuse alveolar capillary damage. It is characterized clinically by the rapid onset of severe life threatening respiratory insufficiency, cyanosis, and severe arterial hypoxemia that is refractory to oxygen therapy and that may progress to extrapulmonary MSOF.

Chest radiographs show diffuse alveolar infiltration. ***Diffuse alveolar damage (DAD) is the histologic manifestation.
125. What is the morphology of ARDS?

1/2
In the acute stage, the lungs are heavy, firm, red, and boggy. They exhibit congestion, interstitial and intra-alveolar edema, inflammation, and fibrin deposition. The alveolar walls become lined with waxy hyaline membranes that are morphologically similar to those seen in hyaline membrane disease of neonates.
126. What is the morphology of ARDS?

2/2
Alveolar hyaline membranes consist of fibrin-rich edema fluid mixed with the cytoplasmic and lipid remnants of necrotic epithelial cells. In the organizing stage, type II epithelial cells undergo proliferation in an attempt to regenerate the alveolar lining. Resolution is unusual; more commonly, there is organization of the fibrin exudate, with resultant intra-alveolar fibrosis.

Marked thickening of the alveolar septa ensues, caused by proliferation of interstitial cells and deposition of collagen. Fatal cases often have superimposed bronchopneumonia.
127. What is the pathogenesis of ARDS?
ARDS and DAD are best viewed as the clinical and pathologic end results, respectively, of acute alveolar injury caused by a variety of insults and initiated by different mechanisms.

***Central to the causation of ARDS is diffuse damage to the alveolar capillary walls; this is followed by a relatively non-specific, often predictable series of morphologic and physiologic alterations leading to respiratory failure.
128. How is ARDS different from the respiratory distress syndrome of neonates?
The mechanism of the respiratory distress syndrome of newborns is a deficiency in pulmonary surfactant.

In contrast, in ARDS, the initial injury is to either capillary endothelium (most frequently) or alveolar epithelium, but eventually both are clearly affected.
129. What are the acute consequences of damage to the alveolar capillary membrane?
These include increased vascular permeability and alveolar flooding, loss of diffusion capacity, and widespread surfactant abnormalities caused by damage to type II pneumocytes.

Importantly, the exudate and diffuse tissue destruction that occur with ARDS cannot be easily resolved, and generally the result is organization with scarring, producing chronic disease.
130. What is a critical component of lung inflammation?
An elevated level or pro-inflammatory mediators combined w/a decreased expression of anti-inflammatory molecules is a critical component of lung inflammation.

This includes expression of pro-inflammatory genes, particularly NF-κB, since it is required for maximal expression of many cytokines involved in the pathogenesis of acute lung injury.
131. What happens within the first 30 minutes following an acute insult (e.g., acid aspiration, trauma, or exposure to bacterial LPS)?
There is increased synthesis of IL-8 by pulmonary macrophages. Release of this and other cytokines like IL-1 and TNF leads to pulmonary microvascular sequestration and activation of neutrophils.

***Neutrophils are thought to play an important role in the pathogenesis of acute lung injury and ARDS.

Activated neutrophils release a variety of products that cause active tissue damage and maintain the inflammatory response.
132. ARDS is mainly associated with what four conditions?
1. Sepsis
2. Diffuse pulmonary infections (vira, mycoplasma, and pneumocystis pneumonia, and tuberculosis)
3. Mechanical trauma, including head injuries
4. Gastric aspriation
133. What is the clinical course of ARDS?
Pts who develop ARDS are usually hospitalized for one or the predisposing conditions that cause it.

Profound dyspnea and tachypnea herald ARDS, but the chest radiograph is initially normal. Subsequently, there are increasing cyanosis and hypoxemia, respiratory failure, and the appearance of diffuse bilateral infiltrates on radiographic examination.

Hypoxemia can then become unresponsive to oxygen therapy, and respiratory acidosis can develop.
134. What are the functional abnormalities of the lungs in ARDS?
The functional abnormalities are not homogeneously distributed throughout the lungs. The lungs are focally stiff and have a decrease in functional volume. In essence, pts lungs can be divided into areas that are infiltrated, consolidated, or collapsed, and regions that have nearly normal levels of compliance and ventilation.

Lungs with ARDS continue to have perfusion of poorly aerated regions, contributing to V/Q mismatching and hypoxemia.
135. What is acute interstitial pneumonia?

What causes it?
Acute interstitial pneumonia is a clinicopathologic term that is used to describe widespread acute lung injury with a rapidly progressive clinical course similar to that seen in ARDS.

Whereas ARDS is associated with known causes such as sepsis, pulmonary infection, gastric aspiration or trauma, acute interstitial pneumonia is of unknown etiology.
136. What are the clinical features of acute interstitial pneumonia?
The mean age is 50. Pts present with acute respiratory failure often following an illness of less than 3 weeks duration that resembles and upper respiratory tract infection. The radiographic and pathologic features are identical to that of ARDS.

The mortality rate is about 50% with most deaths occurring within 1-2 months. In the surviving pts, recurrences and chronic interstitial disease may develop.
137. What does obstructive lung disease mean?
Obstructive disease is characterized by an increase in resistance to airflow owing to partial or complete obstruction at any level, from the trachea and larger bronchi to the terminal and respiratory bronchioles.
138. What does restrictive lung disease mean?
Restrictive disease is characterized by reduced expansion of lung parenchyma, with decreased total lung capacity.
139. What are the four major diffuse obstructive disorders?
1. Emphysema
2. Chronic bronchitis
3. Bronchiectasis
4. Asthma

*In pts with these diseases, pulmonary function tests show limitation of max airflow rates during forced expiration.
140. What are the two general conditions that cause restrictive diseases?
1. Chest wall disorders in the presence of normal lungs (e.g., neuromuscular diseases such as polio, severe obesity, pleural diseases, and kyphoscoliosis)
2. Acute or chronic interstitial and infiltrative diseases (ARDS)

*There is a reduced total lung capacity, while the expiratory flow rate is normal or reduced proportionately.
141. What is emphysema?

What are the four major types?
Emphysema is a condition of the lung characterized by abnormal permanent enlargement of the airspaces distal to the terminal bronchioles, accompanied by destruction of their walls and without obvious fibrosis.

It is classified according to its anatomic distribution within the lobule. There are four major types:
1. Centriacinar
2. Panacinar
3. Paraseptal
4. Irregular
142. What is centriacinar (centrilobular) emphysema?
The distinctive feature of this type of emphysema is the pattern of involvement of the lobules; the central or primal parts of the acini, formed by respiratory bronchioles, are affected, whereas distal alveoli are spared. Thus, both emphysematous and normal airspaces exist within the same acinus and lobule.

The lesions are more common and usually more severe in the upper lobes, particularly in the apical segments. The walls of the emphysematous spaces often contain large amts of black pigment. Inflammation around bronchi and bronchioles is common.
143. In what population is centriacinar emphysema most common?
*Centriacinar emphysema occurs predominantly in heavy smokers, often in association with chronic bronchitis.
144. What is panacinar (panlobular) emphysema?
In this type, the acini are uniformly enlarged from the level of the respiratory bronchiole to the terminal blind alveoli.

In contrast to centriacinar emphysema, panacinar emphysema tends to occur more commonly i the lower zones and in the anterior margins of the lung, and it is usually most severe at the bases.

*This type of emphysema is associated with alpha-1-antitrypsin deficiency.
145. What is distal acinar (paraseptal) emphysema?
In this type, proximal portion of the acinus is normal, but the distal part is predominantly involved.

The emphysema is more striking adjacent to the pleura, along the lobular connective tissue septa, and at the margins of the lobules. It occurs adjacent to areas of fibrosis, scarring, or atelectasis and is usually more severe in the upper half of the lungs.
146. What are the characteristic findings in distal acinar emphysema?
The characteristic findings in distal acinar emphysema are of multiple, continuous, enlarged airspaces from less than 0.5 cm to more than 2.0 cm in diameter, sometimes forming cystlike structures.

This type of emphysema probably underlies many of the cases of spontaneous pneumothorax in young adults.
147. What is irregular emphysema?
AKA airspace enlargement with fibrosis (irregular emphysema). It is so named b/c the acinus is irregularly involved, is almost invariably associated with scarring.

It may be the most common form of emphysema b/c careful search of most lungs at autopsy shows one or more scars from a healed inflammatory process. In most instances, these foci of irregular emphysema are asympatomatic and clinically insignificant.
148. What is the incidence of COPD?
COPD is a major public health problem. It is the fourth leading cause of morbidity and mortality in the US.

There is a clear-cut association between heavy cigarette smoking and emphysema, and the most severe type occurs in men who smoke heavily.
149. What is the most plausible hypothesis to account for the destruction of alveolar walls in emphysema?
The protease antiprotease mechanism, aided and abetted by oxidant-antioxidant imbalance.

This theory holds that alveolar wall destruction results from an imbalance between proteases (mainly elastase) and antiproteases in the lung.
150. What is the principal antielastase activity in serum and interstitial tissue?
Alpha-1-antitrypsin.

Neutrophil elastase is capable of digesting human lung, and this digestion can be inhibited by alpha-1-antitrypsin.

*Thus, emphysema is seen to result from the destructive effect of high protease activity in subjects w/low antiprotease activity.
151. How does the protease-antiprotease hypothesis explain the deleterious effect of smoking?
In smokers, there is both increased elastase availability and decreased antielastase activity.

Neutrophils and macrophages accumulate in alveoli due to the direct chemoattractant of nicotine as well as the ROS in smoke.

Smoking also enhances elastase activity in macrophages. In addition to elastase, MMPs derived from macrophages and neturophils have a role in tissue destruction.
152. How does smoking play a role in the oxidant-antioxidant imbalance in the pathogenesis of emphysema?
Normally the lung contains a healthy complement of antioxidants that keep oxidative damage to a minimum. Tobacco smoke contains abundant ROS that deplete these antioxidant mechanism, thereby inciting tissue damage.

A secondary consequence of oxidative injury is inactivation of native antiproteases, resulting in "function" alpha-1-antitrypsin deficiency even in pts w/o enzyme deficiency.
153. How do these theories explain centriacinar emphysema?
The smoke particles predominately accumulate at the bifurcation of respiratory bronchioles, resulting in influx of neutrophis and macrophages which secrete proteoases.

An increase in protease activity localized in the centriacinar region, together with the smoke induced ROS damage, causes the centriacinar pattern of emphysema that is seen in smokers.
154. What is an explanation for the lower lung destruction of panacinar emphysema in alpha-1-antitrypsin deficient pts?
The panacinar emphysema reflects a total lack of antiprotease throughout the acinus and susceptibility to chronic low-level proteolysis from neutrophils in transit thru the lung ciruclation.

The predominately lower lung distribution (where perfusion and neutrophil numbers are greatest) of panacinar emphysema is also consistent with this explanation.
155. What is the morphology of emphysema?

1/2
Panacinar emphysema, when well developed, produces voluminous lungs, often overlapping the heart and hiding it when the anterior chest wall is removed.

The macroscopic features of centriacinar emphysema are less impressive. The lungs might not appear particularly pale or voluminous unless the disease is well advanced. Generally, the upper 2/3rds of the lungs are more severely affected.

Large apical blebs or bullae are more characteristic of irregular emphysema secondary to scarring and distal acinar emphysema.
156. What is the morphology of emphysema?

2/2
Microscopically, there are abnormally large alveoli separated by thin septa with only focal centriacinar fibrosis. There is loss of attachments of the alveoli to the outer wall of small airways. The pores of Kohn are so large that septa appear to be floating or protrude blinding into alveolar spaces w/a club shaped end.

With advance of the disease, there are even large abnormal airspaces and possibly blebs or bullae. Often, the respiratory bronchioles and vasculature of the lungs are deformed and compressed by the emphysematous distortion of the airspaces, and there is often chronic bronchitis or bronchiolitis.
157. What is the clinical course of emphysema?
Clinnical manifestations do not occur until at least 1/3 of the lung is damged. Dyspnea is the first symptom. Cough and expectoration can occur. Weight loss is common and can be severe. Classically, the pt is barrel-chested and dyspneic, w/obviously prolonged expiration, sits forward in a hunched over position, and breathes thru pursed lips.

***Expiratory airflow limitation, best measured thru spirometry, is the key to Dx.
158. What are pink puffers?

What are blue bloaters?
In pts with severe emphysema, cough is slight, overdistention is sever, diffusion capacity is low, and blood gas values are relatively normal at rest. Such patients may over-ventilate and remain well oxygenated. These are the pink puffers.

Pts with chronic bronchitis more often have a history of recurrent infection, abundant purulent sputum, hypercapnia, and severe hypoxemia, promoting the designation blue bloaters.
159. What are the three most common causes of death in pts with COPD?
1. Respiratory acidosis and coma
2. Right-sided heart failure
3. Massive collapse of the lungs secondary to pneumothorax
160. What is compensatory hyperinflation emphysema?
This term is sometimes used to designate dilation of alveoli but not destruction of septal walls in response to loss of lung substance elsewhere.

It is best exemplified by the hyperexpansion of the residual lung parenchyma that follows surgical removal of a diseased lung or lobe.
161. What is obstructive overinflation emphysema?
Obstructive overinflation refers to the condition in which the lung expands b/c air is trapped within it. A common cause is subtotal obstruction by tumor or foreign objects.

It can be a life-threatening emergency b/c the affected portion distends sufficiently to compress the remaining normal lung.
162. What is congenital lobar overinflation in infants?
congenital lobar overinflation in infants is a good example of obstructive overinflation.

It probably results from hypoplasia of bronchial cartilage and is sometimes associated with other congenital cardiac and lung abnormalities.
163. What are the two things that can cause overinflation in obstructive lesions?
1. B/c of a ball-valve action of the obstructive agent, so that air enters on inspiration but cannot leave on expiration.

2. B/c the bronchus may be totally obstructed but ventilation thru collateral may bring in air from behind the obstruction.
164. What is bullous emphysema?
Bullous emphysema refers merely to any form of emphysema that produces large subpleural blebs or bullae (spaces more than 1 cm in diameter in the distended state).

They represent localized accentuations of one of the four forms of emphysema, are most often subpleural, and occur near the apex, sometimes in relation to old tuberculous scarring. On occasion, rupture of the bullae may give rise to pneumothorax.
165. What is interstitial emphysema?
The entrance of air into the connective tissue stroma of the lung, mediastinum, or subcutaneous tissue is designated interstitial emphysema.

In most instances, alveolar tears in pulmonary emphysema provide the avenue of entrance of air into the stroma of the lung, but rarely a sucking wound in the chest can underlie this disorder.
166. What is chronic bronchitis?
Chronic bronchitis is defined clinically when any pt has a persistent cough with sputum production for at least 3 months in at least 2 consecutive years, in the absence of any other identifiable cause.
167. What is simple chronic bronchitis?

Chronic asthmatic bronchitis?

Obstructive chronic bronchitis?
Simple chronic bronchitis occurs when pts have a productive cough by no physiologic evidence of airflow obstruction.

Chronic asthmatic bronchitis occurs when pts have hyperreactive airways with intermittent bronchospasm and wheezing.

Obstructive chronic bronchitis occurs in heavy smokers who have chronic airflow obstruction, usually with evidence of emphysema.
168. What are the three long-term consequences of chronic bronchitis?
1. May progress to chronic obstructive airway disease
2. Lead to cor pulmonale and heart failure
3. Cause atypical metaplasia and dysplasia of the respiratory epithelium, providing a rich soil for cancerous transformation
169. What is the primary or initiating factor in the pathogenesis of chronic bronchitis?
Chronic irritation by inhaled substances such as tobacco smoke, and gran, cotton, and silica dust. Bacterial and viral infections are important in triggering acute exacerbation of the disease.
170. What is the earliest feature of chronic bronchitis?
Hypersecretion of mucus in the large airways, associated with hypertrophy of the submucosal glands in the trachea and bronchi. Proteases and MMPs stimulate this mucus hypersecretion.
171. What is the next earliest feature of chronic bronchitis?
As chronic bronchitis persists, there is also a marked increase in goblet cells of small airways - small bronchi and bronchioles, leading to excessive mucus production that contributes to airway obstruction.

Many of the respiratory epithelial effects of environmental irritants are believed to be mediated thru the epidermal growth factor (EGF) receptor.
172. Although mucus hypersecretion in large airways is the cause of sputum overproduction, what about alterations in the small airways of the lung?
It is now though that accompanying alterations in the small airways of the lung (small bronchi and bronchioles, less than 2-3 mm in diameter) can result in physiologically important and early manifestations of chronic airway obstruction.
173. What are the four histologic findings in small airways in young smokers?
1. Goblet cell metaplasia w/mucus plugging of the lumen
2. Clustering of pigmented alveolar macrophages
3. Inflammatory infiltration
4. Fibrosis of the bronchiolar wall

Smoke and other irritants which cause the hypertrophy of mucous glands, also result in bronchiolitis, or small airway disease.
174. When bronchitis is accompanied by moderate to severe airflow obstruction, what is the dominant lesion?
Coexistent emphysema is the dominant lesion.
175. What is the morphology of chronic bronchitis?

1/2
Grossly, there may be hyperemia, swelling, and edema of the mucous membranes, frequently accompanied by excessive mucinous to mucopurulent secretions layering the epithelial surfaces. Sometimes, heavy casts of secretions and pus fill the bronchi and bronchioles.

***The characteristic histologic features of chronic bronchitis are chronic inflammation of the airways (predominantly lymphocytes) and enlargement of the mucus secreting glands of the trachea and bronchi.

Although the numbers of goblet cells increase slightly, the major increase is in the size of the mucous glands. (Use Reid index)
176. What is the morphology of chronic bronchitis?

2/2
The bornchial epithelium may exhibit squamous metaplasia and dysplasia. There is marked narrowing of bronchioles caused by goblet cell metaplasia, mucus plugging, inflammation, and fibrosis.

In the most severe cases, there may be oliteration of lumen due to fibrosis (bronchiolitis obliterans).
177. What is the Reid index?
The increase in the size of the mucous glands can be assessed by the ratio of the thickness of the mucous gland layer to the thickness of the wall between the epithelium and the cartilage.

The Reid index (normally 0.4) is increased in chronic bronchitis, usually in proportion to the severity and duration of the disease.
178. What are the clinical features of chronic bronchitis?
The cardinal symptom of chronic bronchitis is a persistent cough productive of sputum.

With the passage of time, dyspnea on exertion appears and later other elements of COPD may appear, including hypercapnia, hypoxemia, and mild cyanosis.
179. What is asthma?
Asthma is a chronic inflammatory disorder of the airways that causes recurrent episodes of wheezing, breathlessness, chest tightness, and cough, particularly at night and/or in the early morning.

These symptoms are usually associated with widespread but variable bronchoconstriction and airflow limitation that is at least partially reversible, either spontaneously or with treatment.

It is thought that inflammation causes an increase in airway responsiveness (bronchospasm) to a variety of stimuli.
180. How is asthma typically categorized?
Typically, asthma is categorized into extrinsic (initiated by a type I hypersensitivity reaction induced by exposure to an extrinsic antigen) and intrinsic (initiated by diverse, nonimmune mechanisms, including ingestion of aspiring; pulmonary infections, esp viral, cold; inhaled irritants; stress; and exercise).
181. What are the major etiologic factors of asthma?
A genetic predisposition to type I hypersensitivity, acute and chronic airway inflammation, and bronchial hyperresponsiveness.

The inflammation involves many cell types and numerous inflammatory cells. TH2 CD4+ cells are prominent components of the bronchial inflammation.
182. How do TH1 cells and TH2 cells interact to cause asthma?
An imbalance in the reciprocal arrangement between TH1 and TH2 cells may be the key to asthma. It appears that in pts with allergic asthma, T cell differentiation is skewed in the direction of TH2 cells.

Research shows that a transcription factor called T-bet is required for TH1 cell differentiation, and studies on lung tissues from asthmatics reveal absence of T-bet in lung lymphocytes.
183. What is the relationship between ADAM-33 and asthma?
An abnormal, perhaps genetically determined, microenvironment in the bronchial wall is an essential cofactor in the pathogenesis of asthma. This view has been bolstered by the linkage of teh ADAM-33 gene to asthma.

ADAM-33 belongs to a MMP subfamily such as collagneases. It is known that ADAM-33 is expressed by lung fibroblasts and bronchial smooth muscle cells. It is speculated that ADAM-33 polymorphisms accelerate proliferation of bronchial smooth muscle cells and fibroblasts, thus contributing to bronchial hyperreactivity and subepithelial fibrosis.
184. What is atopic asthma?
This most common type of asthma usually begins in childhood. The disease is triggered by environmental antigens, such as dusts, pollens, animal dander and foods, but potentially any antigen is implicated.

A positive family history of atopy is common, and asthmatic attacks are often preceded by allergic rhinitis, urticaria, or eczema.
185. What is the pathogenesis of atopic asthma?
T-cell differentiation in these pts appears to be skewed to overproduce TH2-type cells which release IL-4 and IL-5, with subsequent IgE and eosinophil dominated immune responses.

In presensitized pts, repeat antigen exposure causes a classic type I (IgE)-mediated hypersensitivity reaction. It is the IgE-mediated reaction to inhaled allergens that elicits an acute response and a late phase reaction.
186. What occurs in the acute response in atopic asthma?
In the acute phase, antigen binding to IgE-coated mast cells causes primary (e.g., leukotriene) and secondary (e.g., cytokine, neuropeptide) mediator release.

The acute phase mediators cause bronchospasm, edema, mucus secretion, and leukocyte recruitment, along with hypotension in extreme instances.

These inflammatory cells set the stage for the late-phase reaction, which starts 4-8 hours later and may persist for 12-24 hours or more.
197. What occurs in the late-phase reaction in atopic asthma?
A late-phase reaction is mediated by recruited leukocytes (e.g., eosinophils, lymphocytes, neutrophils, monocytes).

It is characterized by persistent bronchospasm and edema, leukocytic infiltration, and epithelial damage and loss.
198. What is eotaxin?
Eotaxin, produced by airway epithelial cells, is a potent chemoattractant and activator of eosinophils. The major basic protein of eosinophils, in turn, causes epithelial damage and airway constriction.
199. What are the three groups of mediators that are implicated in the asthmatic response?
1. This group includes mediators whose role in bronchospasm is clearly supported: leukotrienes C4, D4, and E4; and ACh which also cause smooth muscle constriction
2. These agents present at the scene of the crime with potent asthma-like effects: histamine, prostaglandin D2, and PAF.
3. These are suspects - IL-1, TNF, and IL-6, chemokines (eotaxin), neuropeptides, nitric oxide, bradykinin, and endothelins
200. What is nonatopic asthma?
This is the nonreaginic variety of asthma and is most frequently triggered by respiratory tract infection. Viruses, (e.g., rhinovirus, parainfluenza virus) rather than bacteria are the most common provokers.

A positive family history is uncommon, serum IgE levels are normal, and there are no other associated allergies.

***It is though that virus induced inflammation of the respiratory mucosa lowers the threshold of the subepithelial vagal receptors to irritants. Inhaled air pollutants, such as sulfur dioxide, ozone, and nitrogen dioxide, may also contribute to the chronic airway inflammation and hyperreactivity.
201. What is drug-induced asthma?
Several pharmacologic agents cause asthma. Aspirin-sensitive asthma is an uncommon yet fascinating type, occurring in ts with recurrent rhinitis and nasal polyps. These individuals are exquisitely sensitive to small doses of aspirin, and they experience not only asthmatic attacks but also urticaria.

It is probable that aspirin triggers asthma in these pts by inhibiting the COX pathway of arachidonic acid metabolism w/o affecting the lipoxygenase route, thus tipping the balance toward elaboration of the bronchoconstrictor leukotrienes.
202. What is occupational asthma?
This form of asthma is stimulated by fumes (epoxy resins, plastics), organic and chemical dusts (wood, cotton, platinum), gases (toulene), and other chemicals (formadehyde, penicillin products).

Minute quantities are required to induce the attack, which usually occurs after repeated exposure. The underlying mechanisms vary according to stimulus and include type I reactions, direct liberation of bronchoconstrictor substances, and hypersensitivity responses of unknown origin.
203. What is the morphologic changes in asthma?
Grossly, the lungs are overdistended b/c of overinflation, and there may be small areas of atelectasis.

*The most striking macroscopic finding is occlusion of bronchi and bronchioles by thick, tenacious mucous plugs.

*Histologically, the mucous plugs contain whorls of shed epithelium, which give rise to the well-known Curschmann spirals. Numerous eosinophils and CarcotLeyden crystals are present; the latter are collections of crystalloid made up of eosinophil membrane protein.
204. What are four other characteristic histologic findings of asthma that are collectively termed "airway remodeling"?
1. Thickening of the basement membrane of the bronchial epithelium
2. Edema and an inflammatory infiltrate in the bronchial walls, w/a prominence of eosinophils and mast cells
3. An increase in size of the submucosal glands
4. Hypertrophy of the bronchial wall muscle
205. What is the clinical course of asthma?
The classic asthmatic attack lasts up to several hours and is followed by prolonged coughing; the raising of copious mucous secretions provides considerable relief of the respiratory difficulty. In some pts, these symptoms persist at a low level all the time.

The clinical Dx is aided by the demonstration of an elevated eosinophil count in the peripheral blood and the finding of eosinophils, Curschmann spirals, and Charcot-Leyden crystals in the sputum.
206. What is status asthmaticus?
In its most severe form, status asthmaticus, the severe acute paroxysm persists for days and even weeks, and under these circumstances, ventilatory function might be so impaired as to cause severe cyanosis and even death.
207. What is bronchiectasis?
Bronchiectasis is a disease characterized by permanent dilation of of bronchi and bronchioles caused by destruction of the muscle and elastic tissue, resulting from or associated with chronic necrotizing infections.

To be considered bronchiectasis, the dilation should be permanent; reversible bronchial dilation often accompanies viral and bacterial pneumonia.

It is manifested clinically by cough, fever, and expectoration of copious amts of foul smelling, purulent sputum.
208. What are four types of conditions in in which bronchiectasis develops?
1. Congenital or hereditary conditions, including cystic fibrosis, intralobular sequestration of the lung, immunodeficiency states, and primary ciliary dyskinesia and Kartagener syndromes.

2. Postinfectious conditions, including necrotizing pneumonia caused by bacteria (Mycobacterium tuberculosis, Staph aureus, H. influenzae, Psuedomonas), viruses, and fungi (aspergillus).

3. Other conditions, including RA, SLE, IBD, and post-transplantation (chronic lung rejection, and chronic G vs. H disease after bone marrow transplantation).
209. What are the two major influences associated with bronchiectasis?
Obstruction and infection; it is likely that both are necessary for the development of full-fledged lesions, although either may come first.

*These mechanisms are most readily apparent in the severe form of bronchiectasis associated w/cystic fibrosis.
210. What is primary ciliary dyskinesia?
This is an autosomal-recessive syndrome with variable penetrance and a freq of 1:15,000 live births. In this disease, poorly functioning cilia contribute to the retention of secretions and recurrent infections that in turn lead to bronchiectasis.

There is an absence or shortening of the dynein arms that are responsible for the coordinated bending of the cilia.
211. What is Kartagener syndrome?
Approximately half of the pts with primary ciliary dyskinsia have Kartagener syndrome (bronchiectasis, sinusitis, and situs inversus or partial lateralizing abnormality).

The lack of ciliary activity interferes with bacterial clearance, predisposes the sinuses and bronchi to infection, and affects cell motility during embryogenesis, resulting in the situs inversus.

Males with this condition tend to be infertile, owing to ineffective mobility of the sperm tail.
212. What is allergic bronchopulmonary aspergillosis (ABPA)?
ABPA is a condition that results from a hypersensitivity reaction to the fungus Aspergillus fumigatus.

ABPA is also an important complication of asthma and CF. It is characterized by an intense airway inflammation w/eosinophils and the formation of mucus plugs, which play a primary role in its pathogenesis.

There is evidence that neutrophil-mediated inflammation and the potential deficiency of anti-inflammatory cytokines such as IL-10 may also play a role. Clinically, there are periods of exacerbation and remission that may lead to proximal bronchiectasis and fibrotic lung disease.
213. What is the morphology of bronchiectasis?
Bronchiectasis usually affects the lower lobes bilaterally, particularly air passages that are vertical, and is most severe in the more distal bronchi and bronchiles. When tumors or aspiration of foreign bodies leads to bronchiectasis, the involvement may be sharply localized to a single segment of the lung.

*The airways are dilated, sometimes up to 4x normal size. These dilations may produce long, tube-like enlargements (cylindrical bronchiectasis), or in other cases, may cause fusiform, or even sharply saccular distention (saccular bronchiectasis).
214. What are the characteristic findings of the bronchi and bronchioles in bronchiectasis?
Characteristically, the bronchi and bronchioles are sufficiently dilated that they can be followed, on gross exam, directly out to the pleural surfaces. By contrast, in the normal lung, the bronchioles cannot be followed by ordinary gross dissection beyond a point 2-3 cm removed from the pleural surfaces.

On the cut surface of the lung, the transected dilated bronchi appear as cysts filled w/mucopurulent secretions.
215. What are the histologic findings in bronchiectasis?
The histologic findings vary with the activity and chronicity of the disease. In the full-blown, active case, there is an intense acute and chronic inflammatory exudation within the walls of the bronchi and bronchioles, associated with desquamation of the lining epithelium and extensive areas of necrotizing ulceration. There may be pseudostratification fo the columnar cells or squamous metaplasia of the remaining epithelium.

In some instances, the necrosis completely destroys the bronchial or bronchiolar walls and forms a lung abscess. Fibrosis of the bronchial and bronchiolar walls and peribronchiolar fibrosis develop in the more chronic cases, leading to varying degrees of subtotal or total obliteration of bronchiolar lumina.
216. In the usual case of bronchiectasis, what flora can be cultured from the involved bronchi?
A mixed flora, including staphlococci, streptococci, pneumococci, enteric organisms, anaerobic and microaerophilic bacteria, and (particularly in children) H. influenzae and Pseudomonas aeruginosa.

In ABPA, a few fungal hyphae can be seen on special stains w/in the mucoinflammatory contents of the cylindrically dilated segmented bronchi. In the late stages, the fungus may infiltrate the bronchial wall.
217. What are the clinical features of bronchiectasis?
Bronchietasis causes severe, persistent cough; expectoration of foul-smelling, sometimes blood sputum; dyspnea and orthopnea in severe cases; and occasional life-threatening hemoptysis. A systemic febrile reaction may occur when powerful pathogens are present.

These symptoms are often episodic and are precipitated by upper respiratory tract infections or the introduction of new pathogenic agents.

In the full blown case, the cough is paroxysmal in nature due to frequent positional changes in position. Obstructive ventilatory insufficiency can lead to marked dyspnea and cyanosis.
218. What are diffuse interstitial diseases?
Diffuse interstitial diseases are a heterogeneous group of disorders characterized predominantly by diffuse and usually chronic involvement of the pulmonary connective tissue, principally the most peripheral and delicate interstitium in the alveolar walls.

The interstitium consists of the basement membrane of the endothelial and epithelial cells, collagen fibers, elastic tissue, proteoglycans, fibroblasts, a few mast cells, and occasional lymphocytes and monocytes.
219. In general, what are the clinical and pulmonary functional changes in diffuse interstitial diseases?
The clinical and pulmonary functional changes are those of restrictive rather than obstructive lung disease.

Pts have dyspnea, tachypnea, end-inspiratory crackles, and eventual cyanosis, w/o wheezing or other evidence of airway obstruction.
220. What are the classic physiologic and radiographic features of diffuse interstitial diseases?
*The classic physiologic features are reductions in carbon monoxide diffusing capacity, lung volume, and compliance.

*Chest radiographs show diffuse infiltration by small nodules, irregular lines, or ground glass shadows. Eventually, secondary pulmonary hypertension and right sided HF with cor pulmonale may result.

*The advanced stages are hard to differentiate b/c they result in scarring and gross destruction of the lung, referred to as end-stage lung or honeycomb lung.
221. What are the 5 major categories of chronic interstitial lung disease?
1. Fibrosing
2. Granulomatous
3. Eosinophilic
4. Smoking-related
5. Other
222. What is the earliest common manifestation of most of the interstitial diseases?
Alveolitis, that is, an accumulation fo inflammatory and immune effector cells w/in the alveolar walls and spaces.

The accumulation of leukocytes has two consequences: it distorts the normal alveolar structures, and it results in the release of mediators that can injure parenchymal cells and stimulate fibrosis.
223. What is the initial stimuli for alveolitis?
Some are related to the inhalation of ROS and some chemicals, which are directly toxic to endothelial cells, epithelial cells, or both.

Beyond direct toxicity, a critical event is the recruitment and activation of inflammatory and immune effector cells.

Also, alveolar macrophages release chemotactic factors for neutrophils (IL-8 and leukotriene B4).
224. So, what causes the fibrosis in the later stages of interstitial diseases?
It is thought that interactions among lymphocytes and macrophages and the release of lymphokines and monokines are responsible for the slowly progressive pulmonary fibrosis that ensues.

The alveolar macrophage, in particular, plays a central role in the development of fibrosis.
225. What is idiopathic pulmonary fibrosis?
This disorder with an unknown cause refers to a syndrome with characteristic radiologic, pathologic, and clinical features.

IPF is characterized by progressive pulmonary interstitial fibrosis resulting in hypoxemia.

The histologic pattern of fibrosis is referred to as usual interstitial pneumonia (UIP).
226. What is the pathogenesis of IPF?
IPF is caused by "repeated cycles" of acute lung injury (alveolitis) by some unidentified agent.

Wound healing at these sites gives rise to exuberant fibroblastic proliferation, giving rise to the "fibroblastic foci" that are so characteristic of IPF.

Mediators of wound healing such as TGF-beta are expressed at these sites.
227. What mediators are active in IPF lesions?
May be modified by TH2 inflammatory response. Thus, esoinophils, mast cells, and IL-4 and IL-131 are found in the lesions.

Also, there is an abnormal activation of the Wnt-β-catenin signaling pathway withing the mesenchymal cells of the IPF lesions.
228. What is the morphology of IPF?

1/2
Grossly, the pleural surfaces of the lung are cobblestoned owing to the retraction of scars along the interlobular septa. The cut surface shows fibrosis (firm, rubbery white areas) of the lung parenchyma with lower lobe predominance and a distinctive distribution the subpleural regions and along the interlobular setpa.

***Microscopically, the hallmark of the UIP is patchy interstitial fibrosis, which varies in intensity and with time.
229. What is the morphology of IPF?

2/2
The earliest lesions contain exhuberent fibroblastic proliferation and appear as fibroblastic foci. W/time these areas become more collagenous and less cellular. Quite typical is the coexistence of both early and late lesions.

The dense fibrosis causes collapse of alveolar walls and formation of cystic spaces lined by hyperplastic type II pneumocytes or bronchiolar epithelium (honeycomb fibrosis).
230. What is the clinical course of IPF?
IPF begins insidiously, with gradually increasing DOE and dry cough. Most pts are 40-70 years old at the time of presentation.

Hypoxemia, cyanosis, and clubbing occur late in the course. Most pts have a gradual deterioration of their pulmonary status despite medical treatment.

The mean survival is 3 years or less. Lung transplantation is the only definitive therapy available.
231. What is nonspecific interstitial pneumonia?
NSIP is a diffuse interstitial lung disease of unknown etiology whose lung biopsies fail to show diagnostic features of any of the other well-characterized interstitial diseases. NSIP is divided into cellular and fibrosing patterns.

Pts present with dyspnea and cough of several mos duration. They are typically between 46-55 y/o.

*These pts have a much better prognosis than do those with UIP.
232. What is the morphology of NSIP?
The cellular pattern consists primarily of mild to moderate chronic interstitial inflammation, containing lymphocytes and a few plasma cells, in a uniform or patchy distribution.

The fibrosing pattern consists of diffuse or patchy, interstitial fibrosis w/o the temporal heterogeneity that is characteristic of UIP.

Fibroblastic foci are absent, suggesting that NSIP is not caused by alveolitis.

Those having the cellular pattern somewhat younger and they also have a better outcome than those with the fibrosing pattern or UIP.
233. What is cryptogenic organizing pneumonia?
COP is synonymous w/the term "bronchiolitis obliterans organizing pneunomia".

Pts present w/cough and dyspnea and have subpleural or peribronchial patchy areas of airspace consolidation radiographically. Some pts recover spontaneously but most need treatment with oral steroids for 6 mos or longer for complete recovery.
234. What are the histologic characteristics of COP?
***Histologically, COP is characterized by the presence of polypoid plugs of loose organizing CT w/in alveolar ducts, alveoli, and often bronchioles.

The CT is all of the same age, and the underlying lung architecture is normal.

There is no interstitial fibrosis or honeycomb lung.
235. What is COP with intra-alveolar fibrosis?
COP with intra-alveolar fibrosis can be seen as a response to infections or inflammatory injury of the lungs. These include viral and bacterial pneumonia, inhaled toxins, drugs, collagen vasscular disease, and G vs. H disease in bone marrow transplant recipients.

The prognosis is the same for these pts as that for the underlying disorder.
236. What is the pulmonary involvement in collagen vascular diseases?
Many collagen vascular disease, (e.g., SLE, RA, and scleroderma) can involve the lung.

Patterns include NSIP, UIP, vascular sclerosis, organizing pneumonia, and bronchiolitis.

NSIP pattern occurs classically in scleroderma.

Pulmonary involvement in these disease is usually associated with a poor prognosis.
237. What are the 4 forms of pulmonary involvement in RA?
1. Chronic pleuritis, w/or w/o effusion
2. Diffuse interstitial pneumonia and fibrosis
3. Intrapulmonary rheumatoid nodules
4. Pulmonary hypertension

*30-40% of pts w/classic RA have abnormalities in lung function.
238. What are pneumoconioses?
These disorders refer to the non-neoplastic lung reaction to aerosols, including mineral dusts, organic dusts, fumes and vapors. Air particulates also play a role in their formation.
239. The development of pneumoconiosis depends on what 4 things?
1. The amt of dust retained in the lungs and airways
2. The size, shape, and therefore buoyancy of the particles
3. Particle solubility and physiochemical reactivity
4. The possible additional effects of other irritants such as concommitant cigarette smoking
240. Are the most dangerous particles in pneumoconiosis large or small?
The most dangerous particles range from 1-5 μm in diameter b/c they may reach the terminal small airways and air sacs and settle in their linings.

Also, in general, the smaller the particles, the more likely it is to appear in the pulmonary fluids and reach toxic levels rapidly; therefore, smaller particles tend to cause acute lung injury.
241. Can larger particles cause injury?
Larger particles resist dissolution and so may persist w/in the lung parenchyma for years.

These tend to evoke fibrosing collagenous pneumoconioses, such as is characteristic of silicosis.
242. What can exacerbate the local reaction of lung injury due to asbestos?
Although tobacco smoking worsens the effects of all inhaled mineral dusts, the effects of asbestos are particularly magnified by smoking.
243. What is coal worker's pneumoconiosis?
These symptoms can range from:
(1) Asymptomatic anthracosis to (2) simple CWP with little to no pulmonary dysfunction to (3) complicated CWP, or progressive massive fibrosis, in which lung function is compromised.

Contaminating silica in the coal dust can favor progressive disease. in most cases, carbon dust itself is the major culprit, and studies have shown that complicated lesions contain considerably more dust than simple lesions do.
244. What is anthracosis?

What is the morphology of anthracosis?
Anthracosis is the most innocuous coal-induced pulmonary lesion in coal miners and is commonly seen in all urban dwellers and tobacco smokers. Inhaled carbon pigment is engulfed by alveolar or interstitial macrophages, which ten accumulate in teh CT along the lymphatics, including the pleural lymphatics, or in organized lymphoid tissue along the bronchi or in the lung hilus.

At autopsy, linear streaks and aggregates of anthracotic pigment readily identify pulmonary lymphatics and mark the pulmonary lymph nodes.
245. What is the morphology of simple CWP?
Simple CWP is characterized by coal macules (1-2 mm in diameter) and the somewhat large coal nodules.

The coal macules consist of carbon-laden macrophages; the nodule also contains small amts of a delicate network of collagen fibers.

These lesions can be scattered throughout the lung, but the upper lobes and upper zones of the lower lobes are more heavily involved. They are located primarily adjacent to respiratory bronchioles, the site of initial dust accumulation. In time it can lead to centrilobular emphysema.
246. What is the morphology of complicated CWP?
Complicated CWP occurs on a background of simple CWP and generally requires many years to develop. It is characterized by intensely blackened scars larger than 2 cm, sometimes up to 10 cm in greatest diameter.

They are usually multiple. Microscopically, the lesions consist of dense collagen and pigment. The center of the lesion is often necrotic, resulting most likely from local ischemia.
247. What is the clinical course of CWP?
CWP is usually a benign disease that causes little decrement in lung function. Even mild forms of complicated CWP fail to demonstrate abnormalities of lung function. In a minority of cases (fewer than 10%), complicated CWP develops, leading to increasing pulmonary dysfunction, pulmonary hypertension, and cor pulmonale.

Once that develops, it may become progressive even if further exposure to dust is prevented.
248. What is silicosis?
Silicosis is a lung disease caused by inhalation of crystalline silicon dioxide. Currently, the most preevent chronic occupational disease in the world, silicosis usually presents after decades of expsoure as slowly progressing, nodular, fibrosing pneumoconiosis.
249. What is the pathogenesis of silicosis?
Silica occurs in both crystalline and amorphous forms, but crystalline forms are much more fibrogenic. Of these, quartz is most commonly implicated.

After inhalation, the particles interact w/epithelial cell and macrophages. Although lung macrophages that ingest the silica particles may ultimately succumb to its toxic effects, silica causes activation and release of mediators by viable macrophages. These mediators include IL-1, TNF, fibronectin, lipid mediators, ROS, and fribrogenic cytokines.
250. Interesting note about quartz
It has been noted that when mixed with other minerals, quartz has a reduced fibrogenic effect. This is important b/c quartz in the workplace is rarely pure.
251. What is the morphology of silicosis?
Silicosis is characterized grossly in its early stages by tiny, barely palpable, discrete pale to blackened (if coal dust is also present) nodules in the upper zones of the lungs. As the disease progresses, these nodules may coalesce into hard, collagenous scars. Some nodules may undergo central softening and cavitation. This change may be due to superimposed tuberculosis or ischemia.

Fibrotic lesions may also occur in the hilar lymph nodes and pleura. ****Sometimes, thin sheets of calcification occur in the lymph nodes and are seen radiographically as eggshell calcifications.

Histologically, the nodular lesions consist of concentric layers of hyalinized collagen surrounded by a dense capsule of more condensed collagen.

***Examination of the nodules by polarized microscopy reveals the birefringent silica particles.
252. What is the clinical course of silicosis?
The disease is usually detected when routine chest xray is performed on an asymptomatic worker. The radiographs typically show a fine nodularity in the upper zones of the lung, but pulmonary functions are either normal or only moderately affected.

Most pts do not develop dyspnea until later in the course. At this time, the disease may be progressive even if the pt is no longer exposed.
253. Silicosis is associated with increased susceptibility to...?
Tuberculosis. It is postulated tha tilicosis results in a depression of cell-mediated immunity, and crystalline silica may inhibit the ability of pulmonary macrophages to kill phagocytosed mycobacteria.

Nodules of silicotuberculosis often display a central zone of caseation.
254. Occupational exposure to asbestos is linked to what 6 things...?
1. Localized fibrous plaques or rarely diffuse pleural fibrosis
2. Pleural effusions
3. Parenchymal interstitial fibrosis (asbestosis)
4. Lung CAs
5. Mesotheliomas
6. Laryngeal and perhaps other extrapulmonary neoplasms, including colon CA
255. What are the two distinct geometric forms of asbestos?
Serpentine (curly and flexible fibers) and amphibole (straight, stiff, and brittle fibers.

The serpentine form accounts for most of the asbestos used in industry.

However, it is the amphibole stiff fibers that are delivered deeper into the lungs, where they can penetrate epithelial cells and cause fibrogenic disease.

*Thus, only amphibole exposure correlates with mesothelioma.
256. How is asbestos a tumor initiator/promoter?
Some of the oncogenic effects of asbestos are mediated by ROS generated by asbestos fibers, which preferentially localize in the distal lung, close to the mesothelial layers.

In addition, adsorption of carcinogens in tobacco smoke onto asbestos fibers may well be important in the synergy between tobacco smoking and the development of lung CA.
267. What is the disease like in asbestosis?
Chronic deposition of fibers and persistent release of mediators eventually lead to generalized interstitial pulmonary inflammation and *interstitial fibrosis*
268. What is the morphology of the diffuse pulmonary interstitial fibrosis in asbestosis?
Indistinguishable from other causes, except for the presence of asbestos bodies.

Asbestos bodies appear as golden brown, fusiform or beaded rods w/a translucent center and consist of asbestos fibers coated with an iron containing proteinaceous material.

They arise when macrophages attempt to phagocytose asbestos fibers; the iron is presumably derived from phagocyte ferritin. Other inorganic particulates may become coated with similar iron protein complexes and are called ferruginous bodies.
269. Where does asbestosis begin?
In contrast to CWP and silicosis, asbestosis begins in the lower lobes and subpleurally. The middle and upper lobes of the lungs become affected as fibrosis progresses. The scarring may trap and narrow pulmonary arteries and arterioles causing pulmonary hypertension and cor pulmonale.
270. What are pleural plaques?
Pleural plaques are the most common manifestation of asbestos exposure. They are well-circumscribed plaques of dense collagen, often containing calcium. They develop most frequently on the anterior and posterolateral aspects of the parietal pleura and over the domes of the diaphragm.

They do not contain asbestos bodies; however, only rarely do they occur in individuals w/o asbestos exposure.
271. What is the clinical course of asbestosis?
Dyspnea is usually the first manifestation; at first, it is provoked by exertion, but later it is present even at rest. The dyspnea is usually accompanied by a cough associated w/sputum.

These manifestations rarely appear fewer than 10 years after first exposure and are more common after 20 years or more. The disease may remain static or progress to respiratory failure, cor pulmonale, and death.

With advancement of pneumoconiosis, a honeycomb pattern develops. Asbestosis complicated by lung or pleural CA is associated with a particularly grim prognosis.
272. What are drug-induced lung disease?
Drugs can cause both acute and chronic alterations in respiratory structure and function, including bronchospasm, pulmonary edema, diffuse alveolar damage, organizing pneumonia, interstitial fibrosis, bronchiolitis obliterans, and esoinophilic pneumonia.

For example, cytotoxic drugs used in CA treatment, (e.g., bleomycin) cause pulmonary damage and fibrosis as a result of direct toxicity of the drug and by stimulating the influx of inflammatory cells into the alveoli.

Amiodarone is preferentially concentrated in the lung and causes significant pneumonitis in 5-15% of pts receiving it.
273. What about acute radiation pneumonitis?
After clinical fractionated irradiation, acute radiation pneumonitis occurs in 1020% of pts, 1-6 mos after therapy, manifested by fever, dyspnea out of proportion to the volume of lung irradiated, pleural effusion, and radiologic infiltrates that usually correspond to an area of previous radiation.

With steroid therapy, these symptoms may resolve completely in some pts without long-term effects.
274. What about chronic radiation pneumonitis?
There is increasing evidence that irradiation of the lungs initially causes a lymphocytic alveolitis or hypersentivity pneumonitis that can lead to pulmonary fibrosis (chronic radiation pneumonitis). The latter is a consequence of repair, which is initiated by direct tissue injury to endothelial and epithelial cells w/in the radiation portal.

Morphologic changes are those of DAD, including severe atypia of hyperplastic type II cells and fibroblasts. Form cells and epithelial cell atypia within vessel walls are also characteristic of radiation damage.
275. What is sarcoidosis?
Sarcoidosis is a systemic disease of unknown cause characterized by noncaseating granulomas in many tissues and organs. It presents in many clinical patterns, but bilateral hilar lymphadenopathy or lung involvement is visible on chest xrays in 90% of cases. Eye and skin lesions are next in frequency.

The prevalence of sarcoidosis is higher in women than in men; in the US, the rates are highest in the Southeast; they are 10x higher in blacks than in whites.
276. What are 3 local immunologic factors in the pathogenesis of sarcoidosis?
1. Intra-alveolar and intersitital accumulation of CD4+ T cells; antigen-driven proliferation
2. Increased levels of TH1 cytokines such as IL-2 and IFN-gamma, resulting in T-cell expansion and macrophage activation
3. Increased levels of several cytokines in the local environment (IL-8, TNF, macrophage inflammatory protein 1-alpha)
277. What are 2 systemic immunologic factors in the pathogenesis of sarcoidosis?
1. Anergy to common skin test antigens such as Candida or PPD
2. Polyclonal hypergammaglobulinemia, another manifestation of helper-T cell dysregulation
278. What are the genetic influences in sarcoidosis?
1. Familial and racial clustering of cases
2. Association with certain HLA genotypes (e.g., class I HLA-A1 and HLA-B8)
279. What are the environmental factors associated with sarcoidosis?
Several putative microbes have been proposed as the inciitng agent for sarcoidosis (e.g., mycobacteria, Propionibacterium acnes, and Rickettsia species).

*To date, there is no unequivocal evidence to suggest that sarcoidosis is caused by an infectious agent.
280. What are the histological characteristics of sarcoidosis?
Histologically, all involved tissues show the classic noncaseating granulomas, each composed of an aggregate of tightly clustered epithelioid cells, often with Langhans or foreign body type giant cells. Central necrosis is unusual.

With chronicity the granulomas may become enclosed w/in fibrous rims or may eventually be replaced by hyaline fibrous scares.
281. What are two microscopic features often present in the granulomas of sarcoidosis?
1. Laminated concretions composed of calcium and proteins known as Schaumann bodies

2. Stellate inclusions known as asteroid bodies enclosed within giant cells found in approximately 60% of the granulomas.

*Although characteristic, these microscopic features are not pathognomonic of sarcoidosis b/c asteroid and Schaumann bodies maybe encountered in tuberculosis.
282. What is the morphology of the lungs in sarcoidosis?
The lungs are common sites of involvement. Macroscopically, there is usually no demonstrable alteration, although at times, the coalescence of granulomas may produce small nodules that are palpable or visible as 1-2 cm noncaseating, noncavitated consolidations.

Histologically, the lesions are distributed primarily along the lymphatics, around bronchi and blood vessels, although alveolar lesions are also seen.

*A CD4/CD8 ratio > 2.5 and the CD3/CD4 ratio < 0.31 is bronchoalveolar lavage lymphocytes is commonly seen in sarcoidosis.
283. What is the morphology of the lymph nodes in sarcoidosis?
Lymph nodes are involved in almost all cases specifically the hilar and mediastinal nodes, but any other node in the body may be involved.

Nodes are characteristically enlarged, discrete, and sometimes calcified. The tonsils are affected in about 25-33% of cases.
284. How are the spleen and liver affected in sarcoidosis?
The spleen is affected microscopically in about 75% of cases, but it is enlarged in only one fifth. On occasion, granulomas may coalesce to form small nodules that are barely visible macroscopically. The capsule is not involved.

The liver is affected slightly less often than the spleen. It may also be moderately enlarged and may contain scattered granulomas, more in portal triads than in the lobular parenchyma.
285. What about the bone marrow and sarcoidosis?
The bone marrow is an additional favored site of localization. Roentgenographic changes can be identified in about one firth of cases of systemic involvement.

The radiologically visible bone lesions have a particular tendency to involve phalangeal bones of the hands and feet, creating small circumscribed areas of bone resorption within the marrow cavity and a diffuse reticulated pattern throughout the cavity, with widening of the bony shafts or new bone formation on the outer surfaces.
286. What are the skin lesions in sarcoidosis?
Sin lesions are encountered in one third to one half of cases. Sarcoidosis of the skin assumes a variety of macroscopic appearances (e.g., discrete subcutaneous nodules; focal, slightly elevated, erythematous plaques; or flat lesions that are slightly reddened and scaling and resemble those of SLE).

Lesions may also appear on the mucous membranes of the oral cavity, larynx, and upper respiratory tract.
287. What are the ocular involvements like in sarcoidosis?
The eye, its associated glands, and the salivary glands are involved in one fifth to half of cases.

The ocular involvement takes the form of iritis or iridocyclitis, either bilaterally or unilaterally. Consequently, corneal opacities, glaucoma, and total loss of vision may occur. These ocular lesions are frequently accompanied by inflammation of the lacrimal glands, with suppression of lacrimation.
288. What is Mikulicz syndrome?
Bilateral sarcoidosis of the parotid, submaxillary, and sublingual glands completes the combined uveoparotid involvement designated as Mikulicz syndrome.
289. What are the muscle involvements in sarcoidosis?
Muscle involvement is often undiagnosed, since is may be asymptomatic. Symptoms of muscle weakness, aches, tenderness, and fatigue should prompt consideration of occult sarcoid myositis. Sarcoid noncaseating granulomas can be found in muscle biopsies.

Sarcoid granulomas occasionally occur in the heart, kidneys, CNS, and endocrine glands, particularity in the pituitary, as well as in other body tissues.
290. What is the clinical course of sarcoidosis?
May follow an unpredictable course:
-It can be slowly progressive
-It may pursue a remitting and resolving course (w/ or w/o steroid therapy)
-It can spontaneously resolve
-In 65-70% of pts, there are no or only minimal residual manifestations; 20% have permanent lung or ocular dysfunction; and 10% of pts die, primarily from progressive pulmonary fibrosis
291. What is hypersensitivity pneumonitis?
This immunologically mediated disorder is caused by intense, often prolonged exposure to inhaled organic dusts and related occupational antigens.

Affected individuals have an abnormal sensitivity or heightened reactivity to the antigen, which in contrast to asthma, involves primarily the alveoli.

***It is important to recognize these diseases early in their course b/c progression to serious chronic fibrotic lung disease can be prevented by removal of the environmental agent.
292. What causes the hypersensitivity?
It results from the inhalation of organic dust containing antigens made up of spores of thermophilic bacteria, true fungi, animal proteins, or bacterial products. Uses type III and type IV hypersensitivity reactions.

Examples:
1. Farmer's lung from exposure to dusts from hay that permits actinomycetes sports
2. Pigeon breeder's lung
3. Humidifier or A/C lung is caused by thermophilic bacteria in heated water reservoirs.
293. What are fives lines of evidence supporting that hypersensitivity pneumonitis is an immunologically mediated disease?
1. Bronchoalveolar lavage specimens obtained during the acute phase show increased levels of proinflammatory chemokines such as MIP-1alpha and IL-8.
2. Bronchoalveolar lavage specimens also consistently demonstrate increased numbers of T lymphs of both CD4+ and CD8+ phenotypes.
3. Most pts have specific antibodies in their serum, a feature suggestive of type II (immune complex) hypersensitivity
4. Complement and immunoglobulins have been demonstrated w/in vessel walls, also indicating type III hypersensitivity.
5. The presence of noncaseating granulomas in 2/3's of pts suggests the development of a T cell-mediated (type IV) delayed-type hypersensitivity against the implicated antigens.
294. What is the morphology of hypersensitivity pneumonitis?
Histologic changes in subacute and chronic forms are characteristically centered on bronchioles.

They include (1) interstitial pneumonitis consisting primarily of lymphocytes, plasma cells, and macrophages; (2) noncaseating granulomas in 2/3rds of pts; and (3) interstitial fibrosis and obliterative bronchiolitis (in later stages).
295. What are the clinical features of hypersensitivity pneumonitis?
The clinical manifestations are varied. Acute attacks, which follow inhalation of antigenic dust in sensitized pts, consist of recurring episodes of fever, dyspnea, cough, and leukocytosis. Diffuse and nodular infiltrates appear in the chest Xray and pulmonary function tests show an acute restrictive disorder.

Symptoms usually appear 4-6 hrs after exposure. If exposure is continuous and protracted, a chronic form of the disease supervenes that no long features the acute exacerbations on antigen re-exposure. Instead, there are signs of progressive respiratory failure, dyspnea, and cyanosis and a decrease in total lung capacity and compliance.
296. What is pulmonary esoinophila?
Pulmonary eosinophilia refers to diverse clinicopathologic conditions characterized by eosinophil infiltrates in pulmonary interstitial or alveolar spaces.
297. What are the five types of pulmonary eosinophilia?
1. Acute eosinophilic pneumonia w/respiratory failure
2. Simple pulmonary esoinophilia or Loffler sydnrome
3. Tropical esoinophilia, caused by infection with microfilariae
4. Secondary eosinophilia
5. So called idiopathic chronic eosinophilic pneumonia
298. What is acute eosinophilic pneumonia w/respiratory failure?
This is an acute illness of unknown cause. It has a rapid onset w/fever, dyspnea, and hypoxemic respiratory failure.

The chest X-ray shows diffuse infiltrates and bronchoalveolar lavage fluid contains more than 25% eosinophils.

*There is a prompt response to corticosteroids.
299. What is simple pulmonary eosinophilia or Loffler syndrome?
Simple pulmonary eosinophilia is characterized by transient pulmonary lesions, eosinophilia in the blood, and a benign clinical course. Roentgenograms are often quite striking, w/shadows of varying size and shape in any of the lobes, suggesting irregular intrapulmonary densities.

They alveolar septa are thickened by an infiltrate composed of eosinophils and occasional interspersed giant cells, but there is no vasculitits, fibrosis, or necrosis. In some cases, the eosinophils are found in a background of DAD.
300. What is chronic eosinophilic pneumonia?
Chronic eosinophilic pneumonia is characterized by focal areas of cellular consolidation of the lung substance distributed chiefly in the periphery of the lung fields.

Prominent in these lesions are heavy aggregates of lymphocytes and eosinophils w/in both the septal walls and the alveolar spaces.

These pts have high fever, night sweats, and dyspnea, all of which respond to corticosteroid therapy.

Chronic eosinophilic pneumonia is Dx when other causes of chronic pulmonary eosinophilia are excluded.
301. What is desquamative interstitial pneumonia (DIP)?

What is the morphology?
***Desquamative interstitial pneumonia is characterized by large intra-alveolar collections of macrophages with abundant cytoplasm containing dusty brown pigment (smoker's macrophages) in the airspaces.***

Some of the macrophages contain lammellar bodies (surfactant) w/in phagocytic vacuoles, presumably derived from necrotic type II pneumocytes.

The septa are lined by plump cuboidal pneumocytes. Interstitial fibrosis, when present, is mild. Emphysema is often present.
302. What is the clinical course of DIP?
DIP usually presents in the 4th or 5th decade of life, and it is more common in men than in women by a ratio of 2:1.

Virtually all pts are cigarette smokers. Presenting symptoms include an insidious onset of dyspnea and dry cough over weeks or months, often associated w/clubbing of digits. Pulmonary functions usually show a mild restrictive abnormality w/a moderate reduction of the diffusing capacity of carbon dioxide.

Pts with DIP typically have a good prognosis with excellent response to steroid therapy and cessation of smoking.
303. What is respiratory bronchiolitis-associated interstitial lung disease?
Respiratory bronchiolitis is a common histologic lesion found in cigarette smokers. It is characterized by the presence of pigmented itraluminal macrophages within first and second order respiratory bronchioles.

In its mildest form , it is seen most often as an incidental histologic finding in the lungs of smokers or ex-smokers.
304. What is the morphology of respiratory bronchiolitis-associated interstitial lung disease?
The changes are patchy at low magnification and have a bronchiolocentric distribution. ***Respiratory bronchioles, alveolar ducts, and peribronchiolar spaces containing aggregates of dusty brown macropahges (smoker's macrophages) similar to those seen in DIP.***

There is a patchy submucosal and peribronchiolar infiltrate of lymphocytes and histiocytes. Mild peribronchiolar fibrosis is also seen, which expands contiguous alveolar septa. Centrilobular emphysema is common but not severe.
305. What are the clinical features of respiratory bronchiolitis-associated interstitial lung disease?
Symptoms are usually mild, consisting of gradual onset of dyspnea and cough in pts who are typically current smokers in the 4th or 5th decade of life with average exposures of over 30 pack years of cigarette smoking.

There is a 2:1 male predominance. Cessation of smoking usually results in improvement.
306. What is pulmonary alveolar proteinosis (PAP)?
PAP is a rare disease that is characterized radiologically by bilateral patchy asymmetric pulmonary opacification and histologically by ***accumulation of accelular surfactant in the intra-alveolar and bronchiolar spaces.***

There are three classes of this disease: acquired, congenital, and secondary PAP.
307. What is acquired PAP?
Acquired PAP accounts for 90% of cases.

Autoimmune anti-GM-CSF antibodies may be pathogenic. These antibodies ultimately lead to impaired surfactant clearance by pulmonary macrophages.

Thus, acquired PAP can be considered an autoimmune disorder.
308. What is congenital PAP?
Congenital PAP is a rare cause of immediate-onset neonatal respiratory distress. To date, mutation in three genes have been found for congenital PAP: sufactant protein B (SP-B), GM-CSF, and GM receptor beta chain.

SP-B deficiency is transmitted in an autosomal recessive manner and is most often caused by homozygosity for a frameshift mutation in the SP-B gene. This leads to an unstable SP-B mRNA, reduced or absent SP-B, secondary disturbances of SP-C, and intra-alveolar accumulation of SP-A and SP-C.
309. What is secondary PAP?
Secondary PAP is uncommon. The underlying causes include lysinuric protein intolerance, acute silicosis, and other inhalational syndrome, immunodeficiency disorders, malignancies, and hematopoietic disorders.
310. What is the morphology of PAP?
The disease is characterized by a peculiar homogeneous, granular precipitate within the alveoli, causing focal-to-confluent consolidation of large areas of the lungs w/minimal inflammatory reaction. On section, turbid fluid exudes from these areas. As a consequence, there is a marked increase in the size and weight of the lung.

The alveolar precipitate is PAS positive and also contains cholesterol clefts. Immunohistochemical stains show the presence of surfactant proteins A and C in congenital SP-B deficiency and all three proteins in the acquired form.
311. What are the clinical features of PAP?
Adult pts for the most part, present with nonspecific respiratory difficultly of insidious onset, cough, and abundant sputum that often contains chunks of gelatinous material.

Some pts have symptoms lasting for years, often with febrile illnesses. These pts are at risk for developing secondary infections with a variety of organisms. Progressive dyspnea, cyanosis, and respiratory insufficiency may occur, but some pts tend to have a benign course.

Whole-lung lavage remains the standard of care.
312. What about congenital PAP?
Congenital PAP is a fatal respiratory disorder that is usually apparent in the newborn. Typically, the infant is full term and rapidly develops progressive respiratory distress shortly after birth. W/o lung transplantation, death ensues between 3-6 months of age.
313. Blood clots that occlude the large pulmonary arteries are almost always from where?
Blood clots that occlude the large pulmonary arteries are almost always emoblic in origin.

The usual source of pulmonary emboli is thrombi in the deep veins of the legs in more than 95% of cases.
314. What type of people are predisposed to having pulmonary embolisms?
Pulmonary embolism is a complication principally in pts who are already suffering from some underlying disorder, such as cardiac disease or CA, or who are immobilized for several days or weeks, those w/hip fracture being at high risk.

Hypercoagulable states, either primary (e.g. Factor 5 leiden, prothrombin 20210 A, hyperhomocyteinemia, and antiphospholipid syndrome) or secondary (e.g., obesity, recent surgery, CA, oral contraceptive use, pregnancy) are frequent risk factors.

Also indwelling central venous lines can be a nidus for right atrial thrombus, which can be a source of pulmonary embolism.
315. The pathophysiologic response and clinical significance of pulmonary embolism depends on...?
Depends on the extent to which the pulmonary artery blood flow is obstructed, the size of the occluded vessel(s), the number of emboli, the overall status of the cardiovascular system, and the release of vasoactive factors such as thromboxane A2 from platelets that accumulate at the site of thrombus.
316. Emboli result in what 2 main pathophysiologic consequences?
1. Respiratory compromise owing to the nonperfused, although ventilated, segment.

2. Hemodynamic compromise owing to increased resistance to pulmonary blood flow engendered by the embolic obstruction. This leads to pulmonary hypertension and can cause acute right-sided heart failure.
317. What are the morphologic consequences of embolic occlusion of the pulmonary arteries?
This depends on teh size of the embolic mass and the general stae of the circulation. Large emboli may impact in the main pulmonary artery or its major branches or lodge at the bifurcation as a saddle embolus. Sudden death often ensues, owing largely to the blockage of blood flow thru the lungs. Death may also be caused by acute cor pulmonale.

Smaller emboli can travel out into the more peripheral vessels, where they may cause infarction.
318. When are infarctions not a serous problem?

What occurs instead?
In pts with adequate cardiovascular function, the bronchial arterial supply can often sustain the lung parenchyma despite obstruction to the pulmonary arterial system.

Under these circumstances, hemorrhages may occur, but there is no infarction of the underlying lung parenchyma.

Only about 10% of emboli actually cause infarction. Although the underlying pulmonary architecture may be obscured by the suffusion of blood, ***hemorrhages are distinguished by the preservation of the pulmonary alveolar architecture***; in such cases, resorption of the blood permits reconstitution of the preexisting architecture.
319. So, when do pulmonary embolisms cause infarction?
PEs usually causes infarction only when the circulation is already inadequate, as in pts with heart or lung disease.

About 3/4 of all infarcts affect the lower lobes, and in more than half, multiple lesions occur. They vary in size from lesions that are barely visible to the naked eye to massive involvement of large parts of an entire lobe.

Characteristically, they extend to the periphery of the lung substances as a wedge with the apex pointing toward the hilus of the lung. In many cases, an occluded vessel can be identified near the apex of the infarct.
320. What is the morphology of a pulmonary infarct?
The pulmonary infarct is classically hemorrhagic and appears as a raised, red-blue area in the early stages. Often, the apposed pleural surface is covered by a fibrinous exudate.

The red cells begin to lyse w/in 48 hours, and the infarct becomes paler and eventually red brown as hemosiderin is produced. With the passage of time, fibrous replacement begins at the margins as a gray-white peripheral zone and eventually converts the infarct into a contracted scar.
321. What are the histologic features of pulmonary infarcts?
Histologically, the diagnostic feature of acute pulmonary infarction is the ischemic necrosis of the lung substance within the area of hemorrhage, affecting the alveolar walls, bronchioles, and vessels. If the infarct is caused by an infected embolus, it is modified by a more intense neutrophilic exudation and more intense inflammatory reaction.

Such lesions are referred to as septic infarcts, and some convert to abscesses.
322. What is one of the few causes of virtually instantaneous death?
A large pulmonary embolism.

During CPR resuscitation in such instances, the pt frequently is said to have electromechanical dissociation, in which the EKG has a rhythm but no pulses are palpated b/c of the massive blockage of blood in the systemic venous circulation.

If the pt survives after a sizable pulmonary embolus, however, the clinical syndrome may mimic MI, with severe chest pain, dyspnea, shock, elevation of temperature, and increased levels of serum lactic dehydrogenase.
323. What about small emboli?
Usually, however, in individuals with normal cardiovascular system small emboli induce only transient chest pain and cough or possibly pulmonary hemorrhages w/o infarction. Only in the predisposed, in whom the bronchial circulation itself is inadequate, do small emboli cause small infarcts. Such pts manifest dyspnea, tachypnea, fever, chest pain, cough, and hemoptysis.
324. How are PE's diagnosed?
The chest radiograph may disclose a pulmonary infarct, usually 12-36 hours after it has occurred, as a wedge-shaped infiltrate. Emboli can also be detected by spiral CT angiography and D-dimer testing. *Pulmonary angiography is the most definitive diagnostic technique but entails more risk to the pt.
325. What happens after the initial acute insult from a PE?
The emboli often resolve via contraction and fibrinolysis, particularly in the relatively young. Unresolved, multiple small emboli over the course of time may lead to pulmonary hypertension, pulmonary vascular sclerosis, and chronic cor pulmonale.

*Most important is the fact that a small embolus may presage a large one. In the presence of an underlying predisposing factor, pts with a pulmonary embolus have a 30% chance of developing a second embolus.
326. What is pulmonary hypertension?
Pulmonary hypertension is when the mean pulmonary pressure reaches one fourth of systemic levels.

It is most frequently secondary to structural cardiopulmonary conditions that increase pulmonary blood flow or pressure (or both), pulmonary vascular resistance, or left heart resistance to blood flow.
327. What are these structural cardiopulmonary conditions that can lead to pulmonary hypertension (4 of them)...?
1. Chronic obstructive or interstitial lung diseases
2. Antecedent congenital or acquired heart disease (mitral stenosis)
3. Recurrent thromboemboli
4. Autoimmune disorders (*systemic sclerosis*)
328. What is primary, or idiopathic, pulmonary hypertension?
Uncommonly, pulmonary hypertension is encountered in pts in whom all known causes of increased pulmonary pressure are excluded.

This condition is most commonly sporadic; only 6% of pts have the familial form with autosomal-dominant mode of inheritance. Within these families, there is incomplete penetrance, and only 10% to 20% of the family members actually develop overt disease.
329. What are the genetic causes of primary pulmonary hypertension?
Studies have revealed that primary pulmonary hypertension is caused by mutations in the bone morphogenetic protein receptor type 2 (BMPR2) signaling pathway.

BMPR2 is a cell-surface protein which binds a variety of cytokines, including TFG-beta, BMP, activin, and inhibin.
330. So, how does a mutation in BMPR2 causes primary pulmonary hypertension?
In vascular smooth muscle cells, BMPR2 signaling causes inhibition of proliferation and favors apoptosis.

Thus, in the absence of such signaling, smooth muscle proliferation may be expected.

*Inactivating germ line mutations in the BMPR2 gene are found in 50% of the familial (primary) cases of pulmonary hypertension and 26% of sporadic cases.
331. What is the pathogenesis of secondary forms of pulmonary hypertension?
In secondary forms of pulmonary hypertension, endothelial cell dysfunction is produced by the process that initiates the disorder, such as the increased shear and mechanical injury associated with left-to-right shunts or the biochemical injury produced by fibrin in thromboembolism.

Deceased elaboration of prostacyclin, decreased production of NO, and increased release of endothelin all promote pulmonary vasoconstriction. Also, decreased elaboration of prostacyclin and NO promotes platelet adhesion and activation.
332. What plants or medicines can cause pulmonary hypertension?
Ingestion of the leguminous plant Crotalaria spectabilis, which is indigenous to the tropics and used medicinally in bush tea, the appetite depressant agent, aminorex, adulterated olive oil, and Fenfen obesity drug.

It has been suggested that such substances might act thru effect on serotonin transporter expression or activity.
333. What is the morphology of the vascular lesions in pulmonary hypertension?
The presence of many organizing or recanalized thrombi favors recurrent PE as the cause, and the coexistence of diffuse pulmonary fibrosis, or severe emphysema and chronic bronchitis, points to chronic hypoxia as the initiating event.

The vessel changes can involve the entire arterial tree. In most severe cases, atheromatous deposits form in the pulmonary artery and its major branches, resembling systemic atherosclerosis.

The arterioles and small arteries are most prominently affect, with striking increases in the muscular thickness of the media (medial hypertrophy) and intimal fibrosis. These changes are present in all forms of pulmonary hypertension but are best developed int he primary form.
334. What is plexogenic pulmonary arteriopathy?
One extreme present most prominently in primary pulmonary hypertesnsion ro congenital heart disease with left-to-right shunts is plexogenic pulmonary arteriopathy.

The characteristic features include tufts of capillary formations is present, producing a network, or web, that spans the lumens of dilated thin-walled, small arteries.
335. What is the clinical course of pulmonary hypertension?
Primary pulmonary hypertension is uncommon, typically occurring in women 20-40 years old.

Clinical signs and symptoms of both primary and secondary forms become evdient only with advanced arterial disease. In the course of time, the features generally progresses to severe respiratory insufficiency, cor pulmonale, and death over several years, often superimposed with thromboembolissm and pneumonia.

Therapies include vasodilators, antithrombotic medications, and occasionally lung transplantation.
336. What are the 3 diffuse pulmonary hemorrhage syndromes?
1. Goodpasture syndrome
2. Idiopathic pulmonary hemosiderosis
3. Vasculitis-associated hemorrhage, which is found in conditions such as hypersensitivity angiits, Wegener granulomatosis, and SLE
337. What is Goodpasture syndrome?
Goodpasture syndrome is an uncommon autoimmune disease characterized by the presence of circulating autoantibodies targeted against the noncollageneous domain of the α-3 chain of collagen IV.

The antibodies initiate an inflammatory destruction of the basement membrane in the kidney glomeruli and lung alveoli, giving rise to proliferative, usually RPGN and a necrotizing hemorrhagic interstitial pneumonitis.

Most cases occur in the teens or twenties, and in contrast to many other autoimmune diseases, there is a preponderance among men, and men that smoke.
338. What is the morphology of Goodpasture syndrome?
In the classic case, the lungs are heavy, with areas of red-brown consolidation. Histologically, there is focal necrosis of alveolar walls associated with intra-alveolar hemorrhages. Often, the alveoli contain hemosiderin-laden macrophages.

In later stages there may be fibrous thickening of the septae, hypertrophy of type II pneumoncytes, and organization of blood in alveolar spaces.

The immunofluorescence studies reveal linear deposits of Ig's along the basement membranes.
339. What are the clinical features of Goodpasture syndrome?
Most cases begin clinically with respiratory symptoms, principally hemoptysis, and radiographic evidence of focal pulmonary consolidations. Soon, manifestations of glomerulonephritis appear, leading to rapidly progressive renal failure.

The common cause of death is uremia. Plasma exchange is thought to be beneficial by removing circulating antibasement membrane antibodies as well as chemical mediators of immunologic injury.
340. What is idiopathic pulmonary hemosiderosis?
It is a rare disorder characterized by intermittent, diffuse alveolar hemorrhage. It usually presents with an insidious onset of productive cough, hemotysis, anemia, and weight loss associated with diffuse pulmonary infiltrations similar to Goodpasture syndrome. Most cases occur in children, although the disease occurs in adults as well.
341. What is the morphology of idiopathic pulmonary hemosiderosis?
The lungs are moderately increased in weight, with areas of consolidation that are usually red-brown to red.

*The cardinal histologic features are hemorrhage into the alveolar spaces, and hemosiderosis, both within the alveolar septa and in macrophages lying free within the pulmonary alveoli.

There may also be hyperplasia of type II pneumocytes and varying degrees of interstitial fibrosis.
342. What are the important features in the lungs in Wegener granulomatosis?
The diagnostically important features are capillaritis and scattered, poorly formed granulomas (unlike those of sarcoidosis, which are rounded and well-defined).