• 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

1/339

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;

339 Cards in this Set

  • Front
  • Back
1. How are carbs formed in our diet?
Almost all the carbs of the diet are either large polysaccharides or disaccharides, which are combos of monosaccharides bound to one another by condensation.

In other words, a hydrogen ion has been removed from one of the monosaccharides, and a hdroxyl ion has been removed from the next one.

The two monosaccharides then combine w/each other at these sites of removal, and the hydrogen and hydroxyl ions combine to form water.
2. What happens when carbs are digested?
When carbs are digested, the process is reversed, and the carbs are converted into monosaccharides.

Specific enzymes in the digestive juices of the GI tract return the hydrogen and hydroxyl ions from water to the polysaccharides, and thereby separate the monosaccharides from each other.

This process is called hydrolysis.
3. What is the hydrolysis of fats?
Digestion of TAGS: the fat digesting enzymes return three molecules of water to the TAG molecule and thereby split the fatty acid molecules away from the glycerol.
4. What is the hydolysis of proteins?
The proteolytic enzymes return hydrogen and hydroxyl ions from water molecules to the protein molecules to split them into their constituent AAs.
5. What are the three major sources of carbs in the diet?
1. Sucrose
2. Lactose
3. Starches.

The diet also contain a large amt of cellulose, however; no enzymes are capable of hydrolyzing cellulose in the human GI tract
6. What happens when food is chewed?
When food is chewed, it is mixed w/saliva, which contains ptyalin (an α-amylase) secreted mainly by the parotid glands.

This enzyme hydrolyzes starch into the disaccharide maltose and other small polymers of glucose that contain 3-9 glucose molecules.

However, the food remains in the mouth for only a short time, so that probably not more than 5% of the starches will become hydrolyzed.
7. Can starch digestion occur elsewhere?
Starch digestion sometimes continues in the body and fundus of the sotmach for as long as 1 hour before the food becomes mixed w/the stomach secretions.

Then activity of the salivary amylase is blocked by acid of the gastric secretions b/c the amylase is nonactive when the pH falls below 4.0.

However, as much as 30-40% of the starches will have been hydrolyzed mainly to form maltose in the stomach.
8. What is pancreatic amylase?
Pancreatic secretion, like saliva, contains a large amt of α-amylase that is almost identical to the enzyme in saliva but is more powerful.

Therefore, w/in 15-30 minutes after the chyme empties from the stomach into the duodenum and mixes w/pancreatic juice, virtually all the carbs will have become digested.

In general, the carbs are almost totally converted into maltose and/or very small glucose polymers before passing beyond the duodenum or upper jejunum.
9. What four enzymes in the enterocytes lining the walls of the small intestine hydrolyze disaccharides and small glucose polymers?
1. Lactase
2. Sucrase
3. Maltase
4. α-dextrinase

These enzymes are capable of splitting the disaccharides lactose, sucrose, and maltose, plus other small glucose polymers, into their constituent monosaccharides.

These enzymes are located in the enterocytes covering the intestinal microvilli brush border.
10. What happens to lactose, fructose, maltose, and other small glucose polymers?
Lactose splits into a molecule of galactose and a molecule of glucose.

Sucrose splits into fructose and glucose

Maltose and other small glucose polymers all split into multiple molecules of glucose.

Thus, the final products of carb digestion are all monosaccharides. They are all water soluble and are absorbed immediately into the portal blood.
11. What initiates protein digestion in the stomach?

What is one important feature of this enzyme?
Pepsin. This enzyme requires the acidic pH of the gastric juices to become active.

One of the important features of pepsin digestion is its ability to digest the protein collagen. Collagen is a major part of the intercellular connective tissue of meats; therefore, for the digestive enzymes to penetrate meats and digest other meat proteins, it is first necessary to digest the collagen fibers.
12. What digests proteins in the GI tract?
Pepsin usually provides 10-20% of the total protein digestion in the stomach.

Most protein digestion occurs in the upper small intestine, in the duodenum and jejunum, under the influence of proteolytic enzymes from pancreatic secretion.

The pancreatic secretions contain trypsin, chymotrypsin, carboxypolypeptidase, and proelastase.
13. What do each of these pancreatic enzymes do?
1. Both trypsin and chymotrypsin split protein molecules into small polypeptides
2. Carboxypolypeptidase then cleaves individual AAs from the carboxyl ends of the polypeptides
3. Proelastase, in turn, is converted into elastase, which then digests elastase fibers that partially hold meat together
14. What is the last digestive stage of the proteins in the intestinal lumen?
The last stage is achieved by the enterocytes that line the villi of the small intestine, mainly in the duodenum and jejunum.

These cell have a brush border that has microvilli to increase surface area.

In the membrane of each of these microvilli are multiple peptidases that protrude through the membranes to the exterior where they come into contact w/the intestinal fluids.
15. What are the two types of peptidase enzymes that are important?

Why?
1. Aminopolypeptidase
2. Several dipeptidases

These enzymes succeed in splitting the remaining larger polypeptides into tri- and dipeptides and a few into AAs. Both the AAs plus the di- and tripeptides are easily transported thru the microvillar membrane to the interior of the enterocyte.
16. What happens inside the cytosol of the enterocyte?
There are multiple other peptidases that are specific for the remaining types of linkages between AAs.

Virtally all the last di- and tripeptides are digested to the final stage to form single AAs; these then pass on thru to the other side of the enterocytes and then into the blood.
17. Does fat digestion take place in the stomach?
A small amt of TAGs are digested in the stomach by lingual lipase that is secreted by lingual glands in the mouth and swallowed w/the saliva.

This amt of digestion is less than 10% and is generally unimportant. Instead, all fat digestion occurs in the small intestine.
18. What is the first step of fat digestion?
Phsyically to break the fat globules into very small sizes so that the water-soluble digestive enzymes can act.

This is called emulsification, and it begins by agitation in the stomach to mix the fat w/the products of stomach digestion.
19. Where does most of the emulsification occur?
In the duodenum under the influence of bile,.

Bile contains a large quantity of bile salts as well as the phospholipid lecithin. The polar parts of the bile salts and lecithin molecules are highly soluble in water, whereas most of the remaining portions are highly soluble in fat.

Therefore, these molecules interact w/the fat to greatly decrease the interfacial tension of the fat and make it soluble as well.
20. What is a major function of the bile salts and lecithin?
To make the fat globules readily fragmentable by agitation w/the water in the small bowel. This action is the same as detergents that are used in the wash.

Each time the diameters of the fat globules are significantly decreased as a result of agitation, the total surface area of the fat increases manyfold.

The lipase enzymes are water-soluble compounds and can attack the fat globules only on their surfaces. Thus, this is why this emulsification process is important.
21. What does pancreatic lipase do?

What is enteric lipase?
The most important enzyme for digestion of the TAGs is pancreatic lipase.

Present in pancreatic juice, it can digestion within 1 minute all TAGs that it can reach.

In addition, the enterocytes of the small intestine contain still more lipase, known as enteric lipase, but is is usually not needed.
22. What are the end products of fat digestion?
Free fatty acids and 2-monoglycerides
23. What is another role of bile salts?
TO remove the monoglycerides and free fatty acids from the vicinity of digesting fat globules almost as rapidly as these end products are formed.

Bile salts in water form micelles. The developing sterol nucleus emcompasses the fat digestate, forming a small fat globule in the middle of a resulting micelle, with polar groups of bile salts projecting outward to cover the surface of the micelle.

This allows the entire micelle to dissolve in the water of the digestive fluids and to remain in stable solution until the fat is absorbed into the blood.

Thus, the bile salt micelles also act as a transport medium to carry the monoglycerides and free fatty acids (ferrying)
24. How does digestion of cholesterol esters and phospholipids occur...?
Both cholesterol esters and the phospholipids are hydrolyzed by two other lipases in the pancreatic secretion that free the fatty acids: the enzyme cholesterol ester hydrolase, and phospholipase A2.
25. Does the stomach have a good absorptive area?
No, the stomach is poor in absorption b/c it lacks the typical villus type of absorptive membrane, and also b/c the junctions between the epithelial cells are tight junctions.

Only a few highly lipid-soluble substances, such as alcohol and some drugs like aspirin, can be absorbed in small quantities.
26. What are the valvulae conniventes (or folds of Kerckring)?
These folds are in the small intestinal mucosa villi, and they increase the surface area of the absorptive mucosa about 3x.

These folds extend circularly most of the way around the intestine and are especially well developed in the duodenum and jejunum.
27. Where are the villi located in the GI tract?
On the epithelial surface of the small intestine all the way down to the ileocecal valve.

These project about 1 mm from the surface of the mucosa. They lie so close to one another that they touch in most areas, but their distribution is less profuse in the distal small intestine.

The presence of villi on the mucosal surface enhances the total absorptive areas another 10x.
28. What is the brush border?
Each intestinal epithelial cell on each villus is characterized by a brush border.

This increases the surface area exposed to the intestinal materials at least another 20x.
29. What is three important features of the villus?
1. The advantageous arrangement of the vascular system for absorption of fluid and dissovled material into the portal blood

2. The arrangement of the central lacteal lymph vessel for absorption into the lymph

3. Extending from the epithelial cell body into each microvillus of the brush border are multiple actin filaments that contract rhythmically to cause continual movement of the microvilli, keeping them constantly exposed to new quantities of intestinal fluid.
30. What is greater - absorption or absorptive capacity of the small intestine?
Absorptive capacity.
31. How does absorption of water occur in the small intestine?
Water is transported thru the intestinal membrane entirely by diffusion. It obeys osmosis.

Thus, when the chyme is dilute enough, water is absorbed thru the intestinal mucosa into the blood and vice versa.

Within minutes, sufficient water usually will be transferred by osmosis to make the chyme isosmotic w/the plasma.
32. How does sodium get absorbed?
The motive power for doium absorption is provided by active transport of sodium form inside the epithelial cells thru the basal and side walls of these cells into paracellular spaces.

This active transport obeys the usual laws of active transport; it requires energy, and the energy process is catalyzed by ATPase in the cell membrane.

Part of the sodium is absorbed along w/chloride ions; in fact, the negatively charged chloride ions are mainly passively "dragged" by the positive electrical charges of the sodium ions.
33. How is the sodium concentration in the cell of the enterocytes affected?
It gets reduced to 50 mEq/L

B/c the sodium concentration in the chyme is normally isosmotic with that of plasma (142 mEq/L), sodium moves down this steep electrochemical gradient from the chyme thru the brush border of the epithelial cell into the epithelial cell cytoplasm. This provides still more sodium ions to be transported by the epithelial cells into the paracellular spaces.
34. What is the next step in the transport process of sodium?
Osmosis of water into the paracellular spaces. This occurs b/c a large osmotic gradient has been created by the elevated concentration of ions in the paracellular space.

Much of this osmosis occurs thru the tight junctions between the apical borders of the epithelial cell, but much also occurs thru the cells themselves. Also, osmotic movement of water creates flow of fluid into and thru the paracellular spaces, and finally, into the circulating blood of the villus.
35. How does aldosterone affect sodium absorption?

How is this important in the colon?
When a person becomes dehydrated, excess aldosterone is secreted by the adrenal glands. This increases all aspects of sodium absorption by the intestinal epithelium. And the increased sodium absorption in turn causes secondary increases in absorption of chloride ions, water, and some other substances.

This effect is especially important in the colon b/c it allows virtually no loss of NaCl in the feces and also little water loss.
36. Where does absorption of chloride ions occur?
In the duodenum and jejunum.

In the upper part of the small intestine, chloride ion absorption is rapid and occurs mainly by diffusion. Then chloride ions move along the electrical gradient caused by sodium ions to follow the sodium ions.
37. Where does absorption of bicarbonate ions occur?
Also in the duodenum and jejunum.

Large quantities of bicarb ions must be reabsorbed from the upper small intestine b/c large amts of bicarb have been secreted by the the pancreas and bile.
38. How does bicarbonate ion absorption occur?
Indirectly. When sodium ions are absorbed, moderate amts of hydrogen ions are secreted into the lumen of the gut in exchange for some of the sodium. These hydrogen ions in turn combine with the bicarb ions to form carbonic acid which then dissociates to form water and carbon dioxide.

The water remains as part of the chyme in the intestines, but the CO2 is readily absorbed into the blood and then expired thru the lungs.
39. Where does the bicarbonate-chloride ion exchange take place?
In the ileum and large intestine.

The epithelial cells in the villi of the ileum as well as on all surfaces of the large intestine have a special capability of secreting bicarbonate ions in exchange for absorption of choloride ions.

This is important b/c it provides alkaline bicarbonate ions that neutralize acid products formed by bacteria in the large intestine.
40. Diarrhea and secretion of ions
Deep in the spaces between the intestinal folds are immature epithelial cells that continually divide to form new epithelial cells. These in turn spread outward over the luminal surfaces of the intestine.

While still in the deep folds, the epithelial cells secrete NaCl and water into the intestinal lumen. This secretion in turn is reabsorbed by the older epithelial cells outside the folds, thus providing flow of water for absorbing intestinal digestates.
41. What is cholera?
Extreme diarrheal secretion is initiated by entry of a subunit of cholera toxin into the epithelial cells.

This stimulates the formation of excess cAMP, which opens tremendous numbers of chloride channels, allowing chloride ions to flow rapidly from inside the cell into the intestinal crypts.

In turn, this is believed to activate a sodium pump that pumps sodium ions into the crypts to go along with the chloride ions. Finally, all this extra NaCl causes extreme osmosis of water form the blood, thus providing rapid flow of fluid along with the salt.

All this excess fluid washes away most of the bacteria and is of value to combating the disease; however, too much of a good thing can be lethal
42. Where are calcium ions absorbed?
In the duodenum, and the most of calcium ion absorbed is very controlled to supply exactly the daily need of the body for calcium.

This is controlled by PTH and vitamin D.

PTH activates vitamin D, and the activated vitamin D in turn greatly enhances calcium absorption.
43. Where are iron ions absorbed?
Iron ions are also actively absorbed from the small intestine.
44. Where are potassium, magnesium, phosphate, and other ions absorbed?
These ions are actively absorbed thru the intestinal mucosa.

In general, the monovalent ions are absorbed w/ease and in great quantities. Conversely, bivalent ions are normally absorbed in only small amts; for example, max absorption of calcium ions is only 1/50th as great as the normal absorption of sodium ions.
45. What is the most abundant of the absorbed monosaccharides?

Why?
Glucose, usually accounting for more than 80% of the carb calories absorbed.

The reason for this is that glucose is the final digestion product of our starches in the diet, which is our most abundant carb.

The remaining 20% of absorbed monosaccharides are composed almost entirely of galactose and fructose.
46. How is glucose transported?
In the absence of sodium transport thru the intestinal membrane, virtually no glucose can be absorbed. The reason is that glucose absorption occurs in a co-transport mode w/active transport of sodium.
47. What are the two stages in the transport of sodium thru the intestinal membrane?
1. Active transport of sodium ions thru the basolateral membranes of the intestinal epithelial cells into the blood, thereby depleting sodium inside the epithelial cells.

2. Decrease of sodium inside the cells causes sodium from the intestinal lumen to move thru the brush border of the epithelial cells to the cell interiors by a process of facilitated diffusion.

Thus, a sodium ion combines w/a transport protein, but the transport protein will not transport the sodium to the interior of the cell until the protein itself combines with glucose.

Fortunately intestinal glucose also combines with the same transport protein, and then both the sodium ion and glucose molecule are transported to the interior of the cell.
48. Glucose-sodium transport summary
Thus, the low concentration of sodium inside the cell literally drags sodium to the interior of the cell and along with it the glucose at the same time.

Once inside the epithelial cell, other transport proteins and enzymes cause facilitated diffusion of the glucose thru the cell's basolateral membrane into the paracellular space and from there into the blood.

It is the initial active transport of sodium thru the basolateral membranes of the intestinal epithelial cells that provides the eventual motive force for moving glucose also thru the membranes.
49. How are the other monosaccharides absorbed?
Galactose is transported by almost exactly the same emchanism as glucose.

Conversely, fructose transport does not occur by the sodium co-transport mechanism. Instead, fructose is transported by facilitated diffusion all the way thru the intestinal epithelium but not coupled w/sodium transport.

Much of the fructose, on entering the cell, becomes phosphorylated, then converted to glucose, and finally transported in the form of glucose the rest of the way into the blood. B/c fructose is not co-transported w/sodium, its overall rate of transport is only 1/2 that of glucose or galactose.
50. How are proteins absorbed?
They are absorbed thru the luminal membranes of the intestinal epithelial cells in the form of di- and tripeptides and a few AAs. The energy for most of this transprot process is supplied by a sodium co-transprot mechanism in the same way that sodium co-transport of glucose occurs.

That is, most peptide or AA molecules bind int he cells microvillus membrane w/a specific transport protein that requires sodium binding before transport can occur.

After binding, the sodium ion then moves down the gradient to the interior of the cell and pulls the AA or peptide along with it. This is called co-transport.
51. How are fats absorbed?
After entering the epithelial cell, the fatty acids and monoglycerides are taken up by the cells smooth ER; here, they are mainly used to form new TAGs athat are subsequently released in the form of chylomicrons thru the base of the epithelial cell, to flow upward thru the thoracic duct and empty into the circulating blood.
52. Can some fats be absorbed directly into the protal blood?
Small quantities of short and medium chain fatty acids, such as those from butterfat, are absorbed directly into the portal blood rather than being converted into TAGs first.

The cause of this difference between short and long chain fatty acid absorption is that the short chain fatty acids are more water soluble and mostly are not reconverted into TAGs by the ER.

This allows direct diffusion of these short chain fatty acids from the intestinal epithelial cells directly into the capillary blood of the intestinal villi.
53. Where does absorption occur in the large intestine?
Most of the absorption occurs in the proximal half of the colon, whereas the distal half functions principally for feces storage.
54. How are electrolytes and water absorbed and secreted in the large intestine?
The tight junctions between the epithelial cells of the large intestinal epithelium are much tighter than those of the small intestine. This prevents significant amts of back-diffusion of ions thru these junctions, thus allowing the large intestinal mucosa to absorb sodium ions far more completely - that is, against a much higher concentration gradient than can occur in the small intestine.

In addition, the mucosa of the large intestine secretes bicarb ions while it simultaneously absorbs an equal number of chloride ions in an exchange transport process.

Absorption of sodium and chloride ions creases an osmotic gradient across the large intestinal mucosa, which in turn causes absorption of water.
55. What is the max absorption capacity of the large intestine?
The large intestine can absorb a max of 5-8 L of fluid and electrolytes each day.

When the total quantity entering the large intestine thru the ileocecal valve or by way of large intestine secretion exceeds this amt, the excess appears in the feces as diarrhea.
56. What bacterial are present in the colon?
Numerous bacteria, especially colon bacilli, are present even normally in the absorbing colon. They are capable of digesting small amts of cellulose.

Other important substances formed as a result of bacterial activity are vitamin K, B12, thiamine, riboflavin, and various gases that contribute to gas in the colon.
57. What is the composition of feces?
They are normally 3/4ths water and 1/4th solid matter that itself is composed of about 30% dead bacteria, 10-20% fat, 10-20% inorganic matter, 2-3% protein, and 30% undigested roughage from the food and dried constituents of digestive juices.
58. What gives shit the brown color?

Bad smell?
Color caused by stercobilin and urobilin, derivatives of bilirubin.

The odor is caused by products of baterial action; they include indole, skatole, mercaptans, and hydrogen sulfide.
59. What can cause paralysis of the swallowing mechanism?
Damage to CN V, IX, or X can cause paralysis of significant portions of the swallowing mechanism.

Also, a few diseases, such as polio and encephalitis, can prevent normal swallowing by damaging the swallowing center in the brain.

Finally, paralysis of the swallowing muscles, as occurs in muscular dystrophy or in failure of the NMJ transmission in myasthenia gravis or botulism, can also prevent normal swallowing.
60. When the swallowing mechanism is partially or totally paralyzed, what abnormalities can result?
1. Complete abrogation of the swallowing act so that swallowing cannot occur
2. Failure of the glottis to close so that food passes into the lungs instead of the esophagus
3. Failure of the soft palate and uvula to close the posterior nares so that food refluxes into the nose during swallowing.
61. How is the swallowing mechanism affected during deep anesthesia?
Often, while on the operating table, the patient vomits large quantities of materials from the stomach into the pharynx; then, instead of swallowing the materials again, they simply suck them into the trachea b/c the anesthetic has blocked the reflex mechanism of swallowing. As a result, such patients occasionally choke to death on their own vomit.
62. What is gastritis?
Inflammation of the gastric mucosa.

Mild to moderate chronic gastritis is exceedingly common int he population as a whole, especially in the middle to later years of adult life.

The inflammation may be only superficial, or it can penetrate deeply into the gastric mucosa, in many long-standing cases causing almost complete atrophy of the gastric mucosa. In a few cases, it can be acute and severe, w/ulcerative excoriation of the stomach mucosa by the stomach's own peptic secretions.
63. What can cause gastritis?
Caused by chronic bacterial infection of the gastric mucosa. This often can be treated w/antibacterial therapy.

In addition, certain ingested irritant substances can be damaging to the protective gastric mucosal barrier - often leading to severe acute or chronic gastritis.

Two of the most common of these substances are excesses of EtOH or aspirin.
64. What are two reasons for the low level of absorption in the stomach?
Caused by two specific features of the gastric mucosa:

1. it is lined w/highly resistant mucous cell that secrete a viscid and adherent mucus

2. it has tight junctions between the adjacent epithelial cells

These two together plus other impediments to gastric absorption are called the gastric barrier.
65. What is gastric atrophy?
In many people w/chronic gastritis, the mucosa gradually becomes more and more atrophic until little or no gastric gland digestive secretion remains.

It is also believed that some people develop autoimmunity against the gastric mucosa, this also leading eventually to gastric atrophy.

Loss of the stomach secretions in gastric atrophy leads to achlorhydria and occasionally to pernicious anemia.
66. What is achlorhydria?
Achlorhydria means simply that the stomach fails to secrete HCl.

It is diagnosed when the pH of the gastric secretions fails to decrease below 6.5 after max stimulation.
67. What is hypochlorhydria?
Hypochlorhydria means diminished acid secretion.

When acid is not secreted, pepsin is also usually not secreted; even when it is, the lack of acid prevents it from functioning b/c pepsin requires an acidic medium for activity.
68. Why is pernicious anemia a freq concern in gastric atrophy?
Pernicious anemia is a common feature of gastric atrophy and achlorhydria. Normal gastric secretions contain a glycoprotein called intrinsic factor, secreted by the same parietal cells that secrete HCl.

Intrinsic factor must be present for adequate absorption of vitamin B12 from the ileum. That is, intrinsic factor combines w/B12 in the stomach and protects it from being digested and destroyed as it passes into the small intestine. Then, when the intrinsic factor-vitamin B12 complex reaches the terminal ileum, the intrinsic factor binds w/receptors on the ileal epithelial surface. This in turn makes it possible for the vitamin B12 to be absorbed.

In the absence of intrinsic factor, only 1/50th of the B12 is absorbed. The result is pernicious anemia.
69. What is a peptic ulcer?

Where are they most commonly formed?
A peptic ulcer is an excoriated area of stomach or intestinal mucosa caused principally by the digestive action of gastric juice or upper small intestinal secretions.

The most freq site is within a few cm of the pylorus. In addition, peptic ulcers freq occur along the lesser curvature of the antral end of the stomach, or more rarely, in the lower end of the esophagus where stomach juices freq reflux.

A type of peptic ulcer called a marginal ulcer also often occurs wherever a surgical opening such as a gastrojejunostomy has been made between the stomach and the jejunum of the small intestine.
70. What is the basic cause of a peptic ulceration?
The usual cause is an imbalance between the rate of secreiton of gastric juice and the degree of protection afforded by (1) the gastroduodenal mucosal barrier and (2) the neutralization of the gastric acid by duodenal juices.

In other words, a peptic ulcer is caused by (1) excess secretion of acid and pepsin by the gastric mucosa or (2) diminished ability to protect against the digestive properties of the gastric juices.
71. How is the stomach and upper duodenum protected by a mucosal barrier?
All areas normally exposed to gastric juice are well supplied with mucous glands, beginning with compound mucous glands in the lower esophagus plus the mucous cell coating of the stomach mucosa, the mucous neck cells of the gastric glands, the deep pyloric glands that secrete mainly mucus, and, finally, the glands of Brunner of the upper duodenum, which secrete a highly alkaline mucus.
72. How is the duodenum protected?
Via the alkalinity of the small intestinal secretions. Especially important is pancreatic secretion that neutralizes the HCl, thus also inactivating pepsin and preventing digestion of the mucosa.

In addition, large amts of bicarb ions are provided int the secretions of the large Brunner's glands in the first few cm's of the duodenal wall, and in bile coming form the liver.
73. What two feedback control mechanisms ensure that the neutralization of gastric juices is complete?
1. When excess acid enters the duodenum, it reflexly inhibits gastric secretion and peristalsis in the stomach, both by nervous reflexes and by hormonal feedback from the duodenum, thereby decreasing the rate of gastric emptying.

2. The presence of acid int he small intestine liberates secretin from the intestinal mucosa, which then passes by way of the blood to the pancreas to promote rapid secretion of pancreatic juice. This juice also contain s a high concentration of sodium bicarb, thus making still more sodium bicarb available for neutralization of the acid.
74. What is H. pylori?
Many peptic ulcer patients have chronic infections of the terminal portions of the gastric mucosa and initial portions of the duodenal mucosa most often caused by infection of H. pylori.

Often the bacterium is capable of penetrating the mucosal barrier both by virtue of its physical capability to burrow through the barrier and by releasing bacterial digestive enzymes that liquefy the barrier. As a result, the strong acidic digestive juices of the stomach secretions can then penetrate into the underlying epithelium and literally digest the GI wall.
75. What are other causes of ulceration?
1. Smoking, presumably b/c of increased nervous stimulation of the stomach secretory glands

2. EtOH, b/c it tends to break down the mucosal barrier

3. Aspirin and other NSAIDs that also have a strong propensity for breaking down this barrier.
76. What are two agents that treat peptic ulceration?
1. Use of antibiotics along w/other agents to kill infectious bacteria

2. Administration of an acid-suppressant drug, especially ranitidine, an antihistamine that blocks the stimulatory effect of histamine on gastric gland histamine receptors, thus reducing gastric acid secretion by 70-80%
77. Lack of pancreatic secretion occurs in what three situations?
1. In pancreatitis
2. When pancreatic duct is blocked by a gallstone at the papilla of Vater
3. After the head of the pancreas has been removed b/c of malignancy
78. Loss of pancreatic juice means what...?
Loss of trypsin, chymotrypsin, carboxypolypeptidase, pancreatic amylase, pancreatic lipase, and a few other enzymes. As a result, a large portion of the ingested food cannot be used for nutrition, can copious, fatty feces are excreted.
79. What is the most common cause of pancreatitis?

Second most common?
1. Drinking excess alcohol
2. Blockage of the papilla of Vater by a gallstone

These two together account for more than 90% of all cases.
80. What is nontropical sprue?
AKA celiac disease, results from the toxic effects of gluten present in certain types of grains, especially wheat and rye.

Only some people are susceptible to this effect, but in those who are susceptible, gluten has a direct destructive effect on intestinal enterocytes. In milder forms of the disease, only the microvilli of the absorbing enterocytes on the villi are destroyed, thus decreasing the absorptive surface area by 2x.

In the more severe forms, the villi themselves become blunted or disappear altogether, thus still further reducing the absorptive are of the gut.
81. What is tropical sprue?
A different type of sprue called tropical sprue frequently occurs in the tropics and can often be treated with antibacterial agents.

Even though no specific bacterium has been implicated as the cause, it is believed that this variety of sprue is usually caused by inflammation of the intestinal mucosa resulting from unidentified infectious agents.
82. In the early stages of sprue, what digestive product absorption is most impaired?
In the early stages of sprue, intestinal absorption of fat is more impaired than absorption of other digestive products.

The fat that appears in the stool is almost entirely in the form of salts of fatty acids rather than undigested fat, demonstrating that the problem is one of absorption, not of digestion. This is called steatorrhea.
83. What are four consequences of malabsorption in sprue?
1. Severe nutritional deficiency, often developing wasting of the body
2. Osteomalacia
3. Inadequate blood coagulation caused by lack of vitamin K
4. Macrocytic anemia of the pernicious anemia type, owing to diminished vitamin B12 and folic acid absorption.
84. What can cause constipation?
Any pathology of the intestines that obstructs movement of intestinal contents, such as tumors, adhesions that constrict the intestines, or ulcers, can cause constipation.

A freq functional cause of constipation is irregular bowel habits that have developed thru a lifetime of inhibition of the normal defecation reflexes.

Can also result from spasm of a small segment of the sigmoid colon.
85. What is megacolon?

What is a freq cause?
Occasionally, constipation is so severe that bowel movements occur only once every several days or sometimes only once a week. This allows tremendous quantities of fecal matter to accumulate in the colon, causing the colon sometimes to distend to a diameter of 3 to 4 inches. The condition is called megacolon, or Hirschsprung’s disease.

A frequent cause of megacolon is lack of or deficiency of ganglion cells in the myenteric plexus in a segment of the sigmoid colon. As a consequence, neither defecation reflexes nor strong peristaltic motility can occur in this area of the large intestine. The sigmoid itself becomes small and almost spastic while feces accumulate proximal to this area, causing megacolon in the ascending, transverse, and descending colons.
86. What is enteritis?
Means inflammation usually caused either by a virus or by bacteria in the intestinal tract. In usual infectious diarrhea, the infection is most extensive in the large intestine and the distal end of the ileum.

Everywhere the infection is present, the mucosa becomes extensively irritated, and its rate of secretion becomes greatly enhanced. In addition, motility of the intestinal wall usually increases manyfold. As a result, large quantities of fluid are made available for washing the infectious agent toward the anus, and at the same time strong propulsive movements propel this fluid forward.
87. What is psychogenic diarrhea?
Periods of nervous tension can be accompanied by diarrhea.

This is caused by excessive stimulation of the parasympathetic nervous system, which greatly excites both (1) motility and (2) excess secretion of mucus in the distal colon.
88. What is ulcerative colitis?
Ulcerative colitis is a disease in which extensive areas of the walls of the large intestine become inflamed and ulcerated.

The motility of the ulcerated colon is often so great that mass movements occur much of the day rather than for the usual 10 to 30 minutes.

Also, the colon’s secretions are greatly enhanced. As a result, the patient has repeated diarrheal bowel movements.
89. What causes ulcerative colitis?
The cause of ulcerative colitis is unknown. Some clinicians believe that it results from an allergic or immune destructive effect, but it also could result from chronic bacterial infection not yet understood. Whatever the cause, there is a strong hereditary tendency for susceptibility to ulcerative colitis.

Once the condition has progressed very far, the ulcers seldom will heal until an ileostomy is performed to allow the small intestinal contents to drain to the exterior rather than to pass through the colon.

Even then the ulcers sometimes fail to heal, and the only solution might be surgical removal of the entire colon.
90. What reflex initiates defecation?
It is normally initiated by accumulating feces in the rectum, which causes a spinal cord-mediated defecation reflex passing from the rectum to the conus medullaris of the spinal cord and then back to the descending colon, sigmoid, rectum, and anus.
91. What happens to defecation when the spinal cord is injured somewhere between the conus medullaris and the brain?
The voluntary portion of the defecation act is blocked while the basic cord reflex for defecation is still intact.

But, b/c the cord defecation reflex can still occur, a small enema to excite action of this cord reflex, usually given in the morning, can often cause adequate defecation.
92. What is vomiting?
Vomiting is the means by which the upper gastrointestinal tract rids itself of its contents when almost any part of the upper tract becomes excessively irritated, overdistended, or even overexcitable.

Excessive distention or irritation of the duodenum provides an especially strong stimulus for vomiting.
93. What sensory signals initiate vomiting?
They originate mainly from the pharynx, esophagus, stomach, and upper portions of the small intestines.

The nerve impulses are transmitted by both vagal and sympathetic afferent nerve fibers to multiple distributed nuclei in the brain stem that are together are called the "vomiting center".

From here, motor impulses that cause the actual vomiting are transmitted from the vomiting center by way for the 5th, 7th, 9th, 10th, and 12th cranial nerves to the upper GI tract, through vagal and sympathetic nerves to the lower tract, and through spinal nerves to the diaphragm and abdominal muscles.
94. What is antiperistalsis?
The prelude to vomiting.

In the early stages of excessive GI irritation or overdistention, antiperistalsis begins to occur often many minutes before vomiting appears. Antiperistalsis means peristalsis up the digestive tract rather than downward. This may begin as far down in the intestinal tract as the ileum, and the antiperistaltic wave travels backward up the intestine at a rate of 2-3 cm/sec.

This process can push a large share of the lower small intestinal contents all the way back to the duodenum and stomach within 3-5 minutes. Then, as these upper portions of the GI tract, especially the duodenum, become overly distended, this distention becomes the exciting factor that initiates the actual vomiting act.
95. At the onset of vomiting, what occurs?
Strong intrinsic contractions occur in both the duodenum and the stomach, along w/partial relaxation of the esophageal-stomach sphincter, thus allowing vomitus to being moving form the stomach into the esophagus.

From here, a specific vomiting act involving the abdominal muscles takes over and expels the vomitus to the exterior.
96. What four things occur during the vomiting act?
1. A deep breath
2. Raising of the hyoid bone and larynx to pull the upper esophageal sphincter open
3. Closing of the glottis to prevent vomitus flow into the lungs
4. Lifting of the soft palate to close the posterior nares

Next comes a strong downward contraction of the diaphragm along w/simultaneous contraction of all the abdominal wall muscles. This squeezes the stomach between the diaphragm and the abdominal muscles, building the intragastric pressure to a high level. Finally, the lower esophageal sphincter relaxes completely, allowing expulsion of the gastric contents upward through the esophagus.
97. What is the chemoreceptor trigger zone?
Vomiting can also be caused by nervous signals arising in areas of the brain. This is true for a small area located bilaterally on the floor of the fourth ventricle.

Electrical stimulation of this area can initiate vomiting; but, more important, administration of certain drugs, including apomorphine, morphine, and some digitalis derivatives, can directly stimulate this chemoreceptor trigger zone and initiate vomiting.
98. How can motion sickness cause vomiting?
It is well known that rapidly changing direction or rhythm of motion of the body can cause certain people to vomit.

The mechanism for this is the following: The motion stimulates receptors in the vestibular labyrinth of the inner ear, and from here impulses are transmitted mainly by way of the brain stem vestibular nuclei into the cerebellum, then to the chemoreceptor trigger zone, and finally to the vomiting center to cause vomiting.
99. What is nausea and what three things can cause it?
Nausea is the conscious recognition of subconscious excitation in an area of the medulla closely associated w/or part of the vomiting center.

It can be caused by:
1. Irritative impulses coming from the GI tract
2. Impulses that originate in the lower brain associated w/motion sickness
3. Impulses from the cerebral cortex to initiate vomiting
100. What are four common causes of GI obstructions?
1. Cancer
2. Fibrotic constriction resulting from ulceration or from peritoneal adhesions
3. Spasm of a segment of the gut
4. Paralysis of a segment of the gut
101. What happens if an obstruction occurs at the pylorus?
This results often from fibrotic constriction after peptic ulceration, persistent vomiting of stomach contents occurs.

This depresses bodily nutrition; it also causes excessive loss of hydrogen ions form the stomach and can result in various degrees of whole-body alkalosis.
102. What happens if the obstruction is beyond the stomach?
Antiperistaltic reflux from the small intestine causes intestinal juices to flow backward intot he stomach, and these juices are vomited along w/the stomach secretions.

In this instance, the person loses large amts of water and electrolytes, so that he or she becomes severely dehydrated, but the loss of acid from the stomach and base from the small intestine are about equal so no acid-base balance occurs.
103. What happens if the obstruction is near the distal end of the large intestine?
Feces can accumulate in the colon for a week or more. The patient develops an intense feeling of constipation but at first vomiting is not severe.

After the large intestine has become completely filled and it finally becomes impossible for additional chyme to move from the small intestine into the large intestine, severe vomiting does then occur.

Prolonged obstruction of the large intestine can finally cause rupture of the intestine itself or dehydration and circulatory shock resulting from severe vomiting.
104. Amylose vs. amylopectin
In amylose, the glucosyl residues form a straight chain linked via α-1,4-glycosidic bonds.

In amylopectin, the α-1,4-chains contain branches connected via α-1,6-glycosidic bonds.
105. What is dietary fiber composed of?
Composed principally of plant polysaccharides and a polymer called lignan.
106. Digestion of dietary carbs
In the digestive tract, dietary polysaccharides and disaccharides are converted to monosaccharides by glycosidases, enzymes that hydrolyze the glycosidic bonds between the sugars.

All of these enzymes exhibit some specificity for the sugar, the glycosidic bond (α or β), and the number of saccharide units in the chain.

The monosaccharides formed by glycosidases are transported across the intestinal mucosal cells into the interstitial fluid and subsequently enter the bloodstream.

Undigested carbs enter the colon, where they may be fermented by bacteria.
107. Where does digestion of starch begin?
The digestion of starch (amylopectin and amylose) begins in the mouth, where chewing mixes the food with saliva. The salivary glands secrete salivary α-amylase and other components.
108. What is α-amylase?
α-amylase is an endoglucosidase, which means that it hydrolyzes internal α-1,4-bonds between glucosyl residues at random intervals in the polysaccharide chains.

The shorted polysaccharide chains that are formed are called α-dextrins. Salivary α-amylase is largely inactivated by the acidity of the stomach contents, which contain HCl secreted by the parietal cells.
109. What does pancreatic α-amylase do to starches and glycogen?
It continues to hydrolyze the starches and glycogen, forming the disaccharide maltose, the trisaccharide maltotriose, and oligosaccharides. These oligosaccharides, called limit dextrins, are usually 4-9 glucosyl units long and contain one or more α-1,6-branches. The two glucosyl residues that contain the α-1,6-glycosidic bond eventually become the disaccharide isomaltose, but α-amylase does not cleave these branched oligosaccharides all the way down to isomaltose.
110. Specificity of α-amylase
α-amyalse has no activity toward sugar-containing polymers other than glucose linked by α-1,4-bonds.

α-amylse displays no activity toward the α-1,6-bond at branch points and has little activity for the α-1,4-bond at the nonreducing end of the chain.
111. What disaccharidases are present in the intestinal brush border membrane?
The dietary disaccharides lactose and sucrose, as well as the products of starch digestion, are converted to monosaccharides by glycosidases attached to the membrane in the brush border of absorptive cells.

The different glycosidase activities are found in four glycoproteins; glucoamylase, the sucrase-isomaltase complex, the smaller glycoprotein trehalase, and lactase-glucosylceramidase. These glycosidases are collectively called the small intestinal disaccharidases, although glucoamylase is really an oligosaccharidase.
112. What is glucoamylase?
Glucoamylase and the sucrase-isomaltase complex have similar structures and exhibit a great deal of sequence homogeneity.

A membrane-spanning domain near the N-terminal attaches the protein to the luminal membrane. The long polypeptide chain forms two globular domains, each with a catalytic site.

In glucoamylase, the two catalytic sites have similar activities, w/only small differences in substrate specificity. The protein is heavily glycosylated with oligosaccharides that protect it from digestive proteases.
113. What is the specificity of glucoamylase?
Glucoamylase is an exoglucosidase that is specific for the α-1,4-bonds between glucosyl residues. It begins at the nonreducing end of a polysaccharide or limit dextrin, and sequentially hydrolyzes the bonds to release glucose monosaccharides.

It will digest a limit dextrin down to isomaltose, the glucosyl disaccharide with an α-1,6-branch, which is subsequently hydrolyzed principally by the isomaltase activity in the sucrase-isomaltase complex.
114. What is the sucrase-isomaltase complex?
The structure of the sucrase-isomaltase compelx is very similar to tha tof glucoamylase, and these two proteins have a high degree of sequence homology. Hwoever, after the single polypeptide chain of sucrase-isomaltase is inserted through the membrane and the protein protrudes into the intestinal lumen, an intestinal protease clips it into two separate subunits that remain attached to each other. Each subunit has a catalytic site that differs in substrate specificty from the other through noncovalent interactions.
115. What accounts for all of the intestine's ability to hydrolyze sucrose in addition to maltase activity?
The sucrase-maltase site accounts for approx 100% of the intestine's ability to hydrolyze sucrose in addition to maltase activity; the isomaltase-maltase site accounts for almost all of the intestine's ability to hydrolyze α-1,6-bonds, in addition to maltase activity.

Together, these sites account for approx 80% of the maltase activity of the small intestine. The remainder of the maltase activity is found int he glucoamylase complex.
116. What is trehalase?
Trehalase is only half as long as the other disaccharidases and has only one catalytic site. It hydrolyzes the glycosidic bond in trehalose, a disaccharide composed of two glucosyl units linked by an α-bond between their anomeric carbons.

Trehalose, which is found in insects, algae, etc... is not a major dietary compoennt in the US. However, unwitting consumption of trehalose can cause nausea, vomiting, and other symptoms of severe GI distress if consumed by an individual deficient in the enzyme.
117. What is the β-glycosidase complex (lactase-glucosylceramidase)?
The β-glycosidase complex is another large glycoprotein found in the brush border that has two catalytic sites extending in the lumen of the intestine. However, its primary structure is different, and it is attached to the membrane thru its carboxyl end by a phoshatidylglycan anchor.

The lactase catalytic site hydrolyzes the β-bond connecting glucose and galactose in lactose. The major activity of the other catalytic site in humans is the β-bond between glucose or galactose and ceramide in glycolypids.
118. The production of maltose, maltotriose, and limit dextrins by pancreatic α-amylase occurs where?

Where is sucrase-isomaltase activity highest?

β-glycosidase activity?

Glucoamylase?
Production of those sugars occurs in the duodenum, the most proximal portion of the small intestine.

Sucrase-isomaltase activity is highest in the jejunum, where the enzymes can hydrolyze sucrose and the products of starch digestion

β-glycosidase activity is highest in the jejunum.

Glucoamylase activity increases progressively along the length of the small intestine, and its activity is highest in the ileum.
119. What do the colonic bacteria metabolize?

What major gases are formed?
Colonic bacteria rapidly metabolize the saccharides, forming gases, short-chain fatty acids, and lactate.

The majory short-chain fatty acids formed are acetic acid (2 C's), propionic acid (3 C's), and butyric acid (4 C's).

The short chain fatty acids are absorbed by the colonic mucosal cells and can provide a substantial source of energy for these cells.

The major gases formed are hydrogen gas, carbon dioxide, and methane. These gases are released thru the colon, resulting in flatulence.
120. What is the nonpersistent lactase phenotype?
Lactase activity increases in the human for about 6-8 weeks of gestation and it rises during the late gestational phase (27-32 weeks) thru full term. It remains high for 1 month after birth and then begins to decline.

For most of the world's population, lactase activity decreases to adult levels at approximately 5-7 years of age. Adult levels are less than 10% of those present in infants. These populations have adult hypolactasia and exhibit the lactase nonpersistent phenotype.

This is the normal condition of most of the world's population.
121. What is the persistent lactase phenotype?
In people who are derived mainly from western northern Europeans, and milk-dependent nomadic tribes of Saharan Africa, the levels of lactase remain at, or only slightly below, infant levels throughout adulthood.
122. What intestinal diseases can cause lactase deficiency?
Intestinal diseases that injure the absorptive cells of the intestinal villi diminish lactase activity along the intestine, producing a condition known as secondary lactase deficiency.

Kwashiorkor (protein malnutrition), colitis, gastroenteritis, tropical and nontropical sprue, and excessive alcohol consumption fall into this category.

These disease also affect other disaccharadases, but sucrase, maltase, isomaltase, and glucoamylase activities are usually present at such excessive levels that there are no pathologic effects.

*Lactase is usually the first activity lost and the last to recover.
123. What is one benefit of dietary fiber?
In diverticular disease, in which sacs or pouches may develop in the colon b/c of a weakening of the muscle and submucosal structures. Fiber is thought to soften the stool, thereby reducing pressure on the colonic wall and enhancing expulsion of feces.
124. Why are oats good for our health?

What about pectins?
β-glucan (obtained from oats) has also been shown to reduce cholesterol levels thru a reduction in bile acid resorption in the intestine.

Pectins also may have a beneficial effect in the diet of individuals w/DM by slowing the rate of absorption of simple sugars and preventing high blood glucose levels after meals.
125. What foods have the highest glycemic index?

The lowest?
Glucose and maltose have the highest glycemic indices (142, w/white bread defined as an index of 100).

Cornflakes and potatoes have high glycemic indices, whereas yogurt and skim milk have particularly low glycemic indices.
126. What determines the glycemic response?
Depends not only on the glycemic index of the foods but also on the fiber and fat content of the food, as well as its methods of preparation.
127. Glucose absorption by intestinal epithelium
Glucose is transported thru the absorptive cells of the intestine by facilitated diffusion and by Na⁺-dependent facilitated transport.

Glucose, therefore, enters the absorptive cells by binding to transport proteins, membrane-spanning proteins that bind the glucose molecule on one side of the membrane and release it on the opposite side. Two types of glucose transport proteins are present in the intestinal absorptive cells: the Na⁺-dependent glucose transporters and the facilitative glucose transporters.
128. What are the Na⁺-dependent transporters?
Na⁺-dependent glucose transporters, which are located on the luminal side of the absorptive cells, enable these cells to concentrate glucose from the intestinal lumen. A lower intracellular Na⁺ concentration is maintained by a Na⁺,K⁺-ATPase on the serosal (blood) side of the cell that uses the energy from ATP cleavage to pump Na⁺ out of the cell into the blood.

Thus, the transport of glucose from a low concentration in the lumen to a high concentration in the cell is promoted by the cotransport of Na⁺ from a high concentration in the lumen to a low concentration in the cell (secondary active transport).
129. What are the facilitative glucose transporters?
Facilitative glucose transporters, which do not bind Na⁺, are located on the serosal side of the cells. Glucose moves via the facilitative transporters from the high concentration inside teh cell to the lower concentration in the blood without the expenditure of energy. In addition to the Na⁺-dependent glucose transporters, facilitative transporters for glucose also exist on the luminal side of the absorptive cells.
130. What are the best-characterized facilitative glucose transporters?
Those found in the plasma membranes of cells (referred to as GLUT 1 - 5)

One common structural theme to these proteins is that they all contain 12 membrane-spanning domains.
131. Glucose transport in the kidney
The epithelial cells of the kidney, which reabsorb glucose from the lumen of the renal tubule back into the blood, have Na⁺-dependent glucose transporters similar to those of intestinal epithelial cells.

They are thus also able to transport glucose against its concentration gradient. Other types of cells use mainly facilitative glucose transporters that carry glucose down its concentration gradient.
132. Galactose absorption
Galactose is absorbed thru the same mechanisms as glucose. It enters the absorptive cells on the luminal side via the Na⁺-dependent glucose transporters and facilitative glucose transporters and is transported thru the serosal side on the facilitative glucose transporters.
133. Fructose absorption
Fructose both enters and leaves absorptive epithelial cells by facilitated diffusion, apparently via transport proteins that are part of the GLUT family.

The transporter on the luminal side is GLUT 5. Although this transporter can transport glucose, it has a much higher activity w/fructose. Other fructose transport proteins also may be present. Also, fructose is absorbed at a much more rapid rate when it is ingested as sucrose than when it is ingested as a monosaccharide.
134. How does the transport of monosaccharides differ among tissues?
The properties of the GLUT transport proteins differ among tissues, reflecting the function of glucose metabolism in each tissue. In msot cell types, the rate of glucose transport across the cell membrane is not rate-limiting for glucose metabolism. This is b/c the isoform of transporter present in these cell types has a relatively low Km for glucose (i.e. a low concentration of glucose will result in half the maximal rate of glucose transport) or is present in relatively high concentrations in the cell membrane so that the intracellular glucose concentration reflects that in the blood.

B/c the hexokinase isozyme present in these cells has an even lower Km for glucose, variations in blood glucose levels do not affect the intracellular rate of glucose phosphorylation. However, in several tissues, the rate of transport becomes rate-limiting when the serum level of glucose is low or when low levels of insulin signal the absence of dietary glucose.
135. What tissue is an example of a tissue in which glucose transport is not rate-limiting?
The erythrocyte (RBC). Although the glucose transporter (GLUT 1) has a Km of 1 to 7 mM, it is present in extremely high concentrations, constituting approx 5% of all membrane proteins.

Consequently, as the blood glucose levels fall from a postprandial level to the normal fasting level or even hypoglycemic level, the supply of glucose is still adequate for the rates at which glycolysis and the pentose phosphate pathway operate.
136. What about glucose transport in the liver?
The Km for the glucose tranporter (GLUT 2) in the liver is relatively high compared with that of other tissues. This is in keeping w/the liver's role as the organ that maintains blood glucose levels. Thus, the liver wiill convert glucose into other energy storage molecules only when blood glucose levels are high.
137. What about glucose transport in muscle and adipose tissue?
In muscle and adipose tissue, the transport of glucose is greatly stimulated by insulin.

The mechanism involves the recruitment of glucose transporters (specifically GLUT 4) from intracellular vesicles into the plasma membrane.

In adipose tissue, the stimulation of glucose transport across the plasma membrane by insulin increases its availability for the synthesis of fatty acids and glycerol from the glycolytic pathway.

In skeletal muscle, the stimulation of glucose transport by insulin increases its availability for glycoslysis and glycogen synthesis.
138. What elicits the hypoglycemic response?
A decrease in blood glucose concentration to some point between 18-54 mg/dL.

The hypoglycemic response is a result of a decreased supply of glucose to the brain and starts w/light-headedness and dizziness and may progress to coma.

The slow rate of transport of glucose thru the blood-brain barrier (from the blood into the CSF) at low levels of glucose is thought to be responsible for this neuroglycopenic response.

Glucose transport from the CSF across the plasma membranes of neurons is rapid and is not rate-limiting for ATP generation from glycolysis.
139. How does glucose pass into the brain?
in the brain, the endothelial cells of the capillaries have extremely tight junctions, and glucose must pass from the blood into the extracellular CSF fluid by GLUT 1 transporters in the endothelial cell membranes and then through the basement membrane.

Measurements of the overall process of glucose transport from the blood into the brain (mediated by GLUT 3 on neural cells) shows a Km of 7 - 11 mM and a max velocity not much greater than the rate of glucose utilization by the brain.

Thus, decreases of blood glucose below the fasting level are likely to significantly affect the rate of glucose metabolism int he brain, b/c of reduced glucose transport into the brain.
140. What causes hyperglycemia in poorly controlled diabetics?
This is often attributable to a lack of circulating, active insulin, which sitmulates glucose uptake (through the recruitment of GLUT 4 transporters from the ER to the plasma membrane) by the peripheral tissues (heart, muscle, and adipose tissue).

Without uptake by these tissues, glucose tends to accumulate w/in the bloodstream, leading to hyperglycemia.
141. How does cholera cause illness?
After being ingested, the V. cholerae organisms attach to the brush border of the intestinal epithelium and secrete an exotoxin that binds irreversibly to a specific chemical receptor (Gm1 ganglioside) on the cell surface.

This exotoxin catalyzes an ADP-ribosylation reaction that increases adenylate cyclase activity and thus cAMP levels in the enterocyte.

As a result, the normal absorption of sodium, anions, and water from the gut lumen into the intestinal cell is markedly diminished. The exotoxin also stimulates the crypt cells to secrete chloride, accompanied by cations and water, from the bloodstream into the lumen of the gut.
142. How do starch blockers work and why were they discontinued?
Starch blockers were based on a protein found in beans, which blocked the action of amylase.

They were discontinued b/c they were never shown to be effective in weight loss. This was probably b/c of a combination of factors, such as inactivation of the inhibitor by the low pH in the stomach, and an excess of amylase activity compared w/the amt of starch blocker ingested.
143. Amylase activity and cystic fibrosis
Amylase activity in the gut is abundant and is not normally rate limiting for the process of digestion. EtOH induced pancreatitis or surgical removal of part of the the pancreas can decrease pancreatic secretion.

Pancreatic exocrine secretion into the intestine can also be reduced as a result of CF, in which mucus blocks the pancreatic duct, which eventually degenerates.

However, pancreatic exocrine secretion can be decreased to 10% of normal and still not affect the rate of starch digestion, b/c amylases are secreted in the saliva and pancreatic fluid in excessive amounts. In contrast, protein and fat digestion are more strongly affected in cystic fibrosis.
144. What is sucrase-isomaltase complex deficiency?
Individuals with genetic deficiencies of the sucrase-isomaltase complex show symptoms of sucrose intolerance but are able to digest normal amounts of starch in a meal without problems.

The maltase activity in the glucoamylase complex, and residual activity in the sucrase-isomaltase complex (which is normally present in excess of need), is apparently sufficient to digest normal amts of dietary starch.
145. What is the basis of the hydrogen breath test?
If a sugar is not absorbed, it is metabolized in the intestinal lumen by bacteria that produce various gases, including hydrogen.

The test is often accompanied by measurements of the amt of sugar that appears in the blood or feces, and the acidity of the feces.
146. What can cause lactase intolerance?
Can either be a result of a primary deficiency of lactase production in the small bowel, or it can be secondary to an injury to the intestinal mucosa, where lactase is normally produced.

The osmotic effect of the lactose and lactic acid in the bowel lumen is responsible for the diarrhea that is often seen as part of this syndrome.
147. What malabsorptive disorders are most commonly encountered in the US?
Celiac disease, pancreatic insufficiency, and Crohn disease

Pancreatic insufficiency, primarily from chronic pancreatitis or CF, is a major cause of defective intraluminal digestion.
148. What is celiac disease?
Celiac disease (AKA sprue) is a chronic disease, in which there is a characteristic mucosal lesion of the small intestine and impaired nutrient absorption, which improves on withdrawal of wheat gliadins and related grain proteins from the diet.

It occurs largely in whites and occurs in the range of 1/100 to 1/200.
149. What is the pathogenesis of celiac disease?

What is the hallmark of this disease?
The fundamental disorder is a sensitivity to gluten, which is the alcohol soluble, water insoluble protein component (gliadin) of wheat and closely related gains (oat, barley, and rye).

The hallmark of this disease is a T-cell mediated chronic inflammatory reaction w/an autoimmune component, which most likely develops as a consequence of a loss of tolerance to gluten.

The small intestinal mucosa, when exposed to gluten, accumulates intraepithelial CD8+ T cells and large numbers of lamina propria CD4+ T cells, which are sensitized to gliadin.
150. What genetic factors are associated w/celiac disease?
Almost all individuals w/celiac share the MHC complex class II HLA-DQ2 or HLA-DQ8 haplotype.

It has been suggested that gliadin is deamidated by the enzyme transglutaminase and that deaminadated gliadin peptides bind to DQ2 and DQ8.

Recognition of these peptides by CD4+ T cells leads to secretion of interferon γ, which damages the intestinal wall.
151. Why is celiac disease associated w/lymphoma?
The epithelial cells secrete large amts of IL-15 that activates CD8+ T cells and increases the risk of lymphoma development.
152. What is the morphology of celiac disease?

1/2
By endoscopy, the small intestinal mucosa appears flat or scalloped, or may be visually normal. Biopsies demonstrate diffuse enteritis, with marked atrophy or total loss of villi.

The surface epithelium shows vacuolar degeneration, loss of the microvillus brush border, and an increased number of intraepithelial lymphocytes.

The crypts, on the other hand, exhibit increased mitotic activity and are elongated, hyperplastic and tortuous, so that the overall mucosal thickness remains the same.
153. What is the morphology of celiac disease?

2/2
The lamina propria has an overall increase in plasma cells, lymphocytes, macrophages, eosinophils, and mast cells. All these changes are usually more marked in the proximal small intestine than in the distal, since it is the duodenum and proximal jejunum that are exposed to the highest concentration of dietary gluten.

Although these changes are characteristic of celiac disease, they can be mimicked by other diseases, most notably tropical sprue.

Mucosal histology usually reverts to normal or near-normal following a period of gluten exclusion from the diet.
154. What are the clinical features of celiac disease?
The symptoms vary from patient to patient. Symptomatic diarrhea and failure to thrive may be evident during infancy, yet adults may seek attention much later in life.

The classic presentation includes diarrhea, flatulence, weight loss, and fatigue. However, extraintestinal manifestations of malabsorption may overshadow the intestinal symptoms.

A characteristic skin blistering lesion, dermatitis herpetiformis, can occur in patients w/celiac disease.

Neurologic disorders are occasionally seen.
155. Dx of celiac disease?

Definitive Dx depends on what 3 things?
Detection of circulating anti-gliadin or "anti-endomysial" antibodies strongly favors the Dx; antibodies against tissue transglutaminase also may be detected, as this is the autoantigen recognized by anti-endomysial antibody.

Definitive Dx rests on:
1. Clinical documentation of malabsorption
2. Demonstration of the intestinal lesion by small bowel biopsy
3. Unequivocal improvement in both symptoms and mucosal histology on gluten withdrawal from the diet
156. What is the prognosis of those w/celiac disease?
Most patients w/celiac disease who adhere to a gluten free diet remain well indefinitely and ultimately die of unrelated causes.

However, there is a long term risk of malignant disease, which includes non-Hodgkin lymphoma, small intestinal adenocarcinoma, and esophageal squamous cell carcinoma.
157. What is tropical sprue?
This condition is a celiac like disease that occurs almost exclusively in people living in or visiting the tropics.

No specific causal agent has been clearly associated w/tropical sprue, but bacterial overgrowth by enterotoxigenic organisms (E. coli and Hemophilus) has been implicated.

Malabsorption usually becomes apparent w/in days or a few weeks of an acute diarrheal enteric infection in visitors to endemic locales and may persist if untreated. The mainstay of treatment is broad spectrum antibiotics.
158. What is the morphology of tropical sprue?
Intestinal changes are extremely variable, ranging from near normal to severe diffuse enteritis. Unlike celiac sprue, injury is seen at all levels of the small intestine.

Patients freq have folate and or vitamin B12 deficiency, leading to markedly atypical enlargement of the nuclei of epithelial cells (megaloblastic change), reminiscent of the changes seen in pernicious anemia.
159. What is whipple disease?
Whipple disease is a rare disease caused by the bacterium Tropheryma whippelii. It is a systemic condition that may involve any organ of the body, but principally affects the intestine, CNS, and joints.

The causal organism T. whippelii is a gram positive actinomycete, named on the basis of molecular phylogenetic analysis. The bacteria proliferate preferentially within macrophages and invoke no significant host immune reaction.
160. What is the morphology of whipple disease?

What is the hallmark?
The hallmark is a small intestinal mucosa laden with distended macrophages in the lamina propria. The macrophages contain PAS positive, diastase-resistant granules (which are lysosomes stuffed w/partially digested microorganisms) and rod-shaped bacilli on electron microscopy.

In untreated cases, the bacilli can be seen as well in neutrophils, the extracellular space of the lamina propria, and even in epithelial cells. Expansion of the villi by the dense infiltrate of macrophages imparts a shaggy gross appearance the the intestinal mucosal surface; edema of the mucosa thickens the intestinal wall. Accompanying these changes is involvement of the mesenteric lymph nodes by the same process and lymphatic dilation, suggesting lymphatic obstruction. The lymphatic blockade is believed to be responsible for lipid desposition in the villi, thus the origianl impression of intestinal lipodystrophy.

Bacilli-laden macrophages also can be found in the synovial membranes of affected joints, the brain, cardiac valves, and elsewhere. At each of these sites, inflammation is essentially absent. Functional impairment can be considerable at each affected site.
161. What are the clinical features of Whipple disease?
Whipple disease is principally encountered in whites in the fourth to fifth decade of life, with a strong male predominance of 10:1. Many cases come from rural regions, suggesting an environmental influence.

It usually present as a form of malabsorption with diarrhea and weight loss, sometimes of years' duration.

Arthropathy is often the inital presentation. Atypical presentations, with polyarthritis, obscure psychiatric complaints, cardiac abnormalities, and other symptom complexes are common. Lymphadenopathy and hyperpigmentation are present in over half the cases.

Currently the Dx rests on demonstration of small intestinal PAS-positive macrophages that contain rod-shaped organisms.

Response to antibiotic therapy is usually prompt, although some patients have a protracted, refractory course.
162. What is abetalipoproteinemia?
Inability to synthesize apolipoprotein B is a rare inborn error of metabolism transmitted by autosomal recessive inheritance.

It is characterized by a defect in the synthesis and export of lipoproteins from intestinal mucosal cells. Free fatty acids and monoglycerides that are produced by hydrolysis of dietary fat enter the absorptive epithelial cells and are re-esterified in the normal fashion but cannot be assembled into chylomicrons.

As a consequence, triglycerides are stored within the cells, creating lipid vacuolation that is readily evident under the light microscope, particularly with special fat stains. Concomitantly, there is complete absence in plasma of all lipoproteins containing apolipoprotein B (chylomicrons, very-low-density lipoproteins, and low-density lipoproteins).

The failure to absorb certain essential fatty acids leads to lipid membrane defects, readily evident in the characteristic acanthocytic erythrocytes (burr cells). The disease becomes manifest in infancy and is dominated by failure to thrive, diarrhea, and steatorrhea.
163. What is idiopathic inflammatory bowel disease?

What are the two disorders in this classification?
This is a set of chronic inflammatory conditions resulting from inappropriate and persistent activation of the mucosal immune system, driven by the presence of normal intraluminal flora.

The two disorder known as IBD are Crohn disease and ulcerative colitis.

These diseases share many common features but have distinctly different clinical manifestations.

Both CD and UC are chronic, relapsing inflammatory diseases of obscure origin. CD is an autoimmune disease that may affect any portion of the GI tract from esophagus to anus, but most often involves the distal small intestine and colon. UC is a chronic inflammatory disease limited to the colon and rectum. Both exhibit extraintestinal inflammatory manifestations.
164. What is the pathogenesis of IBD?

What are the two key abnormalities?
In IBD, the normal immune properties of the GI tract are disrupted, leading to two key pathogenic abnormalities: (1) strong immune responses against normal flora, and (2) defects in epithelial barrier function.

It is postulated that IBD results from unregulated and exaggerated local immune responses to commensal microbes in the gut, in genetically susceptible individuals. Thus, it involves failure of immune regulation, genetic susceptibility, and environmental triggers.
165. What are the genetic susceptibility components to IBD?
HLA associations: HLA-DR1/DR1/DQw5 allelic combination has been observed in 27% of white patients with CD, whereas HLA-DR2 is increased in patients with UC.

A gene called NOD2 *nucleotide-binding oligomerization domian) has recently been shown to be associated with CD.
166. What does the NOD2 protein do?
The NOD2 protein is expressed in many types of leukocytes as well as epithelial cells, and is thought to function as an intracellular receptor for microbes.

Upon binding microbial components, it may trigger the NF-κB pathway; recall that NF-κB is a transcription factor that triggers the production of cytokines and other proteins involved in innate immune defense against infectious pathogens.
167. How are NOD2 mutations associated with CD?
The NOD2 mutations that are associated with CD may reduce the activity of the protein, resulting in the persistence of intracellular microbes and uncontrolled, prolonged immune responses.
168. What is the role of intestinal flora in IBD?
Animal studies have definitively established the importance of gut flora in IBD. If gene-knockout mice that normally develop IBD are made germ-free, the disease disappears. However, the hunt for a specific microbe as the underlying cause has been largely fruitless.

There is also no clear evidence that reducing intestinal flora has a beneficial effect on the course of IBD in humans. Microbes could exacerbate immune reactions by providing antigens and inducing costimulators and cytokines, all of which contribute to T-cell activation. Defects in the barrier function of the intestinal epithelium could allow luminal flora to gain access to the mucosal lymphoid tissue, and thus trigger immune responses.
169. What is the responsible party in the exaggerated local immune responses in IBD?
It is believed that the exaggerated local immune response in IBD is a consequence of too much T-cell activation and/or too little control by regulatory T lymphocytes.

In both CD and UC, the prime culprits appear to be T-cells, particular CD4+ T-cells, and the lesions are likely caused by T-cells and their products.

Although antibodies against certain self-antigens, such as tropomyosin, have been detected in some patients with UC, it is not clear if these autoantibodies play a pathogenic role.
170. What is the autoimmune basis behind CD?

Why?
CD appears to be the result of a chronic delayed-type hypersensitivity reaction induced by IFN-γ-producing TH1 cells.

The nature of the inflammatory infiltrate, especially the presence of granulomas, is consistent with a TH1 response.
171. What is the autoimmune basis behind UC?
Although animal models suggest that ulcerative colitis is caused by excessive activation of TH2 cells, in human disease the signature TH2 cytokine, IL-4, has not been found in the lesions.

It may be that the lesions are caused by an atypical TH2 response, or that there is no consistent pattern of T cell activation or dominant cytokine production.
172. How can we Dx IBD?
Since the exact etiology of IBD is not known, the diagnosis of IBD and the distinction between CD and UC are dependent on clinical history, radiographic examination, laboratory findings, and pathologic examination of tissue. There is no single test upon which a diagnosis is made. Even with the best efforts, the distinction between the two diseases still cannot be made in some cases.

Pathologic appearance, both macroscopic and microscopic, plays a central role in establishing a definitive diagnosis.
173. What two antibody tests are useful for the Dx of IBD?
The pANCA (perinuclear antineutrophilic cytoplasmic antibody) is positive in 75% of patients with UC and in only 11% with CD.

Another test detects an antibody against the cell wall mannan polysaccharide of Saccharomyces cerevisiae (ASCA). This antibody appears to be elevated in CD patients. The clinical utility of these tests remains to be proven.
174. What is Crohn disease?

What are the three pathological characterizations?
When fully developed, CD is characterized pathologically by:

1. Sharply delimited and typically transmural involvement of the bowel by an inflammatory process w/mucosal damage
2. The presence of noncaseating granulomas
3. Fissuring w/formation of fistulae
175. What is the prevalence of CD?
3/100,000

It occurs at any age, from young childhood to advanced age, but peak ages of detection are the second and third decades of life with a minor peak in the sixth and seventh decades. Females are affected slightly more often than males. Whites appear to develop the disease two to five times more often than do nonwhites. In the United States, CD occurs three to five times more often among Jews than among non-Jews. Smoking is a strong exogenous risk factor.
176. What is the morphology of CD?
In CD, there is gross involvement of the small intestine alone in about 40% of cases, of small intestine and colon in 30%, and of the colon alone in about 30%. CD may involve the duodenum, stomach, esophagus, and even mouth, but these sites are distinctly uncommon.

In diseased bowel segments, the serosa is granular and dull gray, and often the mesenteric fat wraps around the bowel surface (creeping fat). The mesentery of the involved segment is also thickened, edematous, and sometimes fibrotic.

*The intestinal wall is rubbery and thick, as a consequence of edema, inflammation, fibrosis, and hypertrophy of the muscularis propria. As a result, the lumen is almost always narrowed; in the small intestine this is evidenced on xrays as the "string sign", a thin stream of barium passing thru the diseased segments. Strictures may occur in the colon but are usually less severe.
177. What is a classic feature of Crohn disease?
The sharp demarcation of diseased bowel segments from adjacent uninvolved bowel.

When multiple bowel segments are involved, the intervening bowel is essentially normal ("skip" lesions).
178. What is a characteristic sign of early CD?
Focal mucosal ulcers resembling canker sores (aphthous ulcers), edema, and loss of the normal mucosal texture.
179. What happens as CD develops?
With progressive disease, mucosal ulcers coalesce into long, serpentine linear ulcers, which tend to be oriented along the axis of the bowel. As the intervening mucosa tends to be relatively spared, the mucosa acquires a coarsely textured, cobblestone appearance.

*Narrow fissures develop between the folds of the mucosa, often penetrating deeply thru the bowel wall and leading to bowel adhesions and serositis.

Further extension of fissures leas to fistula or sinus tract formation, either to an adherent viscus, to the outside skin, or into a blind cavity. Free perforation or localized abscesses may also develop.
180. What are the six histologic changes in CD?
1. Mucosal inflammation
2. Chronic mucosal damage
3. Ulceration
4. Transmural inflammation affecting all layers
5. Noncaseating granulomas
6. Other mural changes
181. Mucosal inflammation in CD
The earliest lesion in CD appears to be focal neutrophilic infiltration into the epithelial layer, particularly overlying mucosal lymphoid aggregates. As the disease becomes more established, neutrophils infiltrate isolated crypts; when a sufficient number of neutrophils have traversed the epithelium of a crypt (both in the small and large intestines), a CRYPT ABSCESS is formed, usually w/ultimate destruction of the crypt.
182. Chronic mucosal damage in CD
The hallmark of inflammatory bowel disease, both CD and UC, is chronic mucosal damage.

Architectural distortion is manifested in the small intestine as variable villus blunting; in the colon, crypts exhibit irregularity and branching. The degree of the glandular architectural distortion in CD is usually less severe than in UC.

Crypt destruction leads to progressive atrophy, particularly in the colon. The mucosa may undergo metaplasia; this may take the form of gastric antral type glands (pyloric metaplasia) or the development of Paneth cells in the distal colon, where they are normally absent (Paneth cell metaplasia).
183. Ulceration in CD
Ulceration is the usual outcome of severe active disease. It may be superficial, may undermine adjacent mucosa in a lateral fashion, or may penetrate deeply into underlying tissue layers.

There is often an abrupt transition between ulceration and adjacent normal mucosa.
184. Noncaseating granulomas in CD
In about 1/2 of the cases, sarcoid-like granulomas may be present in all tissue layers, both w/in areas of active disease and in uninvolved regions of the bowel.

Granulomas have been documented throughout the GI tract, from mouth to rectum, in patients with CD limited to one bowel segment.

Conversely, the absence of granulomas does not preclude the Dx of CD.
185. What are the other mural changes in CD?
In diseased segments, the muscularis mucosa usually exhibits reduplication, thickening, and irregularity.

Fibrosis of the submucosa, muscularis propira, and mucosa eventually leads to stricture formation.

Less common findings are mucosal and submucosal lymphangiectasia, hypertrophy of mural nerve fibers, and localized vasculitis.
186. What are the clinical features of CD?
The clinical manifestations of Crohn disease are extremely variable. They are generally more subtle than those of UC. The disease usually begins with intermittent attacks of relatively mild diarrhea, fever, and abdominal pain, spaced by asymptomatic periods lasting for weeks to many months.

Often the attacks are precipitated by periods of physical or emotional stress. Although emotional influences are thought not to have any direct role in the initiation of the disease, they may contribute to flare-ups.

In those with colonic involvement, occult or overt fecal blood loss may lead to anemia over time, but massive bleeding is uncommon.

In about one-fifth of patients the onset is more abrupt, with acute right lower quadrant pain, fever, and diarrhea sometimes suggesting acute appendicitis or an acute bowel perforation. The course of the disease includes bouts of diarrhea with fluid and electrolyte losses, weight loss, and weakness.
187. What complications may arise from long term CD?
Fribrosing strictures, particularly of the terminal ileum, and fistrulas to other loops of bowel, the urinary bladder, vagina, or perianal skin, or into a peritoneal abscess.

Extensive involvement of the small bowel, including the terminal ileum, may cause marked loss of albumin, generalized malabsorption, specific malabsorption of B12, or malabsorption of bile salts, leading to steatorrhea.
188. What are ten extraintestinal manifestations of CD?
1. Migratory polyarthritis
2. Sacroiliitis
3. Ankylosing spondylitis
4. Erythema nodosum
5. Clubbing of the fingertips
6. Hepatic primary sclerosing cholangitis occurs, but the association is not as strong as in UC
7. Uveitis
8. Nonspecific mild hepatic pericholangitis
9. Renal disorders secondary to trapping of the ureters in the inflammatory process sometimes develop
10. Systemic amyloidosis is a rare late complication
189. What is ulcerative colitis?

How is it similar/different to CD?
UC is an ulceroinflammatory disease limited to the colon and affecting only the mucosa and submucosa except in the most severe cases.

Unlike CD, UC extends in a continuous fashion proximally from the rectum. Well-formed granulomas are absent. Like CD, UC is a systemic disorder associated in some patients with migratory polyarthritis, sacroiliitis, akylosing spondylitis, uveitis, hepatic involvement, and skin lesions.
190. What is the prevalence of UC?
4-12/100,000

In the United States it is more common among whites than among blacks, and females are affected more often than males. The onset of disease peaks between ages 20 and 25, but the condition may arise in both younger and considerably older individuals.

Nonsmoking is associated with UC; ex-smokers are at higher risk for developing UC than never-smokers.
191. What is the morphology of UC?
UC involves the rectum and extends proximally in a retrograde fashion to involve the entire colon ("pancolitis") in the more severe cases.

It is a disease of continuity and no skip lesions are found. In 10% of those with severe pancolitis, the distal ileum may develop mucosal inflammation ("backwash ileitis"). This is probably due to the incompetence of the iliocecal valve, resulting in reflux of the inflammatory material from the colon.

In contrast to CD, the ileitis is often diffuse and limited to within 25 cm from the ileocecal valve. The appendix may be involved w/both CD and UC.
192. What is the colonic involvement with UC like as the disease progresses?

1/2
In the course of the colonic involvement w/UC, the mucosa may exhibit slight reddening and granularity w/friability and easy bleeding. W/fully developed severe active inflammation, there may be extensive and broad based ulceration of the mucosa int eh distal colon or throughout its length.

Isolated islands of regenerating mucosa bulge upward to create pseudopolyps. Often the undermined edges of adjacent ulcers interconnect to create tunnels covered by tenuous mucosal bridges.
193. What is the colonic involvement with UC like as the disease progresses?

2/2
As with CD, the ulcers of UC are frequently aligned along the axis of the colon, but rarely do they replicate the linear serpentine ulcers of CD.

With indolent chronic disease or with healing of active disease, progressive mucosal atrophy leads to a flattened and attenuated mucosal surface.

Unlike CD, mural thickening does not occur in UC, and the serosal surface is usually completely normal. Only in the most severe cases of ulcerative disease (UC, CD, and other severe inflammatory diseases) does toxic damage to the muscularis propria and neural plexus lead to complete shutdown of neuromuscular function. In this instance the colon progressively swells and becomes gangrenous (toxic megacolon)
194. What is the course of mucosal inflammation in UC?
First, a diffuse predominantly mononuclear inflammatory infiltrate in the lamina propria is almost universally present, even at the time of clinical presentation. Neutrophilic infiltration of the epithelial layer may produce collections of neutrophils in crypt lumina (crypt abscesses). These are not specific for UC and may be observed in CD or any active inflammatory colitis. Unlike CD, there are no granulomas, although rupture of crypt abscesses may incite a foreign body reaction in the lamina propria.

Second, further destruction of the mucosa leads to outright ulceration, extending into the submucosa and sometimes leaving only the raw, exposed muscularis propria.

Third, with remission of active disease, granulation tissue fills in the ulcer craters, followed by regeneration of the mucosal epithelium. Submucosal fibrosis and mucosal architectural disarray and atrophy remain as residua of healed disease.
195. What is a key feature of UC?
The mucosal damage is continuous from the rectum and extending proximally.

In CD, mucosal damage in the colon may be continuous but is just as likely to skip areas. It should be noted that quiescent UC, particularly treated disease in which active neutrophilic inflammation is not present, may appear virtually normal histologically. This does not preclude risk for dysplasia.
196. What is a significant consequence of UC?
The spectrum of epithelial changes signifying dysplasia and the progression to frank carcinoma. Nuclear atypia and loss of cytoplasmic differentiation may be present in inflamed or uninflamed colonic mucosa. Epithelial dysplasia is referred to as being low-grade or high-grade; cytologic features are the key to evaluating dysplasia.

Distinguishing between regenerative changes and dysplasia can be very difficult and sometimes impossible. Plaque-like dysplastic lesions, overt polypoid dysplasia (adenomas), or invasive carcinoma are the ultimate lesions arising from flat dysplasia. It should be noted that elderly patients with UC are also at risk for sporadic adenomas.

Distinction between IBD-associated dysplasia and a coexistent incidental adenoma may be difficult.
197. What are the clinical features of UC?
UC typically presents as a relapsing disorder marked by attacks of bloody mucoid diarrhea that may persist for days, weeks, or months and then subside, only to recur after an asymptomatic interval of months to years or even decades. If you're lucky, the first attack is your last.

At the other end, the explosive initial attack may lead to such serious bleeding and fluid and electrolyte imbalance as to constitute a medical emergency.

In most patients, bloody diarrhea containing stringy mucus, accompanied by lower abdominal pain and cramps usually relieved by defecation, is the first manifestation of the disease.

In a small number of paitnets, constipation may appear paradoxically due to disruption of normal peristalsis.
198. What can cause UC to begin?
Often the first attack is preceded by a stressful period in the patient's life. Spontaneously, or more often after appropriate therapy, these symptoms abate in the course of days to weeks.

Flare-ups, when they do occur, may be precipitated by emotional or physical stress and rarely by concurrent intraluminal growth of enterotoxin-forming C. difficile.

Sudden cessation of bowel function with toxic dilatation (toxic megacolon) rarely develops with severe acute attacks; perforation is a potentially lethal event.
199. What two factors determine the prognosis of those w/UC?
1. The severity of the active disease
2. Its duration

About 60% of patients have clinically mild disease. In these individuals, the bleeding and diarrhea are not severe, and systemic signs and symptoms are absent. However, almost all patients (97%) have at least one relapse during a 10-year period, and about 30% of patients require colectomy within the first 3 years of onset due to uncontrollable disease. On rare occasion, the disease runs a fulminant course; unless medically or surgically controlled, this toxic form of the disease can lead to death soon after onset.
200. What is the most feared long-term complication of UC?
Cancer.

There is a tendency for dysplasia to arise in multiple sites, and the underlying inflammatory disease may mask the symptoms and signs of carcinoma.

UC is characterized by DNA damage with microsatellite instability in mucosal cells. More recently, genomic instability was detected in non-dysplastic areas of patients with UC, suggesting that these patients have DNA repair deficiency and genomic instability throughout the intestinal tract.

The associated carcinomas are often infiltrative without obvious exophytic masses, further underscoring the importance of early diagnosis.
201. What are the two situations in which hypersensitivity reactions may occur?
1. Responses to foreign antigens may be dysregulated or uncontrolled, resulting in tissue injury

2. The immune responses may be directed against self antigens, as a result of the failure of self-tolerance
202. What are type I hypersensitivity reactions?
Immediate hypersenitivity, or type I, is a type of pathologic reaction that is caused by the release of mediators from mast cells. This reaction most commonly is triggered by the production of IgE antibody against environmental antigens and the binding of IgE to mast cells in various tissues.
202. What are type II hypersensitivity reactions?
Antibodies other than IgE may cause diseases in two ways:

Antibodies directed against cell or tissue antigens can damage these cells or tissues or impair their function. These diseases are said to be antibody mediated and represent type II hypersensitivity.
203. What are type III hypersensitivity reactions?
Sometimes, antibodies against soluble antigens may form complexes w/the antigens, and the immune complexes may deposit in blood vessels in various tissues, causing inflammation and tissue injury.

Such diseases are called type III hypersensitivity reactions.
204. What are type IV hypersensitivity reactions?
Some diseases result from the reactions of T lymphocytes, often against self antigens in tissues.

These T cell-mediated diseases represent type IV hypersensitivity.
205. What is immediate hypersensitivity?
Type I - This is a rapid, IgE antibody, and mast cell - mediated vascular and smooth muscle reaction, often followed by inflammation, that occurs in some individuals on encounter with certain foreign antigens to which they have been exposed previously.

These reactions are also called allergy, or atopy.
206. What is the sequence of events in the development of immediate hypersensitivity?
Consists of the production of IgE antibodies in response to an antigen, binding of IgE to Fc receptors of mast cells, cross-linking of the bound IgE by the antigen, and release of mass cell mediators.
207. What is the late phase reaction?
Some mast cell mediators cause a rapid increase in vascular permeability and smooth muscle contraction.

Other mast cell mediators are cytokines that recruit neutrophils and eosinophils to the site of the reaction over several hours. This inflammatory component is called the late phase reaction, and it is mainly responsible for the tissue injury that results from repeated bouts of immediate hypersensitivity.
208. What causes the production of the IgE antibody?
The individuals w/allergies encounter some antigens which results in the activation of TH2 cells and the production of IgE antibody.

Immediate hypersensitvity develops as a consequence of the activation of TH2 cells in response to protein antigens or chemicals that bind to proteins.
209. What are the cytokines secreted by TH2 cells and what do they do?
TH2 cells secrete interleukin IL-4 and IL-13, and these stimulate B lymphocytes specific for the foreign antigens to switch to IgE-producing plasma cells.
210. How are mast cells activated in type I hypersensitivity reactions?
IgE antibody produced in response to allergen binds to high-affinity Fc receptors specific for the ε heavy chain expressed on mast cells.

Mast cells are present in all connective tissues, and which of the body's mast cells are activated by cross-linking of allergen specific IgE often depends on the route of entry of the allergen.
211. What is the FcεRI?
The high-affinity Fcε receptor, called FcεRI, consists of three chains, one of which binds the Fc portion of the ε heavy chain very strongly. The other two chains of the receptor are signaling proteins.

The same FcεRI also is present on basophils, but the role of basophils in immediate hypersensitivity is not well established as the role of mast cells.
212. What happens when mast cells sensitized by IgE are exposed to the allergen?
The cells are activated to secrete their mediators; Mast cell activation results from binding of the allergen to two or more IgE antibodies on the mast cell.

When this happens, the IgE and the FcεRI molecules that are carrying the IgE are cross-linked, triggering biochemical signals from the signal transducing chains of FcεRI.

The signals lead to three types of responses in the mast cells: degranulation, synthesis and secretion of lipid mediators, and synthesis and secretion of cytokines.
213. What are the most important mediators produced by mast cells?
Vasoactive amines and proteases that are released from granules, products of arachidonic acid metabolism, and cytokines.
214. What is the role of the cytokines produced by the mast cells?
They stimulate the recruitment of leukocytes, which cause the late phase reaction.

The principal leukocytes involved int his reaction are eosinophils, neutrophils, and TH2 cells.

Mast cell-derived TNF and IL-4 promote neutrophil and eosinophil rich inflammation.

Chemokines produced by mast cells and by epithelial cells in the tissues also contribute to leukocyte recruitment.

Eosinophils and neutrophils liberate proteases, which cause tissue damage, and TH2 cells may make this worse by producing more cytokines.
215. What activates eosinophils?

What does IL-13 do?
Eosinophils are activated by the cytokine IL-5, which is produced by TH2 cells and mast cells.

The TH2 cytokine IL-13 acts on airway epithelial cells to stimulate the secretion of mucus.
216. What are the different clinical syndromes of type I hypersensitivity reactions?
1. Allergic rhinitis
2. Food allergies
3. Bronchial asthma
4. Anaphylaxis
217. What is the aim of therapy for immediate hypersensitivity reactions?
Aimed at inhibiting mast cell degranulation, antagonizing the effects of mast cell mediators, and reducing inflammation.

Corticosteroids are used to inhibit inflammation. Epinephrine inhibits further mast cell degranulation.
218. What are type II hypersensitivity diseases?
Antibodies (other than IgE) against cells or extracellular matrix components may deposit in any tissue that expresses the relevant target antigen.

Disease caused by such antibodies usually are specific for a particular tissue. Immune complexes tend to deposit in blood vessels at sites of turbulence or high pressure. Therefore, immune complex diseases tend to be systemic and often manifest as widespread vasculitis, arthritis, and nephritis.
219. What is the etiology of antibody mediated disease?
The antibodies that cause disease most often are autoantibodies against self antigens and less commonly are specific for foreign antigens.

The production of autoantibodies results from a failure of self tolerance.
220. What are the two best examples of antibody mediated diseases that are late sequelae of streptococcal infections??
Rhematic fever and poststroptococcal glomerulonephritis.

Some immune complex disease are cuased by the formation of complexes of antibodies against microbial antigens with the antigens. This may occur in patients with chronic infections with certain viruses, e.g. EBV, malaria.
221. What are the mechanisms of antibody mediated disease?
Antibodies specific for cell and tissue antigens may deposit in tissues and cause injury by inducing local inflammation, or they may interfere with normal cellular functions.

These antibodies induce inflammation by attracting and activating leukocytes. IgG antibodies of the IgG1 and IgG3 subclasses bind to neutrophil and macrophage Fc receptors and activate these leukocytes, resulting in inflammation.

The same antibodies,a s well as IgM, activate the complement system by the classical pathway, resulting in the production of complement byproducts that recruit leukocytes and induce inflammation.
222. What is serum sickness?
Serum sickness is induced by systemic administration of a protein antigen, which elicits an antibody response and leads to the formation of circulating immune complexes.

It usually occurs when a person receives injections of serum from other individuals or species.
223. What is the Arthus reaction?
The Arthus reaction is induced by subcutaneous administration of a protein antigen to a previously immunized animal; it results in the formation of immune complexes at the site of antigen injection, and a local vasculitis.
224. What is the treatment in patients with an antibody specific for CD20, a surface protein of mature B cells, which results in depletion of B cells?
There is interest in inhibiting the production of autoantibodies by treating patients with antagonists that block CD40 ligand and thus inhibit helper T cell dependent B cell activation.
225. What is the etiology of T-cell mediated diseases?
T-cell mediated hypersensitivity reactions are caused by autoimmunity and by responses to environmental antigens.

The autoimmune reactions usually are directed against cellular antigens w/restricted tissue distribution. Therefore, T cell-mediate autoimmune disease tend to be limited to a few organs and usually are not systemic.
226. Tuberculosis, HBV, HCV, and poison ivy
In tuberculosis, a T cell-mediated immune response is raised against M. tuberculosis, and the response becomes chronic b/c the infection is difficult to eradicate. The resultant granulomatous inflammation causes injury to normal tissues at the site of infection.

In the hepatitis virus infection, the virus itself may not be highly cytopathic, but the cytotoxic T lymphocyte response to infected hepatocytes may cause liver injury.
227. What are superantigens?
Excessive polycolonal T cell activation by certain microbial toxins produced by some bacteria and viruses can lead to production of large amts of inflammatory cytokines, causing a syndrome similar to septic shock.

These toxins are called superantigens b/c they stimulate large numbers of T cells.

Superantigens bind to invariant parts of T cell receptors on many different clones of T cells, regardless of antigen specificity, thereby activating these cells.
228. What are the mechanisms of tissue injury in T cell-mediated diseases?
Tissue injury is caused by a delayed type hypersensitivity reaction mediated by CD4+ T cells or by killing of host cells by CD8+ T cells.
229. What are the cytokines involved in T-cell mediated diseases?
CD4+ T cells may react against cell or tissue antigens and secrete cytokines that induce local inflammation and activate macrophages.

Different disease may be associated w/activation of TH1 and TH17 cells.

TH1 cells are the source of IFN-γ, the principal macrophage activating cytokine, and TH17 cells are thought to be responsible for recruitment of leukocytes, including neutrophils.
230. What are the characteristics of T-cell mediated diseases?
Many organ specific autoimmune diseases in humans are believed ot be caused by T cells.

These disorders are typically chronic and progressive, in part b/c T cell-macrophage interactions tend to amplify the reaction.

Also, tissue injury w/release and alteration of self proteins may result in reactions against these proteins, a phenomenon known as "epitope spreading" to indicate that the initial immune response against one or a few self antigen epitopes may expand to include responses against many more self antigens.
231. What are the therapies for T-cell mediated diseases?
Designed to reduce inflammation, using corticosteroids and antagonists against cytokines such as TNF, and to inhibit T cell responses with immunosuppressive drugs such as cyclosporine.

Antagonists of TNF have proved to be beneficial in patients w/RA and inflammatory bowel disease.
233. What is a type I hypersensitivity reaction?

What is it mediated by?
Type I hypersensitivity is a rapidly developing immunologic reaction occurring within minutes after the combination of an antigen w/antibody bound to mast cells in individuals previously sensitized to the antigen.

Immediate hypersensitivity is mediated by IgE antibodies directed against specific antigens (allergens).

Synthesis of IgE antibody requires the induction of CD4+ helper T cells of the TH2 type; these TH2 cells produce multiple cytokines that contribute to various aspects of this response.
234. What cytokine is essential for IgE synthesis?

What is the first step in IgE synthesis?
IL-4, produced by TH2 cells, is essential for IgE synthesis.

The first step in the synthesis of IgE is the presentation of the antigen to naive CD4+ helper T cells by dendritic cells that capture the antigen from its site of entry.
235. What cytokines are responsible for promoting production and survival of eosinophils - important effector cells in type I hypersensitivity reactions?
IL-3, IL-5, and granulocyte-macrophage colony stimulating factor (GM-CSF) promote production and survival of eosinophils.
236. Where are IgE antibodies synthesized in response to prior exposure to allergens are normally bound to where?
Mast cells and basophils via specific surface Fc receptors.

On re-exposure, allergen bind to and cross-links the IgE on mast cells and results in:
1. Degranulation of vesicles containing primary mediators
2. De novo synthesis and release of secondary mediators
237. What six allergens can trigger mast cell and basophil degranulation?
1. Complement fragments (C3a and C5a (anaphylatoxins)
2. Certain drugs i.e. codeine, morphine, adenosine
3. Mellitin (bee venom)
4. Sunlight
5. Trauma
6. Heat/cold
238. What occurs in the initial phase of a type I hypersensitivity reaction?

Four things are released...
Primary mast cell mediators that include the initial rapid response include:
1. Biogenic amines (e.g. histamine) which cause bronchial smooth muscle contraction, increased vascular permeability and dilation, and increased mucous gland secretions.
2. Chemotactic mediators (e.g. eosinophil chemotactic factors and neutrophil chemotactic factors)
3. Enzymes contained in granule matrix (e..g chymase, tryptase) that generate kinins and activated complement by acting on their precursor proteins.
4. Proteoglycans (e.g. heparin)
239. What occurs in the second, delayed phase of a type I hypersensitivity reaction?

What drives this phase?
This stage is characterized by an intense inflammatory cell infiltration with associated tissue damage. This late secondary phase is driven by lipid mediators and cytokines produced by the activated mast cells.
240. What are the four lipid mediators produced by the mast cells in the delayed phase?

What is the role of each lipid mediator?
1. Leukotriene B4 is highly chemcotactic for neutrophils, monocytes, and eosinophils
2. Leukotrienes C4, D4, and E4 are 1000x more potent than histamine in increasing vascular permeability and causing bronchial smooth muscle contraction. These also cause marked mucous gland secretion
3. Prostaglandin D2 causes intense bronchospasm, vasodilation, and mucous secretion
4. Platelet-activating factor causes platelet aggregation, histamine release, bronchoconstriction, vasodilation, and increased vascular permeability. It also has proinflammatory effects, such as chemoattraction and degranulation of neutrophils.
241. In sum, what do the lipid mediators lead to?
The lipid mediators are generated by sequential reactions in the mast-cell membranes that lead to activation of phospholipase A2, an enzyme that acts on the membrane phospholipids to yield arachidonic acid.

This is the parent compound from which leukotrienes and prostaglandins are derived by the 5-lipoxygenase and cyclooxygenase pathways.
242. What are the important cytokine mediators in type I hypersensitivity reactions?
The cytokines include TNF, IL-1, IL-3, IL-4, IL-5, IL-6, and GM-CSF, as well as chemokines, such as macrophage inflammatory protein (MIP)-1α and (MIP)-1β.

Mast cell derived TNF and chemokines are important mediators of the inflammatory response seen at the site of allergic inflammation. Inflammatory cells that accumulate at the sites of type I hypersensitivity reactions are additional sources of cytokines and of histamine-releasing factors that cause further mast-cell degranulation.
243. What cytokines can epithelial cells produce?
IL-6, IL-8 and GM-CSF
244. What is the role of TNF-α in type I hypersensitivity reactions?
TNF-α in particular is a powerful proinflammatory cytokine that recruits and activates many additional inflammatory cells.

Recruited inflammatory cells also release cytokines, and TNF-α-activated epithelial cells secrete chemokines (e.g. eotaxin and RANTES) that recruit eosinophils.
245. What is the role of eosinophils in type I hypersensitivity reactions?
Eosinophils are paritcularly important in late-phase responses; they cause tissue damage by releasing major basic protein and eosinophil cationic protein.
246. What does atopy mean?
Atopy refers to a predisposition to develop localized immediate hypersensitivity reactions to a variety of inhaled and ingested allergens.

Atopic individuals seem to have a higher serum IgE level, and more IL-4 producing TH2 cells, compared w/the general population.
247. What are the genetic components of immediate hypersensitivity?
A positive family history of allergy is found in 50% of atopic individuals.

Candidate genes have been mapped to 5q31 for asthma, where genes for the cytokines IL-3, 4, 5, 9, 13, and GM-CSF are located, consistent with the idea that these cytokines are involved in the reactions.

Linkage has also been noted to 6p, close to the HLA complex, suggesting that the inheritance of certain HLA alleles permits reactivity to certain allergens.

Another asthma associated locus is on chromosome 11q13, the location of the gene encoding the β chain of the high-affinity IgE receptor.
248. What is systemic anaphylaxis?
Systemic anaphylaxis typically follows parenteral or oral administration of an allergen. The severity reflects the level of sensitization, and even minuscule doses may induce anaphylactic shock in an appropriate host.

Pruritus, urticaria, and erythema occur minutes after exposure, followed by bronchoconstriction and laryngeal edema; this can escalate into laryngeal obstruction, hypotensive shock, and death within minutes to hours.
249. What are local immediate hypersensitivity reactions?
These reactions are exemplified by atopic allergies. There is a hereditary predisposition affecting 10% of the population (mapping to 5q31, where many of the TH2 type cytokines are located).

Affected individuals tend to develop local type I responses to common inhaled or ingested allergens.

Symptoms include urticaria, angioedema, rhinitis, and asthma.
250. What is antibody-mediated (type II) hypersensitivity?
Type II hypersensitivity is mediated by antibodies directed toward antigens present on cell surfaces or extracellular matrix.

The antigenic determinants may be intrinsic to the cell membrane or matrix, or they may take the form of an exogenous antigen such as a drug metabolite that is adsorbed on a cell surface or matrix.

In either case, the hypersensitivity reaction results from the binding of antibodies to normal or altered cell surface antigens. Most of these reactions involve the effector mechanisms that are used by antibodies, namely the complement system and phagocytes.
251. What are the three major pathways in type II hypersensitivity reactions?
1. Opsonization and complement- and Fc receptor-mediated phagocytosis

2. Complement- and Fc receptor-mediated inflammation

3. Antibody-mediated cellular dysfunction
252. Opsonization and complement- and Fc receptor-mediated phagocytosis
Cells can be directed lysed via the C5-C9 complement membrane attack complex (MAC) or they can be opsonized (enhanced phagocytosis) as a result of fixation of antibody or C3b fragments.

Low concentrations of bound antibody (IgG or IgE) can also cause cell lysis (w/o phagocytosis) by nonsensitized cells bearing Fc receptors (e.g. NK cells, so called antibody-dependent cell mediated cytotoxicity (ADCC).
253. What is antibody-dependent cell mediated cytotoxicity (ADCC)?
This form of antibody mediated cell injury does not involve fixation of complement but instead requires the cooperation of leukocytes.

Cells that are coated with low concentrations of IgG antibody are killed by a variety of effector cells, which bind to the target by their receptors for the Fc fragment of IgG, and cell lysis proceeds without phagocytosis.
254. What mediates ADCC?
ADCC may be mediated by monocytes, neutrophils, eosinophils, and NK cells.

Although, in most instances, IgG antibodies are involved in ADCC, in certain cases, e.g. eosinophil-mediated cytotoxicity against parasites), IgE antibodies are used.
255. In what four situations does antibody-mediated cell destruction and phagocytosis occur?
1. Transfusion reactions, in which cells from an incompatible donor react w/and are opsonized by preformed antibody in the host.
2. Erythroblastosis fetalis, in which there is an antigenic difference between the mother and the fetus, and antibodies (of the IgG class) from the mother cross the placenta and cause destruction of fetal red cells.
3. Autoimmune hemolytic anemia, agranulocytosis, and thrombocytopenia, in which individuals produce antibodies to their own blood cells, which are then destroyed.
4. Certain drug reactions, in which antibodies are produced that react w/the drug, which may be attached to the surface of erythrocytes or other cells.
256. Complement- and Fc receptor-mediated inflammation
The deposition of antibodies w/subsequent complement activation, e.g. C5a, in the extracellular matrix leads to the recruitment and activation of nonspecific inflammatory cells (neutrophils and macrophages).

These activated cells can release injurious proteases and reactive oxygen species that lead to tissue pathology.
257. Antibody-mediated cellular destruction

What are some diseases that exemplify this mechanism?
In some cases, antibodies directed against cell surface receptors impair or dysregulate function without causing cell injury or inflammation.

For example, in myasthenia gravis, antibodies reactive w/ACh receptors in the motor end-plates of skeletal muscles impair neuromuscular transmission and therefore cause muscle weakness.

In pemphigus vulgaris, antibodies against desmosomes disrupt intercellular junctions in epidermis, leading to the formation of skin vesicles.

The converse (i.e. antibody-mediated stimulation of cell function) is noted in Graves disease. In this disorder, antibodies against the TSH receptor on thyroid epithelial cells stimulate the cells, resulting in hyperthyroidism.
258. What is immune complex-mediate (type III) hypersensitivity?
Antigen-antibody complexes produce tissue damage mainly by eliciting inflammation at the sites of deposition.

The toxic reaction is initiated when antigen combines w/antibody within the circulation and these are deposited, typically in vessel walls, or the complexes are formed at extravascular sites where antigen may have been deposited previously (in situ immune complexes).
259. What are the two general types of antigens that cause cell mediated injury?
1. The antigen may be exogenous, such as a foreign protein, a bacterium, or a virus
2. Under some circumstances, the individual can produce antibody against self-components -endogenous antigens.

The latter can be circulating antigens present in the blood or, more commonly, antigenic components of one's own cells and tissues.
260. What is systemic immune complex disease?
Systemic immune complex disease is characterized by circulating immune complexes that are systemically deposited.

Acute serum sickness is the prototypical systemic immune complex disease. it is caused by administration of large amts of a foreign protein; after inoculation, newly synthesized antibodies complex w/the foreign antigen to form circulating immune complexes
261. Do larger or smaller immune complexes circulate for longer periods of time?
Smaller immune complexes circulate for longer periods of time b/c they bind w/low avidity to mononuclear phagocytes and are ineffectively cleared; these complexes are prone to deposit within a capillary or arteriolar walls, causing vasculitis.

With continued antibody production, large immune complexes eventually form; these are cleared by phagocytes, ending the disease process.
262. What other factors enhance immune complex deposition?
Immune complex deposition is enhanced by increased vascular permeability resulting from inflammatory cell activation by immune complex binding to Fc or C3b receptors.

The activated inflammatory cells release vasoactive mediators, including cytokines.
263. Deposition of immune complexes causes...?
Deposition of immune complexes activates the complement cascade and subsequent tissue injury derives largely from complement-mediated inflammation and cells bearing Fc receptors.
264. C3b does...?

C5a does...?
C3b enhances opsonization

C5a (chemotactic factor) release promotes neutrophil and monoctyte recruitment w/subsequent protease and reactive oxygen species elaboration.
265. C3a and C5a do...?
C3a and C5a release increases vascular permeability and causes smooth muscle contraction
266. What is the relationship between immune complexes and platelet aggregation?
Immune complexes also aggregate platelets (w/subsequent degranulation) and activate factor XII (Hageman factor).

Both of these reactions augment the inflammatory process and initiate the formation of microthrombi. The resultant inflammatory lesion is termed vasculitis if it occurs in blood vessels, glomerulonephritis if it occurs in renal glomeruli, arthritis if it occurs in the joints, and so on.

The coagulation cascade and kinin systems are thus invovled as well.
267. What is the pathogenesis of immune complex disorders?
Complement-fixing antibodies (IgG and IgM) and antibodies that bind to leukocyte Fc receptors (some subclasses of IgG) induce pathologic lesions of immune complex disorders.

B/c IgA can activate complement by the alternative pathway, IgA-containing complexes may also induce tissue injury. The important role of complement in the pathogenesis of the tissue injury is supported by the observations that during the active phase of the disease, consumption of complement decreases the serum levels, and experimental depletion of complement greatly reduces the severity of the lesions.
268. What is the morphology of immune complex injury?
The morphologic consequences of immune complex injury are dominated by acute necrotizing vasculitis, with necrosis of the vessel wall and intense neutrophilic infiltration. The necrotic tissue and deposits of immune complexes, complement, and plasma protein produce a smudgy eosinophilic deposit that obscures the underlying cellular detain *an appearance termed fibrinoid necrosis.

When complexes are deposited in kidney glomeruli, the affected glomeruli are hypercellular b/c of swelling and proliferation of endothelial and mesangial cells, accompanied b neutrophilic and monocytic infiltration. The complexes can be seen on immunofluorescence microscopy as granular lumpy deposits of immunoglobulin and complement and on electron microscopy as electron-dense deposits along the glomerular basement membrane.
269. Disease progression in immune complex injury
With time and clearance of the inciting antigen and immune complex, the lesions resolve.

In chronic serum sickness, resulting from recurrent or prolonged antigen exposures and ongoing immune complex deposition (e.g. systemic lupus erthematosus, SLE), there is intimal thickening and vascular and/or parenchymal scarring.
270. What is the local immune complex disease (Arthus reaction)?
The Arthus reaction is a localized are of tissue necrosis resutling from acute immune complex vasculitis, usually elicited in the skin. The reaction can be produced experimentally by intracutaneous injection of antigen in an immune animal having circulating antibodies against the antigen. As the antigen diffuses into the vascular wall, it binds the preformed antibody, and large immune complexes are formed locally which precipitate in the vessel walls and trigger and inflammatory reaction.

In contrast to IgE mediated type I reactions, which appear immediately, the Arthus lesion develops over a few hours and reaches a peak 4-10 hours after injection.
271. What is the morphology of local immune complex disease?
It can be seen as an area of visible edema w/severe hemorrhage followed occasionally by ulceration.

Immunofluorescent stain reveal complement, immunoglobulins, and fibrinogen deposited in the vessel walls, usually the venules, and histologically the vessels show fibrinoid necrosis and inflammation. Thrombi are formed in the vessels, resulting in local ischemic injury.
272. What is cell mediated (type IV) hypersensitivity?
Cell mediated hypersensitivity is initiated by specifically sensitized T lymphocytes and includes delayed-type hypersensitivity mediated by CD4+ T cells, and T-cell mediated cytotoxicity, mediated by CD8+ T cells.

It is the principal pattern of immunologic response not only to a variety of intracellular microbiologic agents, such as Mycobacterium tuberculosis, but also to many viruses, fungi, protozoa, and parasites.
272. What is delayed type hypersensitivity?

What drives it?
This resposne is largely mediated by CD4+ TH1 cells that secrete specific cytokines after encounter w/processed antigen expressed by APCs.

The TH1 response is driven by IL-12 secreted by activated macrophages.
273. What is the morphology of delayed type hypersensitivity?

1/2
Morphologically, delayed type hypersensitivity is characterized by the accumulation of mononuclear cells around small veins and venules, producing a perivascular "cuffing".

There is an associated increased microvascular permeability caused by mechanisms similar to those in other forms of inflammation. Not unexpectedly, plasma proteins escape, giving rise to dermal edema and deposition of fibrin in the interstitium.
274. What is the morphology of delayed type hypersensitivity?

2/2
A classic example is the tuberculin reaction, which is produced by the intracutaneous injection of tuberculin, a protein LPS component of the tubercle bacillus.

With certain persistent or nondegradable antigens, such as tubercle bacilli colonizing the lungs or other tissues, the initial perivascular lymphatic infiltrate is replaced by macrophages over 2-3 weeks.

The accumulated macrophages often undergo a morphologic transformation into epithelium-like cells and are then referred to as eptheliod cells. A microscopic aggregation of epithleiod cells, usually surrounded by a collar of lymphocytes, is referred to as a granuloma. This pattern of inflammation is called granulomatous inflammation.
275. What are the TH1 cytokines?
TH1 cytokines include IFN-γ, IL-2, and TNF-α; these cytokines mediate injury by recruiting and activating antigen-nonspecific monocytes and macrophages.
276. What is the importance of IL-12?
IL-12, a cytokine produced by macrophages and dendritic cells, is critical for the induction of the TH1 response and hence delayed hypersensitivity. On initial encounter w/a microbe, the macrophages and dendritic cells that are presenting microbial antigens secrete IL-12, which drives the differentiation of naive CD4+ helper cells to TH1 cells.

These, in turn, produce other cytokines. IL-12 is also a potent inducer of IFN-γ secretion by T cells and NK cells. IFN-γ further augments the differentiation of TH1 cells.
277. What is the importance of IFN-γ?
IFN-γ has many effects and is the key mediator of delayed type hypersensitivity. Most importantly, it is a powerful activator of macrophages.

Activated macrophages are altered in several ways: their ability to phagocytose and kill microorganisms is markedly augmented; they express more class II molecules on the surface, thus facilitating further antigen presentation; they secrete several polypeptide growth factors, such as PDGF, which stimulate fibroblast proliferation and augment collagen synthesis; they secrete TNF, IL-1, and chemokines, which promote inflammation, and they produce more IL-12, thereby amplifying the TH1 response.
278. What does IL-2 do?
IL-2 causes autocrine and paracrine proliferation of T cells, which accumulates at sites of delayed hypersensitivity; included in this infiltrate are some antigen specific CD4+ TH1 cells and many more bystander T ells that are recruited to the site.
279. What three things do TNF and lymphotoxin do to endothelial cells?
1. Increased secretion of prostacyclin, which, in turn, favors increased blood flow causing local vasodilation
2. Increased expression of P-E selectins, adhesion molecules that promote attachment of the passing lymphocytes and monocytes
3. Induction and secretion of chemokines such as IL-8

Together, all these changes in the endothelium facilitate the extravasation of lymphocytes and monocytes at the site of the delayed hypersensitivity reaction.
280. What is T cell-mediated cytotoxicity?
Generation of cytotoxic T lymphocytes is the principal pattern of response to many viral infection and to tumor cells.

CTLs also contribute to allograft rejection. CTL-induced injury is mediated by perforin-granzyme and Fas-FasL pathways that ultimately induce apoptosis.
281. What is perforin?
Perforin can perforate the plasma membranes of the target cells that are under attack by CD8+ lymphocytes. At first, CD8+ T cells come in close contact w/the target cells; this is followed by polymerization of the released perforin molecules and their insertion into the target cell membranes, thus drilling holes into the membranes.

In addition, the perforin pores allow water to enter the cells, thus causing osmotic lysis.
282. What are granzymes?
The CTL granules contain proteases called granzymes, which are delivered into the target cells via the perforin-induced pores.

Once within the cells, granzymes activate caspases, which induce apoptosis of the target cells.
283. What is the first step in catecholamine synthesis?
The first step in catecholamine synthesis is the oxidation of tyrosine to dihydroxyphenylalanine (DOPA) by the enzyme tyrosine hydrolase, and is the rate limiting step.
284. How is dopamine created?

How does this lead to norepinephrine?
DOPA is converted to dopamine by a generic aromatic AA decarboxylase.

Dopamine can then be hydroxylated at the 9-position by dopamine-β-hydroxylase to yield norepinephrine.
285. How is epinephrine produced by tissues?
In tissues that produce epinephrine, norepinephrine is then methylated on its amino group by phenylethanolamine-N-methyltransferase (PNMT)
286. How is dopamine transported into synaptic vesicles?
Dopamine is transported into synaptic vesicles by a 12-helix membrane-spanning proton antiporter called the vesicular monamine transporter (VMAT).

Dopamine inside the vesicle is converted to norepinephrine by dopamine-β-hydroxylase.
287. What are hexamethonium and mecamylamine?
Ganglionic blockers such as hexamethonium and mecamylamine block the ganglionic nicotinic ACh receptor, without significant effects on skeletal muscle ACh receptors.
288. What mediates reuptake of catecholamine into the neuronal cytoplasm?
Reuptake of catecholamine into the neuronal cytoplasm is mediated by a selective catecholamine transported (e.g. norepinephrine transporter or NET).

This symporter uses the inward sodium gradient to concentrate catecholamines in the cytoplasm of sympathetic nerve endings, thus limiting the postsynaptic response and allowing neurons to recycle the transmitter for subsequent release.
289. What is MAO and catechol-O-methyltransferase (COMT)?
MAO is a mitochondrial enzyme that is expressed in most neurons. It exists in two isoforms, MAO-A and MAO-B.

The two isoforms have some degree of ligand specificity; MAO-A preferentially degrades serotonin, norepinephrine, and dopamine, while MAO-B degrades dopamine more rapidly than serotonin and norepinephrine.

COMT is a cytosolic enzyme that is expressed primarily in the liver.
290. What are adrenergic receptors?
AKA adrenoceptors; they are selective for norepinephrine and epinephrine.

These receptors are divided into two main classes, termed α & β
291. What are PDZ proteins?
Members of the PDZ family mediate regulatory functions, including receptor internalization and coupling to adaptor proteins such as Grb2 or guanine-nucleotide exchange factors that regulate small monomeric G proteins.
292. What is G protein receptor kinase (GRK)?

What is β-arrestin?
The accumulation of the βγ subunits in the membrane recruits a G protein receptor kinase (GRK), which phosphorylates the receptor at residues in the C-terminus that are important targets for inactivator proteins.

The phosphorylated state of a G protein can bind to another protein called β-arrestin that sterically inhibits the receptor-G protein interaction, effectively silencing receptor signaling.
293. What are the epinephrine receptors?
Epinephrine is an agonist at both α & β adrenoceptors.

At low concentrations, epinephrine has predominantly β₁ and β₂ effects, while at high concentrations, its α₁ effects predominate.
294. What are the norepinephrine receptors?
Norepinephrine is an agonist at α₁ and β₁ receptors, but has relatively little effect at β₂ receptors.
295. What are the inhibitors of catecholamine synthesis, and what are they used for?
Inhibitors of catecholamine synthesis have limited clinical utility b/c such agents nonspecifically inhibit the formation of all catecholamines.

α-Methyltyrosine is a structural analogue of tyrosine that is transported into nerve terminals, where it inhibits tyrosine hydroxylase, the first enzyme int he catecholamine biosynthesis pathway.

This agent is used occasionally in the treatment of hypertension associated with pheochromocytoma.
296. α-Methyltyrosine
MOA: Inhibits tyrosine hydroxylase.

PURPOSE: pheochromocytoma associated hypertension

ADVERSE: Orthostatic hypertension and sedation

CONTRA: Hypersensitivity to α-Methyltyrosine

NOTES: *Used rarely.
297. What are inhibitors of catecholamine storage?
MOA: These agents inhibit catecholamine storage in vesicles, resulting short term increase in release of catecholamines from the synaptic terminal ("sympathomimetic") but long-term depletion of available pool of catecholamines ("sympatholytic").

Includes:
1. Reserpine
2. Guanethidine
3. Guanadrel
4. Amphetamine
5. Methylphenidate
6. Pseudoephedrine
298. Reserpine
MOA: Reserpine binds tightly to the vesicular antiporter VMAT. The drug irreversibly damages VMAT, resulting in vesicles that lose their ability to concentrate and store norepinephrine and dopamine.

PURPOSE: Hypertension

ADVERSE: cardiac arrhythmia, GI hemorrhage, thrombocytopenia, dream anxiety disorder, impotence, psychotic depression, dizziness, and nasal congestion.

CONTRA: Active GI disease, depression, electroshock therapy, and renal failure

NOTES: Used experimentally to assess whether effect of drug requires its concentration in presynaptic terminals. Rarely used as a therapeutic agent due to its irreversible action and its association with psychotic depression.
299. Tyramine
Tyramine is a dietary amine that is ordinarily metabolized in the GI tract and liver by MAO. In patients taking MAO inhibitors, tyramine is absorbed in the gut, transported through the blood, and taken up by sympathetic neurosn, where it is transproted into synaptic vesciles by VMAT.

By this mechanism, an acute challenge with large amts of tyramine can cause acute displacement of vesicular NE and massive non-vesicular release of NE from the nerve terminal.
300. Octopamine
Although tyramine itself is poorly retained in synaptic vesicles, its hydroxylated metabolite octopamine can be stored at high concentrations in the vesicles.

B/c octopamine has little agonist activity at most mammalian adrenoceptors, postsynaptic response to sympathetic stimulation may gradually be diminished, leading ultimately to postural hypertension.
301. Guanethidine
MOA: Like tyramine, guanethidine concentrates in transmitter vesicles and displaces NE, leading to gradual depletion of NE.

PURPOSE: Hypertension

ADVERSE: Kidney disease, apnea, orthostatic hypotension, fluid retention, dizziness, blurred vision, impotence.

CONTRA: MAOI therapy, heart failure and pheochromocytoma.

NOTES: Inhibition of cardiac sympathetic nerves leads to reduced cardiac output; inhibition of sympathetic response leads to symptomatic hypotension following exercise.
302. Guanadrel
Guanadrel has similar MOA as guanethidine. Guanadrel also acts as a false neurotransmitter.

As with guanethidine, this agent can be used in the treatment of hypertension, but it is no longer a first line agent.

Adverse effects similar to guanethidine.
303. Amphetamine
MOA: (1) it displaces endogenous catecholamines from storage vesicles; (2) it is a weak inhibitor of MAO; (3) it blocks catecholamine reuptake mediated by NET and DAT.

PURPOSE: ADHD and narcolepsy

ADVERSE: hyperension, tachyarrhythmia, Tourette's syndrome, seizure, psychotic disorder w/prolonged use, restlessness, dysphoric mood, rebound fatigue, addiction potential, loss of appetite, irratibility, erectile dysfunction

CONTRA: advanced cardiovascular disease, glaucoma, hyperthyroidism, MAOI therapy, and severe hypertension.
304. Methylphenidate
Methylphenidate is a structural analogue of amphetamine, and is widely used in psychiatry to treat ADHD in children; its major effect is though to be related to enhanced attention.
305. Pseudoephedrine
MAO: Structurally related agents with actions less marked than those of amphetamine

PURPOSE: Allergic rhinitis and nasal congestion

ADVERSE: atrial fibrillation, ventricular premature beats, myocardial ischemia, hypertension, tachyarrhythmia, rebound congestion, insomnia

CONTRA: advanced cardiovascular disease, MAOI therapy and severe hypertension

NOTES: Used as an OTC decongestant; often found in cold remedies and appetite suppressants. Ephedrine and phenylpropanolamine have been restricted in the US.
306. How do inhibitors of catecholamine reuptake work, and what are they?
These agents inhibit NE transporter (NET) mediated reuptake of catecholamine, potentiating catecholamine action.

In other words, they prolong the time that released neurotransmitter remains in the synaptic cleft.

Includes:
1. Cocaine
2. Imipramine
3. Amitriptyline
307. Cocaine
Cocaine is a potent inhibitor of NET; unlike other uptake inhibitors, cocaine essentially eliminates catecholamine transport.

Cocaine is a controlled substance with high abuse potential. It is used occasionally as a local anesthetic but its most important role is as an agent of abuse.
308. TCAs such as imipramine and amitriptyline
MOA: These agents inhibit NET-mediated reuptake of NE into presynatic terminals, and thus allow accumulation NE in the synaptic cleft. The TCAs also inhibit serotonin as well as NE reuptake into presynaptic terminals and blocking serotonergic, α-adrenergic, histaminergic, and muscarinic receptors at therapeutic doses.

PURPOSE: Depression

ADVERSE: Postural hypotension (through α-adrenergic blockage), sinus tachycardia (through potentiation of NE action on cardiac sympathetic nerves), and arrhythmia

NOTES: Although these drugs begin inhibiting NE and serotonin reuptake immediately, there is a latency period of several weeks before improvement in symptoms is seen.
309. What are MAOIs?
MAOIs prevent secondary deamination following reuptake of catecholamines into presynaptic terminals by inhibiting MAO.

Therefore, more catecholamine accumulates in presynaptic vesicles for release during each action potential.
310. What are the non-selective MAOIs (Agents that inhibit both MAO-A and MAO-B)?
Agents that inhibit both MAO-A and MAO-B:

1. Phenelzine
2. Iproniazid
3. Tranylcypromine
311. What are the selective MAOIs?
Clorgyline is selective for MAO-A

Selegiline is selective for MAO-B
312. What are the newer, reversible inhibitors of MAO-A?
1. Brofaromine
2. Befloxatone
3. Moclobemide
313. Purpose of MAOIs

What about Selegiline?
MAOIs are used to treat depression.

Selegiline is also approved for the treatment of Parkinson's disease; its mechanism fo action may include both potentiation of dopamine in the remaining nigrostriatal neurons and decreased formation of neurotoxic intermediates.
314. MAOI contraindications
Patients taking MAOIs should avoid eating certain fermented food containing large amounts of tyramine and other monoamines, b/c MAOIs block oxidative deamination of these monoamines int eh GI tract and liver, causing a hypertensive crisis.

Concomitant use of MAOIs and SSRIs is also contraindicated b/c it may lead to the serotonin syndrome, characterized by restlessness, tremors, seizures, and possibly coma and death.
315. What are α₁-adrenergic agonists?

What are the names of the α₁-adrenergic agonists?
The α₁-selective adrenergic agonists increase peripheral vascular resistance and thereby maintain or elevate blood pressure.

These drugs may also cause sinus bradycardia through activation of reflex vagal responses.

These include:
1. Methoxamine
2. Phenylephrine
3. Oxymetazoline
4. Tetrahydrazoline
316. Methoxamine
MOA: Selectively activates α₁-adrenergic receptors to increase peripheral vascular resistance

PURPOSE: Hypotension, shock

ADVERSE: Bradycardia (vagal reflex), ventricular ectopic beat, hypertension, vasoconstriction, nausea, headache, and anxiety.

CONTRA: Severe hypertension

NOTES: very limited clinical use in the treatment of shock
317. Phenylephrine, oxymetazoline, and tetrahydrazoline
PURPOSE: Opthalmic hyperemia, nasal congestion, hypotension (phenylephrine only)

ADVERSE: Cardiac arrhythmia, hypertension, headache, insomnia, nervousness, rebound nasal congestion

CONTRA: narrow angle glaucoma, severe hypertension or tachycardia (for IV form of phenylephrine)

NOTES: Used in the OTC remedies Afrin and Visine for relief of nasal congestion and ophthalmic hyperemia; rebound of symptoms often accompanies use of these drugs. Phenylephrine is also used intravenously in the treatment of shock.
318. What are α₂-adrenergic agonists?

What drug is the best characterized α₂-agonist?

What are the names of the α₂-agonists?
MOA: These agents selectively activate central α₂-adrenergic receptors, and thereby inhibit sympathetic outflow from CNS.

Clonidine is the best characterized α₂-agonist.

Includes:
1. Clonidine
2. Guanabenz
3. Guanfacine
4. Methyldopa
319. Clonidine
MOA: Selectively activates central α₂-adrenergic receptors, and thereby inhibit sympathetic outflow from CNS.

PURPOSE: Hypertension, opioid withdrawal, cancer pain

ADVERSE: Bradycardia, heart failure, hypotension, constipation, xerostomia, sedation, dizziness

NOTES: Clonidine is used for treatment of hypertension and symptoms associated with opioid withdrawal.
320. Guanabenz, guanfacine, and methyldopa
MOA: Selectively activates central α₂-adrenergic receptors, and thereby inhibit sympathetic outflow from CNS.

PURPOSE: Hypertension

ADVERSE: Bradycardia, hepatotoxcity (methyldopa), autoimmune hemolytic anemia (methyldopa), hypotension, constipation, xerostomia, sedation, dizziness.

CONTRA: MAOI therapy and active liver disease (contraindications for use of methyldopa)

NOTES: Methyldopa is drug of choice for treatment of hypertension during pregnancy
321. What are β-adrenergic agonists?
Stimulation of β₁-adrenergic receptors causes an increase in heart rate and the force of contraction, resulting in increased cardiac output, while stimulation of β₂-adrenergic receptors causes relaxation of vascular, bronchial, and GI smooth muscle.
322. Isoproterenol
MOA: A nonselective β-agonist; it lowers peripheral vascular resistance and diastolic BP (a β₂ effect), while systolic BP remains unchanged or slightly increased (a β₁ effect). It increases cardiac contractility and cardiac output.

PURPOSE: Bronchoconstriction in asthma, arrhythmia, bradycardia (atropine resistant)

ADVERSE: Stokes-Adams seizures, arrhythmias, cardiac arrest, bronchospasm, hypotension, nervousness, tachycardia, angina, nausea

CONTRA: Angina, cardiovascular disease, hyperthyroidism, DM, tachycardia

NOTES: Causes less hyperglycemia than does epinephrine, because the former agent stimulates β-adrenergic activation of insulin secretion. This drug has been mostly supplanted by newer β₂-selective agonists.
323. Dobutamine isomers
MOA: The (-) isomer acts as both an α₁-agonist and a weak β₁-agonist, whereas the (+) isomer acts as both an α₁-antagonist and a potent β₁-agonist. The α₁-agonist and α₁-antagonist effectively cancer each other out when the racemic mixture is administered, and the observed clinical result is that of a selective β₁-agonist.
324. What is dobutamine used for?
This agent has more prominent inotropic than chronotropic effects, resulting in increased contractility and cardiac output.

Dobutamine is used clinically in the acute management of heart failure.
325. What are the other β-adrenergic agonists?
Metaproterenol is the prototype β₂ selective agonist. This drug is used to treat obstructive airway disease and acute bronchospasm.

Terbutaline and albuterol are two other agents in this class that have similar efficacy and duration of action.

Salmeterol is a long acting β₂-agonist -its effects last for about 12 hours.
326. What are α-adrenergic antagonists?
α-adrenergic antagonists block endogenous catecholamines from binding to α₁ and α₂ adrenoceptors. These agents cause vasodilation, decreased BP, and decreased peripheral resistance.

The baroreceptor reflex usually attempts to compensate for the fall in BP, resulting in reflex increases in HR and CO.
327. Phenoxybenzamine
MOA: Blocks both α₁ and α₂ receptors irreversibly.

PURPOSE: Pheochromocytoma associated hypertension and sweating

ADVERSE: Seizure, postural hypotension, tachycardia, palpitations, xerostomia, sedation, miosis, absence of ejaculation

CONTRA: Severe hypotension

NOTES: Used only in preoperative management of pheochromocytoma
328. Phentolamine
MOA: Phentolamine is a reversible, nonselective α-adrenoceptor antagonist.

This drug can also be used in the preoperative management of pheochromocytoma.

ADVERSE: same as phenoxybenzamine

CONTRA: Severe hypotension and coronary artery disease
329. Prazosin
MOA: Selective blocker of α₁-receptors in arterioles and veins; results in decreased peripheral vascular resistance and dilation of the venous vessels. Has little tendency to increase cardiac output and HR.

PURPOSE: Hypertension, BPH

ADVERSE: Pancreatitis, hepatotoxicity, SLE, marked first dose hypotension, palpitations, dyspepsia, dizziness, sedation, increased urinary freq, nasal congestion

NOTES: TCAs may increase postural hypotension. Due to potential severe postural hypotension, the dose is generally prescribed for small quantities around bed time.
330. Terazosin and doxazosin
These agents are similar to prazosin but have longer half-life, allowing less frequent dosing.
331. Tamsulosin
MOA: Specific antagonist of α₁ₐ-receptors.

PURPOSE: BPH

ADVERSE: same as prazosin, except less postural hypotension

CONTRA: hypersensitivity to tamsulosin

NOTES: Tamsulin has more specificity for genitourinary smooth muscle, thus it has lower incidence of orthostatic hypotension.
332. Yohimbe
MOA: Selective blockage of α₂-autoreceptors leads to increased release of NE with subsequent stimulation of cardiac β₁-receptors and peripheral vasculature α₁-receptors.

PURPOSE: organic and psychogenic impotence

ADVERSE: Bronchospasm, nervousness, tremor, anxiety, agitation, increased BP, antidiuresis

CONTRA: Chronic inflammation of sexual organs or prostate, concurrent use with mood altering drugs, gastric and duodenal ulcers, pregnancy, psychiatric patients, renal and liver disease.

NOTES: Also leads to increased insulin release due to blockage of α₂-receptors in pancreatic islets.
333. Propanolol, nadolol, and timolol
MOA: Propanolol, nadolol, and timolol interact with β₁ and β₂ receptors equally and do not block α receptors.

PURPOSE: Hypertension, angina, heart failure, pheochromocytoma, glaucoma

ADVERSE: Bronchospasm, AV block, bradyarrhythmia, sedation, decreased libido, mask symptoms of hypoglycemia, depression, dyspnea, wheezing.

CONTRA: Bronchial asthma or COPD, cardiogenic shock, uncompensated cardiac failure, 2nd and 3rd degree AV block, severe sinus bradycardia

NOTES: Propanolol is extremely lipophilic, its concentration is sufficiently high that sedation and decreased libido may result. An ocular formulation of timolol is used for the treatment of glaucoma.
334. Labetalol and carvedilol
MOA: Labetalol and carvedilol block α₁, β₁, and β₂ receptors (labetalol also acts as a weak partial agonist at β₂ receptors but has a 5-10x greater effect as a β blocker). These drugs decrease blood pressure.

PURPOSE: Hypertension and angina

ADVERSE: Same as propanolol, additionally, labetalol can cause hepatotoxicity - liver enzymes must be monitored
335. Pindolol
MOA: Pindolol is a partial agonist at β₁ and β₂ receptors. The drug blocks the action of endogenous norepinephrine at β₁ receptors and is useful in treating hypertension.

As a partial agonist, pindolol also causes partial stimulation of β₁ receptors, leading to overall smaller decreases in resting heart rate and BP than those caused by pure β atagonists.

***Therefore, the drug may be preferable in hypertensive patients who have bradycardia or decreased cardiac reserve.
336. Acebutolol
Acebutolol is a partial agonist at β₁ adrenoceptors but has no effect at β₂ receptors.

This drug is also used to treat hypertension.
337. Esmolol, metoprolol, and atenolol
MOA: Esmolol, metoprolol, and atenolol are β₁-selective adrenergic antagonists. The elimination half life is the main feature that distinguishes these agents. Esmolol has the shortest (3-4 min), metoprolol and antenolol have intermediate (4-9 hrs).

PURPOSE: B/c of its short half-life, esmolol is used for emergency β-blockade as in thyroid storm. Also used in hypertension, angina, and heart failure.
338. Celiprolol
Celiprolol is a β₁-selective antagonist and β₂-selective agonist.

Also used in treating hypertension, angina, and heart failure.