• 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

1/798

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;

798 Cards in this Set

  • Front
  • Back
  • 3rd side (hint)
Based on Michaelis-Menton kinetics (linear) at the saturation point of a reaction when the enzyme is working as fast as it can (Vmax), what is the only thing that varies

A. Substrate concentration
B. Enzyme concentration
B. Enzyme concentration

Thus using excess substrate one can determine?
Enzyme concentration
NADH absorbs light at what wavelength?
340 nm

How is this useful in measuring enzyme activity?
It makes it easy to measure the formation or disappearance of NADH.

If a reaction does not utilize NAD/NADH then it can be coupled to the reaction (coupled enzyme assay)
What is the purpose of a cofactor?
Cofactors are substances that bind to an enzyme and enhance their activity.

There are inorganic cofactors = zinc, calcium, mag, iron

And organic cofactors, or co enzymes = NAD, protein S, pyridoxine (vit B6), etc.
If a macroenzyme is bound to an antibody it has two effects on the enzyme, name them.
It makes in incapable of functioning

And

Prevents it from being cleared in the blood
Accordingly, awareness of the presence of macroenzymes and procedures for their determination should be an integral part of diagnostic work-up, in order to obviate the use of unnecessary expensive and invasive diagnostic procedures. The patient’s physician should be informed on the presence of a macroenzyme in the patient’s serum. These data should be entered in the patient’s medical records and history form. On the other hand, the patient should be properly reassured that the occurrence of this enzyme form requires no specific therapeutic intervention.
It should be noted that data on the possible diagnostic significance of the macroenzyme occurrence remain scarce, vague and questionable. This fact should not be neglected, but the information on the reasons for the presence of macroenzymes and on the mechanisms of their formation should be continuously collected and reconsidered. Indeed, future research into the formation and detection of macroenzymes may hopefully result in defining some new diagnostic markers.
Noncompetitive inhibition can not be overcome by increasing the substrate concentration. What type of inhibition can be overcome in the way?
Competitive inhibition
An uncompetitive inhibitor alters two parts of the kinetics, name them.
What is the definition of an international unit?
The amount of enzyme that catalyzes the conversion of 1 micro mole of substrate per minute.
Define katal.
1katal = the amount of enzyme that catalyzes te conversion of 1 mole of substrate per minute

1IU= 16.7 nanokatals
The mole is a unit of measurement used in chemistry to express amounts of a chemical substance, defined as an amount of a substance that contains as many elementary entities (e.g., atoms, molecules, ions, electrons) as there are atoms in 12 grams of pure carbon-12 (12C), the isotope of carbon with atomic weight 12. This corresponds to a value of 6.02214179(30)×1023 elementary entities of that substance. It is one of the base units in the International System of Units, and has the unit symbol mol.[1]
The mole is widely used in chemistry, instead of units of mass or volume, as a convenient way to express the amounts of reagents and products of chemical reactions. For example, the chemical equation 2 H2 + O2 → 2 H2O implies that 2 mol of dihydrogen and 1 mol of dioxygen react to form 2 mol of water. The mole may also be used to express the number of atoms, ions, or other elementary entities in some sample. The concentration of a solution is commonly expressed by its molarity, the number of moles of the dissolved substance per liter of solution.
The number of molecules in a mole (known as Avogadro's number) is defined so that the mass of one mole of a substance, expressed in grams, is exactly equal to the substance's mean molecular weight. For example, the mean molecular weight of natural water is about 18.015, so one mole of water is about 18.015 grams. This property considerably simplifies many chemical and physical computations.
The name gram-molecule was formerly used for essentially the same concept.[1] The name gram-atom (abbreviated gat.) has been used for related but distinct concept, namely a quantity of a substance that contains Avogadro's number of atoms, whether isolated or combined in molecules. Thus, for example, 1 mole of MgB2 is 1 gram-molecule of MgB2 but 3 gram-atoms of MgB2.[2][3]
Name the two liver transaminases.
Aspartate aminotransferase (AST/SGOT)

Alanine aminotransferase (ALT/SGPT )

Which one is more specific?
ALT.
Confined mainly to the liver and kidney.
AST is located in what organs (7)?
Cardiac muscle
Liver
Skeletal muscle
Kidney
Brain
Lung
Pancreas

Where in the cell is AST found?
Cytoplasm (20%)
And
Mitochondria (80%)

ALT is only located in the cytoplasm.
Heparin can elevate both liver transaminases to around how many times the baseline?
3x
In RENAL failure liver transaminases become lower or higher than healthy individuals.
Significantly LOWER
AST and ALT are both higher in men and African Americans, hemolysis raises both.

In children AST is slightly higher than ALT, this reverses by age 20.
This may reverse again with old age.
The ratio Of AST:ALT is called
The deRitis ratio.
LDH is present in numerous tissues and is traditionally separable into how many isoenzymes by electrophoresis?
5 isoenzymes
The fastest isoenzymes are LD1 and LD2.

These are found in greatest abundance in what tissues ( 3)?
Heart
Red blood cells
Kidney

There is much more LD1 than 2.
List the isotopes of LDH in order from highest to lowest concentration in normal CSF.
LD1>LD2>LD3>LD4>LD5

1
2
3
4
5
The slowest LDH isoenzymes are what (2)?
And where are they found (2)?
The slowest are LD4 and LD5

They are found in liver and skeletal muscle

Therefore they will be elevated in liver damage.
Where is LD3 found?
Lymphocytes

and

Pancreas
If an isoenzyme "6" is seen on electrophoresis of LDH its presence is thought to be associated with what?
Hepatic vascular insufficiency( usually in the setting of cardiovascular collapse)
The concentration of LDH isoenzymes in normal serum is
LD2>LD1>3>4>5

When LD1 is greater than LD2 three possibilities exist, what are they?
Acute myocardial infarction
Hemolysis
Renal infarction
If LD1and LD5 isoenzymes of LDH are elevated what are two explanations?
1. Acute MI ith liver congestion

2. Chronic alcoholism that has been complicated by liver damage and megaloblastic anemia
The best test to confirm that an elevated alkaline phosphatase is from the biliary tree?
Gamma-glutamyl transferase or GGT
Where is gamma-glutamyl transferase (GGT) found
In the biliary epithelial cell, particularly those that line the small inter lobular bile ducts and bile ductules.

Thus, it is exquisitely sensitive to biliary injury.

GGT can also be elevated in (6)?
Steatosis
Diabetes
Hyperthyroid
Rheumatoid arthritis
Acute MI
COPD
GTT is found to be around 2-3x the upper limit in heavy drinkers.

How long after abstaining from alcohol will levels be normal?

This is a good marker of alcohol consumption.
3 weeks

Note: GGT is lower in women and higher if African-Americans
GGT is a great marker to confirm that alkaline phosphatase is from the billiard tree, due to GGT in the biliary tree.

However, GGT is also in the smooth ER of hepatocytes. So GGT can be elevated due to excess toxins. In particular certain drugs, name a few (6).
Warfarin
Barbiturates
Dilantin
Valproate
Methotrexate
Alcohol
What is an alternative to GGT that is also found in the biliary epithelium.
It's highest levels are in cholestatic conditions.

It is specific, but not sensitive.mso it is stil second to GGT.
5'-Nucleotidase
Hyperammonemia is nearly always due to what?
Liver failure.

However, in children one should suspect?
An inborn error of metabolism

Especially urea cycle enzyme deficiencies
The sources, name two, of the body are?
Skeletal muscles and gut

Bacterial in the GI tract breakdown protein and release ammonia. The liver removes the ammonia and discards it in the form of urea in urine.
Name causes of elevated ammonia (3).
Hepatic yet dysfunction
Too much collateral circulation
Excess protein in the gut ( for example, excess hemoglobin from a bleed)

Ammonia is neurotoxic.
Ammonia measurement requires?
A fresh blood sample, chilled during transport, and no hemolysis.

Patients should also refrain from smoking.
There are two types of phosphatases: alkaline (optimum pH is 9)
and acid ( pH optimum is 5)

Acid phosphatases are found in greatest concentrations where (3 places)?
Prostate
Red cells
Bone

Red cell phosphatase is distinguished from other Acid phosphatases by?
It's susceptibility to inhibition by 2% formaldehyde and resistance to inhibition by tartrate.

This is the same tartrate-resistant acid phosphatase (TRAP) that is found in hairy cell lukemia.

Non-TRAP (or prostate acid phosphatase) has been used as a serum marker for prostatic adenocarcinoma, but has been largely replaced.
Alkaline phosphatase ( pH 9) is produced by four tissues, name them.
Bone
Bile ducts
Intestine
Placenta

Therefore elevated alk phos is seen in biliary disease, bone disease, bone growth ( children), and pregnancy.

Separate reference ranges are needed in children and pregnancy.

When do we see decreased levels (2)?
Hypophosphatasia ( an inborn deficiency)
And malnutrition
The regan isoenzyme is alk phos is observed in 5% of individuals with what condition?
Carcinoma

It appears to be identical to placental alk phos.
Bone alkaline phosphatase is produced by what cell type?
Osteoblasts

And reflects bone-forming activity.
It is important to get a fasting alkaline phosphatase for what reason?
Intestinal alk phos can falsely elevate levels. Especially in Lewis positive type B or O secretors.

Ingesting a meal can elevate alk phos by 30% for 2-12 hours.
Which type of bilirubin is water-insoluble?
Unconjugated
Another name for unconjugated bilirubin is:
Indirect
Which type of bilirubin is produced directly from the breakdown of heme?
Unconjugated/Indirect bilirubin
The process that occurs in the liver to cause bilirubin to be water-soluble is called?
Glucuronidation
Which type of bilirubin is secreted in bile?
Conjugated/Direct
What effect does intestinal bacteria have on bilirubin?
Conversion to Urobilinogen

(which is yellow and gives stool/urine color)
A patient with a cholangiocarcinoma causing complete biliary obstruction may demonstrate what characteristic stool finding?
White, acholic stools

(Siver stool of Thompson)--how lame to have your eponym be about poop
Bilirubinuria is always associated with which type of hyperbilirubinemia?
If a patient has bilirubinuria, then the elevated bilirubin is CONJUGATED b/c indirect bili is not water soluble.
A patient presents with jaundice and bright yellow urine that is 3+ positive for bilirubin on dipstick. A likely clinical scenario that could accompany this is:
a) Gilbert syndrome
b) Choledocholelithiasis
c) Crigler Najjar syndrome
d) loss of hepatocyte function due to drug effect
b) A stone obstructing the bile duct is the only scenario listed that will cause conjugated bilirubinemia.

Crigler Najjar (failure of conjugation in hepatocyte), Gilbert (failure of hepatocyte to take up unconjug bili), and loss of hepatocyte function all cause indirect/unconjugated bilirubinemia
Which of the bilirubin measurements is calculated, not measured?
Indirect (Unconjugated): remember-this is the early stuff.
The diazo-colorimetric method of hemoglobin measurement uses which of the following to calculate findings:
a) temperature
b) light scatter
c) Light transmisison at 540 nm
d) Turbidity
e) Color dye reaction
Color dye reaction.
What is purpose of adding an accelerant to the diazo-colorimetric method?
Without accel, you measure mainly conjugated. With addition of accelerant, you maeasure conjug + unconjug (total). Subtract conjug from total and get (estimated) unconjugated.
One more time:

Conjugated is to _________ as Unconjugated is to _______

a) direct, total
b) indirect, direct
c) direct, indirect
d) total, indirect
Remeber: conjugated/unconjugated has to do with how the bili is in the body, while direct/indirect is the method of measurement. Indirect (Unconjugated) is the one you calculate *indirectly*

It's nice, because Indirect and Unconjugated both have prefixes, so you can remember they go together.
Measuring absorbance by direct spectrophotmetry at 455nm can determine what?
Total bilirubin
What is the limitation of measuring bilirubin by direct spectrophotometry and how do we correct for this?
Interference by hemoglobin can occur so we simultaneously measure hemoglobin level at its other peak and subtract them to get real answer
What is delta hemoglobin?
With prolonged elevation, conjugated bilirubin can actually bind to albumin (delta-bili). This sticks around longer, as it cannot be excreted by liver/kidney. Makes it look like hyperbili is still present.
A patient with alcoholic cirrhosis and jaundice would be expected to have what changes in below parameters:

Type of hyperbili, AST/ALT, Alk Phos, Cholesterol, Pruritis
Some unconjugated, but some conjugated; really high AST/ALT, not super-high Alk Phos, Normal cholesterol, no pruritis
A patient with a bile duct obstruction would be expected to have what changes in below parameters:

Type of hyperbili, AST/ALT, Alk Phos, Cholesterol, Pruritis
Almost all conjugated, Not super-high AST/ALT, really high Alk Phos, Increased Cholesterol, Itching +
What is the half-life of Factor VII?
12 hours
A patient presents to an ER without CT or ultrasound equipment with jaundice and elevated PT. Before the workup can reveal whether he has cirrhosis or a bile duct obstruction, an simple medication can be given to help discern. What is it?
Parenteral Vitamin K.

If the patient has cirrhosis, then Vit K should not help correct the factor levels much. If the patient has a stone, then the PT prolongation is likely due to a lack of absorption of Vit K (lack of bile salts) and should correct with IV Vit K.
What pattern of gamma globulins do you expect to see in autoimmune hepatitis?
polyclonal IgG increase
What pattern of gamma globulins do you expect to see in primary biliary cirrhosis?
polyclonal IgM increase
What pattern of gamma globulins is common in all liver diseases?
Impaired hepatic synthesis of protein with increased immunoglobulin synthesis results in an albumin to globulin ration <1.
What leads to so-called physiologic jaundice of the newborn? What type of hyperbilirubinemia is it?
Unconjugated hyperbilirubinemia. The gut lacks the bacteria to convert Conjugated Bili to urobilinogen and it can't be excreted via stool. Conjug bili is thus converted back to Unconjug by beta-gluconaridase in gut. Fetal clearance of the Unconjug bili is via placenta. At birth, if the baby cannot pass enough bili through stool then becomes jaundiced. Also, hepatic enzymes aren't full-force yet.
Name the following characteristics of neonatal jaundice:
a) time of onset
b) time of peak
c) peak level and rate of increase
a) onset at day 2-3 days
b) peaks at 4-5 days
c) peak of 5-6 mg/dL (usually) and at a rate of <5mg/dL/day
Two common causes of marked elevation in newborn bilirubin:
Hemolytic disease of newborn, sepsis
What is the CNS damage caused by hyperbilirubinemia termed? Why does this happen in infants?
Kernicterus

Underdeveloped blood-brain barrier
A 7 day-old infant dies after severe hyperbilirubinemia. At autopsy, the cause is found to be due to biliary atresia. In addition, what CNS findings would you see?
Yellow-stained subthalamic nucleus, hippocampus, thalamus, globus pallidus, cerebellar nuclei, and cranial nerve nuclei.
Features in neonatal jaundice that raise the concern that it is not "physiologic:"

Timing of appearance and peak, length of jaudice, levels, type
Appearance at <24 hours, rising bili past 1 week, persistance of jaundice past 10 days, level >12mg/dL, increase >5mg/dL/day, conjugated bili >2mg/dL. (Remember, Physiologic jaundice is more unconjugated--due to crappy liver enzymes).
A newborn with biliary atresia develops jaundice. What type of jaundice is most likely, and why will phototherapy not work?
The bilirubin is most likely conjugated (it's already been through the liver), and phototherapy works by converting unconjugated bili to conjug bili for water solubility and excretion.
When should phototherapy be considered in the treatment of neonatal jaundice?
As age increases, the threshold level increases. But it basically starts at 10mg/dL at 12 hours then increases 2mg/dL for each 12 hours of life.
When should exchange transfusion be considered in the treatment of neonatal jaundice?
>20mg/dL
Timing and type of hyperbilirubinemia related to:
Physiologic jaundice of Newborn
>24 hours but less than 10 days; unconjugated
Timing and type of hyperbilirubinemia related to:
Breast milk Jaundice
>7 days; unconjugated
Timing and type of hyperbilirubinemia related to:
Polycythemia, Hemolytic disease of newborn, erythroblastosis
<24 hours, unconjugated
Timing and type of hyperbilirubinemia related to:
Hemoglobinopathies or RBC enzyme defects
>7 days, unconjugated
Timing and type of hyperbilirubinemia related to:
Bowel obstructions like CF, Hirschsprung, ileal atresis
>7 days, unconjugated

(*unconjugated* because the gut bacteria don't see the conjugated bili to help it get changed to urobilinogen and excreted, but the enzyme β-glucuronidase is present to "unconjugate" the bili to get it reabsorbed.
Timing and type of hyperbilirubinemia related to:

Inherited disorders of bilirubin metabolism

Extra: what are these disorders? (2)
Unconjugated, >7days

Crigler-Najjar, Gilbert
Timing and type of hyperbilirubinemia related to:

Biliary obstruction
Conjugated, >7days
Timing and type of hyperbilirubinemia related to:

TORCH infections

Extra: What are they?
<24 hours, conjugated

Toxo, Rubella, CMV, Herpes, Others (Coxsackievirus, Syphilis, Varicella-Zoster Virus, HIV, and Parvovirus B19)
Timing and type of hyperbilirubinemia related to:

Metabolic disorders such as storage diseases
>7days, conjugated
Timing and type of hyperbilirubinemia related to:

Disorders of bilirubin transport

Extra: What are they? (2)
Conjug, >7 days

Rotor, Dubin-Johnson
Timing and type of hyperbilirubinemia related to:

Allagille Syndrome
Conjugated, >7days
What is the main use of transcutaneous bilirubin measurements?
Screening otherwise healthy newborn infants for elevations of bilirubin to evaluate those at risk for jaundice.
In regards to drug-induced hepatitis, when do most events occur in relation to the drug dosage?
Within first 4 months of administration.
Which type of viral hepatitis is the most likely to become chronic?
HCV
What type of acute hepatic injury will raise the AST/ALT the most?
Ischemic or toxic injury
What level of AST will give you a 90% chance that the etiology of injury is toxic?
>3,000 U/L
A patient presents with AST of 600 U/L and an ALT of 600. Is the cause more likely alcohol or viral?
Viral. Viral infections often raise the AST to greater than 10x the upper limit of normal, while EtOH rarely does this.
A patient presents with AST of 300 and ALT of 145. Is the cause more likely alcohol or viral?
Alcohol. EtOH often raises the AST 2x's higher than the ALT. Viral patterns are usually closer to 1:1.
Which patients are more likely to have jaundice? Hep A, Hep b, Hep C, EtOH
Hep A, EtOH
Which levels correct first, bili or transaminases?
Transaminases
What is probably the best indicator of prognosis in acute hepatic injury?
PT >4.0s
What two isoenzymes make up serum amylase?

How do we differentiate?
Salivary and pancreatic amylase

Electrophoresis, inhibition, or monoclonal antibodies
If you put amylase on an electrophoresis gel how many bands do you see? Which are pancreatic and which are salivary?
6, first three are salivary, second 3 are pancreatic
How can we differentiate between salivary and pancreatic amylase using inhibition tests?
Salivary amylase is inhibited by wheat germ lectin.
When do serum amylase levels rise and normalize in acute pancreatitis?
Rise within 2-24 hours, fall to normal in 2-3 days.
Do high levels of amylase mean anything in the diagnosis of pancreatitis? What about prolonged elevation?
High levels don't reflect severity, but do increase liklihood that pancreatitis is the cause. Prolonged elevation suggests pseudocyst formation or complication
Why do most pancreatitis patients also have increased urine amylase?
The amylase is normally cleared by glomerulus. (Keep in mind--kidney failure patients might have increased plasma amylase...) Can calculate fractional excretion of amylase.
A 42 year-old gentleman presents with severe acute epigastric pain, nausea, and "stranding" (evidence of inflammation) of the pancreas on CT scan. Pancreatitis is the top of the differential, but serum amylase is barely elevated. What could be the cause?
If his pancreatitis is caused by hypertriglyceridemia, TGs may interfere with measure of amylase assay.
Name 10 non-pancreatic causes of hyper-amylasemia:
diabetic ketoacidosis, peptic ulcer disease, acute cholecystitis, ectopic pregnancy, salpingitis, bowel ischemia, intestinal obstruction, macromaylasemia, renal insufficiency, opioid administration (?)
What is macroamylasemia? What is the incidence?
What is the cause?
1% incidence where apparently healthy individuals have elevated amylase and low urine amylase due to Ig-amylase complexes that are not cleared.
Is lipase specific for the pancreas?
Yes
When does lipase rise and fall in acute pancreatitis?
Rises like amylase in 2-24 hours of onset and remains elevated for 14 days.
How is lipase better than amylase for pancreatitis diagnosis?
Less renal clearance, so more predictable. Only pancreatic form exists.
What is Ranson criteria used for in treatment of pancreatitis?
Predict severity

Note: Lipase/Amylase levels do not predict severity.
What two time frames are used to evaluate Ranson criteria?
Admission and 48 hours following
What criteria are used for Ranson criteria at admission?
Age, WBC, Glucose, AST, LDH
What criteria are used for Ranson criteria at 48 hours?
Urea, Calcium, PaO2, Base deficit, fluid sequestration, hematocrit
Most common causes of acute pancreatitis? (2)
Alcohol and Gallstones
What are some other causes of pancreatitis besides EtOH and gall stones?
Hypertriglyceridemia, Hypercalcemia, Viral, inherited, CF, scorpion bite
secretin-cholecystokinin (CCK) test is for
pancreatic exocrine function
endoscope is introduced and the duodenal concentrations of pancreatic exocrine products (bicarbonate, amylase, lipase, trypsin) are measured after the intravenous administration of secretin and CCK
fecal fat test is for
pancreatic exocrine function, other causes of positive results
Severe ileal diseases (eg, Crohn disease) or ileal resection
which is a better test, fecal chymotrypsin (bentiromide) or fecal elastase-1?
The sensitivity and specificity of elastase-1 appears superior to that of chymotrypsin.
Is the D-xylose test a measure of the pancreatic exocrine function?
no, D-xylose test is a measure of small bowel mucosal absorptive capacity
Which type of pancreatic cyst: Associated with pancreatitis, ovoid unilocular lesion with thick wall adjacent to pancreas
pseudocyst
Which type of pancreatic cyst: ↑Amylase ↓CEA ↑CA 19-9
pseudocyst
Which type of pancreatic cyst:
↓Amylase
↓CEA
↓CA 19-9
Serous cystadenoma or Solid-cystic (solid-pseudopapillary tumor)
Which type of pancreatic cyst:
↓Amylase
↑CEA
n1-↑CA 19-9
Mucinous cystadenoma (mucinous cystic neoplasm) or Intraductal papillary mucinous tumor
In normal subjects serum CK is of which isoform?
MM, which comes from?
skeletal muscle
CK-BB (CK1) is found primarily in the?
Brain
CK-MB (CK2) is found in which tissue(s)?
Cardiac muscle is about 30% MB, skeletal about 1% MB.
What is the ‘relative index’ that adds to the ability of the CK-MB assay to distinguish myocardial infarction?
serum, CK-MB/total CK, 2% is the cutoff
The immunoinhibition type of CK-MB immunoassay is susceptible to falsely elevated CK-MB due to
hemolysis, the presence of CK-BB (in stroke), and macro-CK.
Which type of serum CK is elevated in stroke?
CK-BB
What is macro-CK?
a CK-Ig complex. On electrophoresis, it migrates between MM and MB. It is found in completely healthy elderly women.
What is the significance of Mitochondrial CK?
It is seen in patients with advanced, often disseminated, malignancies and is associated with a poor prognosis.
Are assays for CK isoforms better than CK-MB?
Assays for CK isoforms are not widely available, and published reports do not appear to display significant advantages over CK-MB.
Why there is both an immediate release of cytoplasmic troponin (4-8 hours) and a sustained release (10-14 days)?
While a small proportion of cardiac muscle troponin is free in the cytoplasm, the vast majority of it is bound to actin and myosin.
What is the most sensitive of the cardiac markers and is the earliest marker of acute myocardial infarction?
Myoglobin (Mgb)
why the Altered Cobalt Binding (ACB) assay shows promise as a marker of transient ischemia?
Ischemia-modified albumin has altered Cobalt Binding. It rises within minutes of myocardial ischemia and returns to baseline within 6 hours due to rapid hepatic clearance.
What is the value of CRP in acute coronary syndrome?
C-reactive protein (CRP) and high-sensitivity CRP (hsCRP) appear to be strong predictors for the development of ACS in healthy individuals, and to predict short-term prognosis following non-AMI ACS.
the sensitivity of a single troponin test at 4 hours after onset of symptoms is
50%, how to increase sensitivity?
two tests at 6 and 12 hours have a combined sensitivity of approaching 100%.
If the cardiac troponin I is so sensitive and specific, why are we still measuring CK-MB?
it is still important to measure serial CK-MBs to determine:
(1) whether the MB is going up or down; that is, whether the infarct is acute or resolving, and
(2) whether the MB comes down appropriately; that is, whether there is a complication of MI such as extension of the infarct
How does serum CK-MB level change during an MI?
Increased serum CK is detectable within 3–6 hours of an AMI, with a peak at 20 to 24 hours. Assuming there is not ongoing injury, CK returns to normal within 72 hours.
How does serum cardiac markers change after a successful reperfusion?
In successful reperfusion, all of the cardiac markers peak earlier than normal; although, their sequence of peaks parallels that of normal MI.
What is the Biuret technique for?
for determining total protein, how does it work?
In an alkaline medium copper salts form a purple complex with proteins.
Proteins absorb ultraviolet light at which wave length?
210 and 280 nm
What are the bands on serum protein electrophoresis?
albumin, α1, α2, β and γ.
What will happen if albumin is congenitally absent?
hyperlipidemia and mild edema
Which is the fastest migrating protein in SPEP?
Prealbumin
What are major functions of prealbumin?
It binds to T3, T4 (transthyretin) and vit A complex
Elevations of prealbumin are seen in what conditions?
Chronic alcohol abuse, Steroid therapy
Elevations of alpha2 macroglobulin are seen in what conditions?
Liver disease, nephrotic syndrome
Decreased Ceruloplasmin is seen in which condition?
Wilson disease,hepatic failure, Menke's syndrome
Which protein is decreased in acute intravascular hemolysis?
Haptoglobulin
Transferrin can cross blood brain barrier as?
tau-protein ( modified as asialated transferrin), normal transferrin
what is the half life of prealbumin?
48 hrs
CSF protein electrophoresis will show?
Sharp prealbumin band, double transferrin peaks,dim albumin
In what conditions fibrinogen is present in serum?
Dysfibrinogenemia,APL syndrome, heparin, vit K def
Fibrogen appears at which location in SPEP?
gamma and beta interface, may be misinterpreted as M protein
Beta-gamma bridging is seen in which condition?
Liver cirrhosis
from which organ is CRP produced?
liver
what fraction of the population has CRP > 2mg/L?
1/2
what fraction of the population has CRP > 3mg/L?
1/3
Does the population distribution of CRP follow a Gaussian curve?
No, It is a curve significantly skewed, with a dense cluster in the very lowest CRP levels and a long tail extending into the > 10 range.
What the cutoff values for the three levels of CRP level?
Normal <3mg/L, low-level elevation 3-10, high level >10.
What is the high level CRP indicative of?
acute inflammation
Low level elevations of CRP are predictive of (2)?
1. poor outcomes of cardiovascular events;
2. also correlated with mortality in non-cardiac diseases as well as in apparently healthy individuals
Why is transferrin increased in an individual who does not have iron deficiency (2)?
1. Pregnancy, 2.estrogen therapy
Which form of serun transferrin may be superior to GGT as a marker for alcohol use?
carbohydrate-deficient transferrin
How many transferrin peaks do we observe in the CSF?
Two peaks. Why?
transferrin and asialated transferrin (Tau protein)
Is transferrin an acute phase reactant?
no. It decreases in the acute phase, but later increases if the inflammation persists.
What's the characteristic feature of serum protein electrophoresis in monoclonal gammopathy? (1)
discrete, dark band (M-spike) usually in gamma region but can be seen in beta or alpha 2 regions.
What is a monoclonal gammopathy and what's a another name for it?
condition in which there's an immunochemically homogenous immunoglobulin in the serum. Another name is paraproteinemia
What are some causes of a mononclonal gammopathy? (5)
multiple myeloma, solitary plasmocytoma, MGUS, Waldenstroms macroglobulinemia, CLL/SLL
How often do biclonal gammopathies occur and what's the cause?
3-4% of cases, usually composed of IgA spikes having a single light chain, should be considered monoclonal
What percentage of multiple myeloma patients have a hypogammaglobulinemia and what other finding are they likely to have?
About 10%, they're more likely to have Bence-Jones protein (free light chains) in their urine
When is immunofixation or immunosubstraction when the SPEP is negative for an M-protein? (3)
When there's a strong clinical suspicion for multiple myeloma, there's systemic amyloidosis, or there's hypogammaglobulinemia
What method is used to characterize the M-spike when it's found on SPEP?
immunofixation or immunosubstraction
What is the usual composition of an M protein?
an intact immunoglobulin composed of 2 heavy and 2 light chains. Sometimes a light chain only and rarely a heavy chain only.
What's the connection between systemic AL amyloidosis and monoclonal gammopathy?
AL amyloidosis can occur when the M-protein produced has unusual (amyloidogenic) properties
What test is used to monitor monoclonal gammopathy disease progression or efficacy of treatment?
serum electrophoresis
How is M protein quantification done and why is it useful? (2)
carried by nephelometry (or turbidimetry) and used to classify disease and and follow disease
Why are major immunoglobulins quantitated in multiple myeloma patients?
to determine the degree of suppression
Why is the quantity of serum free light chains important in monitoring multiple myeloma? (3)
useful to follow individuals with light chain only disease, detect abnormalities in the kappa:lambda, and is extremely sensitive in detecting disease recurrence
When does hyperviscosity syndrome develop, what are the symptoms (4) and when should serum viscosity be measured?
-syndrome develops when serum viscosity is <3 cp -symptoms: nasal bleeding, blurred vision, retinal vein dilation (flame shaped retinal hemorrhages), and neurologic symptoms - serum viscosity should be measured when IgM M-protein > 4 g/dL or IgA/IgG > 6 g/dL
What else should be looked for in patients with M proteins?
cryoglobulinemia
What's the difference between CSF and serum proteins?
CSF normally has all of the same proteins but in smaller quantities
What are the most characteristic features of normal CSF electrophoresis? (3)
prominent pre-albumin band, double beta transferrin) band, dim albumin and alpha2 band
What's the main purpose of CSF electrophoresis?
support diagnosis of multiple sclerosis (oligoclonal bands) these bands should be absent from the patient's serum in a sample run concurrently
What's the utility of urine protein electrophoresis in proteinuria?
to determine the source of the protein
What is a glomerular proteinuria pattern in urine electrophoresis and what type of renal pathology does it suggest?
urine contains strong albumin, alpha1 and beta bands. Indicates glomerulonephritis where very large and very small proteins don't enter the urine (some preservation filtering and tubular resorption)
What is a tubular proteinuria pattern in UPEP and what renal pathology does it suggest?
urine has a weak albumin band, strong alpha1 and beta bands. Implies tubulointerstitial nephritis or acute tubular necrosis (impaired resorption of smaller proteins, normal glomerular filtration of large proteins)
What is an overflow proteinuria pattern in UPEP and what renal pathology does it suggest?
most commonly a monoclonal light chain (Bence Jones) but can also be due to myoglobin and hemoglobin. Implies a serum protein in excess of kidney's filtration/resorption capacity leading to overflow
Define cryoglobulins and what's the method to detect them?
-defined as immunoglobulins that precipitate reversibly at low temperatures. -to detect blood drawn,kept, clotted, and centrifuged at 37C, remaining serum stored at 37C (for at least 3 days) then centrifuged at 4C any precipitate is a cryoglobulin
What are the 3 types of cryoglobulinemia recognized?
Type I - monoclonal immunoglobulins Type II - mixture of monoclonal IgM and polyclonal IgG Type III - mixture of 2 polyclonal immunoglobulins
What is the most common clinical condition associated with cryoglobulins? Name 4 other less common conditions
#1 is hepatitis C infections others include lymphoproliferative diseases, chronic infections, autoimmune diseases (esp SLE), chronic liver diseases
What are the clinical manifestations of cryoglobulinemia? (7)
palpable purpura (constant feature usually over lower extremities), arthralgias, hepatosplenomegaly, lymphadenopathy, anemia, sensorineural deficits, and glomerulonephritis
What are the renal complications of cryoglobulinemia and when do they usally manifest?
nephrotic or nephritic syndrome with type II membranoproliferative glomerulonephritis on renal bx. Renal involvement trails by 4-5 years
What's the basic pathologic lesion in cyroglobulinemia?
vasculitis with large subendothelial immune complex deposition with a fibrillary or tubular structure on electron microscopy
What's the goal and method of treatment of cryoglobulinemia? What's the success rate?
Goal is immune modulation methods include alpha interferon, corticosteroids, and plasmapheresis. Remisssion achieved in 75% but 50% relapse
Define hyponatremia
serum sodium <135 mmol/L, considered severe when <120 mmol/L
Under what clinical conditions is the diagnosis of SIADH most accurate? (5)
when hyponatremia is irrefutable, urine sodium is >20 mmol/L, patient is euvolemic and has normal renal function, there's been no recent diuretic use, normal adrenal function
What are the possible complications of correction hyponatremia too quickly? Too slowly?
Central pontine myelinosis, cerebral edema
What is pseudohyponatremia and what are 3 possible causes?
serum osmolarity >280, may be due to hyperglycemia, hyperlipidemia, or hyperproteinemia
What's the differential for hyponatremia when patient is hypovolemic? (5)
Renal losses (urine Na+ >30 mmol/L), diuretics, medullary renal disease, Addison, and RTA type I
What's the differential for hyponatremia when patient is euvolemic? (3)
SIADH, psychogenic polydipsia, and drugs with ADH-like effect
What drugs are known to have an ADH/vasopressin n-like effect? (4)
desmopressin, SSRIs, TCAs, ectasy
What's the differential for hyponatremia when patient is hypervolemic? (3)
congestive heart failure, cirrhosis, nephrotic syndrome
Under what conditions is hypernatremia usually found?
A dehydrated person with physical inability to respond to their thirst response.
What's the differential diagnosis for hypernatremia?
iatrogenic sodium administration or water loss
What is the differential diagnosis for hypokalemia? (3)
GI losses, renal losses, transcellular shifts
What urine K+ value characterizes hypokalemia due to GI loss and what are some possible etiologies? (4)
Urine K < 30 mEq/day vomiting, NG tube, diarrhea, and villous adenoma
What urine K+ value characterizes hypokalemia due to renal loss and what are some possible etiologies? (9)
Urine K > 30 mEq/day diuretics, hypomagnesemia, antibiotics, mineralocorticoid excess, RTA types I and II, Cushing syndrome, congenital adrenal hyperplasia, and hyperrenism
What's the possible etiology of transcellular shifts leading to hypokalemia? (2)
alkalosis, correction of diabetic ketoacidosis
What's a complication of correcting diabetic ketoacidosis?
profound hypokalemia unless supplemental K is given
What is pseudohyperkalemia?
elevated potassium levels in absence of clinical evidence of hyperkalemia (doesn't need to be treated)
What's a clue that hyperkalemia is due to in vitro release from red cells?
visible hemolysis in the tube
What chromosomal abnormality has been associated with passive transmembrane leak of potassium from red cells, without concomitant hemolysis?
autosomal dominant abnormal gene on chromosome 16 (familial pseudohyperkalemia)
What's the differential diagnosis for hyperkalemia? (6)
acidosis, renal failure, potassium sparing diuretics, adrenal insufficiency, iatrogenic, rhabomyolysis
Nearly all case of acidosis are associated with hyperkalemia except? (2)
RTA types I and II in which K+ is low
What electrolyte abnormalities characterize primary hypeparathyroidism? (4)
increased Ca2+, decreased PO4, increased Cl-, and increased nephrogenous cAMP
What's the effect o f PTH on renal tubules? (2)
increasing Ca2+ resorption while increasing phosphate excretion
What's the differential diagnosis of primary hyperparathyroidism? (3)
parathyroid adenoma (most common), 4-gland hyperplasia, parathyroid carcinoma
What is tertiary hyperparathyroidism?
hyperparathyroidism that persists in post-renal transplant patients (parathyroids become autonomous)
T/F hypercalcemia of malignancy is always caused by bone metastases
F- can also occur in absence of bone metastases (humor hypercalcemia of malignancy)
What is the most common mediator of humoral hypercalcemia of malignancy?
PTH related protein (PTHrp)
What lab findings are most suggestive of hypercalcemia of malignancy?
increased nephrogenous cAMP in the presence of normal PTH
What is the gene associated with familial hypocalciuric hypercalcemia?
CASR gene on 3q
What drugs are associated with hypercalcemia? (4)
thazides, calcium containing antacids, calcium supplements, hypervitaminosis
What endocrine disorders are associated with hypercalcemia? (3)
hyperthyroidism, Addison, acromegaly
What causes hypervitaminosis D and what is its effect on the kidneys?
Caused by long term ingestion of vitamin supplements. Vit D enhances resorption of both calcium and phospate in the kidney (can lead to calciphylaxis)
What granulomatous disease is associated with hypercalcemia and why?
Sarcoid, the histiocytes of sarcoid have the capacity to activate vit D to the active form (1,25 di-hydroxy vitamin D)
What are the clinical signs and symptoms of hypercalcemia? (8)
nephrolithiasis, lethargy, hypo-reflexia, slowed mentation, nausea, vomiting, constipation, depression, and high peaked T waves on EKG
What is calciphylaxis and what electrolyte abnormalities are associated with it?
metastatic calcification of vessel walls and soft tissue, occurs when long term hypercalcemia and hyperphosphatemia occur concomitantly
What forms of PTH are modern immunoassays able to detect? (4)
Intact PTH (84 amino acids) N-terminal PTH, mid terminal PTH, and C-terminal PTH (all three of which are breakdown products produced in the parathyroids and in the blood)
Which parathyroid forms have biological activity? (2)
intact and N-terminal PTH
Parathyroid Hormone (PTH) forms
Biologic Activity Intact PTH + N- terminal PTH + Mid- region PTH - C-terminal PTH -
What are the effect of PH change on ionized calcium measurement?
Acidosis Increase Alkalosis Decrease
What are the characteristics of the sample for ionized calcium measurement(5)?
1) Must be from an artery 2) Must not be exposed to the air ( metabolism will change the PH) 3) Must not be drawn in calcium-chelating anticoagulants (EDTA, citrate) 4) Avoid prolong tourniquet 5) Must be kept cool and deliver to the lab rapidly
What is the rule- of- thumb correction for Ca measurement in hypoproteinemia?
0.8 mg/dl Ca per 1 g/ dl protein lost.
Which tumors can secret PTH- related protein (PTHrP) (6)?
SCC lung, head & neck, skin, cervix, esophagus and breast carcinoma.
Secondary hyperparathyroidism
Is due to peripheral resistance to the action of PTH (comonly seen in chronic renal insufficiency or vit.D deficiency), the Ca is low while the PTH is normal or high. It produces activation of osteoclasts, leading to brown tumors of bone (renal osteodystrophy).
The effect of hypomagnesemia on PTH
Acute hypomagnesemia cases increased PTH secretion but persistent or marked hypomagnesemia inhibits PTH secretion
EKG changes in hypocalcemia(3)
Lengthening of the QT interval, low voltage T waves, and dysarythmias
D.D of Hypocalcemia (9)
1) Vit. D deficiency 2) CRF 3) Drugs ( Heparin, glucagon, diuretics, aminoglycosides) 4) Hypoparathyroidism 5) Medulary thyroid cancer 6) Hypoproteinemia 7) Hyperphosphatemia 8) Pancreatitis 9) Massive transfusion
Anion gap
AG= Na - [ Cl + Hco3 ] Normal <12 Hypoalbuminemia can mask the anion gap Corrected anion gap= Na - [Cl + Hco3] + 2.5 (4 - albumin)
Low anion gap causes (6)
Paraproteinemia, hypoalbuminemia, hypermagnesemia, hypercalcemia, Lithium therapy, and hypophosphatemia
Causes of metabolic acidosis with increased Anion Gap (7)
Methanol, uremia, ketoacidosis( diabetic, starvation or Etoh), paraldehyde, lactic acidosis, ethylene glycol, salicylate
Causes of metabolic acidosis with normal Anion Gap (7)
RTA, diarrhea, recovery phase DKA, uterosigmoidostomy, NH4Cl, TPN, carbonic anhydrase inhibitors
" BUN always underestimate GFR" False or True?
True. Urea is freely filtered and partially reabsorbed by the nephrone; this reabsorption has a consequence that BUN always underestimate GFR. Reabsorption increases with hypovolemia, thus causes even more BUN understimation
Creatinine underestimate GFR. False or True?
False. Creatinine is freely filtered through the glomerulus, also a small amount of Creatinine is secreted by the tubules. Thus creatinine slightly overestimates GFR
simplified GFR calculation formula
CLcr = Ucr x Vur / Pcr,
Ucr: urine creatinine
Vur: volume of urine
Pcr: plasma creatinine
The inverse relationship between GFR and creatinins is more linear in higher GFR. True or False?
False. This relationship is more linear , when GFR is about half normal. Mild to moderate degrees of GFR impairment do not cause appreciable increase in the creatinine concentration.
The Modificiation of Diet in Renal Disease (MDRD) formula for estimated GFR (eGFR) includes which variables?
creatinine, age, sex, race
True or False: Azotemia refer to a very high level of nonprotein nitrogen in the serum.
False. It's BUN, not NPN.
True or False: acidemia refers to arterial PH <7.0
False, it's arterial pH<7.4
True or False: alkalemia refer to arterial pH>7
False, it's 7.44.
Normal range of arterial blood pCO2
40-44 mmHg
Normal range of arterial blood bicarbonate
24-28 mEq/L
Normal range of anion gap
3-10
Normal arterial PaO2
90-100 mmHg
Normal arterial O2 saturation
95%-98%
Respiratory acidosis is due to
too little elimination of CO2 by the lungs (hypoventilation).
Respiratory alkalosis is due to
too much elimination of CO2 by the lungs (hyperventilation).
True or False: Compensation in primary respiratory disorders involves buffers and alterations in pulmonary handling of CO2.
False, It's the renal handling of bicarbonate (HCO− 3).
True or False: Compensation in primary metabolic disorders involves buffers and alterations in renal handling of bicarbonate (HCO− 3).
False, It's the pulmonary handling of CO2.
write out the Henderson-Hasselbach equation
pH=pK+log(base/acid), what mainly constitutes the acid and base in the equation?
base: bicarb, acid, dissolved PaCO2
co-oximeter measures which two serum proteins that pulse oximeter doesn't?
methemoglobin and carbozyhemoglobin
what form of hemoglobin that the all-mighty co-oximeter does not measure?
sulfhemoglobin
Is pulse oximeter the method of choice for detecting carbon monoxide poisoning?
No, it's co-oximeter
what serum measures are used in calculating the osmolal gap? (4)
measured blood osmolarity, Na, Glucose, BUN
Determine what type of acid-base disorder is this: pH and [HCO3] go same direction (bicarb usually < 25 mEq/L)
Metabolic acidosis
Determine what type of acid-base disorder is this: pH and [HCO3] go opposite direction (pCO2 usually > 44 mm Hg)
Respiratory acidosis
Determine what type of acid-base disorder is this: pH and [HCO3] go same direction (bicarb usually > 25 mEq/L)
Metabolic alkalosis
Determine what type of acid-base disorder is this: pH and [HCO3] go opposite direction (pCO2 usually < 40 mm Hg)
Respiratory alkalosis
how does the calculated anion gap change when there is hypoalbuminemia?
the anion gap decreases, how to calculate the corrected anion gap?
corrected anion gap = anion gap +2.5(4-albumin)
What is the normal BUN/creatinine ratio?
10:1

When both BUN and creatinine are elevated maintaining this ratio is suggestive of what?
Intra-renal disease - glomerulonephritis and tubulointerstitial
Often refered to as renal azotemia
When the BUN/creatinine ratio is elevated this suggests two possible problems, name them?
Prerenal azotemia

And

Postrenal azotemia

Define these terms
Prerenal - when there is poor renal perfusion- hypovolemia, hypotension - and results are from BUN reabsorbtion being increased

Postrenal - results from renal obstruction
A decreased BUN/creatinine ratio is rare and results from what two causes?
Dietary insufficiency

And

Severe liver disease
Name a protease inhibitor that is a better predictor than creatinine of future cardiac events in renal disease. It is independent of age, sex, or muscle mass.
Cyststin C
Normal proteinuria does not exceed how many mg/day?
150 mg/day

This is mainly Tamm-Horsfall protein and small amounts of albumin.
The normal glomerulus filters protein based on what two properties?
Size and charge

Normally albumin does not freely cross the glomerular membrane.
Significant proteinuria is usually defined as exceeding how many mg/day?
300 mg/day

This is traditionally based on a 24-hour urine collection.
What test can be used when a 24-hour urine is not possible?
Urine protein:creatinine ratio

The urine protein assay is sensitive to all kinds of protein ( albumin, globulins, Bence-Jones), and has a low detection limit of 3 mg/dL.

A random urine sample, or spot urine, can be misleading, but the ratio is considered to be as good as the 24 hour urine)
A urine dipstick is most sensitive to what protein?
Albumin, the lower limit of detection is 18mg/dL

It is not sensitive to microalbuminuria, globulins, or Bence-Jones proteins.
What two assays can be used to detect renal tubular dysfunction?
B2-microglobulin
and
Lysozyme

These are freely filtered by the glomerulus and completely reabsorbed by the proximal convoluted tubule.

Their presence in urine suggests?
B2-microglobulin
and
Lysozyme

These are freely filtered then completely reabsorbed by the proximal convoluted tubule.
Microalbuminuria is currently defined in terms of the albumin:creatinine ratio using what kind of sample?
Spot urine, not 24-hour urine

Can detect as little as 0.3 mg/dL of albumin

Since the microalbumin assay is a homogeneous immunometric one, it is subject to what effect?
The hook effect. Or prozone effect.


In an agglutination or precipitation reaction, prozone or prezone is the zone of relatively high antibody concentrations within which no reaction occurs. In an agglutination test, a person's serum (which contains antibodies) is added to a test tube which contains a particular antigen. There are many types of different antibodies present in a persons serum, and there might be one particular antibody that will bind to antigen present in the tube. There are also other types of antibodies which would not bind the antigen. The antibody-antigen complex forms an agglutinate. In some cases, the concentration of antibody specific for a particular antigen is too low compared to other types of antibodies, and when this occurs, the small portion of agglutination that took place will be masked by larger aggregate of non-binding antibodies, and the mixture remain as fluid. The agglutination which took place is invisible, and this is called prozone. This can lead to false negative result. Therefore in each agglutination test, dilution of the antibody-antigen mixture is done to a certain level, until agglutination can be seen, or otherwise the test is negative.



How can one check this effect?
The current recommendation of the national kidney foundation is annual testing for those individuals who are high risk, they include?
People with diabetes, hypertension, or a family history of renal disease

Recommended screening includes what two measurements?
eGFR

And

Microalbuminuria screen (urine albumin:creatinine ratio), albumin is measured by the microalbumin assay.
Chronic kidney disease is defined by one of two ways, name them.
GFR > 60mL per 1.73m3 of body surface area, or

Albuminuria for 3 or more consecutive months
There are 5 stages of chronic renal disease:
Stage one is proteinuria without decreased GFR
Stage two mild decrease in GFR
Stage three moderate
Stage four severe
Stage five?
Renal failure, defined as?
GFR < 15 mL per minute per 1.73m3 or

Dialysis dependent
The most common cause of renal cases acute renal failure is?
Acute tubular necrosis

Acute glomerulonephritis may also cause renal ARF
New onset of acute renal failure must have a battery of tests, these include a renal ultrasound, initial and serial BUN and creatinine, urine volume monitoring, BUN/creatinine ratio, urine osmolarity, and fractional excretion of sodium (FENa), urinalysis.
Non-oliguric renal failure can occur in renal or Postrenal, but rarely Prerenal.
Indications of dialysis includes (five)?
Volume overload
Hyperkakemia
Metabolic acidosis
End organ damage due to uremia (pericarditis)
BUN > 100 gm/dL
BUN/creatinine ratio, urine osmolarity, and fractional excretion of sodium (FENa)- this combination is useful is distinguishing renal from Prerenal. The FENa is almost always w (<1%) in Prerenal ARF. What is the exception?
When the patient has been given a diuretic or has glycosuria.

The fractional excretion of urea can be an alternative.
A lack of findings in urinalysis (or isolated byline casts) suggest?
Prerenal ARF

Dysmorphic red blood cells and red blood cell casts suggest?
Glomerulonephritis


Pigmented casts suggest?
Determination of the amniotic fluid bilirubin is part of the clinical evaluation of the pregnancy complicated by alloimmunization. The amniotic fluid bilirubin reflects?
The degree of fetal hemolysis
The concentration of bilirubin in amniotic fluid is proportional to absorbance. The maximal absorbance of bilirubin is at?
450 nm

The difference between the actual absorbance and the norm 450) is plotted against the estimated gestational age.

Liley chart
This chart correlates with the previous chart as far as zone 1-3.

Unless the result falls in zone 3, serial measurements are taken.

If they are in zone 3 ( or in 2 and rising) then deliver if baby is term.
What is hepatorenal syndrome?
Development of progressive renal impairment in patients with severe end-stage liver disease, tere should be no other identifiable cause.

HRS has an incidence of about 5% of admissions for decompensated cirrhosis and about 50% over the course of cirrhosis.

HRS it thought to be due to what reason?
Dis regulation of renal blood flow.
What is the most common cause of renal failure in a cirrhotic patient?
Spontaneous bacterial peritonitis (SBP)

Nephrotoxic agents are also common ( NSAIDs, contrast)
Renal biopsy in hepatorenal syndrome is essentially normal. An abnormal biopsy must be interpreted cautiously - many pts with cirrhosis have hepatitis viruses that can cause associated glomerulonephritis, and cirrhosis itself can lead to?
An IgA like nephropathy
What is the level of hCG in women with gestational trophoblastic disease compared to normal?
Higher (usually complete moles hCG> partial moles)
Which is more likely to be clinically diagnosed rather than histologically, partial or complete moles?
Complete
What's the characteristic clinical presentation for a complete mole? (5)
Uterine enlargement greater than gestational age, vaginal bleeding, hypertension, absence of fetal heart tones, characteristic ultrasound
Why must both types of molar pregnancies be monitored? (2)
for 1) persistence of trophoblastic disease 2) development of malignant trophoblastic disease
What's the risk of malignancy for molar pregnancies?
<5% for partial moles and ~20% for complete moles
1) What is the hCG monitoring schedule after the evacuation of a molar pregnancy? 2) How long after evacuation does hCG normally remain detectable?
1) monitor weekly until hCG is undetectable for 3 consecutive weeks then monitor monthly for 1 year 2) hCG remains detectable for up to 10 weeks (in uncomplicated molar pregnancy)
When is intevention warranted when monitoring hCG following evacuation of a molar pregnancy?
If hCG levels plateau or rise then persistent GTD is suspected and may need chemotherapy following a metastatic workup
1. T/F uterine choriocarcinomas are always preceeded by a molar pregnancy
1. False, 1/2 follow a normal term pregnancy, 25% follow a normal spontaneous abortion, 25% follow a molar pregnancy
What are the components of a "triple screen"? (3)
hCG, AFP, and unconjugated estriol (uE)
When is the triple screen normally performed and on what type of specimen?
perfomed on serum drawn during 2nd trimester, ideally around 18 weeks
What's the sensitivity of the triple screen for Down's syndrome?
0.7
1) What are the components of the "Quad test"? (4) 2) What is its sensitivity for detection of Down's syndrome?
1) hCG, AFP, unconjugated estriol, and dimeric inhibin A 2) 80%
What's the advantage of quad test over triple screen?
Concentration of dimeric inhibin A is fairly stable throughout the 2nd trimester so it is less prone to error based on inaccurate gestational age
1) What is the "integrated screen" for detection of trisomy and neural tube defects? 2) What is its sensitivity in detecting Downs?
1) Pregnancy associated plasma protein A (PAPP-A) and hCG measured 1st trimester then AFP, unconj. estriol and inhibin A measured 2nd trimester and data combined 2) sensitivity 85% (90% if combined with measurement of nuchal fold thickness)
What effects do the following maternal conditions have on prenatal screening for trisomy and neural tube defects? 1. diabetes 2. smoking
1. both unconj. estradiol (uE) and hCG are decreased mildly 2. MSAFP increased, uE and hCG decreased
What is the sensitivity of prenatal screening for Down's syndrome in twin gestations?
50-70%
1. What are the characteristic serum findings in prenatal screening for a fetus with trisomy 18 (Edward syndrome)? (3) 2. What percentage of cases with this pattern are actually affected? 3. What percentage of cases are detected with screening?
1. decreased AFP, decreased hCG, decreased unconj. estriol 2. 10% 3. 80%
What are the characteristic serum findings in prenatal screening for a fetus with neural tube defects?
increased AFP, normal hCG, decreased unconj. estradiol
What percentage of cases with increased maternal serum AFP are due to neural tube defects?
0.1
If an elevated maternal serum AFP is deemed to be caused by a neural tube defect what's the next step in the patient's workup?
obtain amniotic fluid for AFP and acetylcholinesterase
What is the sensitivity of maternal serum AFP for detecting neural tube defects in singleton and multiple gestations?
singleton 90% multiple 30%
1. What are the characteristic maternal serum findings when fetus has Down's syndrome? (4) 2. What follow up is recommended when they're found?
1. decreased AFP, increased hCG (roughly 2 times normal), decreased unconjugated estradiol, decreased inhibin 2. high resolution ultrasound and amniocentesis for cytogenetics
1.What is alpha fetoprotein (AFP)? 2. How do levels change in normal pregnancy? 3. What maternal characteristics require adjustments in AFP interpretation? (4)
1. principle plasma protein in the fetus 2. rises progressively during 1st and 2nd trimesters 3. maternal weight, race, number of fetuses, and maternal diabetes
How do the following affect AFP? 1. maternal weight 2. number of fetuses 3. maternal diabetes
1. has a dilutional effect (falsely low values) 2. levels tend to be much higher 3. levels are much lower
1. What unit is used to compare maternal serum AFP (MSAFP)? 2. what value indicates increased risk of neural tube defect? 3. What value is considered abnormal in maternal diabetes? multiple gestation?
1. multiples of the median (MOM) 2. >2.5 MOM (present in >80% of cases) 3. diabetes >2.0 MOM, multiple gestations >4.5 MOM
What conditions are associated with an increased maternal serum AFP? (13)
neural tube defects, omphalocele, gastroschisis, renal anomalies, sacrococcygeal teratoma, cystic hygroma, hydrops fetalis, Turner syndrome, bowel obstruction, twins, wrong gestational age, fetal demise, fetal-maternal hemorrhage
1. T/F decreased unconjugated estriol is sensitive for both Down syndrome and trisomy 18 2. What other congenital abnormalities is it associated with? (2)
1. False weakly sensitive for Down syndrome but a good indicator for trisomy 18 2. also decreased in Smith-Lemli-Optiz syndrome (SLOS) and inherited (fetal) deficencies of steroid sulfatase
1. Where is dimeric inhibin A produced? 2. what level of elevation is associated with Down syndrome
1. it's a glycoprotein produced by the placenta 2. increased an average of 1.9 MOM in Down syndrome fetus
What markers have demonstrated some (although limited) utility in predicting preterm labor? (3)
serum estradiol, salivary estriol, screening for bacterial vaginosis
Where is fetal fibronectin normally found and when can it be detected in cervicovaginal fluid?
normally found at placental fetomaternal inferface. Can be detected briefly during early gestation then absent until just before labor
1. What's the utility of fetal fibronectin in detecting preterm birth? 2. When should it be collected?
1. its absence has a very high negative predictive value and can exclude preterm labor, its presence suggests onset of preterm labor but positive predictive is very low 2. greater than 24 hours after the last cervical exam or sexual intercourse
What is the major phospholipid produced by mature type II pneumocytes and what are its major components?
lecithin- majority is disaturated phosphatidylcholine (DPC), lesser amounts of phosphatidylglycerol (PG), phosphatidylinositol (PI), phosphatidylethanolamine (PE), and sphingomyelin
1. During which gestational ages is fetal lung maturity assesment most critical? 2. When might it be indicated for term pregnancies?
1. between 34-37 weeks gestation 2. poorly controlled maternal diabetes
1. What is the best specimen for fetal lung maturity? 2. What would be considered a suboptimal specimen?
1. Uncontaminated amniotic fluid obtained by amniocentesis 2. suboptimal specimen vaginal pool specimens which may be contaminated by blood or meconium
T/F mosts tests for fetal lung maturity are better at predicting maturity than immaturity
True - mature result is fairly reliable, immature result needs confirmation with another technique
How do the the following change with increasing gestational age: 1. lecithin (L) concentration? 2. sphingomyelin (S) concentration? 3. L:S ratio?
1. increases 2. remains the same 3. around 1:1 until 26 weeks then ration increases until ratio of 2:1 reached around 35 weeks
1. What lecithin/sphingomyelin ratio indicates fetal lung maturity? 2. above the ratio in #1 what percentage of of premature infants will develop RDS? 3. below the ratio in #1 what percentage of of premature infants will develop RDS?
1. 2:1 2. 2% 3. 60%
How do the following influence the interpretation of the lecithin/sphingomyelin ratio? 1. maternal diabetes mellitus 2. presence of meconium 3. presence of blood
1. ratio 2:1 doesn't ensure maturity (phosphatidylglycerol should be used) 2. falsely decrease the L:S ratio 3. normalizes the L:S to around 1.5
Phosphatidylglycerol 1. when can it first be detected in the aminotic fluid? 2. what does its presence indicate? 3. T/F Its concentration is not effected by blood or meconium. 4. measurable by what 2 methods?
1. around 36 weeks 2. fetal lung maturity 3. True (thus it is test of choice for contaminated specimens) 4. TLC or agglutination (equally reliable)
1. What is the foam stability index? 2. How is it performed? 3. What value is considered indicative of fetal lung maturity?
I1. ndication of concentration of surfactant in amniotic fluid (AF), which forms a film that can support the structure of a foam. 2. AF serially diluted with ethanol, highest concentration at which a complete ring of bubbles is seen is the foam stability index. 3. > 0.47
1. How is surfactant lamellar body number density (LBND) measured? 2. What value is predictive of fetal lung maturity?
1. the platelet channel of a cell counter can quantify them since they're about the same size as platelets 2. >50,000
1. What is the disaturated phosphatidylcholine concentration used for? 2. what does it measure 3. In what clinical scenario is useful?
1. it's an alternative to the lecithin/sphingomyelin ratio 2. determines the major component of lecithin directly 3. useful when only contaminated amniotic fluid specimen is available (unaffected by blood or meconium)
1. What test is used most commonly to predict fetal lung maturity? 2. what value is considered mature? immature? 3. what effect does blood have on the values? 4. what value is considered mature even in the presence of blood?
1. fluorescence polarization assay (FPA) 2. <260 considered mature, >290 considered immature 3. <0.5% blood values unaffected, greater amounts tend to have a parodoxical effect on the FPA values 4. <230 considered mature
What effect does pregnancy have on maternal: 1. serum triglycerides? 2. albumin and total protein? 3. GFR? 4. transport proteins?
1. increased by 40% (often causes a low-level ketosis) 2. decreased (hemodilution) 3. increased (due to increased blood volume) 4. increased (due to increased estrogen)
What effect does pregnancy have on maternal: 1. BUN? 2. creatinine? 3. urate?
1. decreased 2. decreased 3. decreased
1. How does insulin resistance change during pregnancy? 2. What mediates this change?
1. improved glucose tolerance until mid 2nd trimester then relative insulin resistance 2. human placental lactogen
1. What is the incidence of DVT during the antepartum and postpartum period? 2. PE?
1. 1/2000 antepartum and 1/700 postpartum 2. 1/2500
1. What effect do autoimmune diseases have on pregnancy in general? 2. SLE? 3. Graves? 4. myasthenia gravis
1. generally no effect 2. often exacerbated 3. common to have post-partum flareups 4. common to have post-partum flareups
1. What neonatal complication can result from maternal idiopathic thrombocytopenic purpura (ITP)? 2. What must this complication be distinguished from? 3. What's a life threatening complication of either?
1. ITP antibodies can cross the placenta causing neonatal thrombocytopenia 2. Neonatal alloimmune thrombocytopenia 3. neonatal intracranial hemorrhage
1. What affect does pregnancy have on systemic lupus erythematosis? 2. Does it change throughout pregnancy?
1. increases risk of flare ups 2. risk higher in early pregnancy and during puerperium, relative quiescence in latter half of pregnancy
Why might it be difficult to distinguish a lupus flare up from pregnancy induced hypertension (PIH) and what test can aid the distinction?
both can cause hypertension, edema, and proteinuria. Distinction can be aided by complement levels (low in SLE flare, normal in PIH)
T/F mortality is increased in pregnant women with SLE
true (most deaths result of pulmonary hemorrhage due to lupus pneumonitis)
Why is it important to distinguish a lupus flare up from pregnancy induced hypertension (PIH)
treatments very different (delivery for PIH and steroids for SLE)
1. What is thought to be responsible for recurrent miscarriage, abortion, and preterm labor in SLE patients? 2. What's the treatment?
1. lupus anticoagulant 2. anticoagulation (not corticosteroids)
What conditions are neonates born to mothers with SLE at greater risk for? (3)
IUGR, preterm labor, congenital heart block (mediated by Ro and La antibodies)
1. How is the diagnosis of asymptomatic bacteriuria made? 2. what's the incidence in pregnant women? 3. what's the associated complication? 4. what's the most common causative agent?
1. clean catch voided urine specimen with >100,000 bacterial colonies/mL 2. 10-20% (same as in nonpregnant women) 3. associated with development of UTI with high risk of pyelonephritis 4. Escherichia coli
1. What is Sheehan syndrome? 2. What causes it? 3. What are the clinical symptoms?
1. postpartum hypopituitarism 2. pregancy-associated pituitary enlargement and severe blood loss duing delivery 3. inability to lactate, lethargy, weakness, and weight loss
1. Why is there an increased incidence of thyroid disorders during pregnancy? 2. What is responsible for the increase? 3. Which is more common in pregnancy hyper- or hypothyroidism? 3. What the best test of thyroid status in pregnancy?
1. increased demand on thyroid due to increased thyroid-binding globulin and TSH like effect of hCG 2. hypothyroidism (esp in patients with borderline thyroid function or availability to iodine) 3. serum TSH
What causes transient hyperthyroidism of hyperemesis gravidarum?
high levels of hCG (thought to be most common cause of hyperthyroidism in pregnancy)
A 25y/o female in late preganancy presents with nausea, RUQ pain, and jaundice. She eventually becomes confused. If acute fatty liver of pregnancy is suspected, what do you expect to see on histologic examination of the liver? What is the treatment of choice?
Widespread microvesicular steatosis (especially around Zone 3-pericentrally), with little inflammation or hepatocellular necrosis.

Delivery of the baby
What hematologic problem is common in acute fatty liver of pregnancy?
DIC
A woman in her 3rd trimester of pregnancy presents with jaundice and itching. If cholestasis of pregnancy is suspected, what laboratory findings can you expect? (6 things)
Elevated Alk Phos (massively), increased GGT, increased 5'-nucleotidase, direct bilirubinemia (but less than 5mg/dL), normal or mildly elevated transaminases, increased bile acids *** (most characteristic)
What are the typical histological findings in cholestasis of pregnancy?
dilated canaliculi containing bile plugs, especially in pericentral (zone 3) region
Name 4 tests that might be undertaken to evaluate "recurrent pregnancy loss:"
Parental or abortus karyotyping, endometrial biopsy, thyroid function, lupus anticoagulant
What is luteal phase defect defined as in regards to endometrial biopsy?
endometrial histology that is 2 or more days discrepant with dates
3 Clinical applications of Toxicology?
Drugs of abuse screening, overdose management, therapeutic drug monitoring
Define half-life:
The time it takes for the drug concentration to reach 1/2 of the starting amount
What is first-order kinetics?
The rate of loss is exponential during drug elimination
What is steady-state, when speaking of drug metabolism? (conceptually)

After how many doses does this usually occur?
the amount of drug leaving the body equals the drug entering the body.

Typically reached after 5 doses given at an interval of 1 half-life each.
When do the peak and trough of drug dosages occur?
Peak: soon after a dose (typically)

Trough: just before a dose
What is the most common thing that circulating drugs are bound to?
Albumin
Which part is the active component of the drug, free or bound?
Free
Increasing the amount of a protein that binds drugs into the circulation will do what to a drug's availability?
Less free drug is available, and thus may have a weaker effect than anticipated for a dose
A patient is taking Drug A, a drug with a narrow therapeutic window. He is prescribed a new Drug B. Three days later, he is admitted to the ER with toxicity of Drug A. What could be a mechanism involved in this that includes binding proteins?
Likely Drug B displaced Drug A off of a binding protein, increasing the amount of Drug A available
Which drugs remain tightly confined in the vascular space, lipophobic (aka-hydrophilic) or lipophilic (hydrophobic)?
Lipophobic/hydrophilic
The degree to which a drug can distribute into various body tissues (vasculature, adipose tissue, interstitium, etc) is defined as its:
Volume of distribution
What equation can calculate a drugs volume of distribution?
Vd=D/C

where D= dose and C=resulting measured plasma concentration
What is the most typical specimen received for drug screening (drugs of abuse)?
Urine
Urine drug of abuse screens have a low or high sensitivity?

Low or high specificity?
High sensitivity

Low specificity
What method is most frequently employed in drug of abuse screens?
Immunoassay
Why does every positive drug of abuse screen require confirmation?
False positives due to cross-reactivity can occur
What methods are typically used for confirmatory testing for a positive drug of abuse screen?
Mass spectrometry and or gas chromatography
What is "chain of custody?"
A method insuring that a specimen that will have potential consequences is under control of someone or in locked storage at all times.
Name 7 different checks that can be performed to evaluate a urine sample for adulteration:
Color, Odor, Temperature, pH, Specific gravity, creatinine, nitrite
What is the half-life of Cocaine?
1:00:00
How long is cocaine detectable in testing?
24-72 hours
Key metabolites of cocaine:
benzoyl ecgonine methyl ester
What is the half-life of heroin?
0:03:00
How long is heroin detectable in testing?
72:00:00
Key metabolite of heroin:
6-acetyl morphine
What is the half-life of amphetamine?
0:30:00
How long are amphetamines detectable in testing?
72:00:00
2 key metabolites of amphetamines:
norepinephrine and phenylacetone
What is the half-life of PCP?
0:30:00
How long is PCP detectable in testing?
72:00:00
2 metabolites of PCP:
hyroxylated and gluconarated PCP
What is the half-life of cannabis?
8 hours (wow!)
How long is cannabis detectable in testing?
Weeks
What is the key metabolite of cannabis?
delta-9-THC-COOH
What is physiologic basis of chest pain in cocaine users?
Vasoconstriction with concurrent increased rate and blood pressure
What happens to the specificity of myoglobin, CK-MB, and troponin I in cocaine-induced myocardial infarction?
CK-MB and myoglobin specificity decrease due to skeletal muscle effects, but Troponin I stays the same
Acute intoxication with opiates can lead to: (name 5 symptoms)
Sedation, pinpoint pupils, constipation, bradycardia, and hypotension (also respiratory depression)
What drug is commonly used to counteract opioid intoxication?
Narcan (Naloxone) (also Nalmefene) are synthetic opioid antagonists that counteract the opioid effects
Withdrawl from opiates commonly causes the following: (name 7 symptoms)
increased lacrimation, rhinorrhea, diaphoresis, dilated pupils, tachycardia, irritability, restlessness
What is methadone?
A long-acting opioid that can be used to help withdrawl symptoms (less high, more sustained treatment of all withdrawl symptoms)
What is clonidine's role in opioid withdrawl?
It does not interact with the opiod receptors (like methadone does) but counteracts the symptoms through other mechanisms.
Why does propoxyphene cause more symptoms than other opioids?
It (and its metabolite) have a interference with calcium channels of the heart, very similar to quinidine.

Can cause cardiac conduction abnormalities and even seizures
How do barbituates act on the CNS?
They facilitate GABA (a depressant) effect in the CNS, especially in the medulla
How do amphetamines (and methamphetamines) act on the CNS?
Mediate release of dopamine
What are long-term effects of meth and amphetamine use that are related to their mechanism of action?
The release of dopamine eventually depletes and destroys the dopamine secreting cells of the substantia nigra and cause Parkinsonian syndrome (it's irreversible!)
How does phencyclidine (PCP) act on the CNS?
It acts on blocking catecholamine re-uptake
What unique symptomatology often occurs with PCP use? (2 things)
Its especially known for psychiatric effects. Horizontal nystagmus is very often seen.
What other lab testing, besides a drug screen, must be carried out in those individuals suspected of having PCP intoxication?
Glucose monitoring: often can cause hypoglycemia
CK and BUN: can cause rhabdomyolysis
At what blood alcohol level does coma/death occur?
>0.4 % BAC
Outline the steps of alcohol metabolism:

In what organ does this occur?
Liver

Ethanol metabolized by alcohol dehydrogenase to acetaldehyde.

Acetaldehyde is then converted by aldehyde dehydrogenase to acetic acid
What type of tube should whole blood for alcohol testing be submitted in?
Sodium fluoride and potassium oxalate will prevent increases (from fermentation) or decreases in the level
What methodology is used for analyzing blood for alcohol content?
Enzymatic method utilizing alcohol dehydrogenase
What is the short-coming is using an alcohol dehydrogenase method for analyzing blood alcohol content?
Only measures ethanol, will not pick up methanol
What principle is used for analyzing breath alcohol?
Since blood alcohol diffuses across alveolar septa and is excreted in expiration, the level can be measured in breath
What is the ratio of blood:breath alcohol?
12/31/1899 11:00:01
What liver function test is often increased in chronic alcohol users?

When does it increase?
GGT (gamma glutamyl transferase)

>4 drinks per day for more than 4 weeks

4 weeks of abstinence to decrease
Wat is CDT? What is it being used for evaluating?
Carbohydrate deficient transferrin: being investigated as a marker of heavy alcohol consumption. More specific than GGT, at least as sensitive as GGT. Raises sooner than GGT.
Which 2 populations may not be as easy to monitor with CDT levels? Why?
Women: they have naturally higher CDT, and it doesn't rise as much with alcohol.

Those with chronic liver disease--they have elevated levels of CDT without drinking being involved
What RBC parameter measured by a CBC (or peripheral smear review!) is elevated with heavy alcohol consumption?
MCV
How is anion gap calculated?
Sodium - (Chloride + BIcarb) = anion gap
When is an increase in anion gap significant?
20 mEq/L or higher is significant
What is an important cause of decreased anion gap?

Why is this important?
Hypoalbuminemia. For every 1gram decrease, there is a 2.5 mEq decrease in anion gap.

Important because this could "mask" an increase in anion gap!
An increase in anion gap can lead typically to acidosis or alkalosis?
Acidosis
Numerous toxins cause anion gap metabolic acidosis: (listed are 12)
Acetominophen, Salicylates, ascorbate, hydrogen sulfide, ethylene glycol, methanol, ethanol, formaldehyde, carbon monoxide, nitroprusside, epinephrine, paraldehyde
What is a toxidrome?
A set of symptoms that can suggest a particular agent or group of agents have been ingested
toxidrome: Hyperthermia, dry skin, flushing, altered mental status, psychosis, mydriasis, constipation, what acronym?
“hot as a hare, dry as a bone, red as a beet, mad as a hatter”, what category?
Anticholinergic, which drugs besides Atropine?
Salivation, lacrimation, urination, diarrhea, GI cramps, emesis (); diaphoresis, miosis, and wheezing, what acronym?
“SLUDGE”, what category?
Cholinergic, which drugs?
Hypertension, tachycardia, mydriasis, anxiety, hyperthermia, what category?
Adrenergic, which drugs?
Amphetamines, Cocaine, Pseudoephedrine Ephedrine, PCP
Altered mental status, slurred speech, hypopnea/apnea, what type of drugs?
1. Sedative, 2. Narcotic, names?
Barbiturates, Opiates
Hallucinations, anxiety, hyperthermia, what type of drugs?
Hallucinogenic, names?
LSD, PCP, Amphetamines, Cocaine
In an overdose case, workup includes?
1. tox screening; 2. calculate anion gap; 3. calculate osmal gap; 4. measure blood gas
When is the anion gap significant in a overdose case?
An increase in the anion gap to >20 mEq/L is significant.
Roughly, how does alterations in albumin affect anion gap?
for every 1 gram decrease in albumin, there is a 2.5 mEq decrease in the anion gap
What are the chemicals that you will never realize that they will cause increased anion gap metabolic acidosis?
ethylene glycol, methanol, ethanol, formaldehyde, nitroprusside, epinephrine
In general, what type of agents will cause increased osmolal gap?
all kinds of alcohols, ehtanol, methanol, glycerol, acetone, etc.
When the same sample from a alcohol intoxication case was sent to lab A and lab B, lab A detected increased osmalol gap but not lab B?
Lab A used freezing point depression osmometry, while Lab B has less-used vapor pressure osmometry.
What do you call the difference between the saturation given by co-oximetry and the saturation given by the ABG analyzer?
oxygen saturation gap
Causes of an increased oxygen saturation gap (4)
carbon monoxide poisoning (carboxyhemoglobin), methemoglobin, hydrogen sulfide poisoning (sulfmethemoglobin), and cyanide poisoning
Why does the ABG analyzer falsely report a high oxygen saturation when there is carbon monoxide poisoning or sulfide poisoning?
The ABG analyzer consider them to be oxyhemoglobin!
An abnormally high venous oxygen content (arteriolization of venous blood) is seen in (2)
cyanide and hydrogen sulfide poisoning
How useful is the oxygen saturation gap when it refers to the difference between the percent oxyhemoglobin given by the ABG analyzer and that given by the pulse oximeter?
it might reflect the presence of carboxyhemoglobin or methemoglobin, but the precise numbers are hard to predict
Antifreeze contains which alcohol?
Ethylene glycol
rubbing alcohol contains which alcohol?
isopropanol
windshield washer fluid contains which alcohol?
Methanol
common Toxic Alcohol Poisoning agents all cause increased anion gap, true or false?
False. Isopropanol doesn't.
most common Toxic Alcohol Poisoning agents cause increased anion gap, true or false?
False. Only ethanol might.
in suspected methanol or ethylene glycol intoxication, what surrogate marker to use if direct tests are not available?
Osmalol gap
Which alcohol intoxication does not cause acidosis but does create an osmolal gap
isopropanol
What does the urine crystal look like in ethylene glycol intoxication?
envelope-shaped, translucent, and birefringent, what's in it?
calcium oxalate
which alcohol intoxication result in ocular toxicity?
Methanol
Lead is toxic to cells in which 2 ways?
nonspecifically binds to and inhibits enzymes bearing sulfhydryl groups, many in the heme synthesis, and it is directly toxic to mitochondria.
Iron deficiency and lead toxicity frequently coexist. The effect of iron deficiency is to enhance the toxic effects of lead in two ways:
1.The final step in biosynthesis of heme, in which iron is incorporated into protoporphyrin, is further inhibited by a deficiency of iron.
2.In an attempt to upregulate intestinal absorption of iron, there is the unintended effect of increased absorption of lead.
What shows in the peripheral smear of a lead poisoning?
microcytic, hypochromic anemia with basophilic stippling
What renal function changes (other than elevated BUN and creatinine) are seen in long-term lead toxicity?
aminoaciduria, glycosuria, and phosphaturia, what causes these?
Mitochondrial toxicity leads to reduced ATP available to drive the numerous ATP-dependent channels involved in tubular epithelial function
What is the cutoff for the blood lead level?
10ug/dL
If blood lead levels ‘rebound’ during treatment, what tests to use to distinguish false increase from true increase?
free erythrocyte protoporphyrin or (FEP) or zinc protoporphyrin (ZPP) should not be increased in false rebound; they are intermediate products in heme synthesis
Can we use capillary blood obtained from heel- or finger-sticks for lead levels?
No. It's not accurate. Unless you test for FEP or ZPP, which is only sensitive for high level toxicity.
name a few chelators used in treatment for lead poisoning
dimercaprol (also known as British antilewisite or BAL), CaNa-EDTA, D-penicillamine, and succimer
What level of blood CO will lead to coma and death?
>50%
Why infants are at greater risk for CO poisoning?
CO has higher avidity for fetal hemoglobin
venous blood is as good as arterial for CO level determination, true or false?
TRUE
The half life T½ of CO on 100% O2 is:
1 hour
What is the major organ disfunction of Acetaminophen toxicity?
liver
What is the Rumack-Matthew nomogram used for?
To predict the risk in acetaminophen toxicity
For an acetaminophen toxicity case, should be the initial blook sample drawn immediately after the patient arrives in the ER?
No, it should be drawn 4 hours after the ingestion, for full absorbtion.
When to administer N-acetylcysteine for acetaminophen toxicity cases?
When the risk falls into the probable and possible hepatic toxicity categories in the Rumack-Matthew nomogram.
In acetaminophen toxicity cases presenting late, samples drawn greater than 2–3 hours apart can be used to estimate the elimination half-life of acetaminophen in the patient. If the half-life is >4 hours, then hepatic necrosis is likely, why?
patients with hepatic damage after overdose may exhibit a more prolonged acetaminophen half-life
How does the serum bilirubin level affect the measured acetaminophen level?
Hyperbilirubin cause elevated acetaminophen readings
How does chronic ethanol use affect acetaminophen toxicity?
Chronic alcohol use induces the P450 system, increases the proportion of acetaminophen processed to NAPQI, enhancing toxicity.
True or False: Hyperbilirubinemia causes false negative Acetaminophen level?
False.Hyperbilirobinemia causes "False Positive" Acetaminophen level mainly because of its broad spectrophotometric absorbanse.
In which organ Acetaminophen is metabolized?
Liver
What are the two main pathways for handling acetaminophen in Liver?
1) Mostly conjugation with glucuronide or sulfate to forme nontoxic metabolite. 2) A small amount is metabolized by P450 system into the toxic metabolite N-acetyl-p-enzoquinoneimine(NAPQI)
How the liver detoxified the acetaminophen toxic metabolite(N-acetyl-p-enzoquinoneimine)?
NAPQI is detoxified by glutathion
True or False: Any agent that induces P450 system decreases acetaminophen toxicity?
False.They increase the proportion of acetaminophen processed to NAPQI, enhancing toxicity, for example chronic ethanol use has this effect.
What kind of liver pathology can we see in Acetaminophen toxicity?
Centrilobular (zone 3) hepatic necrosis with periportal sparing that causes mostly by NAPQI metabolite.
False or true: N-acetyl-p-enzoquinoneimine is only toxic for liver?
False. Its formation within hepatocytes makes the liver primary target, but other organs may be affected.
What is the mainstay of acetaminophen toxicity treatment?
N-acetylcysteine (Mucomyst)
What is N-acetylcysteine (Mucomyst) mechanism in acetaminophen toxicity? treatment?
It promotes acetaminophen metabolism via the conjugation pathways, thereby decreasing the formation of the toxic metabolite NAPQI
What are the usual ways for Cyanide poisoning?
1) inhalation of smoke from a fire (present in insulation) 2) industrial exposure (present in pesticide nad other industrial materials) 3) Suicide or homicide attempt
What is the Cyanide poisoning mechanism?
It bindes to and inhibites cytochrome a3, thus uncoupling the electron transport system.
True or False: Cyanide poisoning causes non anion-gap metabolic acidosis?
False,it causes anion-gap metabolic (lactic) acidosis.
What are clinical presentation of acute Cyanide poisoning?
Cherry-red skin color, hyperpnea followed by apnea and loss of consciousness and bitter almond odor on patient's breathin.
True or False: Cyanide is rapidly metabolized to thiocyanate which persists for a long period of time and is more reliable for the diagnosis?
TRUE
True or False:? A normal serum Lactate exclude cyanide poisoning?
True, all patients exposed to Cyanide should have an elevated serum lactate and an anion gap metabolic acidosis
False or True: Cyanide-poisoned patients have hypoglycemia?
False, they have hperglycemia due to decreased utilization.
How blood gases, performed by co-oximetry can be informative in Cyanide poisoning?
First, if compared to venous blood it shows a decrease in the arterial- venous oxygen gap ( due to decrease utilization). Second, the co- oximeter can specifically measure carboxyhemoglobin and methemoglobin, removing them from the differential diagnosis.
Cyanide poisoning treatment?
Administration of sodium nitrate and amylnitrate leads to formation of methemoglobin, which binds available cyanide. Then sodium thiosulfate, which reacts with cyanomethemoglobin to form thiocyanate that can be cleared by kidney.
Aspirin effects on acid-base balance?
First, it directly stimulate the respiratory center within the medula oblaganta, promoting respiratory alkalosis. Second, it uncouples oxidative phosphorilation and inhibits Krebs cycle, shunting energy production towards anaerobic pathways with the development of a metabolic acidosis.
The earliest sign of salicylate toxicity?
Tinnitus and dizziness
True or False:The acid- base disorder in salicylate toxicity is triphasic?
True, 1) between 3-8 hours following ingestion, respiratory alkalosis due to stimulation of the respiratory center 2) between 12-24 hours creates a compensatory metabolic acidosis 3) alteration in metabolic pathways causes an increased anion gap metabolic acidosis
True or false: Salicylate toxicity causes CNS stimulation?
False, Salicylate toxicity causes CNS depression at third phase of toxicity that may contribute to hypoventilation and a compounding respiratory acidosis
True or false: Mortality in salicylate toxicity is best correlated with the 8-hour plasma salicylate concentration?
False, mortality is best correlated with the 6-hour plasma salicylate concentration
Which salicylate concentration value has a high fatality rate?
>130 mg/dl
Chronic salicylate intoxication manifestations(3)
Chronic metabolic acidosis, hypoglycemia, and possible hearing loss
Whre are the possible ways to get an accidental arsenic toxicity?
Through exposure to various pesticides, wood preservations, and leather tanning.
In which industries there is a risk for arsenic gas intoxication?
Production of metal alloys and semiconductor manufacture.
What are the manifestations of arsenic gas inhalations?
Arsenic gas is the most toxic form, capable of producing acute renal failure, hemolysis, and death within 24-48 hours.
True or false: Ingested arsenic excreted in GI tract?
False, Ingested arsenic excreted in urine
In which tissues, arsenic can be distributed?
Skin, nails, and hair
Acute arsenic toxicity manifestation?
Arsenic inhibits oxidative production of ATP. Thus intial toxicity is manifested in dividing tissue such as GI mocusa ( nasesa, vomiting, bloody diarrea, and abdominal pain), bone marrow (cytopenia with erythrocyte basofilic stippling similar to that seen in lead toxicity.
Chronic arsenic toxicity manifestations
Periferal neuropathy, skin hyperpigmentation, nephropathy, and hyperkeratosis ( particularly palms and soles, and transverse Mees lines in the nails.
Which samples can be used for the diagnosis of chronic arsenic intoxication?
Finger nails, hair, or urine
True or false: A blood arsenic level is the most reliable test in arsenic intoxication?
False, A blood arsenic level is highly unreliable, as the substance is rapidly cleared from circulation.
Which test is the most reliable test in arsenic intoxication?
Quantitative 24-hour urinary arsenic excretion: however, this result may be misleadingly elevated due to recent seafood ingestion
TCAs mechanism?
Blocking reuptake of dopamine and epinephrine from the synaptic space.
True or false: many of the TCAs adverse effects relate to their cholinergic effect?
False, it relates to their anticholinergic effect (not cholinergic)
TCAs anticholinergic manifestations
Dry mouth, constipation, urinary retension, papillary dilatation, hyperthermia, lethargy, confusion
The major that are affected by tricyclic overdose
The CNS and cardiac conduction system
ECG manifestation of tricyclic overdose
Widening of the QRS complex (QRS prolongation) that can lead to ventricular arrhythmias.
True or false: the duration of the QRS is predictive of the likelihood of seizures?
True, QRS interval longer than 0.1 seconds associated with a high risk of seizure
Organophosphates and carbamates mechanism
they inhibit acetylcholinesterase (that breaks acetylcholine) resulting in over stimulation of cholinergic process.
The classicmanifestation of organophosphates and carbamates toxicity?
Miosis, diaphoresis, excess salivation, lacrimation, GI hypermotility, bradycardia, and bronchospasm
Lab diagnosis of organophosphates and carbamates toxicity?
The level of erythrocyte cholinesterase activity.
How long can we detect organophosphates metabolite in urine?
Up to one week after exposure
True or false: elemental mercury can easily absorb from GI tract
False, Ingestion of elemental mercury is largely inconsequential, as GI absorption does not occure to any significant degree. In contrast, organic mercury (from the ingestion of fish) is readilly absorbed in the GI tract.
Acute elemental mercury toxicity manifestations?
Respiratory distress and renal failure
Chronic mercury toxicity syndromes?
Chronic elemental mercury toxicity causes two charactristic syndroms:acrodynia and erethism
Acrodynia syndrome?
Acrodynia (feer syndrome) in chronic elemental mercury toxicity manifest with autonomic symptoms (sweating, hemodynamic instability) and a desquamative erythematous rash on the palms and soles It is associated with incraesed urinary catecholamines and can in many ways mimic pheochromocytoma.
Erethism syndrome?
Erethism in chronic elemental mercury toxicity manifest with a central nervous disorder manifesting as personality change, irritability, and fine motor disturbances.
Organic mercury toxicity manifestations?
Visual field consriction, peripheral neuropathy, tremor, and hearing loss.
True or false: inorganic mercury crosses the placenta, resulting in cardiac malformations
False, inorganic mercury crosses the placenta, resulting in infants with sever neurologic impairment.
True or fase: 24-hour urine collection is essential for diagnosis of organic mercury poisoning?
False, 24-hour urine collection is essential for diagnosis of elemental (not organic) mercury poisoning. Organic mercury is not significantly excreted in urine.
Suitable samples in organic mercury toxicity?
Whole blood or hair analysis.
Digoxin half life?
36 hours
True or false: Digoxin is primarily excreted by the kidney?
True.
Suitable time for Digoxin level samplng?
8-12 hours after the last does
True or false: serum digoxin level is the best way to evaluate the eficacy and the toxicity of digoxin?
While serum digoxin level is important, both eficacy and the toxicity of digoxin may best measured by clinical parameters (heart rate, etc)
Factors that incraese digoxin toxicity?
Hypokalemia, hypercalcemia, hypomagnesemia, hypoxia, hypothyroidism, quinidine, and calcium chanel blockers.
True or false: Quinidine can decrease the risk of digoxin toxicity?
False. Quinidin in addition to enhancing end-organ effects of digoxin, frees digoxin from protein binding sites and impires digoxin clearance and enhances the risk of digoxin toxicity.
Digoxin-like Immunoreactive substances (DLIS)
DLIS or endogenous digoxin-like substance (EDLS) are found in the blood of some individual who are not taking digoxin and cause cross-reactivity. It is particularly common in neonates, pregnant women, liver and renal failure.
True or false: Procainamid is cleared by the kidney
False, it cleared by the liver
What is the procainamid metabolite in the liver?
N-acetylprocainamide (NAPA). The rate of procainamid conversion to NAPA is determined by concentration of hepatic acetyltransferase.
True or false: High levels of NAPA are in slow acetylators
False, In fast acetylator, who have genetically high levels of acetyltransferase, will higher level of NAPA NAPA
True or false: The clearance of NAPA is prdominantly renal
TRUE
True or false: Only the metabolite of procainamide, NAPA is biologically active
False, both procainamide and NAPA have bilologic activity, and the level of both should be measured.
True or false: the drug-induced lupus effect of procainamide is dose-dependent
Fase, the drug-induced lupus effect of procainamide is not dose-dependent
Which organ is mainly responsible for clearing aminoglycosides from blood?
The Kidneys
The ideal time for the gentamycin trough and peak specimens drawning?
The trough specimen is drawn immidietely before a dose, and the peak is drawn 30minutes after the completion of a does.
The class IA antiarrythmic agents?
Quinidine, disopyramide, and procainamide.
The mechanism of class IA antiarrythmic agents?
Blocking the sodium and potassium channels in cardiac conduction fibers.
The typical manifestations of quinidine toxicity?
Altered mental status, cinchonism (tinnitus, vertigo, blurred vision), a widening of the QRS complex, aprolonged QT interval, and hypotension ( without compensatory tachycardia)
Common conditions related to the prolonged QT interval, CNS changes, and hypotension?
Quinidine, TCAs, phenothiazines toxicity, and electrolyte abnormalities.
Which organ clears quinidine?
Liver
The most reliable tool to predict quinidine toxicity?
ECG. Quinidine levels are not routinely used to predict quinidine toxicity. IT is important to check and correct electrolyte levels.
Phenytoin toxicity manifestations?
Early manifestations of phenytoin toxicity include ocular dismotility (particularly horizontal gaze nystagmus), ataxia, and incoordination. More sever toxicity is associated with altered mental status and cardiac conduction disturbances.
True or fase: oral phenytoin overdose has not been associated with significant cardiac conduction abnormality?
True, in contrust to oral phenytoin, IV phenytoin overdose often produces a prolonged PR interval (AV block), hypotension and the arrhythmia.
True or false: Arrhythmogenic effects of IV phenytoin intoxication is due to the propylene glycol diluent of phenytoin.
TRUE
Fetal hydantoin syndrome?
The result of intrauterine exposure to phenytoin. It is carachterized by intrauterine growth retardation, microcephaly, mental retardation, midfacial hypoplasia, hypertelorism, a flattened philtrum, and shortened nose.
The therapeutic range of lithium?
0.4-1.2 mmol/L
The lithium level at which the risk of adverse effects becomes extemely high?
1.5 mmol/L
True or false: Some toxicity can be seen in the lithium therapeutic range
True, the margin between therapeutic effect and toxicity is a narrow one for lithium, and some toxicity can be seen in the upper reahces of lithium therapeutic range.
True of false: For routine lithium monitoring, a sample should be taken at 12 hours following the last does?
TRUE
The litium half-life?
It varies from 8-40 hours (depending largely upon age and renal function)
When steady state would be expected after starting lithium therapy?
Between 2 and 8 days.Checking levels after a period of 3 days to 1 week is recommended.
True or false: Amiodarone levels should not necessarily be routinely monitored.
TRUE
The main Amiodarone metabolite?
Desethylamiodarone
The major risks due to amiodarone?
Pulmonary toxicity (incidence of 1% annually), thyroid toxicity, hepatotoxicity (0.6%), and peripheral neuropathy (0.3%),
Hyperthyroidism is the most common manifestation of the amiodarone thyroid toxicity?
Fase, amiodarone thyroid toxicity apears with hypothyroidism in 5%-15%, and hyperthyroidism in 1%-2% of cases.
Routine tests for monitoring the patients who are taking amiodarone?
Thyroid and liver function tests, in addition chest X-ray and ECG
The recommended intervals for the routine monitoring tests in amiodarone therapy?
Thyroid and liver function tests, at baseline and every 6 months, chest X-ray and ECG annually.
True or false: amiodarone increases the clearance of warfarin?
False, it decreases the clearance of warfarin and can thereby prolong the INR.
How amiodarone can affect the digoxin concentrations?
Amiodarone increases digoxin concentration, and doses of digoxin must be reduced.
The major lipids found in plasma?
Cholestrol, triglyceride, and phospholipid.
Triglyceride composition?
TG is composed of 3 fatty acids extending from a glycerol group.
Short, medium, and long fatty acids chain definition?
Fatty acids are linear hydrocarbons that come in several lengths: short chain (4-6 carbons), mediumchain (8-12 carbons), and long chain (>12 carbons)
Saturated, monounsaturated, polyunsaturated fatty acids definition?
Saturated fatty acids: have only single bonds between carbons and thus have maximum number oa attached hydrogen bonds Monounsaturated FAs: have one double bond and thus one fewer hydrogen Polyunsaturated: have several double bonds and less hydrogen
Phospholipid composition?
Two fatty acids extending from a glycerol group, with the third spot on glycerol occupied by a phosphatidyl group ( such as phosphatidylcholine).
Cholestrol composition?
it is composed of 4 rings, a hydrogen side chain, and a hydroxyl group.
Lipoprotein constituents?
Cholestrol, triglyceride, and phospholipid, and apolipoproteins
What kind of lipoproteins is made by enterocytes?
Chylomicrons
The chylomicrons function?
It transports lipid from entrocytes to other somatic cells, particularly hepatocytes.
The chylomicrons apolipoproteins?
B-48, A-1, C, E
The VLDL apolipoproteins?
B-100, C, E
The VLDL function?
It is produced in the liver and is the vehicle for transporting of TG to somatic cells.
What are major risks with amiodarone therapy?
Pulmonary toxicity, hepatotoxicity, thyroid toxicity, peripheral neuropathy
What test are recommended every 6 months of amiodarone therapy?
Thyroid function tests, LFTs
What test are recommended every year on amiodarone therapy?
Chest Xray, EKG
What effect does amiodarone have on warfarin treatment?
It reduces Warfarin clearance and Prolong INR
How much time is required for above mentioned effect to occur when used with warfain?
7 weeks
What effect does amiodarone have on digoxin treatment?
digoxin concentartion is increased
What major lipids are found in plasma?
Cholestrol, triglycerides, phospholipids
What are Triglycerides composed of?
3 fatty acid extending from glycerol group
What are saturated fatty acids?
Fatty acids having single bonds between carbons and hydrogen ions
What are monosaturated fatty acids?
there is one double bond btw carbon and hydrogen
Which lipoprotein transfers lipids from enterocytes to somatic cells?
Chylomicron
Lipids are ingested by hepatocytes by which receptor?
apolipoprotein E
What happen to ingested lipis in hepatocytes?
They are converted to VLDL
What apolioproteins are associated with VLDL?
B-100,C and E
What is the function of VLDL?
Transports triglycerides to somatic cells
What happens to VLDL in blood?
It is converted to IDL the LDL by action of lipoprotein lipase
What is the function of LDL?
Transports cholestrol to somatic cells
what apolioproteins are associated with LDL?
B-100
LCAT( lecithin cholestrol acyl transferase) is produced by which organ?
Liver
what apolipoproteins are associated with chylomicrons?
B-48,A-1, C,E
How is cholestrol measured?
spectrophotometric method
Calculate VLDL from TG?
TG/5 if expressed in mg/dl, TG/2.2 if expressed in mmol/dl
At what value of TG is this equation invalid?
If TG isgreater than 400mg/dL, chylomicrons or beta VLDL is present
How is HDL measured?
By enzymatic sequence after removal of non HDL cholestrol
What is Friedewald equation?
LDL cholestrol= total cholesterol-HDL-TG/5
When Is this equation invalid?
If TG isgreater than 400mg/dL, chylomicrons or beta VLDL is present
What are direct methods of LDL measurement?
the methods are tedious. Ultracentrifugation, electrophoresis,homogenous assays
What is the principle involved in ultracentrifugation?
Liporoteins can be separated on the basis of difference in densities
what is the pattern of separation of lipoproteins in electrophoresis?
Chylomicron donot move from point of application,LDL migrates to beta region, VLDL in pre beta region, HDL goes to alpha region
After refrigeration if plasma has creamy top layer, what does this indicate?
Increased chylomicrons
After refrigeration if plasma looks opaque, what does this indicate?
increased HDL,VLDL or LDL
What is major apolipoprotein in HDL?
Apo-A1
What is the consequence of high LDL or IDL?
premature atherosclerosis
What is the consequence of high TG?
eruptive xanthomas
tendinous xanthomas are seen in elevation of what lipoproteins?
IDL ( raised TG and cholesterol)
Xanthelesma are seen in elevation of what lipoproteins?
LDL
Acute pancreatitis is seen in elevation of which lipoproteins?
chylomicrons, VLDL(TG= >5-10mmol/L)
What are clinical features of familial LPL deficiency?
xanthomas, pancreatitis
What are clinical features of familial apo C-II deficiency?
panreatitis
what is phenotype of apolipoprotein deficiency?
Type II b
What is phenotype of dysbetalipoproteinemia ?
Type III
What are clinical features of familial combined hyperlipidemia?
premature atherosclerosis
what is predominant hypercholestrolemia?
total cholestrol >200 mg/dL
Which lipoprotein is predominantly elevated in predominant hypercholestrolemia?
LDL
What is the cause of familial hypercholestrolemia?
LDL receptors deficiency
What is low levell of HDL?
< 35 mg/dL
What is Tangier diease?
It is autosomal recessive, low cholesterol, nl to increase TG, absent HDL and APO-A1
What happen in Tangier disease?
Cholestrol esters deposit in tonsils,lymphnodes, vessels, spleen with development of corneal opacities
What are recommendations of 3rd Adult Treatment Panel Report?
Fasting lipoprotein profile including total cholesterol, HDL, LDL and TG
What is desirable total cholestrol?
< 200
What is desirable LDLl?
<100
What is desirable HDL?
>60
What is principle of capillary electrophoresis?
Capillary tube is immersed in sample, volatge is applied and elution time is noted.
What are advantages of capillary zone electrophoresis?
small sample size, ability to automate procedure, very sensitive
Which is equivalent to albumin in fetal blood?
alpha feto protein
What happens to albumin in inflammatory conditions?
It decreases.
Which protein is amyloid precursor protein in cardiac amyloidosis?
prealbumin
How does prealbumin enter CSF?
It is secreted by choroid plexus
Why prealbumin band becomes prominent in heparinsed patients?
There is alteration of B-lipoproteins that migrates in prealbumin region
What happens to pre-albumin in inflammatory conditions?
It is decreased
what is alpha2- macroglobulin?
Protease inhibitor
What happens to alpha2- macroglobulin in nephrotic syndrome and why?
It is retained due to its large size so its concentration increase 10 fold
A false poistive ceruloplasmin level is seen in what conditions?
pregnancy, inflammatory conditions
Does haptoglobulin binds to myoglobin?
No
Which form of transferrin is superior to GGT in diagnosing chronic alcholism
Carbohydrate deficient transferrin
C reactive protein in found in which region of SPEP?
gamma region
what vaues of CRP areassociated with cardiac events
. 2-3 mg/L
Symptoms of hypoglycemia can be classified into two types of responses: Fasting or Reactive

What is the cause of each?
Fasting: caused by brain hypoglycemia, not rapid in onset

Reactive: caused by adrenergic activity, tends to be rapid
How do the symptoms differ in reactive vs. fasting hypoglycemia?
Fasting: mental status changes

Reactive: sweating, tachycardia, nervousness
What are 4 different drugs that can induce hypoglycemia (classes, not specifics)?
Alcohol, sulfonylureas and other oral hypoglycemics, insulin, and quinine
What is nesidioblastosis?
islet Beta cell proliferation
What is Whipple's triad and what is it associated with?
plasma glucose <45mg/dL, hypoglycemic changes, and correction of symptoms by glucose administration
In a patient with a suspected insulinoma, what do you expect the insulin level to be?
The absolute insulin level may actually be normal, but will be inappropriately high for the degree of hypoglycemia. Or it might be high.
What should the insulin:glucose ration be in insulinoma?
>180
What are some causes of reactive hypoglycemia?
The abnormal rapid-onset hypoglycemia following a meal can be caused by heriditary fructose intolerance, galactosemia, dumping syndrome (post vagotomy), rarely early type 2 diabetes
Pancreatic islet cells secrete what two substances in equimolar amounts?
Insulin and C-peptide
Which substance secreted by pancreatic islet cells is metabolized faster?
Insulin is metabolized faster than c-peptide.
What is the primary clinical use of C-peptide?
Since it hangs around so much longer than insulin but is secreted in equal amounts you can determine if insulin is being naturally released or exogenously administered.
What is the shortcoming in using the C-peptide to evaluate for exogenous administration of insulin?
Renal failure impedes C-peptide clearance and will result in elevated C-peptide levels
How can proinsulin be used clinically?
Proinsulin should represent less than 10% of the insulin level, so an elevated level may represent insulinoma
Anti-insulin antibodies can be seen in what 3 clinical settings?
Administration of insulin produces (rare), autoimmune insulin syndrome, and rarely insulinoma (actually all of these are rare)
What is the cause of hypoinsulinemic hypoglycemia?
Too much insulin (insulinoma or exogenous)
What are the two types of hyperinsulinemic hypoglycemia?
Ketotic and non-ketotic
Non-ketotic hypoglycemia suggests what clinical settings?

How about ketotic hypoglycemia?
presence of insulin-like activity such as in autoimmune hypoglycemia, starvation, liver failure

Ketotic: all other causes
Since insulinoma may be associated with MEN1, what additional test should be performed?
Serum calcium (remember PTH adenomas?)
How can you diagnose hypoglycemia after death?
Virtually impossible

You can detect insulin, peptide C, and proinsulin for several days after death
What is the cause of Type 1 diabetes?
autoimmune destruction of islet B cells
What percentage of diabetics are type 1?
~10%
T/F

Autoantibodies are not part of the clinical picture in Type I Diabetes
False

anti-GAD65, anti-ICA512, Anti-IAA can each be present
What is the cause of Type 2 Diabetes?
progressive insulin resistance with subseuent burn-out of islet cells
What is the recommeded test to diagnose diabetes in nonpregnant persons?

What is the diagnostic level?
Fasting plasma glucose

> or = 126 mg/dL is considered diagnostic
A 60yo obese man presents with frequent urination, recent weight loss, poorly healing wounds, and fatigue. You suspect diabetes, however he has eaten breakfast that morning and a fasting glucose cannot be performed. What can you do?
A random plasma glucose of >200mg/dL with symptoms of diabetes is diagnostic
What is the diagnostic level for diabetes with the oral glucose tolerance test?
75g of glucose is given and a 2 hour reading is taken. >200 mg/dL is diagnostic.
What fasting plasma glucose level is defined as "impaired fasting glucose/pre-diabetic"
99mg/dL
What is the diagnostic level in the glucose tolerance test for "Impaired glucose tolerance"/prediabetes
>139mg/dL at 2 hours after 75 grams
How is the gestational diabetes oral glucose tolerance test different than the normal fasting glucose tolerance test?
The amount of glucose is different
What types of women can avoid glucose tolerance testing totally?
young (<25y), healthy, normal pre-pregnancy weight, low-risk ethnicity, no family history and no prior OB complications
What is the earliest screening for high risk women for GDM?
a fasting plasma glucose (>126mg/dL) is diagnostic
Non-high risk women and high risk women who "pass" their first screening should undergo what test next?
At 24-28 weeks they should have a 100g load oral glucose tolerance test.
What are the criteria for a positive 100g OGTT?
Need two of the following:
>95 mg/dL fasting
>180 mg/dL 1hour
>155 mg/dL 2 hour
>140 mg/dL 3 hour
Often, women are "pretested" before undergoing the full 3 hour/100g OGTT? What does this consist of?
a 50g 1 hour OGTT: A measurement of >140mg/dL at one hour necessitates the 3 hour test.
A 28 year-old woman is diagnoses with gestational diabetes and goes on to deliver a (large) healthy baby with no complications. What additional followup does mom need?
Testing for GDM at 6-12 weeks postpartum
Should the HbA1c test be used for diabetes diagnosis?
NO
What does the HbA1c test monitor?
Long term glycemic control
What is the American Diabetes Association recommended goal of therapy level of HbA1C?
<7%
Name 4 other lab values that are important to check in diabetics?
Lipid disorders, microalbunemia, serum creatinine for estimated glomerular filtration rate, hypomagnesemia
Who gets diabetic ketoacidosis?
Insulin-dependent diabetics
What are the 3 usual requirements for the diagnosis of DKA?
hyperglycemia (>200mg/dL), ketosis, metabolic acidosis (venous pH < 7.3 or bicarb less than 15mmol/L)
What are the urinalysis findings usually seen with DKA?
glycosuria and ketonuria
T/F: DKA is often associated with leukopenia.
False. DKA is often associated with neutrophilia (that is not necessarily infectious in nature--unknown mechanism) But remember--DKA can be prompted by infection
What are the three major serum ketones in DKA?
acetone, acetoacetic acid, B-hydroxybutyrate
What laboratory technique is used for measuring ketones?
nitroprusside technique sensitive to acetone and acetoacetic acid but not B-hydroxybutyrate.
What is the shortcoming in our laboratory analysis of ketones?
Our nitroprusside technique measures only acetone and acetoacetic acid which accounts for only 20% of the serum ketones. B-hydroxybutyrate is converted to the other two measurable forms during treatment, so an apparent "bump" might be seen in the lab levels even with successful treatment
What do you expect glucose level to be in DKA?
>200mg/dL
Is bicarbonate decreased or increased in DKA?

Is pH decreased or increased in DKA?
Bicarb is down, pH is down (acidosis)
Why is Potassium elevated in DKA?
The Potassium is extracellular with the low pH, but the urinary excretion is increased, so true body potassium can be very LOW. When given insulin, the potassium can shift very quickly.
What is the typical BUN level during DKA?
increased due to severe volume depletion resulting in prerenal azotemia.
Which patients get Hyperglycemic hyperosmolar nonketotic coma?
Type 2 (non-insulin dependent) diabetics
Which is more common, DKA or HHNC?
DKA
Which has higher mortality, DKA or HHNC?
HHNC
What is the typical glucose level in HHNC?
at least 600mg/dL (may be >1000!)
What is the typical osmolarity in HHNC?
>330 mOsm/L
What is the pH/Bicarb level in HHNC?
normal
What is the ketone level in HHNC?
normal
What is the potassium level in HHNC?
may be elevated with true total body deficit (as in DKA)
What is the BUN level in HHNC?
Very elevated secondary to dehydration
What is Syndrome X?
Metabolic syndrome, the insulin resistance syndrome referring to the cluster of hyperlipidmia, impaired glucose tolerance, central obesity, increased CRP, HTN--association with cardiac disease
Why is it not cost-effective to screen for cancer with tumor markers?
The sensitivity of a test is dependent on the prevelence in the population, thus if you screen everyone you negate the good points of the test
What is the "Hook effect" in immunoassay?
When very high concentration overwhelm binding capacity of both capture and signal antibodies, thus yielding a low or even negative result.
Why is the Hook effect a concern in tumor marker testing?
Widely metastatic disease can have super high tumor marker levels, and lead to the Hook effect
How can you adjust for the Hook effect?
Perform the assay at multiple dilutions
What are heterophile antibodies?
Antiblodies that may be present in the patient that have wide reactivity with antibodies of other species.
How can one adjust for heterophile antibodies?
heterophile blocking reagents, remove immunoglobulins, serial dilutions
What are other situations that may show elevated PSA (besides cancer)?
BPH, prostatitis, prostatic infarct, and following needle biopsy of the prostate
What is the level of PSA that, beyond which, is rarely associated with benign disease?
10ng/mL--above this level you will typically not see a benign process
What is the approximate percentage of men that will be found to have prostatic carcinoma with elevated PSA (>4.0 ng/mL)?
30-40% of men
1. What is the benefit of age specific PSA? 2. At what expense is the benefit obtained?
1. increases sensitivity of PSA for younger men and increases specificity for older men 2. decreased sensitivity in older men
1. What is the definition of PSA density? 2. What value is considered abnormal? 3. T/F PSA density improves the performance of PSA
1. PSA divided by the estimated prostatic volume 2. > 0.15 3. false
1. What is PSA velocity? 2. What level is considered abnormal? 3. PSA velocity does increase sensitivity and specificity but what is its limitation?
1. rate of change of successive PSA measurements 2. >0.75 ng/mL/year 3. biologic variation of PSA from non-malignant causes
In what form does PSA normally circulate?
1. 75-90% is bound to protease inhibitors mostly alpha-1-antichymotrypsin but also alpha-2-macroglobulin and alpha-1-protein inhibitor
Free PSA can be measured, how does it correlate with prostate cancer?
low free PSA fraction (increased bound fraction) = low likelihood of cancer, high likelihood when free PSA < 10%
Which PSA measurement should be taken only after several weeks have passed since urethral instrumentation and digital instrumentation
% Free PSA
What volume of prostatic fluid is the advantage of % free PSA seen over total serum PSA?
< 60 cc
T/F Free PSA is stable and specimens need not be separated and frozen.
False- specimens that can't be processed within 2 hrs should be separated and frozen
In what form is PSA released from glandular cells and how is it broken down?
Initially pro-PSA --> 7 aa leader peptide clipped --> mature PSA. Anomalous clipping can lead to several additonal pro-PSA forms.
Detection of what molecules has led to improved diagnosis of prostate cancer in men with PSA in 2.5-4.0 range?
pro-PSA and truncated forms due to anomalous clipping (both are increased in prostate cancer)
What is benign PSA (bPSA) and why is it clinically useful?
form of PSA first isolated in transistion zone of benign prostatic hyperplasia (BPH). Can discriminate BPH from non-BPH prostate enlargement
In prostate cancer what does preoperative serum PSA quantity reflect?
tumor volume and stage (roughly)
1. What is the expected serum PSA level following radical prostatectomy? 2. How are levels used after surgery?
1. should drop to undetectable levels 2. used to screen for tumor recurrence or metastases
After what time period does the correlation between PSA and tumor recurrence strengthen?
after 5 years
What is the definition of PSA recurrence per the American Society for Therapeutic Radiology and Oncology (ASTRO)? (2)
3 consecutive increases PSA or a single rise so great as to trigger the initiation of hormone therapy
When is the date of treatment failure in prostate cancer?
the midpoint between the post-radiation nadir PSA and the first of three consecutive increases
T/F Biochemical treatment failure and clinical failure in prostate cancer occur at approximately the same time.
False - biochemical failure precedes clinical failure by approximately 6-18 months
What may be a better indicator of pending treatment failure in prostate cancer than serum PSA quantity?
PSA doubling time
What factors affect CEA levels in patients with colorectal cancer? (7)
tumor stage, tumor grade, tumor ploidy, tumor site, obstruction, liver function, and smoking
What's the relationship between CEA level and colon cancer tumor grade?
Inversely related i.e. well differentiated tumors = higher CEA and vice versa
What percent of tumors at the following stages have elevated CEA levels? 1. confined to colon 2. positive nodes 3. distant metastases
1. 25% 2. 50% 3. 75%
T/F Left sided colon cancers have higher CEA levels than right sided tumors.
TRUE
How does bowel obstruction affect CEA?
higher CEA levels
T/F diploid colorectal tumors produce higher CEA levels than aneuploid
FALSE
How does liver dysfunction affect CEA?
increases CEA
How do the median CEA levels of smokers compare to nonsmokers?
higher
How does preoperative CEA affect patient outcome?
higher CEA = worse predicted outcome
Post-treatment surveillance of CEA (serial measurements): 1) sensitivity and specificity to detect recurrence? 2) sensitivity to liver metastases? 3)overall sensitivity to locoregional metastases?
1) 80% and 70% 2) 94% 3)60%
What other neoplastic conditions are associated with CEA elevations? (7)
gastric adenoCA (esp. well-differentiated intestinal type), breast cancer, lung cancer, pancreatic adenoCA, medullary thyroid CA, cervical adenoCA, and urothelial carcinoma
What nonneoplastic conditions are associated with elevated CEA? (7)
smoking, peptic ulcer disease, inflammatory bowel disease, pancreatitis, hypothyroidism, biliary obstruction, and cirrhosis
CEA levels above __ ng/mL are rarely due to benign disease?
10
What's the mainstay for detection of follicular and papillary thyroid carcinomas (so called differentiated)?
serum thyroglobulin
Which thyroid tumors are NOT associated with increased thyroglobulin?
anaplastic and medullary
What's the half-life of circulating thyroglobulin and how long before it becomes undectectable following total thyroidectomy?
65 hours, approximately 1 month
How is thyroglobulin normally cleared from circulation?
Catabolism in the liver and recycling in the thyroid
1. What percentage of normal individuals have anti-thyroglobulin antibodies? % with thyroid carcinoma? 2. how does this affect estimation of serum thyroglobulin and what assay allays this problem?
1. 10% normal, >20% with carcinoma 2. usually results in underestimation, assays are available to detect thyroglobulin antibodies
1. What is an alternative to serum thyroglobulin measurement as a tumor marker? 2. What would an increase of the above answer suggest?
1. serial quantitative serum anti-thyroglobulin antibody 2. antigenic stimulation, suggesting recurrence
1. Under what conditions is thyroglobulin assay sufficiently specific? 2. Addition of what can enhance the assay's specificity after the above requirement(s) have been met?
1. ablation of all functional thyroid tissue (total thyroidectomy followed by radioactive iodine) 2. TSH can be administered or thyroxine replacement can be withheld prior to testing
Tumor-associated trypsin inhibitor (TATI) has been used as a marker with what neoplasms?
mucinous ovarian carcinoma, urothelial carcinoma, and renal cell carcinoma
Tumor-associated trypsin inhibitor (TATI): 1. method of clearance? 2. affect of renal failure on serum levels? 3. how does degree of elevation correlate with severity of disease? 4. poor prognosis associated with what value? 5. What inflammatory condition is associated with elevated levels?
1. kidneys 2. elevated 3. direct relationship i.e. higher levels = greater severity 4. >70 micrograms/L 5. pancreatitis
1) What percentage of stage I ovarian mucinous tumors are associated with increased Tumor-associated trypsin inhibitor (TATI)? 2) Stage IV?
1) 40% 2) nearly 100%
1) What percentage of pancreatic adenoCA are associated with increased Tumor-associated trypsin inhibitor (TATI)? 2) What condition limits its specificity in pancreatic adenoCA?
1) 85-95% 2) pancreatitis
1. Tumor-associated trypsin inhibitor (TATI) is elevated in what percentage of gastric carcinomas? 2. TATI can complement which tumor marker in gastric CA?
1. 60% (infiltrative signet ring type) 2. CEA which is only elevated in intestinal type
1) Elevated tumor-associated trypsin inhibitor (TATI) what percentage of urothelial carcinoma 2)sensitivity for renal cell carcinoma
1) varies with tumor stage from 20% low stage to 80% advanced stage 2) 70% (most likely elevated in advanced stage)
CA-125 1) elevated and used for monitoring in what neoplasm? 2) why hasn't it proven useful for screening?
1) non-mucinous epithelial ovarian neoplasms 2) elevated in only 50% of pts with stage I disease, poor positive predictive value in unselected women
CA 125 elevations associated with which: 1) nonneoplastic conditions? (6) 2) non-ovarian neoplasms?(5)
1) pregnancy, fibroids, benign ovarian cysts, pelvic inflammation, ascites, endometriosis 2) endometrium, fallopian tube, pancreas, breast, and colon
CA-125 1) what level is considered normal? 2) what happens to levels in postmenopausal women? 3) normal values in black and Asian women compared to others? 4) how does it fluctuate with menstrual cycle?
1) < 35 U/mL 2) decreases 3) lower 4) increases slightly during follicular (proliferative) phase
How does CA-125 help differentiate pelvic masses in postmenopausal women?
levels > 65 U/mL has a positive predictive value of > 95% for ovarian malignancy
Rising CA-125 following treatment suggests relapse. This precedes clinical evidence of disease by how long?
3-6 months
What preoperative CA 125 level is considered unfavorable in terms of prognosis?
> 65 U/mL
How does CA 125 level correlate with prognosis... 1) half-life of CA 125 following chemotherapy? 2) time to normalization after chemo?
1) patients with half-life >20 days have improved survival 2) patients whose level normalized within 3 cycles have improved survival
CA-125 greatest utility is in women with known ovarian malignancy 1) what trend in CA 125 following initial treatment suggests treatment efficacy? 2) rate of fall correlates best with? 3) increase during treatment suggests? 4) increase following treatment suggests?
1) falling levels 2) duration of disease-free survival 3) treatment failure 4) disease recurrence
1) Persistent elevation of CA 125 above what level predicts residual disease? 2) With what specificity?
1) > 35 U/mL 2) 95%
How does CA 125 level correlate with prognosis... 1) half-life of CA 125 following chemotherapy? 2) time to normalization after chemo?
1) patients with half-life >20 days have improved survival 2) patients whose level normalized within 3 cycles have improved survival
What are CA 27.29 (aka BR 27.29) and CA 15-3?
they're different epitopes of a single antigen - the protein product of breast cancer associated MUC1 gene
Which is the preferred serum marker for breast cancer CA27.9 or CA15-3 and why?
CA27.9 due to enhanced sensitivity and specificity. Elevated in ~30% low stage disease and 60-70% of advanced-stage disease
What benign conditions are associated with elevated: 1) CA27.9 and CA15-3? (3) 2) CA 15-3 only?
1) benign ovarian cysts, benign breast disease, and benign liver disease 2) cirhosis, sarcoidosis, and lupus
CA 27.9 may be elevated in what non-breast malignancies? (5)
colon, stomach, pancreas, prostate, and lung
ER and PR assays: 1) performed almost exclusively by what method? 2) how are they graded?
1) immunohistochemistry 2) according to the proportion of cells expressing intense nuclear staining
1. What are the 2 methods for determining Her-2 status? 2. What's the purpose of testing?
1. immunohistochemistry and FISH 2. determine suitability for treatment with trantuzumab
1) What's the best marker for pancreatico-biliary adenoCA? 2) what is its greatest utility in the above disease? 3) what other malignancies can be associated with elevations of this marker? (2) 4) in what benign conditions might it be elevated? (3)
1) Cancer Antigen (CA) 19-9 2) monitoring treatment response 3) esophagus and intestines 4) pancreatitis, cholestasis, cholangitis, and cirrhosis
1) What percentage of individuals with CA 19-9 levels > 100 U/mL have benign disease? 2) > 1000 U/mL
1) ~3% 2) virtually none
1) What blood antigen is CA 19-9 identical to? 2) what group of individuals don't produce it?
1) Lewis blood group antigen 2) Lewis-negative
1) What is the major component of fetal serum? 2) Where is it synthesized?
1) Alpha Fetoprotein (AFP) 2) yolk sac, fetal liver, and fetal GI tract
1) What's the normal level of AFP in adults? 2) Values above ___ are rarely associated with benign disease?
1) under 5.4 ng/mL 2) 500 ng/mL
In what benign conditions is AFP normally elevated? (3)
pregnancy (usually not >100 ng/mL), hepatitis, and cirrhosis
1) AFP has utility in detection of what tumors? (2) 2) it may be sporadically elevated in other tumors, most notably what?
1) hepatocellular carcinoma and yolk sac tumors 2) hepatoid variant of gastric cancer
How is AFP used in yolk sac tumors?
magnitude of AFP correlates with prognosis (levels > 10,000 ng/mL associated with poor prognosis)
1) What's the reported rate of AFP elevation in hepatocellular carcinoma? 2) is this true in the Western world? why or why not?
1) 80% 2) no, in other areas tumors may be detected when quite large at time of diagnosis
What conditions are associated with elevated Human Chorionic Gonadotropin (HCG)? (4)
pregnancy, trophoblastic disease, and choriocarcinoma, and marijuana use (low level elevation)
In what percentage of seminomas is HCG elevated?
0.15
1) Where is Beta2-Microglobulin (B2M) expressed? 2) when and where is it released? 3) why is it a good but nonspecific tumor marker?
1) surface of most nucleated cells, non-covalently linked to class I MCH molecules 2) released into the extracellular fluid when cells die 4) elevated whenever there's increased cell turnover
Beta2-Microglobulin 1) independent prognostic factor in what disease? 2) can be used to monitor what process? 3) how is cleared?
1) multiple myeloma 2) renal transplant rejection 3) by kidneys (nonspecifically elevated in renal insufficiency)
Lipid-Associated Sialic Acid in Plasma (LASA-P) is a nonspecific tumor marker elevated in what kind of malignancies? (5)
hematolymphoid neoplasms, tumors of the breast, GI tract, lung and ovary
Name three conditions in which alkaline phosphatase would be elevated?
Osteogenic sarcoma
Bone metastisis
Paget disease of the bone
Alkaline phosphatase is the most sensitive marker for what condition concerning the liver?
Hepatic metastisis
In the case of primary intra-abdominal carcinoids alkaline phosphatase correlates with what?
Prognosis
Which of the following types of carcinoid tumors is most commonly associated with high levels of serotonin?

A. Foregut
B. Midgut
C. Hindgut
D. Atypical carcinoids
E. no specific type is associated with high serotonin
B. Midgut
Which of the following tumors is associated with elevated plasma chromogranin A levels?

A. Pheochromocytoma
B. Carcinoid tumor
C. Pancreatic islet tumor
D. Small cell neuroendocrine tumor
E. all of the above
E. all of the above
Name several neuroendocrine tumors (5- list not all inclusive)
Carcinoids
Islet tumors
Medullary thyroid carcinomas
Pheochromocytomas
Neuroendocrine
What cells are carcinoid neoplasms composed of?
Enterchromaffin cells

Endocrine cells (EC). The secretions are seen here as eosinophilic granules (SG).

Also shown in this image are two Paneth cells, an entirely different type of secretory resident cell of the intestine; and one that's not any sort of endocrine cell at all, despite some similarities of appearance. Paneth cells secrete their product into the lumen; and their granules (PCG) are much larger and coarser than those of the true endocrine cells.
Which immunohistochemical pattern is most consistent with medullary thyroid carcinoma?

A. Calcitonin (-), CEA (-), thyroglobulin (+)
B. Calcitonin (+), CEA (-), thyroglobulin (+)
C. Calcitonin (+), CEA (+), thyroglobulin (+)
D. Calcitonin (+), CEA (+), thyroglobulin (-)
E. Calcitonin (-), CEA (+), thyroglobulin (+)
D. Calcitonin (+), CEA (+), thyroglobulin (-)
Carcinoid neoplasms are capable of producing what neurotransmitter?
Serotonin

What is the rate limiting step in the biosynthesis of seratonin?
Conversion of tryptophan to 5-hydroxytryptamine by tryptophan hydroxylase
What cell takes up 5-hydroxytryptamine and stores it in its secretory (dense) granules?
Platelets
Is seratonin cleared by the kidneys or liver?
Liver
Seratonin is dumped into the portal system.
What are the symptoms of carcinoid syndrome?
Diarrhea
Flushing
Wheezing

What two chemicals can be assayed in a urine sample to detect possible seratonin excess?
5-HIAA (a serotonin metabolite)
Serotonin

These can be falsely elevated if the patient has been consuming what food?
Platelet serotonin appears to be the most accurate marker for the detection of what tumor?
Carcinoid
Plasma calcitonin is useful to detect what type of carcinoma?
Medullary carcinoma


At the center and to the right is a medullary carcinoma of thyroid. At the far right is pink hyaline material with the appearance of amyloid. These neoplasms are derived from the thyroid "C" cells and, therefore, have neuroendocrine features such as secretion of calcitonin.
What is the normal level of calcitonin in a individual?
<10ng/L, some affected individuals have leves this low, provocative testing using what type of drugs (3) can make the test more sensitive?
Pentagastrin
Omeprazole
Or calcium
This test may need to be done in families with MEN II

Current assays for calcitonin measure what form?
Thyroglobulin is not elevated in medullary carcinoma.

True or false?
TRUE
CEA is commonly elevated in __________ carcinoma. Higher values correlate with greater dedifferentiation and suggests a worse prognosis.
Medullary

A 28 year old male has suffered from FUO, high blood pressure, diarrhea, and rapid heart rate for several years. Examination revealed enlarged cervical lymph nodes and thyroid nodularity. A biopsy was taken, as were several blood tests. After determining he carried a specific genetic mutation he was diagnosed with Sipple Syndrome (MEN 11-B).



A mutation in what gene was found?

RET
STAT 1
BCR/ABL
RET

The cause of most medullary thyroid carcinoma cases is unknown. However, some are associated with MEN (multiple endocrine neoplasia). One of these diseases is Sipple Syndrome (MEN 11-B). It is an autosomal dominant inherited syndrome that includes bilateral C cell (medullary) carcinoma, pheochromocytoma and hyperthyroidism. Both males and females carrying this gene usually become symptomatic in their thirties. The cancer cell of origin is the C cell which is the producer of calcitonin.
Paragangliomas and pheochromocytomas originate from what cell type?
Chromaffin cells

These cells are capable of secreting what molecules?
Catecholamines
Norepinephrine cen be converted to epinephrine by the action of what enzyme?
PNMT

Phenolethanolamine-N-methylTRANSFERASE

Tumors also have the capability to convert norepinephrine to epinephrine. Therefore adrenal tumors will secrete both norepinephrine and epinephrine. Is this true for extra-adrenal tumors?
No.

What do they secrete?
Norepinephrine is metaboilzed to normetanephrine. Most normetanephrine is conjugated to sulfate and excreted in urine, and some is metabolized to what compound?
VMA. Or. Vanillylmandelic acid

Epinephrine is also metabolized to VMA, however the intermediate is what compound?
Metanephrine
Fractionation of catecholamines or metanephrines can be useful in what way in diagnosing a pheochromocytoma?
It can help with suggesting a location (paraganglioma ((extra-adrenal mainly secrete norepinephrine)) vs. pheochromocytoma)
Certain anti hypertensive drugs can interfere with the following assays: urinary VMA, urinary metanephrines, urinary catecholamines, plasma metanephrines, or plasma catecholamines, which of the following are included in this list?


A. Imipramine
B. Reserpine
C. Guanethidine
D. Nitroglycerine
E. MAO inhibitors
F. All of the above
F. All of the above

Imipramine
Reserpine
Guanethidine
Nitroglycerine
MAO inhibitors
Which of the following tests is considered most accurate for initial screening for a pheochromocytoma?

A. urinary VMA
B. urinary metanephrines
C. urinary catecholamines
D. plasma metanephrines
F. plasma catecholamines
D. Free (unconjugated) Plasma metanephrines

Why is this?
Plasma metanephrines better reflect long-term catecholamine secretion.

This is because catecholamines are released in an episodic nature, so plasma catecholamines have poor sensitivity.

What test can be done to clarify equivocal serum and urine tests?
What two metabolites are found in most cases of neuroblastoma?
Vanillylmandelic acid
and
Homovanillic acid

HVA is the final metabolic product of what two compounds?
DOPA and dopamine