• 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

1/260

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;

260 Cards in this Set

  • Front
  • Back
1. What are astrocytes?
The astrocytes are found in the CNS and are star-shaped cells that provide physical and nutritional support for neurons. During development of the CNS, the astrocytes guide neuronal migration to their final adult position, and form a matrix that keeps them in place.

These cells phagocytose debris left behind by cells, provide lactate (from glucose metabolism) as a carbon source for the neurons), and control the brain EC ionic environment.

Astrocytes help to regulate the content of the ECF by taking up, processing, and metabolizing nutrients and waste products.
2. What are oligodendrocytes?
Oligodendrocytes provide the myelin sheath that surrounds the axon, acting as insulation for many neurons in the CNS.

Oligodendrocytes can form myelin sheaths around multiple neurons in the CNS by sending out processes that bind to the axons on target neurons. Along with astrocytes, they also form a supporting matrix for the neurons.

They have a limited capacity for mitosis, and if damaged, do not replicated. If this occurs, demyelination of the axons may occur.
3. What are Schwann cells?
Schwann cells are the supporting cells of the PNS. Like oligodendrocytes, they also form myelin sheaths around the axons, but they can only myelinate one axon.

Schwann cells also clean up cellular debris in the PNS. The Schwann cells also provide a means for peripheral axons to regenerate if damage. There is a synergistic interaction among the Schwann cells, secreted growth factors, and the axon that allows damaged axons to reconnect to the appropriate target axon.
4. What are microglial cells?
The microglial cells are the smallest glial cells in the nervous system. They serve as immunologically responsive cells that function similarly to the action of macrophages in the circulation.

Microglial cells destroy invading micro-organisms and phagocytose cellular debris.
5. What are ependymal cells?
The ependymal cells are ciliated cells that line the vacities (ventricles) of the CNS and the spinal cord.

In some areas of the brain, the ependymal cells are functionally specialized to elaborate and secrete CSF into the ventricular system. The beating of the ependymal cilia allow for efficient circulation of the CSF throughout the CNS.

Recent data has found that cells within the ependymal layer can act as neural stem cells which can regenerate neurons.
6. What is the function of the CNS?
The CSF acts as both a shock absorber protecting the CNS from mechanical trauma and as a system for the removal of metabolic wastes.

The CSF can be aspirated from the spinal canal and analyzed to determine whether disorder of CNS function, with their characteristic CSF changes, are present.
7. What is the BBB?
The BBB begins with the endothelial cells that form the inner lining of the vessels supplying blood to the CNS.

These cells are joined by tight junctions that do not permit the movement of polar molecules from the blood into the interstitial fluid bathing the neurons. They also lack mechanisms for transendothelial transport that are present in other capillaries of the body. These mechanisms include fenestrations or transpinocytosis.
8. What are the five important components of the BBB?
1. Tight junctions btwn endothelial cells
2. Narrow intercellular spaces
3. Lack of pinocytosis
4. Continuous basement membrane
5. Astrocyte extension
9. What is the role of the endothelial cells in the BBB?
These cells serve actively and passively to protect the brain. B/c they contain a variety of drug-metabolizing enzyme systems similar to those in the liver, the endothelial cells can metabolize neurotransmitters and toxic chemicals and thus form an enzymatic barrier to entry of these substances.

They actively pump hydrophobic molecules that diffuse into endothelial cells back into the blood (esp xenobiotics) with P-glycoproteins, which act as transmembranous, ATP-dependent efflux pumps).
10. What molecules can passively diffuse across the BBB?
Lipophilic substances, water, oxygen, and carbon dioxide can readily cross the BBB via passive diffusion.

Other molecules depend on specific transport systems. Differential transporters on the luminal and abluminal endothelial membranes can transport compounds into as well as out of the brain.
11. What is the role of the basement membrane in the BBB?
Further protection against the free entry of blood-borne compounds into the CNS is provided by a continuous collagen-containing basement membrane that completely surrounds the capillaries.

The BM appears to be surrounded by the foot processes of astrocytes.
12. How is glucose transported to the brain?
GLucose is transported thru both endothelial membranes by facilitated diffusion via the GLUT-1 transproter.

GLUT-3 transporters present on the neurons then allow the neurons to transport the glucose from the ECF.

Glial cells express GLUT-1 transporters.
13. What happens to the energy metabolism of the brain when blood glucose levels fall?
Normally the rate of glucose transport into the ECF normally exceeds the rate required for energy metabolism by the brain. However, glucose transport may become rate-limiting as blood glucose levels fall below the normal range.

Thus, people begin to experience hypoglycemic symptoms at approx 60 mg/dL, as the glucose levels are reduced to or below the Km of the GLUT-1 transporters in the endothelial cells of the BBB barrier.
14. How are large neutral amino acids transported into the brain?
Large neutral AAs, such as phenylalanine, leucine, tyrosine, isoleucine, valine, tryptophan, methionine, and histidine, enter the CSF rapidly via a single AA transporter (L-leucine-preferring system AA transporter).

B/c a single transporter is involved, these AAs compete with each other for transport into the brain.
15. What about the entry of small neutral AAs?
The entry of small neutral AAs, such as alanine, glycine, proline, and GABA, is marked restricted b/c their influx could dramatically change the content of neurotransmitters.

They are synthesized in the brain, and some are transported out of the CNS and into the blood via the A(alanine)-preferring system carrier.
16. What is receptor mediated transcytosis?
Certain proteins, such as insulin, transferrin, and insulinlike growth factors, cross the blood-brain barrier by receptor-mediated transcytosis.

Once the protein binds to its membrane receptor, the membrane containing the receptor-protein complex is endocytosed into the endothelial cell to form a vesicle. It is released on the other side of the endothelial cell.
17. What are the two basic structural categories of molecules that serve as neurotransmitters?
1. Small nitrogen-containing molecules
-glutamate, GABA, glycine, ACh, dopamine, norepinephrine, serotonin, and histamine, NO

2. Neuropeptides
-endorphins, GH, TSH
18. What are the general features of neurotransmitter synthesis?
Most of the transmitters are synthesized from AAs, intermediates of glycolysis and the TCA cycle, and O2 in the cytoplasm of the presynaptic terminal. The rate of synthesis is generally regulated to correspond to the rate of firing of the neuron.

Once synthesized, the transmitters are stored in storage vesicles by an ATP requiring pump linked with the proton gradient. Release from the vesicle is triggered by an action potential that depolarizes the membrane and causes an influx of calcium ions. This causes the fusion of the vesicle with the synaptic membrane and release of the neurotransmitter.
19. What determines the action of the neurotransmitter?
The action of the neurotransmitter is terminated thru reuptake into the presynaptic terminal, uptake into glial cells, diffusion away from the synapse, or enzymatic inactivation.

The enzymatic inactivation may occur in the postsynaptic terminal, the presynaptic terminal or an adjacent astrocyte microglial cell, or in endothelial cells in the brain capillaries.
20. How is dopamine, norepinephrine, and epinephrine synthesized?
These three neurotransmitters are synthesized in a common pathway from the AA L-tyrosine.

*The first and rate limiting step in the synthesis of these neurotransmitters from tyrosine is the hydroxylation of the tyrosine ring by tyrosine hydrolase. The product formed is called dopa.

The second step is the decarboxylation of dopa to form dopamine.

Most stop here, but to get to norepinephrine, dopamine is hydroxylated by dopamine beta hydroxylase.

Epinephrine synthesis is dependent on the presence of adequate levels of vitamin B12 and folate.
21. Where does the synthesis of the catecholamines take place?
Ordinarily, only low concentrations of catecholamines are free in the cytosol, whereas high concentrations are found within the storage vesicles.

Conversion of tyrosine to L-dopa and that of L-dopa to dopamine occurs in the cytosol. Dopamine is then taken up into the storage vesicles.

In norepinephrine containing neurons, the final beta-hydroxylation reaction occurs in the vesicles.
22. Where does the storage and release of catecholamines occur?
The catecholamines are transported into VMAT. The mechanism that concentrates the catecholamines in the storage vesciles is an ATP-dependent process linked to a proton pump. Protons are pumped into the vesicles via a vesicular ATPase (V-ATPase).

The protons then exchange for the positively charged catecholamine via the transporter VMAT2. The influx of the catecholamine is thus driven by the H+ gradient across the membrane.

In the vesicles, the catecholamines exist in a complex with ATP and acidic proteins known as chromogranins.
23. What are the two roles of the vesicles?
1. They maintain a steady supply of catecholamines at the nerve terminal that is available for immediate release

2. They mediate the process of release When an action potential reaches the nerve terminal, the calcium influx promotes the fusion of vesicles with the neuronal membrane. The vesicles then discharge their soluble contents, including the neurotransmitters, ATP, chromogranins, and DBH, into the extraneuronal space by the process of exocytosis.
24. What are two of the major reactions in the process of inactivation and degradation of catecholamines?
1. Monoamine oxidase
-present on the outer mitochondrial membrane of many cells

2. Catechol-O-methoyltransferase (COMT)
-also found in many cells, including RBCs
25. What is MAO?
MAO is present on the outer mitochondrial membrane of many cells and oxidizes the carbon containing amino group to an aldehyde, thereby releasing ammonium ion. In the presynaptic terminal MOA inactivates catecholamines that are not protected in storage vesicles. Thus, drugs that deplete storage vesicles indirectly increase catecholamine degradation.

There are two isoforms of MAO. MAO in the liver and other sites protects against the ingestion of dietary biogenic amines such as the tyramine found in many cheeses.
26. What are the two isoforms of MAO?
MAO-A preferentially deaminates norepinephrine and serotonin

MAO-B acts on a wide spectrum of phylethylamines (this refers to a -CH2- group linked to a phenyl ring).
27. What is COMT?
COMT works on a broad spectrum of extraneuronal catechols and those that have diffused away from the synapse. COMT transfers a methyl group from SAM to a hydroxyl group on the catecholamine or its degradation product.

B/c the inactivation reaction requires SAM, it is indirectly dependent on vitamin B12 and folate.
28. In the CSF, what is an indicator of dopamine degradation?
CSF homovanillylmandelic acid (HVA) is an indicator of dopamine degradation.

Its concentration is decreased in the brain of pts with Parkinson's disease.
29. What regulates tyrosine hydroxylase?
Tyrosine hydroxylase, the first committed step and rate-limiting enzyme in the pathway, is regulated by feedback inhibition that is coordinated with depolarization of the nerve terminal.

Tyrosine hydroxylase is inhibited by free cytosolic catecholamines that compete at the binding site on the enzyme for the pterin cofactor (tetrahydrobiopterin).

Depolarization of the nerve terminal activates tyrosine hydroxylase, along with other protein kinases, (PK-C and PK-A) can CAM kinases that phosphorylate tyrosine hydroxylase and cause it to bind BH4 more tightly.
30.. What about the long-term regulation of tyrosine hydroxylase?
A long-term process involves alterations in the amts of tyrosine hydroylase and dopamine beta-hydroxylase present in the nerve terminals.

When sympathetic neuronal activity is increased for a prolonged period, the amts of mRNA that code for tyrosine hydroxylase and dopamine-beta-hydroxylase are increased.

This increased gene transcription may be the result of phosphorylation of CREB by protein kinase A. CREB then binds to the CRE in the promoter region of the gene.
31. How is serotonin synthesized and degraded?
The first enzyme of the pathway, tryptophan hydroxylase, uses an enzymic mechanism to hydroxylate the ring structure of tryptophan.

The second step of the pathway is a decarboxylation reaction catalyzed by the same enzyme that decarboxylates DOPA.

Serotonin, like the catecholamines, can be inactivated by MAO.
32. How is melatonin produced?
The neurotransmitter melatonin is also synthesized from tryoptophan.

Melatonin is produce din the pineal gland in response to the light-dark cycle, it level in the blood rising in a dark environment. It is probably thru melatonin that the pineal gland conveys info about light-dark cycles to the body. Melatonin also may be involved in regulating reproductive functions.
33. What cells release histamine in the brain?
Within the brain, histamine is produced both by mast cells and by certain neuronal fibers. Mast cells are a family of bone marrow-derived secretory cells that store and release high concentrations of histamine.

They are prevalent in the thalamus, hypothalamus, dura mater, leptomeninges, and choroid plexus.

Histaminergic neuronal cell bodies in the human are found in the tuberomamillary nucleus of the posterior basal hypothalamus. The fibers project into nearly all areas of the CNS, including the cerebral cortex, the brainstem, and the spinal cord.
34. How is histamine synthesized?

Where is it stored?
Histamine is synthesized form histidine in a single enzymatic step. The enzyme histidine decarboxylase requires pyridoxal phosphate, and its mechanism is very similar to that of dopa decarboxylase.

Histamine is stored in the nerve terminal vesicles. Depolarization of nerve terminals activates the exocytotic release of histamine by a voltage-dependent as well as a calcium dependent mechanism.
35. Once released from neurons, what happens to histamine?
Histamine is thought to activate both postsynaptic and presynaptic receptors.

Unlike other neurotransmitters, histamine does not appear to be recycled into the presynpatic terminal to any great extent.

However, astrocytes have a specific high-affinity uptake system for histamine and may be the major sites of the inactivation and degradation of this monoamine.
36. How is histamine degraded?
The first step in the inactivation of histamine in the brain is methylation. The enzyme histamine methyltransferase transfers a methyl group from SAM to a ring nitrogen of histamine to form methylhistamine.

The second step is oxidation by MAO-B, followed by an additional oxidation step.

In peripheral tissues, histamine undergoes deamination by diamine oxidase, followed by oxidation to a carboxylic acid.
37. How is ACh synthesized?
The synthesis of ACh from acetyl coenzyme A and choline is catalyzed by the enzyme choline acetyltransferase (ChAT). This synthetic step occurs in the presynaptic terminal. The compound is stored in vesicles and later released thru calcium-mediated exocytosis.

Choline is taken up by the presynaptic terminal from the blood via a low-affinity transport system and from the synaptic cleft via a high-affinity transport mechanism.

It is also derived from the hydrolysis of phosphatidylcholine in membrane lipids.
38. How is choline made?
Choline is a common component of the diet but also can be synthesized in the human as part of the pathway of the synthesis of phospholipids. The only route for choline synthesis is via the sequential addition of three methyl groups from SAM to the ethanolamine portion of phosphatidylethanolamine to form phosphatidylcholine.

Phosphatidylcholine is subsequently hydrolyzed to release choline or phosphocholine. This conversion is folate and vitamin B12 dependent.
39. Where does the acetyl group used for ACh synthesis come from?
The acetyl group is derived principally form glucose oxidation to pyruvate and decarboxylation fo pyruvate to form acetyl CoA via the pyruvate dehdyrogenase reaction.

This is b/c the neuronal tissues have only a limited capacity to oxidize fatty acids to acetyl CoA, so glucose oxidation is the major source of acetyl groups. Pyruvate dehydrogenase is found only in mitochondria.
40. How is ACh inactivated?
Via acetylcholinesterase, which is a serine esterase that forms a covalent bond with the acetyl group. The enzyme is inhibited by a wide range of compounds that form a covalent bond with this reactive serine group. This is how Sarin nerve gas works.

ACh is the major neurotransmitter at the neuromuscular junctions; inability to inactivate this molecule leads to constant activation of the nerve-muscle synapses, a condition that leads to varying degrees of paralysis.
41. How is glutamate synthesized?
Glutamate = excitatory neurotransmitter within the CNS. Within nerve terminals, glutamate is generally synthesized de novo from glucose rather than taken up from the blood, b/c its plasma concentration is low and it does not readily cross the BBB.

It is synthesized primarily from the TCA cycle intermediate alpha-ketoglutarate. via glutamate dehydrogenase or via transamination.
42. What is GABA?
GABA is the major inhibitory neurotransmitter in the CNS.

It is synthesized by the decarboxylation of glutamate via glutamic acid decarboxylase. GABA is recycled in the CNS by a series of reactions called the GABA shunt, which conserves glutamate and GABA.
43. Much of the uptake of GABA occurs where?
In glial cells. The GABA shunt in glial cells produces glutamate, which is converted to glutamine and transported out of the glial cells to neurons, where it is converted back to glutamate.

Glutamine thus serves as a transporter of glutamate between cells in the CNS.

Glial cells lack GAD and cannot synthesize GABA.
44. What is aspartate?
Aspartate, like glutamate, is an excitatory neurotransmitter, but it functions in far fewer pathways. It is synthesized from the TCA cycle intermediate oxaloacetate via transamination reactions.

Like glutamate syntehsis, aspartate synthesis uses oxaloacetate that must be replaced thru anaplerotic reactions.

Aspartate cannot pass thru the BBB.
45. What is glycine?
Glycine is the major inhibitory neurotransmitter in the spinal cord. Most of the glycine in neurons is synthesized de novo within the nerve terminals from serine by the enzyme serine hydroxymethyltransferase, which requires folic acid.

Serine in turn is synthesized from the intermediate 3-phosphoglycerate in the glycolytic pathway. The action of glycine is probably terminated via uptake by a high affinity transporter.
46. What is the action of NO?

How is it made?
NO is synthesized from arginine in a reaction catalyzed by NO synthase.

The synthesis of NO occurs in response to a stimulator binding to a receptor on some cells or to a nerve impulse in neurons. NO enters smooth muscle cells, stimulating guanylate cyclase to produce cGMP, which causes smooth muscle cell relaxation. When the smooth muscle cells relax, blood vessels dilate.

It can readily cross cell membranes b/c it is a gas.
47. What is hypoglycemic encephalopathy?
Hypoglycemia signs reflect the appearance of physiologic protective mechanisms initiated by hypothalamic sensory nuclei such as sweating, palpitations, anxiety, and hunger. If these symptoms are ignored, they proceed to a more serious CNS disorder, progressing thru confusion and lethargy to seizures and eventually coma.

Prolonged hypoglycemia can lead to irreversible brain damage.
48. What occurs during hypoglycemic encephalopathy when the blood sugar falls to 2.5 - 2.0 mM?
As blood glucose falls below 2.5 mM, the brain attempts to use internal substrates such as glutamate and TCA cycle intermediates as fuels. B/c the pool size of these substrates is quite small, they are quickly depleted. If blood glucose levels continue to fall below 1 mM, ATP levels become depleted.

As the blood glucose drops from 2.5 to 2.0 mM, the symptoms appear to arise from decreased synthesis of neurotransmitters in particular regions of the brain rather than a global energy deficit.
49. What occurs during hypoglycemic encephalopathy when the blood sugar falls below 1 mM?
The EEG becomes isoelectric, and neuronal cell death ensues.

Cell death is not global in distribution; rather, certain brain structures, in particular hippocampal and cortical structures, are selectively vulnerable to hypoglycemic insult.
50. The pathophysiologic mechanisms responsible for neuronal cell death in hypoglycemia include...?
Glutamate excitotoxicity. Glutamate excitotoxicity occurs when the cellular energy reserves are depleted.

The failure of the energy-dependent reuptake pumps results in a buildup of glutamate in the synaptic cleft and overstimulation of the postsynaptic glutamate receptors. The prolonged glutamate receptor activation leads to prolonged opening of the receptor ion channel and the influx of lethal amounts of calcium ion, which can activate cytotoxic intracellular pathways in the postsynaptic neuron.
51. What is hypoxic encephalopathy?
When mild to moderate hypoxia occurs, there is diminished cognitive function due to impaired neurotransmitter synthesis. In mild hypoxia, cerebral blood flow increases to maintain oxygen delivery to the brain.

In addition, anaerobic glycolysis is accelerated, resulting in maintenance of ATP levels. This occurs, however, at the expense of an increase of lactate production and a fall of pH. Acute hypoxia (PaO2 < 20 mmHg) generally results in a coma.
52. What occurs during hypoxic encephalopathy?
All forms of hypoxia result in diminished neurotransmitter synthesis.

Inhibition of pyruvate dehydrogenase diminished ACh synthesis, which is acutely sensitive to hypoxia.

Glutamate and GABA synthesis, which depend on a functioning TCA cycle, are decreased as result of elevated NADH levels, which inhibit TCA cycle enzymes. NADH levels are increased when oxygen is unavailable to accept electrons from the ETC.

Even the synthesis of catecholamine neurotransmitters may be decreased b/c the hydroxylase reactions require O2.
53. What is the relationship between glutamate synthesis and the anaplerotic pathways of pyruvate carboxylase and methylmalonyl CoA mutase?
Synthesis of glutamate removes alpha-ketoglutarate from the TCA cycle, thereby decreasing the regeneration of oxaloacetate in the TCA cycle. B/c oxaloacetate is necessary for the oxidation of acetyl CoA, oxaloacetate must be replaced by anaplerotic reactions.

There are two major types of anaplerotic reactions: (1) pyruvate carboxylase and (2) the degradative pathway of the branched chain AAs, valine and isoleucine, which contribute succinyl CoA to the TCA cycle. This pathway uses vitamin B12 but not folate in the reaction catalyzed by methylmalonyl CoA mutase.
54. What types of fatty acids does the BBB restrict?
The BBB restricts the entry of nonessential FAs, such as palmitate, that are released from adipose tissue or present in the diet. B/c the BBB significantly inhibits the entry of certain FAs and lipids into the CNS, virtually all lipids found there must be synthesized within the CNS.

Conversely, essential FAs are taken up by the brain (e.g., linoleic and linolenic acid).
55. What is an important type fatty acid that is synthesized in the brain?
Very long chain fatty acids are synthesized int he brain, where they play a major role in myelin formation.

As a result, peroxisomal fatty acid oxidation is important in the brain b/c the brain contains very long-chain fatty acids and phytanic acid, both of which are oxidized in the peroxisomes by alpha oxidation.
56. What is Refsum disease?
Disorders that affect peroxisome biogenesis (such as Refsum) severely affect brain cells b/c of the inability to metabolize both branched chain and very long chain FAs.
57. What is the predominant cerebroside?
Galactosylcerebroside, which has a single sugar attached to the hydroxyl group of the sphingosine.

Galactocerebrosides pack more tightly together than phosphatidylcholine; the sugar, although polar, carries no positively charged AA or negatively charged phosphate. The high cholesterol content of the membrane also contributes to the tight packing.
58. What are the two major myelin structural proteins in the CNS?
1. Proteolipid protein (PLP)
2. Myelin basic proteins (MBPs)
59. What is PLP?
The PLP is a very hydrophobic protein that forms large aggregates in aqueous solution and is relatively resistant to proteolysis.

*It is highly conserved in sequence among species. Its role is thought to be one of promoting the formation and stabilization of the multilayered myelin structure.
60. What are the MBPs?
The MBPs are a family of proteins. Unlike PLP, MBPs are easily extracted from the membrane and are soluble in aqueous solution. The major MBP has no tertiary structure; it is located on the cytoplasmic face of myelin membranes.

***Antibodies directed against MBPs elicit experimental allergic encephalomyelitis.
61. What is the major myelin structural protein in the PNS?
Po, a glycoprotein that accounts for > 50% of the PNS myelin protein content. Po is thought to play a similar structural role in maintaining myelin structure as PLP does in the CNS.

MBPs are also found in the PNS. The major PNS-specific MBP is P2.
62. Most of the signs and symptoms of catecholamine excess can be masked by...?
Phenoxybenzamine, a long acting α1- and α2-receptor antagonist, combined with a β1- and β2-receptor blocker such as propranolol.

This therapy alone is reserved for pts with inoperable pheochromocytomas.
63. What is the primary injury to the CNS in MS?
The primary injury to the CNS in MS is the loss of myelin in the white matter, which interferes w/nerve conduction along the demyelinated area.

The CNS compensates by stimulating oligodendrocytes to remyelinate the damaged axon, and when this occurs, remission is achieved.
64. What is a consequence of the remyelination in MS?
Often remyelination leads to a slowing in conduction velocity b/c of a reduced myelin thickness (speed is proportional to myelin thickness) or a shortening of the internodal distances (the action potential has to be propagated more times).

Eventually, when it becomes too difficult to remyelinate large areas of the CNS, the neuron adapts by upregulating and redistributing along its membrane ion pumps, to allow nerve conductance along demyelinated axons. Eventually this adaptation also fails, and the disease progresses.
65. Treatment of MS is based on...?
Treatment of MS is now based on blocking the action of the immune system. B/c antibodies directed against cellular components appear to be responsible for the progression of the disease, agents that interfere with immune responses have had various levels of success in keeping pts in remission for extended periods.
66. What mutation leads to Charcot-Marie-Tooth polyneuropathy syndrome?
Inherited mutations in Po (the major PNS myelin protein) leads to a version of Charcot-Marie-Tooth polyneuropathy syndrome.

The inheritance pattern for this disease is autosomal dominant, indicating that the expression of one mutated allele leads to the expression of the disease.
67. What mutation leads to Pelizaeus-Merzbacher disease and X-linked spastic paraplegia type 2 disease?
Mutations in PLP (the major myelin protein in the CNS) lead to Pelizaeus-Merzbacher disease and X-linked spastic paraplegia type 2 disease.

These diseases display a wide range of phenotypes, from a lack of motor development and early death (most severe) to mild gait disturbances. The phenotype displayed depends on the precise location of the mutation within the protein.

An altered function of either Po or PLP leads to demyelination and its subsequent clinical manifestations.
68. What is facilitated glucose transporter protein type 1 (GLUT1) deficiency syndrome?
In this disorder, GLUT1 transporters are impaired, which results in a low glucose concentration in the CSF (called hypoglycorrhachia).

A diagnostic indication of this disorder is that in the presence of normal blood glucose levels, the ratio of CSF glucose to blood glucose levels is < 0.4.

Clinical features are variable but include seizures, developmental delay, and a complex motor disorder. These symptoms are the result of inadequate glucose levels in the brain. The disorder can be treated by prescribing a ketogenic diet. This will force the pt to produce ketone bodies, which can spare the brain's requirement for glucose as an energy source.
69. The mental retardation in untreated PKU and maple syrup urine disease may be attributable to...?
High levels of either phenylalanine or branched-chain amino acids in the blood.

These high levels overwhelm the large, neutral AA carrier, so excessive levels of the damaging AA enter the CNS.

In support of this theory is the finding that treatment of PKU pts with large doses of LNAAs that lack phenylalanine resulted in a decrease of phenylalanine levels in the CSF and brain, with an improvement in the pts cognitive functions as well.
70. What is reserpine?

How did it help the understanding of depression?
Reserpine blocks catecholamine uptake into vesicles and had been used as an antihypertensive and antiepileptic drug for many years, however, it was noted that a small percentage of pts on the drug become depressed and even suicidal.

Thus, a link was forged between monoamine release and depression in humans.
71. Why are chromogranins necessary?
Chromogranins are required for the biogenesis of the secretory vesicles. When chromogranins are released from the vesicles, they can be proteolytically clipped to form bioactive peptides.

*Elevated levels of chromogranins in the circulation may be found in pts who have neuroendocrine tumors, such as pheochromocytomas.
72. In albinism, what enzymes may be defective?
In albinism, either the copper-dependent tyrosine hydroxylase of melanocytes (which is distinct from the tyrosine hydroxylase found in the adrenal medulla) or other enzymes that convert tyrosine to melanins may me defective.
73. What is tyramine?
Tyramine is a degradation product of tyrosine that can lead to headaches, palpitations, nausea and vomiting, and elevated blood pressure if it is present in large quantities.

Tyramine mimics norepinephrine and binds to norepinephrine receptors, stimulating them.

Tyramine is inactivated by MAO-A, but if a person is taking and MAOI, foods containing tyramine should be avoided.
74. What are the first generation of MAOIs?
The first generation of MAOIs, exemplified by iproniazid, which was originally used as an antituberculosis drug and was found to induce mood swings in pts) were irreversible inhibitors of both the A and B forms of MAO.

Although they did reduce the severity of depression (by maintaining higher levels of serotonin), these drugs suffered from the cheese effect.

Cheese and other foods contain tyramine. Usually tyramine is inactivated by MAO-A, but if an individual is taking an MAOI, tyramine levels increase.
75. What about the next generation of MAOIs?
When it was realized that MAO existed in two forms, selective irreversible inhibitors were developed; exampled include clorgyline for MAO-A, and deprenyl for MAO-B.
76. What is the role of deprenyl?
Deprenyl - MAO-B inhibitor; has been used to treat Parkinson's disease. Deprenyl, however, is not an antidepressant.
77. What is the role of clorgyline?
Clorgyline (MAO-A inhibitor) is an antidepressant but suffers from the cheese effect.
78. What is moclobemide?
This is a third generation MAOI. Moclobemide is a specific, reversible inhibitor of MAO-A and is effective as an antidepressant.

More important, b/c of the reversible nature of the drug, the cheese effect is not observed, b/c as tyramine levels increase, they displace the drug from MAO, and the tyramine is safely inactivated.
79. What is Redux?
Redux increases the secretion of serotonin. Drugs that can increase serotonin levels may be able to control appetite and depression.

Redux was a second generation drug developed from fenfluramine, a known appetite suppressant. Redux acts as an SSRI but also increase the secretion of serotonin, leading to elevated levels of this compound in the synapse.
80. Why was fenfluramine ineffective?
When it was first used, it could not be resolved between two distinct optical isomers. The l-isomer induced sleepiness, so to counteract this effect, fenfluramine was often given with phentermine, which elevated epinephrine levels to counteract the drowsiness (known as fen/phen).

Once the two optical isomers of fenfluramine could be resolved, Redux, dexfenfluramine, was developed.
81. What is the result of histamine secretion?
Histamine is the major mediator of the allergic response. It leads to vasodilation and an increase in the permeability of blood vessel walls. When it is released in the lungs, the airways constrict in an attempt to reduce the intake of the allergic material. The ultimate result of this, however, is bronchospasm, which can lead to difficult in breathing.
82. Is histamine excitatory or inhibitory in the brain?
In the brain, histamine is an excitatory neurotransmitter. This is why antihistamines cause drowsiness.
83. What causes the neurologic symptoms in vitamin B12 deficiency?
It is believed that the vitamin B12 requirement for choline synthesis contributes to the neurolgic symptoms.

The methyl groups for choline synthesis are donated by SAM, which is converted to S-adenosyl homocytsteine in the reaction.

The formation of SAM thru recycling of homocysteine requires both tetrahydrofolate and vitamin B12 (unless extra amounts of methionine are available to bypass the B12-dependent methionine synthase step).
84. What can be supplemented in the diet to increase choline synthesis?
Supplementation of the diet with lecithin (phosphatidylcholine) has been sued to increased brain ACh in pts suffering from tardive dyskinesia (often persistent involuntary movements of the facial muscles and tongue).

The neonate has a very high demand for choline. High levels of phosphatidylcholine in maternal milk and a high activity of a high affinity transport system thru the BBB for choline in the neonate help to maintain brain choline requirements.
85. How does the fetus obtain choline?
There is a high affinity transport system for choline across the placenta. The choline is derived from maternal stores, maternal diet, and synthesis primarily in the maternal liver.

B/c choline synthesis is dependent on folate and B12, the high fetal demand may contribute to the increased maternal requirement for both vitamins during pregnancy.
86. What are the results of an inherited pyruvate dehydrogenase deficiency, a thiamine deficiency, or hypoxia?
In all these conditions, the brain is deprived of a source of acetyl CoA for ACh synthesis, as well as a source of acetyl CoA for ATP generation from the TCA cycle.
87. What is tiagabine?
Tiagabine is a drug that inhibits the reuptake of GABA from the synapse, and it has been used to treat epilepsy as well as other convulsant disorders.

B/c GABA is an inhibitory neurotransmitter in the brain, its prolonged presence can block neurotransmission by other agents, thereby reducing the frequency of convulsions.
88. What is the normal rate of cerebral blood flow?
The normal blood flow thru the brain of the adult person averages 50-65 mL per 100 g of brain tissue per minute.

For the entire brain, this amts to 750-900 ml/min, or 15% of the resting cardiac output.
89. What three metabolic factors have potent effects in controlling cerebral blood flow?
1. Carbon dioxide concentration
2. Hydrogen ion concentration
3. Oxygen concentration
90. How does carbon dioxide concentration in the arterial blood perfusing the brain effect cerebral blood flow?
An increase in carbon dioxide concentration greatly increases the cerebral blood flow.

Carbon dioxide is believed to increase cerebral blood flow by combining first with water in the body fluids to form carbonic acid, with subsequent dissociation of this acid to form hydrogen ions.

The hydrogen ions then cause vasodilation of the cerebral vessels - the dilation being almost directly proportional to the increase in hydrogen ion concentration up to a blood flow limit of about 2x normal.
91. How does the acidity of the brain tissue (hydrogen ion concentration) affect cerebral blood flow?
Any other substance that increases the acidity of the brain tissues will likewise increase cerebral blood flow.

Such substances include lactic acid, pyruvic acid, and any other acidic material formed during the course of tissue metabolism.
92. What is the importance of cerebral blood flow control by carbon dioxide and hydrogen ions?
Increased hydrogen ion concentration greatly depresses neuronal activity. Therefore, an increase in hydrogen ion concentration causes an increase in blood flow, which in turn carries hydrogen ions, carbon dioxide and other acid forming substances away from the brain tissues.

Thus, this mechanism helps maintain a constant hydrogen ion concentration in the cerebral fluids and thereby helps to maintain a normal, constant level of neuronal activity.
93. Oxygen deficiency in the brain causes does...?
If blood flow to the brain ever becomes insufficient to supply the needed amt of oxygen, the oxygen deficiency mechanism for causing vasodilation immediately causes vasodilation, returning the brain blood flow and transport of oxygen to the cerebral tissues to near normal.

Thus, this local blood flow regulatory mechanism is almost exactly the same in the brain as in coronary vessels, in skeletal muscle, and in most other ares of the body.
94. A decrease in cerebral tissue PO2 below ____ begins to increase cerebral blood flow?
Below about 30 mm Hg (normal value is 35 to 40 mm Hg).

This is important b/c brain function becomes deranged at not much lower values of PO2, esp so at PO2 levels below 20 mm Hg. Even coma can result at these low levels.
95. How does the blood flow in the brain change in response to neuronal activity?
It has become clear that blood flow in each segment of the brain changes as much as 100-150% within seconds in response to changes in local neuronal activity.
96. How is the cerebral blood flow regulated when the arterial pressure changes?
Cerebral blood flow is autoregulated extremely well between arterial pressure limits of 60-140 mm Hg. That is the mean arterial pressure can be between these values without significant changes in cerebral blood flow.

But, if the arterial pressure falls below 60 mm Hg, the cerebral blood flow then does becomes severely depressed.
97. What is the sympathetic innervation to the brain?
The cerebral circulatory system has strong sympathetic innervation that passes upward from the superior cervical ganglia in the neck and then into the brain along with the cerebral arteries. This innervation supplies both the large brain arteries and the arteries that penetrate into the substance of the brain.

However, transection of the sympathetic nerves or mild to moderate stimulation of them usually causes very little change in cerebral blood flow b/c the blood flow autoregulation mechanism can override the nervous effects.
98. What is the role of the sympathetic nervous system in controlling cerebral blood flow?
When mean arterial pressure rises acutely to an exceptionally high level, such as during strenuous exercise, the sympathetic nervous system normally constricts the large- and intermediate-sized brain arteries enough to prevent the high pressure from reaching the smaller brain blood vessels.

*This is important in preventing vascular hemorrhages in the brain (cerebral stroke).
99. What is the cerebral microcirculation?
The number of blood capillaries in the brain is greatest where the metabolic needs are greatest.

The overall metabolic rate of the brain gray matter where the neuronal cell bodies lie is about 4x as great as that of white matter; correspondingly, the number of capillaries and rate of blood flow are also about 4x as great in the gray matter.
100. What is an important structural characteristic of the brain capillaries?
They are much less "leaky" than the blood capillaries in almost any other tissue of the body.

A reason for this is that the capillaries are supported on all sides by glial feet, which are small projections from the surrounding glial cells that abut against all surfaces of the capillaries and provide physical support to prevent overstretching of the capillaries in case of high capillary blood pressure.
101. What happens to the walls of the small arterioles leading to the brain capillaries in people with HTN?
The walls of the small arterioles leading to the brain capillaries become greatly thickened in ppl with HTN. These arterioles remain significantly constricted all the time to prevent transmission of the high pressure to the capillaries.
102. What causes most strokes?
Cerebral strokes occur when cerebral blood vessels are blocked.

Most strokes are caused by arteriosclerotic plaques that occur in one or more of the feeder arteries to the brain. The plaques can activate the clotting mechanism of the blood, causing a blood clot to occur and block blood flow in the artery, thereby leading to acute loss of brain function in the localized area.
103. One of the most common types of stroke is blockage of what artery?
The middle cerebral artery that supplies the midportion of one brain hemisphere. For instance, if the MCA is blocked on the left side of the brain, the person is likely to become almost totally demented b/c of lost function in Wernicke's speech comprehension areas in teh left cerebral hemisphere, and he or she also becomes unable to speak words b/c of loss of Broca's motor area for word formation. In addition, loss of function of neural motor control areas of the left hemisphere can create spastic paralysis of most muscle son the opposite side of the body.
104. What about blockage of the PCA?
Blockage of a PCA will cause infarction of the occipital pole of the hemisphere on the same side as the blockage, which causes loss of vision in both eyes in the half of the retina on the same side as the stroke lesion.
105. Why are strokes involving the midbrain especially devastating?
This can block nerve conduction in major pathways between the brain and spinal cord, causing both sensory and motor abnormalities.
106. What is the capacity of the cerebral cavity? How much of it is taken up by the CSF?
Capacity of entire cerebral cavity = 1600-1700 mL

About 150 mL of this is occupied by the CSF and the remainder by the brain and cord.

The CSF is present in the ventricles of the brain, in the cisterns around the outside of the brain, and in the subarachnoid space around both the brain and the spinal cord.
107. What is a major function of the CSF?
To cushion the brain within its solid vault. The brain simply floats in the CSF.
108. What is a contrecoup injury?
When a blow to the head is extremely severe it may not damage the brain on the side of the head where the low is struck but on the opposite side.

If on same side as impact injury = coup injury
109. What parts of the brain are often the sites of injury and contusions after a severe blow to the head?
The poles and the inferior surfaces of the frontal and temporal lobes, where the brain comes into contact with bony protuberances in the base of the skull.
110. What is the rate of formation fo teh CSF?
The CSF is formed at a rate of about 500 mL each day, which is 3-4x as much as the total volume of fluid in the CSF system.

About 2/3's or more of this fluid originates as secretion from the chorioid plexuses in the four ventricles, *mainly in the two lateral ventricles.
111. After the CSF flows into the two lateral foramina of Luschka and a midline foramen of Magendie, where does it go?
It enters the cisterna magna, a fluid spaces that lies behind the medulla and beneath the cerebellum.

The cisterna magna is continuous with the subarachnoid space that surrounds the entire brain and spinal cord. Almost all the CSF then flows upward from the cisterna magna thru the subarachnoid spaces surrounding the cerebrum. From here, the fluid flows into and thru multiple arachnoidal villi that project into the large sinuses.
112. The choroid plexus projects into what four areas?
1. Temporal horn of right lateral ventricle
2. Temporal horn of left lateral ventricle
3. Posterior portion of the third ventricle
4. Roof of the fourth ventricle
113. Secretion of fluid into the ventricles by the choroid plexus depends mainly on...?
Active transport of sodium ions thru the epithelial cells lining the outside of the plexus.

The sodium ions in turn pull along large amts of chloride ions as well b/c the positive charge of the sodium ions attracts the chloride ion's negative charge.

The two of these together increase the quantity of osmotically active sodium chloride in the CSF, which then causes almost immediate osmosis of water thru the membrane, thus providing the fluid of the secretion.
114. What are the characteristics of the composition of CSF in comparison to the plasma?
Osmotic pressure, approx equal to that of plasma

Sodium ion concentration; also approx equal to that of plasma

Chloride ion, about 15% greater than in plasma

Potassium ion: approx 40% less

Glucose: about 30% less
115. Absorption of CSF occurs through...?
The arachnoidal villi are microscopic fingerlike inward projections of the arachnoidal membrane thru the walls and into the venous sinuses.

Conglomerates of these villi form macroscopic structures called arachnoidal granulations that can be seen protruding into the sinuses.
116. What three things are allowed relatively free flow in the arachnoidal villi?
1. CSF
2. Dissolved protein molecules
3. Particles as large as red and white blood cells into the venous blood
117. What is the pia mater and the perivascular space?
The large arteries and veins of the brain lie on the surface of the brain but their ends penetrate inward, carrying with them a layer of pia mater, the membrane that covers the brain.

The pia is only loosely adherent to the vessels, so that a space, the perivascular space, exists between it and each vessel.

Therefore, perivascular spaces follow both the arteries and the veins into the brain as far as the arterioles and venules go.
118. What is the lymphatic function of the vascular space?
A small amt of protein leaks out of the brain capillaries into the interstitial spaces of the brain. B/c no true lymphatics are present in brain tissue, excess protein in the brain tissue leaves the tissue flowing with fluid thru the perivascular spaces into the subarachnoid spaces. On reaching the subarachnoid spaces, the protein then flows with the CSF and is absorbed thru the arachnoidal villi into the large cerebral veins.

Therefore, perivascular spaces are a specialized lymphatic system for the brain.
119. What is the normal CSF pressure when one is lying in a horizontal position?
Avgs 130 mm of water (100 mm Hg), although is may be as low as 65 mm or as high as 195 even in the normal healthy person.
120. What regulates the CSF pressure?
The arachnoidal villi. They function like valves that allow CSF and its contents to flow readily into the blood of the venous sinuses while not allowing blood to flow backward in the opposite direction.

Normally, this valve action allows CSF to flow into the blood when CSF pressure is about 1.5 mm Hg greater than the pressure of blood in the venous sinuses. Then, if the pressure still rises, the valves open more widely.
121. What can block the villi?
In some disease state, the villi sometimes become blocked by large particulate matter, by fibrosis, or by excesses of blood cells that have leaked into the CSF in brain disease. Such blockage can cause high CSF pressure.
122. What can increase the CSF pressure?
1. Large brain tumors
2. Hemorrhage or infarction due to large numbers of RBCs and WBCs
3. Some babies are born with hydrocephalus due to an abnormally high resistance to fluid reabsorption thru the arachnoidal villi.
123. How does one measure the CSF pressure?
The person lies horizontal so the pressure in the spinal canal is equal to that in the cranial vault.

A spinal needle is then inserted into the lumbar spinal canal and the needle is connected to a vertical glass tube that is open to the air at its top. The spinal fluid is allowed to rise in the tube as high as it will.

Whatever level it rises to above the level of the needle, is indicative of the actual pressure.
124. High CSF pressure causes...?
Papilledema. When the pressure in the CSF rises, ti also rises inside the optic nerve sheath. The retinal artery and vein pierce this sheath a few mm behind the eye and then pass along with the optic nerve fibers into the eye itself.
125. What are three things that cause papilledema due to high CSF pressure?
1. High CSF pressure pushes fluid first into the optic nerve sheath and then along the spaces between the optic nerve fibers to the interior of the eyeball.
2. The high pressure decreases outward fluid flow in the optic nerves, causing accumulation of excess fluid in the optic disc
3. The pressure in the sheath also impeded flow of blood in the retinal vein, thereby increasing the retinal capillary pressure throughout the eye, which results in still more retinal edema.
126. What is noncommunicating hydrocephalus?
Usually the noncommunicating type is caused by a block in the aqueduct of Sylvius, resulting from atresia before birth in many babies or from blockage by a brain tumor at any age.

As fluid is formed in the 2 lateral and third ventricles, the volumes of these three ventricles increase greatly. This flattens the brain into a thin shell against the skull.
127. What is the communicating type of hydrocephalus?
The communicating type is usually caused by blockage of fluid flow in the subarachnoid spaces around the basal regions of the brain or by blockage of the arachnoidal villi where the fluid is normally absorbed into the venous sinuses.

Fluid therefore collects both on the outside of the brain and to a lesser extent inside the ventricles.
128. BBBs exist at the choroid plexus and at the tissue capillary membranes in essentially all areas of the brain parenchyma except...?
Except in some areas of the hypothalamus, pineal gland, and area postrema, where substances diffuse with greater ease into the tissue spaces.

The ease of diffusion in these areas is important b/c they have sensory receptors that respond to specific changes in the body fluids such as changes in osmolality and in glucose concentration, as well as receptors for peptide hormones that regulate thirst, such as angiotensin II. It also has specific carrier molecules that facilitate transport of hormones, such as leptin, to control appetite.
129. The BBBs are highly permeable/impermeable to...?
Permeable to water, carbon dioxide, oxygen, and most lipid soluble substances such as alcohol and anesthetics.

It has slight permeability to electrolytes such as sodium, chloride, and potassium.

It is totally impermeable to plasma proteins and most non-lipid-soluble large organic molecules.
130. What is the cause of the low permeability of the BBB?
The endothelial cells are joined together by tight junctions.
131. What is the usual cause of brain edema?
The usual cause is either greatly increased capillary pressure or damage to the capillary wall that makes the wall leaky to fluid.

A very common cause is a serious blow to the head, leading to brain concussion, in which the brain tissues and capillaries are traumatized so that capillary fluid leaks into the traumatized tissues.
132. What are the two vicious circles of brain edema once it begins?
1. Edema compresses the vasculature, which causes brain ischemia. The ischemia in turn causes dilation with sill further increase in capillary pressure. The increased capillary pressure then causes more edema fluid.

2. The decreased cerebral blood flow also decreases oxygen delivery. This increases the permeability of the capillaries, allowing still more fluid leakage. It also turns off the sodium pumps of the neuronal tissue cells, thus allowing these cells to swell in addition.

*Concentrated mannitol solution can be used to stop this.
133. What is the brain metabolic rate under resting but awake conditions?
The resting brain accounts for 15% of the total metabolism in the body. Therefore, even under resting conditions, brain metabolism per unit mass of tissue is about 7.5x the avg metabolism in non-nervous system tissues.

*Most of this excess metabolism of the brain occurs in the neurons not in the glial supportive tissues.
134. What is the major need for metabolism in the neurons?
Mainly to pump ions through their membranes; transport sodium and calcium ions to the outside of the neuronal membrane and potassium ions to the interior.
135. Is the brain capable of anaerobic metabolism?
The brain is not capable of much anaerobic metabolism. One of the reasons for this is the high metabolic rate of the neurons, so that most neuronal activity depends on second-by-second delivery of oxygen from the blood.
136. Is glucose delivery to the neurons dependent on insulin?
No. Therefore, there is no loss of mental function in diabetic pts.

However, when a diabetic pt is overtreated with insulin, the blood glucose levels fall so low that not enough glucose is left in the blood to supply the neurons properly and mental function does then become seriously deranged, sometimes leading to coma.
137. What do Neisseria look like?
Neisseria are Gram-negative diplococci that are flattened on the adjoining sides, giving the pair the shape of a coffee bean.

These aerobic bacteria have stringent nutritional requirements and grow best on enriched media such as lysed sheep's blood agar (chocolate agar).

The two clinically significant Neisseria are N. meningitidis and N. gonorrhoeae.
138. What is N. meningitidis?
N. meningitidis is a significant cause of bacterial meningitis, particularly among people ages 5-19. The organism is a common colonizer of the oropharynx and is spread by the respiratory route.

There are at least 13 serotypes of N. meningitidis. Invasive disease mainly occurs when people living in crowded quarters encounter new strains to which they are not immune..
139. Do all people get meningitis in the absence of an immune response to N. meningitidis?
Even in the absence of an immune response, only a small fraction of those infected get meningitis. The bacteria must invade respiratory epithelial cells and travel to the basolateral side of the cells to enter the blood.

Once in the blood, the capsule of the bacteria reduces opsonization and destruction of the bacteria by complement proteins. Regardless, the efficacy of complement against N. meningitidis is very good. Perople without MAC (C5-C9) have high rates of disease.
140. What is N. gonorrhoeae?
N. gonorrhoeae is an important cause of STD,. It is second only to Chlamydia as a causative agent of STD.

Infection in men causes urethritis. In women, infection is often asymptomatic and so might go untreated. Untreated infection can lead to pelvic inflammatory disease, which can cause infertility or ectopic pregnancy.
141. How does N. gonorrhoeae usually manifest?
As a local infection in the genital or cervical mucosa, pharynx, or anorectum, however, disseminated infections may occur. In ppl with deficient MAC, disseminated infection is highly likely.

Disseminated infection of adults and adolescents usually causes septic arthritis accompanied by a rash of hemorrhagic papules and pustules. Neonatal N. gonorrhoeae infection causes blindness, and rarely sepsis.
142. What are the virulence factors for Neisseria?
Neisseria organisms use antigenic variation to evade immune responses. Neisseria adhere to and invade non-ciliated epithelial cells at the site of entry. Bacterial persistence or invasion depends on escape from immune defenses.

The antigenic variation arises by expression of alternative genes for adhesive pili (which bind to CD46) and OPA proteins. OPA proteins increase binding of Neisseria to epithelial cells and promote entry of bacteria.
143. What is whooping cough?
Whooping cough is caused by the Gram-negative coccobacillus Bordetella pertussis, and is an acute, highly communicable illness characterized by paroxysms of violent coughing followed by a loud inspiratory "whoop".
144. What is the pathogenesis of whooping cough?
B. pertussis colonizes the brush border of the bronchial epithelium and also invades macrophages. *Coordinated expression of virulence factors is regulated by the Bordetella virulence gene locus (bvg). BVGS is a transmembrane protein that sense signals that induce expression of virulence factors.

Pertussis toxin ADP ribosylates and inactivates guanine nucleotide-binding proteins. As a result, G-proteins cannot transduce plasma membrane receptor signals. The toxins produced by B. pertussis paralyze the cilia, thus impairing an important pulmonary defense.
145. What is the morphology of whooping cough?
Bordetella bacteria cause a laryngotracheobronchitis that in severe cases features bronchial mucosal erosion, hyperemia, and copious mucopurulent exudate. Unless superinfected, the lung alveoli remain open and intact.

In parallel with a striking peripheral lymphocytosis, there is hypercellularity and enlargement of the mucosal lymph follicles and peribronchial lymph nodes.
146. What is pseudomonas infection?
Pseudomonas aeruginosa is an opportunistic aerobic Gram-negative bacillus that is a frequent, deadly pathogen of pts with CF, severe burns, or neutropenia. P. aeruginosa is a common cause of hospital acquired infections.

P. aeruginosa also causes a corneal kertatitis in wearers of contact lenses, endocarditis and osteomyelitis in IV drug abusers, external otitis in healthy individuals, and severe external otitis in diabetics.
147. What is the pathogenesis of pseudomonas infection?
P. aeruginosa has pili and adherence proteins that bind to eptihelial cells and lung mucin, as well as an endotoxin that causes the symptoms of gram-negative sepsis.

In the lungs of pts with CF, these bacteria secrete a mucoid exopolysaccharide called alginate, forming a slimy biofilm in which bacteria are protected from antibodies, complement, phagocytes, and antibiotics.

The organisms also secrete exotoxin A, which inhibits protein synthesis by ADP-ribosylating EF-2.
148. What else does pseudomonas secrete?
P. aeruginosa also releases exoenzyme S, which may interfere with host cell growth.

They also secrete phospholipase C that lyses RBCs and degrades pulmonary sufactant, and an elastase that degrades IgGs and ECM proteins. These enzymes may be important in tissue invasion and destruction of the cornea in keratitis.
149. What is the morphology of Pseudomonas pneumonia?
Pseudomona pneumonia is the prototype of necrotizing inflammation, distributing thru the terminal airways in a fleur-de-lis pattern, w/striking whitish necrotic centers and red, hemorrhagic peripheral areas. On microscopic exam, masses of organisms cloud the tissue w/a blush haze, concentrating in the wall of blood vessels.

This picture of Gram-negative vasculititis accompanied by thrombosis and hemorrhage is highly suggestive of P. aeruginosa infection.
150. What is the morphology CF pts with pseudomonas?
Bronchial obstruction caused by mucous plugging and subsequent P. aeruginosa infection are frequent complications of CF. Despite antibiotic treatment, chronic P. aeruginosa infection may result in bronchiectasis and pulmonary fibrosis.
151. What is the morphology of P. aeruginosa in skin burns?
In skin burns, P. aeruginosa proliferates widely, penetrating deeply into the veins and spreading to cause massive bacteremias.

Well demarcated and necrotic and hemorrhagic skin lesions of oval shape often arise during these bacteremias, called ecthyma gangrenosum. DIC is a frequent complication of bacteremia.
152. What is the plague?
Yersinia pestis is a gram-negative facultative intracellular bacterium that is transmitted by fleabites or aerosols and causes a highly invasive, frequently fatal systemic infection called plague.
153. How does the Yersinia infect?
The pathogenic Yersinia proliferate w/in lymphoid tissue.

These organisms have a complex of genes, called the Yop virulon, which enable the bacteria to kill host phagocytes, weakening the immune system. The Yop virulon includes a type III secretion system, which is a hollow syringe-like structure that injects bacterial toxins, called Yops into the cell.

The toxins block phagocytosis and cytokine production.
154. What is the morphology of Y. pestis infection?

What are 4 distinctive histologic feature?
Y. pestis causes lymph node enlargement (buboes), pneumonia, or sepsis, all with a striking neutrophilia.

The distinctive histologic features are:
1. Massive proliferation of the organisms
2. Early appearance of protein rich and polysaccharide-rich effusions with few inflammatory cells but with marked tissue swelling
3. Necrosis of tissues and blood vessels with hemorrhage and thrombosis
4. Neutrophilic infiltrates that accumulate adjacent to necrotic areas as healing begins.
155. What is the morphology of bubonic plague?

Pneumonic and septicemic plague?
In bubonic plague, the infected fleabite is usually on the legs and is marked by a small pustule or ulceration. The draining lymph nodes enlarge dramatically within a few days and become soft, pulpy, and plum colored and may infarct or rupture thru the skin.

In pneumonic plaque, there is a severe, confluent, hemorrhagic, and necrotizing bronchopenumonia, often with fibrinous pleuritis.

In septicemic plague, lymph nodes throughout the body as well as organs rich in mononuclear phagocytes develop foci of necrosis. Fulminant bacteremias also induce DIC with widespread hemorrhages and thrombi.
156. What is a chancroid?
Chancroid is an acute, sexually transmitted, ulcerative infection caused by Hemophilus ducreyi. This disease is most common in tropical and subtropical areas and is more prevalent in lower socioeconomic groups and among men who have regular contact with prostitutes.

*Chancroid is one of the most common causes of genital ulcers in Africa and Southeast Asia, which it probably serves as an important cofactor in the transmission of HIV-1 infection.
157. What is the development of a chancroid like?
4-7 days after inoculation, the pt develops a tender, erythematous papule involving the external genitalia. In males, the primary lesion is usually on the penis; in females, most lesions occur in the vagina or the periurethral area.

Over the course of several days, the surface of the primary lesion erodes to produce an irregular ulcer, which is more apt to be PAINFUL in males than in females.
158. What is the morphology of a chancroid?
In contrast to the primary chancre of syphilis, the ulcer of chancroid is not indurated, and multiple lesions may be present. The base of the ulcer is covered by shaggy, yellow-gray exudate. The regional lymph nodes in the inguinal region become enlarged and tender and can progress to erode or ulcerate and drain.
159. What are the microscopic features of a chancroid ulcer?
Microscopically, the ulcer contains a superficial zone of neutrophilic debris and fibrin, w/an underlying zone of granulation tissue containing areas of necrosis and thrombosed vessels.

A dense, lymphoplasmacytic inflammatory infiltrate is present beneath the layer of granulation tissue.
160. What is a granuloma inguinale?
Granuloma inguinale, or donovanosis, is a chronic inflammatory disease caused by Calymmatobacterium donovani, a minute, encapsulated, coccobacillus that is related to the Klebsiella genus. The organism is sexually transmitted.

Untreated cases are characterized by the development of extensive scarring, often associated with lymphatic obstruction and lymphedema (elephantitis).
161. What is the morphology of granuloma inguinale?
Granuloma inguinale begins as a raised, papular lesion involving the moist, stratified squamous epithelium of the genitalia, or the extragenital sites include the oral mucosa or pharynx. The lesion eventually undergoes ulceration, accompanied by the development of abundant granulation tissue, which is manifested grossly as a protuberant, soft, painless mass.
162. What are the microscopic features of C. donovani infection?
Microscopic exam of active lesions reveals marked epithelial hyperplasia at the borders of the ulcer, sometimes mimicking CA (pseudoepithleiiomatous hyperplasia).

A mixture of neutrophils and mononuclear inflammatory cells is present at the base of the ulcer beneath the surrounding epithelium. The organisms are demonstrable in Giemsa stained smears of the exudate as minute, encapsulated coccobacilli (Donovan bodies) in macrophages.
163. What are mycobacteria?
Bacteria in the genus Mycobacterium are slender, aerobic rods that grow in straight or branching chains. Mycobacterium have a waxy cell wall composed of mycolic acid, meaning they are acid fast and will retain stains even on treatment with a mixture of acid and alcohol. Mycobacteria stain weakly positive with Gram stain.
164. In what areas/populations is tuberculosis more common?
Tuberculosis flourishes wherever there is poverty, crowding and chronic debilitating illness. In the US, tuberculosis is mainly a disease of the elderly, the urban poor and people with AIDS.

*Certain disease states also increase the risk: DM, Hodgkin's lymphoma, chronic lung disease (particularly silicosis), chronic renal failure, malnutrition, alcoholism, and immunosuppression.
165. What is the difference between tuberculosis infection and disease?
Infection is the presence of organisms, which may or may not cause clinically significant disease. Most infections are acquired by person-to-person transmission of airborne droplets of organisms from an active case to a susceptible host. In most people, primary tuberculosis is asymptomatic, although it may cause fever and pleural effusion.

Generally, the only evidence of infection, if any remains, is a tiny, fibrocalcific nodule at the site of the infection. Viable organisms may remain dormant in such lesions for decades. When the person's immune defenses are lowered, the infection may reactivate to produce communicable and potentially life-threatening disease.
166. How is tuberculosis infection detected?
Infection w/M. tuberculosis typically leads to the development of delayed hypersensitivity to M. tuberculosis antigens, which can be detected by the tuberculin (Mantoux) test. About 2-4 weeks after infection, intracutaneous injection of purified protein derivative (PPD) of M. tuberculosis induces a visible and palpable induration that peaks in 48-72 hours.

A positive tuberculin test result signifies cell-mediated hypersensitivity to tubercular antigens. It does not, however, differentiate between infection and disease.
167. What is the pathogenesis of tuberculosis?
The pathoglogical manifestations of tuberculosis, such as caseating granulomas and cavitation, are the result of the hypersensitivity that is part and parcel of the host immune response. B/c the effector cells that mediate immunity also mediate hypersensitivity and tissue destruction, the appearance of hypersensitivity also signals the acquisition of immunity to the organism.

*Macrophages are the primary cells infected by M. tuberculosis. Early in infection, tuberculosis bacilli replicate essentially unchecked, while later in infection, the T-helper response stimulates macrophages to contain the proliferation of the bacteria.
168. How does M. tuberulosis enter macrophages?
Via endocytosis mediated by several macrophage receptors: mannose receptors bind liparabinomannan, and complement receptors bind opsonized mycobacteria.
169. Once in the macrophage, what does the mycobacteria do?
Within macrophages, M. tuberculosis blocks phagosome-lysosome fusion, allowing unchecked bacterial proliferation in the phagosome.

Thus the earliest stage of primary tuberculosis (<3 weeks) in the nonsensitized individual is characterized by the proliferation of bacteria in the pulmonary alveolar macrophages and air-spaces, with resulting bacteremia and seeding of multiple sites. Despite the bacteremia, most pts at this stage are asymptomatic or have a mild flulike illness.
170. People with a polymorphism in what gene may allow disease development without an effective immune response?
Polymorphisms in the NRAMP1 gene. NRAMP1 protein is a transmembrane protein found in endosomes and lysosomes that pumps divalent cations into the lysosome. This may have a role in generation of anti-microbial oxygen radicals.
171. What happens about 2 weeks after infection with tuberculosis?
A TH1 response against M. tuberculosis is mounted that activates macrophages to become bactericidal. TH1 cells are stimulated by mycobacterial antigens drained to the lymph node, which are presented with MHC II proteins by APCs.

Differentiation of TH1 cells depends on the presence of IL-12, which is produced by APCs that have encountered the mycobacteria.
172. What is the critical mediator which drives the macrophages to become competent to contain the M. tuberculosis infection?
IFN-γ, produced by mature TH1 cells.

IFN-γ stimulates formation of the phagolysosomes in infected macrophages, exposing the bacteria to an acidic environment. IFN-γ also induces the expression of iNOS, which produces NO to help destroy the mycobacteria.
173. In addition to stimulates macrophages to kill mycobacteria, what else does the TH1 response do?
The TH1 response orchestrates the formation of granulomas and caseous necrosis.

Activated macrophages, stimualted by IFN-γ, produce TNF, which recruits monocytes. These monocytes differentiate into the "epithelioid histiocytes" that characterize the granulomatous response.
174. Pts with RA who are treated with a TNF antagonist have an increased/decreased risk of tuberculosis reactivation?
Pts with RA who are treated with a TNF antagonist have an increased risk of tuberculosis reactivation.
175. Defects in any of the steps in generating a TH1 response results in...?
Results in absence of resistance, and disease progression.
176. What are the features of reactivation of the infection or re-exposure?
Results in rapid mobilization of a defensive reaction but also increased tissue necrosis.
177. What is primary tuberculosis?
Primary tuberculosis is the form of disease that develops in a previously unexposed, and therefore unsensitized person.

95% of people have asymptomatic infections with a persistent, latent lung infection focus.

5% have symptomatic infections with lobar consolidation, hilar adenopathy, and pleural effusion (progressive primary tuberculosis).
178. What is the difference between primary tuberculosis and progressive primary tuberculosis?
Progressive primary tuberculosis more often resembles an acute bacterial pneumonia, with lower and middle lobe consolidation, hilar adenopathy, and pleural effusion.

*Cavitation is rare, especially in pts with severe immunosuppresison.
179. What causes tuberculous meningitis and miliary tuberculosis?
Lymphohematogenous dissemination is a dreaded complication and may result in the development of tuberculos meningitis and miliary tuberculosis.
180. What is secondary tuberculosis?
Secondary tuberculosis is the pattern of disease that arises in a previously sensitized host.

Infectious usually occurs from reactivation of latent infection when immune resistance is weakened.

Typically, infection causes cavitation in apex of upper lung lobes, with associated low-grade fever, night sweats, and weight loss.
181. Secondary pulmonary tuberculosis is classically localized to what part of the lungs...?
The apex of the upper lobes or one or both lungs. This may be b/c the high oxygen tension in the apices promotes growth of the bacteria. As a result of this localization, the regional lymph nodes are less prominently involved early in the secondary disease that they are in primary tuberculosis.

On the other hand, cavitation occurs readily in the secondary form, resulting in dissemination of mycobacteria along the airways.
182. HIV pts with CD4+ T cell counts greater than 300 cells/mm present with...?
Usual secondary tuberculosis (apical disease w/cavitation)
183. HIV pts with CD4+ T cell counts less than 200 cells/mm present with...?
A clinical picture that resembles progressive primary tuberculosis (lower and middle lobe consolidation, hilar lymphadenopathy, and non-cavitary disease).

The extent of immunodeficiency also increases the freq of extrapulmonary involvement,.

*However, there is alack of characteristic granulomas in tissues, particularly in the late stages of HIV.
184. What is the morphology of primary tuberculosis?
As sensitization develops to an inhaled bacilli in the lung, a 1 to 1.5 cm area of gray-white inflammatory consolidation emerges, known as the Ghon focus.

In most cases, the center of this focus undergoes caseous necrosis. Tubbercule bacilli drain to the regional lymph nodes, which also often caseate.

*This combination of parenchymal lung lesion and nodal involvement is referred to as the Ghon complex.

The Ghon complex undergoes progressive fibrosis, often followed by radiologically detectable calcification (Ranke complex).
185. What are the histological characteristics of primary tuberculosis?
The sites of active involvement are marked by a characteristic granulomatous inflammatory reaction that forms both caseating and non-caseating tubercules.

Individual tubercles are microscopic; it is only when they coalesce into granulomas that they become macroscopically visible.
186. What is the morphology of secondary tuberculosis?
The initial lesion is usually a small focus of consolidation, less than 2 cm in diameter, within 1-2 cm of the apical pleura. Such foci are sharply circumscribed, firm, gray-white to yellow areas that have a variable amt of central caseation and peripheral fibrosis.

Histologically, the active lesions show characteristic coalescent tubercles with central caseation.

Localized apical, secondary pulmonary tuberculosis may head with fibrosis either spontaneously or after therapy, or the disease may progress and extend along several different pathways.
187. What is the morphology of progressive pulmonary tuberculosis?
Progressive pulmonary tuberculosis may ensue in the elderly and immunosuppressed. The apical lesion enlarges w/expansion of the area of caseation. Erosion into a bronchus evacuates the caseous center, creating a ragged, irregular cavity lined by caseous material that is poorly walled off by fibrous tissue.

Erosion of blood vessels results in hemoptysis.

***The pleural cavity is invariably involved, and serous pleural effusions, tuberculous empyema, or obliterative fibrous pleuritis may develop.
188. What is miliary pulmonary disease?
Miliary pulmonary disease occurs when organisms drain thru lymphatics into the lymphatic ducts, which empty into the venous return to the right side of the heart and thence into the pulmonary arteries.

Individual lesions are either microscopic or small, visible (2 mm) foci of yellow-white consolidation scattered thru the lung parenchyma.

Miliary lesions may expand and coalesce to yield almost total consolidation of large regions or even whole lobes of the lung.
189. What is endobronchial, endotracheal, and laryngeal tuberculosis?
These may develop when infective material is spread either thru lymphatic channels or from expectorated infectious material.

The mucosal lining may be studded with minute granulomatous lesions, sometimes apparent only on microscopic exam.
190. What is systemic miliary tuberculosis?
Systemic miliary tuberculosis ensues when infective foci in the lungs seed the pulmonary venous return to the heart; the organisms subsequently disseminate thru the systemic arterial system. Almost every organ in the body can be seeded.

Lesions resemble those in the lung. Miliary tuberculosis is most prominent in the liver, bone marrow, spleen, adrenals, meninges, kidneys, fallopian tubes, and epididymis.
191. What is isolated organ tuberculosis?
Isolated organ tuberculosis may appear in any of the organs or tissues seeded hematogenously and may be the presenting manifestation of tuberculosis.

Organs that are typically involved include the meninges, kidneys, adrenals, bones, and fallopian tubes.

*When the vertebrae are affected, the disease is referred to as Pott's disease. Paraspinal "cold" abscesses in these pts may track along the tissue planes to present as an abdominal or pelvic mass.
192. What id the most frequent form of extrapulmonary tuberculosis?
Lymphadenitis is the most frequent form of extrapulmonary tuberculosis, usually occurring in the cervical region ("scrofula").

In HIV negative individuals, lymphadenopathy tends to be unifocal, and most pts do not have evidence of extranodal disease.

HIV positive pts, on the other hand, almost always demonstrate multifocal disease, systemic symptoms, and either pulmonary or other organ involvement by active tuberculosis.
193. What is mycobacterium avium-intracellulare complex?
Mycobacterium avium (which includes three subspecies), and Mycobacterium intracellulare are separate species, but the infection they cause are very similar, and are called MAC>

MAC is common in soil, water, dust, adn domestic animals. Clinically significant infection w/MAC is uncommon except among people with AIDS and low levels of CD4+ lymphocytes (< 60 cells/mm)
194. What are the symptoms of MAC in AIDS pts?
In AIDS pts, MAC causes widely disseminated infections, and organisms proliferate abundantly in many organs, commonly including the lungs and GI system.

Unchecked by the immune response, the organissm reach very high levels. Pts are feverish, with drenching night sweats and weight loss.

In the rare cases of MAC in a pt w/o HIV, the organisms primarily infect the lung, causing a productive cough and sometimes fever and weight loss.
195. What is the morphological hallmark of MAC infections in pts with HIV?
*Abundant acid-fast bacilli within macrophages.

MAC infections are usually widely disseminated throughout the mononuclear systems, causing enlargement of the involved lymph nodes, liver, and spleen. There may be a yellowish pigmentation to these organs secondary to the large number of organisms present in swollen macrophages.

Granulomas, lymphocytes, and tissue destruction are rare.
196. What is leprosy?
Leprosy, or Hansen disease, is a slowly progressive infection caused by Mycobacterium leprae, affecting the skin and peripheral nerves and resulting in disabling deformities.

M. leprae is, for the most part, contained w/in the skin, but leprosy is likely to be transmitted from person-to-person thru aerosols from lesions in the upper respiratory tract.

Inhaled M. leprae, like M. tuberculosis, is taken up by alveolar macrophages and disseminates thru the blood, but grows only in relatively cool tissues of the skin and extremities.
197. What are the important characteristics of M. leprae?
M. leprae is an acid-fast obligate intracellular organism that grows very poorly in culture but can be grown in the armadillo. It grows more slowly than other mycobacteria and grows best at 32-34 degrees C, the temp of the human skin and the core temp of armadillos.

Like M. tuberculosis, M. leprae secretes no toxins,and its virulence is based on properties of its cell wall.

Cell mediated immunity is reflected by delayed type hypersensitivity reactions to dermal injections of a bacterial extract called lepromin.
198. What are the two different patterns of leprosy?
Tuberculous (less severe)

and

Lepromatous (more severe)
199. What is tuberculous leprosy?
Pts with this form have dry scaly skin lesions that lack sensation. They often have large, asymmetric peripheral nerve involvement.

*This form is caused by a TH1 response, with production of IFN-γ and IL-2. Also, IL-12 is produced by APCs and is important to the generation of TH1 cells.
200. What is lepromatous leprosy?
The more sever form of leprosy includes symmetric skin thickening and nodules. This is also called anergic leprosy, b/c of the unresponsiveness of the host immune system.

Cooler areas of skin, including the earlobes and feet, are more severely affected than warmer areas. Damage to the nervous system comes from widespread invasion of the mycobacteria into Schwann cells and into endoneural and perineural macrophages.

*This form is caused by a TH2 response, with production of IL-4, IL-5, and IL-10, which may suppress macrophage activation in response to M. leprae.
201. What is the morphology of tuberculoid leprosy?
Tuberculoid leprosy begins w/localized skin lesions that are first flat and red but enlarge and develop irregular shapes w/indurated, elevated, hyperpigmented margins and depressed pale centers (central healing). Neuronal involvement dominates tuberculoid leprosy. Nerves become enclosed within granulomatous inflammatory reactions, and if small enough, are destroyed.

Nerve degeneration causes skin anesthesias and skin and muscle atrophy, with the development of indolent skin ulcers.

*On microscopic exam, all sites of involvement disclose granulomatous lesions closely resembling those found in tuberculosis, and bacilli are almost never found.* The presence of granulomas and absence of bacteria reflect strong T-cell immunity.
202. What is the morphology of lepromatous leprosy?
Lepromatous leprosy involves the skin, peripheral nerves, anterior chamber of the eye, upper airways (down to the larynx), testes, hands and feet. The vital organs and CNS are rarely affected (b/c the core temp is too high). Peripheral nerves are symmetrically invaded.

***Lepromatous lesions contain large aggregates of lipid-lade macrophages (lepra cells), often filled w/masses of acid-fast bacilli (globi).***

The failure to contain the infection and to form granulomas reflects failure of the TH1 response. With progression, nodular lesions coalesce to form distinctive leonine facies. Lymph nodes show aggregation of foamy macrophages in the paracortical (t-cell) areas.
203. What are spirochetes?
Spirochetes are Gram-negative, slender corkscrew-shaped bacteria w/axial periplasmic flagella wound around a helical protoplasm.

The bacteria are covered in a membrane called an outer sheath, which may mask bacterial antigens from the host immune response.
204. What is the causative organism of syphilis?

What are the three stages of syphilis?
Treponema pallidum.

1. Primary syphilis
2. Secondary syphilis
3. Tertiary syphilis
205. What is primary syphilis?
The primary stage occurs 3 weeks after contact, features a single firm, *nontender*, raised, red lesion (chancre) located at the site of treponemal invasion on the penis, cervix, vaginal wall, or anus. The chancre heals in 3-6 weeks with or w/o therapy.

*Spirochetes are plentiful w/in the chancre and can be seen by dark-field microscopy or immunofluorescent stains of serous exudate. Treponemes spread throughout the body by hematologic and lymphatic dissemination even before he appearance of the chancre.
206. What is secondary syphilis?
Secondary syphilis occurs 2-10 weeks later in 75% of untreated pts, with spread and proliferation of spirochetes within the skin and mucocutaneous tissues.

*The skin lesions, which frequently occur on the palms or soles of the feet, may be maculopapular, scaly, or pustular*. Moist areas of the skin may have condylomata lata.

All these painless superficial lesions contain spirochetes and are infectious.

Lymphadenopathy, mild fever, malaise, and weight loss are also common. The symptoms of secondary syphilis last several weeks, after which the pt enter the latent phase of the disease.
207. What is tertiary syphilis?
Tertiary syphilis is rare but occurs in 1/3 of untreated pts, usually after a latent period of 5 years or more. There are three main manifestations: (1) cardiovascular syphilis, (2), neurosyphilis, and (3) benign tertiary syphilis.
208. What is cardiovascular syphilis?
Cardiovascular syphilis, in the form of syphilitic aortitis, accounts for more than 80% of tertiary disease.

*The aortitis leads to slowly progressive dilation of the aortic root and arch, which causes aortic valve insufficiency and aneurysms of the proximal aorta.
209. What is neurosyphilis?
May be symptomatic or asymptomatic. Symptomatic disease manifests in either chronic meningovascular disease, tabes dorsalis, and a generalized brain parenchymal disease called general paresis.
210. How is asymptomatic neurosyphilis detected?
It is detected when a pts CSF exhibits abnormalities such as pleocytosis, elevated protein levels, or decreased glucose.

Antibodies stimulated by the spirochetes can also be detected in CSF, this is the most specific test for neurosyphilis.
211. What is benign tertiary syphilis?
Benign tertiary syphilis is characterized by the formation of gummas in various sites. These are nodular lesions probably related to the development of delayed type hypersensitivity to the bacteria.

Gummas occur most commonly in bone, skin, and the mucous membranes of the upper airway and mouth.
212. What is congenital syphilis?
Congenital syphilis usually occurs with maternal primary or secondary syphilis. Intrauterine and perinatal death each occur in 25% of cases.

Early congenital syphilis includes nasal discharge, skin sloughing, hepatomegaly, and skeletal abnormalities.

Late (tardive) syphilis manifests as notched central incisors, deafness, and interstitial keratitis with blindness (Hutchinson triad).
213. What is the morphology of primary syphilis?
The chancre occurs on the penis or scrotum in 70% of men and on the vulva or cervix of 50% of women.

The chancre is a slightly elevated, firm, reddened papule up to several cm in diameter, that erodes to create a clean based shallow ulcer. The contiguous induration creates a buttonlike mass directly adjacent to the eroded skin.

On histologic exam, treponemes are visible at the surface of the ulcer with silver stains (e.g., Warthin-Starry stain).

The regional lymph nodes are usually enlarged and may show a nonspecific acute or chronic lymphadenitis.
214. What is the morphology of secondary syphilis?
Widespread mucocutaneous lesions involve the oral cavity, palms of hands, and soles of feet. The rash is frequently macular, w/discrete red-brown spots less than 5 mm in diameter, but it may be follicular, pustular, annular, or scaling.

Histologically, the lesions show the same plasma cell infiltrate and obliterative endarteritis as the primary chancre, although the inflammation is often less intense.
215. What is the morphology of the aortitis in tertiary syphilis?
The aortitis is caused by endarteritis of the vasa vasorum of the proximal aorta. Occlusion of the vasa vasorum results in scarring of the media of the proximal aortic wall, causing a loss of elasticity.

There may be narrowing of the coronary artery ostia caused by subintimal scarring w/resulting myocardial ischemia.
216. What is the morphology of neurosyphilis?
Syphilitic gummas are white-gray and rubbery, occur singly or multiply, and vary in size from microscopic defects resembling tubercles to large tumorlike masses.

They occur in most organs but particularly in skin, subcutaneous tissue, bone and joints.

*In the liver, scarring as a result of gummas may cause a distinctive hepatic lesion known as hepar lobatum*.

On histologic exam, the gummas contain a center of coagulated, necrotic material and margins composed of plump or palisaded macrophages and fibroblasts surrounded by large numbers of mononuclear leukocytes, chiefly plasma cells.
217. What are the skeletal deformities in early congenital syphilis?
Syphilitic osteochondritis and periostitis affect all bones, although lesions of the nose and lower legs are most distinctive. Destruction of the vomer causes collapse of the bridge of the nose and later on, the characteristic saddle nose deformity.

Periostitis of the tibia leads to excessive new bone growth on the anterior surfaces and anterior bowing, or saber shin. There is also widespread disturbance in endochrondral bone formation.
218. What are the lung deformities in congenital syphilis?
The lungs may be affected by a diffuse interstitial fibrosis. In the syphilitic stillborn, the lungs appear as pale, airless organs (pneumonia alba).
219. What is the triad of late occurring congenital syphilis?
The triad consists of:

1. Interstitial keratitis
2. Hutchinson teeth
3. CN 8 nerve deafeness
220. What is the pathogenesis of T. pallidum infection?
The immune response to T. pallidum reduces the burden of bacteria, but it may also have a central role in the pathogenesis of the disease.

The T-helper cells that infiltrate the chancre are TH1 cells, suggesting that activation of macrophages to kill bacteria may cause resolution of the local infection. However, the antibody response does not eliminate the infection. *The outer membrane of T. pallidum appears to protect the bacteria from antibody binding.
221. What is relapsing fever?
Relapsing fever is an insect-transmitted disease characterized by recurrent fevers with spirochetemia.

Epidemic relapsing fever is caused by body louse-transmitted Borrelia recurrentis, which only infects humans.

Endemic relapsing fever is caused by several Borrelia species, which are transmitted from small animals to humans by Ornithodorus (soft-bodied) ticks.
222. What are the clinical features of relapsing fever?
In both louse-and tick-transmitted borreliosis, there is a 1- to 2-week incubation period after the bite as the spirochetes multiply in the blood.

Clinical infection is heralded by shaking chills, fever, headache, and fatigue, followed by DIC and multiorgan failure.
223. What is the pathogenesis of relapsing fever?
Spirochetes are temporarily cleared from teh body by anti-Borrelia antibodies, which target a single major surface protein called the variable major protein.

After a few days, bacteria bearing a different surface antigen emerge and reach high densities in the blood, and symptoms return until a second set of host antibodies clears these organisms.
224. What is the Jarish-Herxheimer reaction?
Antibiotic treatment of Borrelia infections may cause a massive release of endotoxin, resulting in the production of cytokines that cause fevers with rigors, fall in blood pressure, and leukopenia.
225. What is the morphology of relapsing fever?
Dx can be made by identification of spirochetes in blood smears obtained during febrile periods. In fatal louse-borne disease, the spleen is moderately enlarged and contains focal necrosis and miliary collections of leukocytes, including neutrophils, and numerous borreliae. There is congestion and hypercellularity of the red pulp.

The liver may also be enlarged and congested with prominent Kupffer cells and septic foci.

Scattered hemorrhages resulting from DIC may be found in serosal and mucosal surfaces, skin, and viscera. Pulmonary bacterial superinfection is a frequent complication.
226. What is Lyme disease?
Lyme disease is caused by the spirochete Borrelia burdorferi, transmitted from rodents to people by Ixodes deer ticks.
227. What is the first stage of Lyme disease?
Spirochetes multiply at the site of the tick bite, causing an expanding area of redness, often with a pale center (erythema chronicum migrans), fever, and lymphadenopathy.
228. What is the second stage of Lyme disease?
Spirochetes spread hematogenously, causing secondary skin lesions, lymphadenopathy, migratory joint and muscle pain, cardiac arrhythmias, and meningitis.
229. What is the third stage of Lyme disease?
A few years later after the initial bite, Lyme borreliae cause a chronic arthritis sometimes with severe damage to large joints and an encephalitis that varies from mild to debilitating.
230. What is the pathogenesis of B. burdorferi infection?
The initial immune response is stimulated by binding of bacterial lipoproteins to TLR-2 expressed by macrophages. In response, these cells release IL-6 and TNF and generate bactericidal NO, reducing but not eliminating the infection.

***B. burgdorferi escapes the antibody response through antigenic variation.
231. What is the morphology of B. burgdorferi infection?
Skin lesions are characterized by edema and a lymphocytic plasma cell infiltrate. In early Lyme arthritis, the synovium resembles that of early RA.

*A distinctive feature of Lyme arthritis is an arteritis, with onionskin-like lesions resembling those seen in lupus.*

In late Lyme disease, there may be extensive erosion of the cartilage in large joints.
232. What are the usual causes of abscesses?
Commensal bacteria from adjacent sites (oropharynx, intestine, and female genital tract) are the usual cause of abscesses, so the species found in the abscess reflect the species found in the normal flora.
233. What causes head and neck abscesses?
Oral flora: (Prevotella and Porphromonas, mixed with S. auereus and S. pyogenes).
234. What causes abdominal abscesses?
1. Bacteroides fragilis
2. Peptostreptococcus
3. Clostridium spp.
4. E. coli
235. What causes genital tract abscesses?
Caused by anaerobic gram negative bacilli, including Prevotella spp, often mixed with E. coli or S. agalactiae.
236. What is Lemierre syndrome?
Lemierre syndrome is characterized by infection of the lateral pharyngeal space and spetic jugular vein thrombosis.

*It is caused by Fusobacterium necrophorum, an oral commensal.
237. What is the morphology of abscesses?
The pus of the abscesses is discolored and foul smelling owing to the presence of anaerobes, especially in lung abscesses, and the suppuration is often poorly walled off.
238. What does C. perfringens infection cause?
C. perfringens causes cellulitis and myonecrosis of traumatic and surgical wounds (gas gangrene), uterine myonecrosis often associated with illegal abortions, mild food poisoning and infection o f the small bowel of ischemic or neutropenic pts often leading to severe sepsis.
239. What does C. tetani cause?
C. tetani proliferates in puncture wounds and in the umbilical stump of newborn infants in developing countries and releases a potent neutrotoxin, called tetanospasmin, that causes convulsive contractions of skeletal muscles (lockjaw).
240. What is the major virulence factor in clostridial infections?
Their most powerful virulence facotrs are the many toxins they produce.

*C. perfringens secretes 14 toxins, the most important of which is α-toxin. This toxin is a phospholipase C, a phingomyelinase, and it releases IP3, prostaglandins and thromboxanes.
241. How does tetanus toxin cause violent spastic paralysis?
It blocks the release of the neurotransmitter GABA from motor neurons.
242. C. difficle produces what toxins?
Produces toxin A that stimulates chemokine production and thus attracts leukocytes.

It also secretes toxin B, which causes distinctive cytopathic effects in cultures cells.

Both toxins are glucosyl transferases.
243. What is associated with dusk-colored, wedge-shaped infarcts in the small bowel, particularly in neutropenic pts?
C. perfringens.
244. What is the morphology of chlamydia infection?
The morphologic features of C. trachomatis urethritis are virtually identical to those of gonorrhea. The primary infection is characterized by a mucopurlent discharge containing a predominance of neutrophils. Organisms are not visible in Gram-stained smears or sections.
245. What causes chlamydial infection?
Chlamydiae trachomatis serotypes D-K cause sexually transmitted genital infections.

The chlamydiae are very small bacteria that are obligate intracellular parasites. They have a more complicated life cycle than free living bacteria b/c they can exist in different forms.
246. What are the two forms of chlamydiae?
1. The elementary body (EB) is adapted for extracellular survival and for initiation of infection

2. The reticulate body (RB) is adapted for intracellular multiplication
247. What are the three groups of serotypes of Chlamydia trachomatis?
1. Serotypes A, B, and C are the causes of the serious eye infection trachoma.
2. Serotypes D-K are the cause of genital infections and associated ocular and respiratory infections.
3. Serotypes L1, L2, and L3 cause the systemic disease lymphogranuloma venereum (LFV).
248. How do chlamydiae enter the host?
Chlamydiae enter the host thru minute abrasions in the mucosal surface. They bind to specific receptors on the host cells and enter the cells via parasite-induced endocytosis.

Once in the cell, fusion of the chlamydia-containing vesicle w/lysosomes is inhibited and the EB begins its developmental cycle.

Within 9-10 hr or cell invasion, the EBs differentiate into metabolically active RBs, which divide via binary fission and produce fresh Eb progeny. These are then released into the EC environment.
249. What is lyphogranuloma venereum (LGV)?
LGV is caused by C. trachomatis serotypes L1, L2, and L3.

LGV is a serious disease especially common in Africa, Asia, and South America.

LGV is a systemic infection involving lymphoid tissue and is treated with doxycline or erythromycin.
250. What does LGV infection look like?
The primary lesion is an ulcerating papule at the site of inoculation and may be accompanied by fever, headache, and myalgia. The lesion heals rapidly, but the chlamydiae proceed to infect the draining lymph nodes, causing characteristic inguinal buboes which gradually enlarge, and can coalesce and rupture.

The infection may disseminate from the lymph nodes to cause proctitis, or rectal strictures.
251. What is the morphology of lympogranuloma venereum?
The lesions of lympogranuloma venereum contain a mixed granulomatous and neutrophilic inflammatory response. Regional lymphadenopathy is common.

***Lymph node involvement is characterized by a granulomatous inflammatory reaction associated with irregularly shaped foci of necrosis and neutrophilic infiltration (stellate abscesses).***
252. What are the three main Rickettsial infections?
1. Epidemic typhus (R. prowazekii
2. Scrub typhus (Orienta tsutsugamushi)
3. Spotted fevers (R. rickettsii)

These organisms have the structure of Gram-negative, rod shaped bacteria, although they stain poorly with Gram stain.
253. What is epidemic typhus?
Epidemic typhus, which is transmitted from by body lice, is associated with wars and human suffering.

In mild cases, the gross changes are limited to a rash and small hemorrhages due to the vascular lesions. In more severe cases, there may be areas of necrosis of the skin with gangrene of the tips of the fingers, nose, earlobes, scrotum, penis and vulva.
254. What are the most prominent microscopic changes in typhus fever?
The small vessel lesions that underlie the rash and the focal areas of hemorrhage and inflammation in the various organs and tissues affected.

Endothelial swelling in the capillaries, arterioles, and venules may narrow the lumina of these vessels. A cuff of mononuclear inflammatory cells usually surrounds the affected vessels. Necrosis of the vessel wall is unusual in typhus compared to RMSF.
255. What is scrub typhus?
Scrub typhus, transmitted by chiggers, was a major problem for US. solders in WWII.

It is a mite-borne infection, and is usually a milder form of typhus fever. The rash is usually transitory or might not appear.

Vascular thrombosis is rare, but there may be a prominent inflammatory lymphadenopathy.
256. What is ehrlichiosis?
*Human ehrlichiosis is an acute, febrile illness, fever, headache, and malaise, which may progress to respiratory insufficiency, renal failure, and shock.

It is caused by infection in macrophages by Ehrlichia chaffeensis, or infection of neutrophils by Anaplasma phagocytophilum.*
257. What are the characteristic Dx features of ehrlichiosis?
Characteristic cytoplasmic inclusions (morulae), occasionally shaped like mulberries and composed of masses of bacteria, can be seen in the appropriate leukocytes in ehrlichiosis.

Rash occurs in approx 40% of pts with E. chaffeensis infection.
258. What is RMSF?
The causative organism, Rickettsia rickettsii, is transmitted from dogs or small wild animals to ticks and then to humans. Infection occurs primarily during warmer months, the period of greatest tick activity.

The fulminant onset of severe frontal headache, chills, fever, myalgias, and conjunctivitis occur after 2-14 days; cough and SOB develop in 25% of patients.

*Typically occurs in the southeastern US.
259. What does the rash look like in RMSF?
The hemorrhagic rash characteristically begins on day 3-5 of illness as 1-4 mm erythematous macules on the hands, wrists, feet and ankles. Palms and soles may also be involved. The rash may be transient, but it usually spreads to the trunk and may become petechial.

Dx is via specific complement fixation test which shows a rise in titers and allows retrospective confirmation of Dx.
260. What is the major cause of death with RMSF?
A noncardiogenic pulmonary edema causing adult respiratory distress syndrome is the major cause of death with RMSF.