• 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

1/261

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;

261 Cards in this Set

  • Front
  • Back
1. What are the characteristics of basal ganglia disorders?
Pts with basal ganglia lesions can have either hyperkinetic or hypokinetic movement disorders.

Hyperkinetic = Huntington's disease

Hypokinetic = Parkinson's disease
2. What are the components of the basal ganglia?
THe main components are:

1. Caudate*
2. Putamen*^
3. Globus pallidus^
4. Subthalamic nucleus
5. Substantia nigra
3. What is the striatum?
The caudate and putamen are histologically and embryologically related and can be though of as a single large nucleus called the striatum, which receives all inputs to the basal ganglia.
4. What is the caudate nucleus?
The caudate is divided into three parts, the head, body, and tail. The amygdala lies just anterior to the tip of the caudate tail, in the temporal lobe.
5. What is the putamen?
The putamen is a large nucleus forming the lateral portion fot he basal ganglia. Anteriors and ventrally the putamen fuses w/the head of the caudate. This region, called the ventral striatum, is important in limbic circuitry and is often considered part of the striatum b/c of its similar embryological development and I/O connections. Most of the vental stritum consists fo the nucleus accumbens.
6. What is the globus pallidus?
Just medial to the putamen lies the globus pallidus, which has many myelinated fibers. The globus pallidus has an internal segment and an external segment.
7. What is the lentiform or lenticular nucleus?
The putamen and globus pallidus together make up the lenticular or lentiform nucleus. These nuclei more or less resemble an ice cream cone lying on its side, w/the putamen representing the ice cream and the globus pallidus the cone.
8. Moving from lateral to medial in the horizontal brain section, name the 10 structures you will encounter
1. Insula
2. Extreme capsule
3. Claustrum
4. External capsule
5. Putamen
6. External medullary lamina
7. External segment of the globus pallidus
8. Internal medullary lamina
9. Internal segment of the globus pallidus
10. Internal capsule
9. What is the internal capsule?
The internal capsule is a V shaped collection of fibers going to and from the cortex.

The anterior limb of the internal capsule passes btwn the lentiform nucleus and the head of the caudate. The posterior limb of the internal capsule passes btwn the lentiform nucleus and the thalamus.
10. Where is the caudate, thalamus, and lentiform nucleus in relation to the internal capsule?
The caudate and thalamus are always medial to the internal capsule while the lentiform nucleus is always lateral to the internal capsule.
11. The head and body of the caudate form a bulge where...?
The head and body of the caudate form a bulge on the lateral wall of the lateral ventricle, while the tail of the caudate runs along the roof of the temporal horn.
12. Where is the thalamus again in relation to the ventricles?
The thalamus forms the lateral walls of the third ventricle and lies along the floor of the body of the lateral ventricle.

In a coronal section, if the thalamus is not yet visible, we are still in the anterior limb of the internal capsule.

On the other hand, fi the thalamus can be seen, we are at the level fo the posterior limb of the internal capsule, separating thalamus from lentiform nucleus.
13. Where is the substantia niga and the substantia nigra pars reticulata?
By following the internal capsule downward, we see the beginnings of the cerebral peduncles of the midbrain. The substantia nigra is visible, just dorsal to the cerebral peduncles.

The substantia nigra has a ventral portion called the substantia nigra parts reticulata, which contains cells very similar to those of the internal segment of the globus pallidus.
14. What separates the internal segment of the globus pallidus from the sustantia nigra pars reticulata?
The internal capsule.
15. Review: what borders the anterior limb of the internal capsule?
The anterior limb of the internal capsule is bordered by the head of the caudate medially and the lentiform nucleus laterally.
16. Review: what borders the posterior limb of the internal capsule?
The posterior limb of the internal capsule is bordered by the thalamus medially and by the lentiform nucleus laterally.
17. What is the subthalamic nucleus?
Under the thalamus lies the spindle shaped subthalamic nucleus. Unlike the thalamus, the subthalamic nucleus is derived embryologically from the midbrain rather than the forebrain.
18. Where do the striatum and globus pallidus get their blood supply?
The blood supply to the striatum and globus pallidus is mainly from the lenticulostriate branches of the MCA, although the medial globus pallidus is often supplied by the anterior choroidal atery (branch of internal carotid artery), and the caudate head and anterior potions of the lentiform nucleus are often supplied by the recurrent artery of Heubner (branch of ACA).
19. Virtually all inputs to the basal ganglia arrive where?

Outputs leave where?
Vitually all inputs to the basal ganglia arrive via the striatum (caudate, putamen, and nucleus accumbens).

Outputs leave the basal ganglia via the internal segment of the globus pallidus and the closely related substantia nigra pars reticulata.

I/O's of the basal ganglia are thus easily visualized as a funnel, w/the spout pointing medially.
20. What are the 4 major sources of input to the basal ganglia? For each input, list the major neurotransmitter.
1. Projections from entire cerebral cortex (putamen is the most important for motor control) - excitatory pathway and uses glutamate TO STRIATUM
2. Substantia nigra pars compacta (dopaminergic nigrostriatal pathway) excitatory and inhibitory TO STRIATUM
3. Glutamatergic inputs from intralaminar nuclei (centromedian and parafascicular nuclei) - excitatory TO STRIATUM
4. Raphe nuclei provides serotonergic inputs TO BASAL GANGLIA
21. Where do basal ganglia outputs arise from?

Which controls which?
From the internal segment of the globus pallidus and from the substantia nigra parts reticulata. These pathways use GABA and they are inhibitory.

For motor control, the substantia nigra pars reticulata appears to convey info for the head and neck, while the internal segment of the globus pallidus conveys info for the rest of the body.
22. What is the main output of these pathways?
The main output of these pathways are to the ventral lateral (VL) and ventral anterior (VA) nuclei of the thalamus via the thalamic fasciculus.

*The more anterior parts of the thalamic fasciculus carry outputs from teh basal ganglia to the anterior portion of VL, whiel the more psoterior parts of the thalamic fasciculus carry cerebellar outputs of the posterior VL.

*Caudal parts of VL receives inputs from Cerebellum
23. What are the other thalamic nuclei in which basal ganglia outputs also travel to?
The intralaminar nuclei, which project back to teh striatum, and the mediodorsal nucleus, which is inolved primarily in limbic pathways.

In addition, the internal segment fo the globus pallidus and the substantia nigra pars reticulata project to the pontomedullary reticular formation, thereby influencing the descending reticulospinal tract. The substantia nigra pars reticulata also projects to the superior colliculus, to influenc etectospinal pathways.
24. What are the 2 predominant pathways from input to output nuclei through the basal ganglia?
1. The direct pathway travels from the striatum directly to the internal segment of the globus pallidus or the substantia nigra pars reticulata.

2. The indirect pathway takes a detour from the striatum, first to the external segment of the globus pallidus and then to the subthalamic nucleus, before finally reaching the internal segment of the globus pallidus or the substantia nigra pars reticulata.
25. What is the net effect of excitatory input from the cortex thru the direct pathway?
Excitation of the thalamus, which will facilitate movements through its connections w/the motor and premotor cortex.
26. What is the net effect of excitatory input from the cortex thru the indirect pathway?
On the other hand, the net effect of excitation of the indirect pathway will be inhibtion of the thalamus, resulting in inhibition of movements thru connections back to the cortex

*Indirect inhibits
27. What are the neurotransmitters in the direct pathway?
Striatal projection neurons for both pathways are primarily inhibitory spiny neurons, which contain GABA. In the direct pathway, spiny striatal neurons project to the internal segemtn of the globus pallidus and contain the peptide substance P in addition to GABA.

Output neurons from the internal globus pallidus and substantia nigra pars reticulata to the thalamus are also inhibitory and contain GABA.
28. What are the neurotransmitters in the indirect pathway?
In the indirect pathway, striatal neurons project to the external segment of the globus pallidus and contain the inhibitory neurotransmitter GABA, plus the peptide enkephalin.

Neurons of the external globus pallidus, in turn, send inhibitory GABAergic projections to the subthalamic nucleus. Excitatory neurons in the subthalamic nucleus containing glutamate then project to the internal segment of the globus pallidus and to the substantia nigra pars reticulata.
29. What is dopamine's effect on the direct pathway?

Indirect pathway?
Dopamine appears to have excitatory effects on the neurons of the direct pathway, but inhibitory effects on the indirect pathway, which normally has a net excitatory effect on the thalamus.

*Therefore, loss of dopamine will result in net inhibition of the thalamus, through both the direct and the indirect pathways, which may account for the paucity of movement seen in Parkinson's disease.
30. What is acetylcholine's effect on the indirect pathway?
There are large interneurons in the striatum calls aspiny neurons, some of which contain ACh. Some evidence suggests that these cholinergic interneurons preferentially form excitatory synapses onto striatal neurons of the indirect pathway.

*Removal of cholinergic excitation of the indirect pathway produces a net decrease in inhibition of the thalamus, which may account for the beneficial effects of anticholinergic agents in parkinsonism.
31. What is the importance of hemiballismus?
In hemiballismus, there are unilateral wild flinging movements of the extremities contralateral to a lesion in the basal ganglia, typically involving the subthalamic nucleus.

*Damage to the subthalamic nucleus could decrease excitation of the internal segment of the globus pallidus, resulting in less inhibition of the thalamus, causing a hyperkinetic movement disorder (Huntingtons).
32. What are the 4 channels that the basal ganglia contains for info processing?
1. Motor channel
2. Oculomotor channel
3. Prefrontal channel
4. Limbic channel
33. Which ones are the dorsal striatal pathways?

Ventral?
The motor, oculomotor, and prefrontal channels are the dorsal striatal pathways, while the limbic is the ventral striatal pathways.
34. What is the role of the motor channel?
The motor is the best known; Cortical inputs travel mainly to the putamen, and output targets are the VL and VA of the thalamus.

From the thalamus, the motor channel continues to the supplementary motor area, premotor cortex, and primary motor cortex.
35. What is the role of the oculomotor channel?
This channel subserves basal ganglia regulation of eye movements.

The input is from the body of the caudate nucleus. Output is to the frontal eye fields and supplementary eye fields of the frontal lobes; areas important for the higher control of eye movements.
36. Prefrontal channel?
The prefrontal channel is probably important in cognitive processes involving the frontal lobes.

Input is primarily from the head of the caudate, and output reaches the prefrontal cortex.
37. Limbic channel?
The limbic channel is an important ventral pathway thru the basal ganglia that is involved in limbic regulation of emotions and motivational drives.

Inputs arise from major areas for the limbic system, such as the nucleus accumbens, ventral caudate, and ventral putamen. Output targets are the anterior cingulate gyrus and medial orbital frontal gyri.
38. What is an important relay station for the limbic channel?
The mediodorsal nucleus is particularly important to limbic circuitry. Projections from these thalamic nuclei reach the limbic cortex of the anterior cingulate gyrus and medial orbital frontal gyri.
39. What is another role of the limbic channel?
The limbic channel through the basal ganglia is likely to play a central role in many neurobehavioral and psychiatric disorders.
40. What is the importance of the ventral tegmental areas?
Another component of this pathway is the dopaminergic projection from the ventral tegmental area, which lies just medial and dorsal to the substantia nigra of the midbrain, at the base of the interpeduncular fossa.

The ventral tegmental area provides dopaminergic inputs to the nucleus accumbens, as well as to other limbic structures and to the frontal lobes. The dopaminergic projections of the ventral tegmental area may be affected in the pathophysiology of schizophrenia and other psychotic disorders.
41. The internal segment of the globus pallidus sends outputs to the thalamus through two different pathways. What are they?
1. Ansa lenticular
2. Lenticular fasciculus
42. What is the ansa lenticularis?
This pathway is so named b/c it takes a loop ventrally under the internal capsule before passing dorsally to reach the thalamus.

The ansa lenticularis actually passes slightly rostrally as it loops around the inferior medial edge of the internal capsule, and then it turns back toward the thalamus.
43. What is the lenticular fasciculus?
Instead of taking a looping course, fibers of the lenticular fasciculus penetrate straight thru the internal capsule. They then pass dorsal to the subthalamic nucleus and ventral to the zona incerta before turning superiorly and laterally to enter the thalamus.
44. OK then, what is the zona incerta?
The zona incerta is the inferior extension of the reticular nucleus of the thalamus (not the reticular formation).
45. Where do the fibers of the ansa lenticularis and lenticular fasciculus join?
They join together to form the thalamic fasciculus, which enters the thalamus.

The thalamic fasciculus also contains fibers ascending to the thalamus from the deep cerebellar nuclei.
46. What is the H1 field of Forel?

H2?
H1 is the thalamic fasiculus

H2 is the lenticular fasciculus, where it lies dorsal to the subthalamic nucleus.
47. What is the H field of Forel?
This is the region where the ansa lenticularis and lenticular fasciculus join together.
48. What is the subthalamic fasciculus?
In addition to the ansa lenticularis, lenticular fasciculus, and thalamic fasciculus, there is the subthalamic fasiculus.

This carries fibers of the indirect pathway from the external segment of the globus pallidus to the subthalamic nucleus, and from the subthalamic nucleus to the internal segment of the globus pallidus.
49. Again, what is hemiballismus?
In hemiballismus there are unilateral wild flinging movements of the extremities contralateral to a lesion in the basal ganglia, typically involving the subthalamic nucleus.

Damage to the subthalamic nucleus could decrease excitation of the internal segment of the globus pallidus, resulting in less inhibition of the thalamus, causing a hyperkinetic movement disorder.
50. What is chorea?
Chorea is multiple quick, random movements, usually most prominent in the appendicular muscles.

Caused by atrophy of the striatum as in Huntington chorea.
51. What is athetosis?
Athetosis is slow, swriting movements, which are usually more severe in the appendicular muscles.

Caused by diffuse hypermyelinization of the corpus striatum and thalamus cerebral palsy.
52. What are the movement disorders ranked from slowest to fasted movement speed?
SLOWEST

1. Bradykinesia, hypokinesia
2. Rigidity
3. Dystonia
4. Athetosis
5. Chorea
6. Ballismus
7. Tics
8. Myoclonus
9. Tremor

FASTEST
53. How can dopamine neurons effect both the direct and indirect pathways?
The effect of dopamine excites or drives the direct pathway, increasing cortical excitation. Dopamine excites the direct pathway through D1 receptors and inhibits the indirect pathway through D2 receptors.
54. What effect do cholinergic neurons have on the indirect pathway?
Cholinergic neurons have the opposite effect. ACh drives the indirect pathway, decreasing cortical excitation.
55. What is dystonia?
Dystonia refers to a slow, prolonged movement involving predominantly the truncal musculature. Dystonia often occurs with athetosis.

Blepharospasm and writer's cramp are all examples of dystonic movements.
56. What causes palatal myoclonus (clinking sounds)?
Palatal myoclonus is typically caused by lesions of the central tegmental tract, most commonly w/brainstem infarcts.
57. What is pallidotomy?
For Parkinson's disease that has progressed to severe on-off swings despite medical therapy, pallidotomy can provide substantial improvement in bradykinesia, rigidity, and medication -related dyskinesias. In this procedure a lesion is placed stereotactically in the ventral posterior portion of the globus pallidus. The lesion interrupts the inhibitory output pathway carried by the ansa lenticularis from the medial globus pallidus to the ventral lateral nucleus of the thalamus.

Removal of this inhibition may explain why bradykinesia and rigidity are improved.
58. What is a thalmotomy?
For debilitating tremor, especially in cases of severe essential tremor not responding to medications, thalamotomy can be beneficial. In this procedure a lesion is placed sterotactically in the posterior portion of the ventral lateral nucleus of the thalamus.
59. Involuntary, wild flinging movements of the right arm and leg
The most likely clinical localization is left subthalamic nucleus or left stratum.
60. Irregular jerking movements, slightly decreased tone, and unsteady gait; moderately slowed saccadic eye movements; flat affect, argumentative, and denied having nay involuntary movements
This pt has a bilateral hyperkinetic movement disorder, best described as mild tic or chorea. The most likely Dx is Huntington's disease.
61. Asymmetric bradykinesia, cogwheel rigidity, and resting tremor; stooped gait w/short steps, decreased arm swing, en bloc turning, and retropulsion; significant benefit from levodopa; gradual progression over a period of years
Idiopathic Parkinson's disease.
62. Bilateral bradykinesia and waxy rigidity; no tremor; gait slow, shuffling w/retropulsion; no significant benefit from levodopa; gradual progression over a period of years; masked facies, slow saccades, slow dysarthric speech
Atypical Parkinsonism. ***Atypical parkinsonism alone, w/o other significant abnormalities, can be seen in striatonigral degeneration (a form of multisystem atrophy)
63. How does one differentiate striatonigral degeneration from Parkinson's?
In striatonigral degeneration, as in idiopathic Parkinson's disease, there is loss of dopaminergic neurons from the substantia nigra pars compacta.

*In striatonigral degeneration, however, the striatal neurons degenerate as well. Therefore, administration of levodopa is usually not as beneficial to these pts as in Parkinson's disease.
64. What are demyelinating diseases?
Demyelinating diseases of the CNS are acquired conditions characterized by preferential damage to myelin, with relative preservation of axons.

The clinical deficits are due to the effect of myelin loss on the transmission of electrical impulses along axons.
65. What is MS?
MS is an autoimmune demyelinating disorder characterized by distinct episodes of neurological deficits separated in time, attributable to white matter lesions that are separated in space.

Affects 1/1,000 persons in most of US and Europe. Women are affected 2x more than men.
66. What is the typical presentation of a pt w/MS?
The clinical course of the illness evlolves as relapsing and remitting episodes of neurologic deficit during variable interval of time (wks to mos to yrs) followed by gradual, partial recovery of neurologic function.

The frequency of relapses tends to decrease during the course of time, but there is a steady neurologic deterioration in most patients.
67. What causes the lesions in MS?
The lesions of MS are caused by a cellular immune response that is inappropriately directed against the components of the myelin sheath.
68. Genetic susceptibility of MS
The likelihood of developing this autoimmune process is influenced by genetic and environmental factors.

Risk is 15x higher when the disease is present in a first degree relative and higher for twins.

Genetic linkage of MS susceptibility to the DR2 extended haplotype of the MHC is also well established.

The molecular basis for influence of this particular haplotype on the risk of developing MS is unknown, other genetic local are involved as well.
69. What initiates the immune response in MS?
The available evidence indicates that the disease is initiated by CD4+ TH1 cells that react against self myelin antigens and secrete cytokines, such as IFN-γ, that activate macrophages.

The demyelination is caused by these activated macrophages and their injurious products.

The infiltrate in plaques and surrounding regions of the brain consists of T cells (mainly CD4+, some CD8+), and macrophages.

Antibodies are also freq present. Therapies are being developed that modulate or inhibit TH1 responses and block the recruitment of T cells into the brain.
70. Morphology of MS
Since MS is a white matter disease and gray matter covers much of the surface of hemispheres, macroscopic exam of the outer cortex is unremarkable.

Evidence of disease may be found on the surface of the brainstem or along the spinal cord, where myelinated fiber tracts course superficially; here lesions appear as multiple, well-circumscribed, somewhat depressed, glassy, gray-tan, irregularly shaped plaques, both on external exam and on section.

In the fresh state, these have firmer consistency than the surrounding white matter (sclerosis).
71. Morphology of the MS plaques
Lesions (plaques) are sharply defined areas of gray discoloration of white matter occurring esp around the ventricles but potentially located anywhere in the CNS.

Active plaques show myelin breakdown, lipid-laden macrophages, and relative axonal preservation. Lymphocytes and mononuclear cells are prominent at the edges of plaques and around venules in and around plaques.

Inactive plaques lack the inflammatory cell infiltrate and show gliosis; most axons within the lesion persist but remain unmyelinated.
72. Location of MS plaques
Plaques can be found throughout the white matter of the neuraxis; they may also extend into the gray matter structures, as these have myelinated fibers running through them.

Plaques commonly occur beside the lateral ventricles and may be demonstrated to follow the course of paraventricular veins when the surface of the ventricle is inspected en face. They are also frequent in the optic nerves and chiasm, brain stem ascending and descending fiber tracts, cerebellum, and spinal cord.
73. What is the morphology of active plaques?
In an active plaque, there is evidence of ongoing myelin breakdown with abundant macrophages containing lipid-rich, PAS-positive debris.

Inflammatory cells, including both lymphocytes and monocytes, are present, mostly as perivascular cuffs, especially at the outer edge of the lesion.

Small active plaques are often centered on small veins.
74. What happens w/in an active plaque?
W/in a plaque, there is relative preservation of axons and depletion of oligodendrocytes. In time, astrocytes undergo reactive changes.
75. What about an inactive plaque?
As lesions become quiescent, there is lessening of the inflammatory cell infiltrate and of macrophages.

W/in the center of an inactive plaque, ***little to no myelin is found, and there is a reduction in the number of oligodendrocyte nuclei; instead astrocytic proliferation and gliosis are prominent.
76. What are shadow plaques? What is their significance?
In some MS plaques known as shadow plaques, the border btwn normal and affected white matter is not sharply circumscribed.

In this type of lesion, some abnormally thinned-out myelin sheaths can be demonstrated at the outer edges. This has been interpreted either as evidence of partial and incomplete myelin loss or as remyelination by surviving oligodendrocytes.
77. What are the 4 patterns of active plaques?
I. Those that are sharply demarcated and centered on blood vessels, WITH deposition of Ig and complement
II. Those that are sharply demarcated and centered on blood vessels, W/O deposition of Ig and complement
III. Those that are less demarcated and are not centered on vessels with widespread oligocendrocyte apoptosis
IV. Those that are less demarcated and are not centered on vessels with only central oligocendrocyte apoptosis
78. What is the significance of these 4 patterns of active plaques?
Autopsy studies have shown that only one pair of patterns (I/II or III/IV) is present in a given individual.
79. Clinical features of MS
Although MS lesions can occur anywhere in the CNS and, as a consequence, may induce a wide range of manifestations, certain patterns of neurologic symptoms are commonly observed.

Unilateral visual impairment during the course of a few days, due to involvement of the optic nerve (optic neuritis) is a frequent initial manifestation of MS.

Involvement of the brainstem produces cranial nerve signs, ataxia, nystagmus, and internuclear ophthalmoplegia from interruption of the fibers of the medial longitudinal fasciculus (MLF).

Spinal cord lesions give rise to motor and sensory impairment of trunk and limbs, spasticity, and difficulties w/the voluntary control of bladder function.
80. CSF in patients with MS shows...?
Exam of the CSF in MS patients shows a mildly elevated protein level, and in one third of cases, there is moderate pleocytosis. The proportion of gamma globulin in increased, and most MS patients show ***oligoclonal bands***.

This increase in CSF immunoglobulin is the result of proliferation of B cells within the nervous system; the target epitopes of these antibodies are wildly variable.
81. What is neruromyelitis optica (Devic disease)?
Some individuals, especially Asians, develop a demyelinating disease similar to MS with presenting symptoms of bilateral optic neuritis and prominent spinal cord involvement.

This disease is referred to as neuromyelitis optica or Devic disease.

It may be rapidly and relentlessly progressive, following a relapsing-remitting course, or manifest as a single episode without subsequent relapses.

The lesions in Devic disease are similar in histologic appearance to MS, *although they are considerably more destructive, and gray matter involvement of the spinal cord can be striking*.
82. What is acute MS (Marburg form)?
Acute MS (Marburg form) tends to occur in *young individuals and is characterized clinically by a fulminant course during a period of several months.*

On pathological examination, the plaques are large and numerous, and there is widespread destruction of myelin with some axonal loss.
83. What is acute disseminated encephalomyelitis (ADEM)?
ADEM is a monophasic demyelinating disease that follow either a viral infection or, rarely, a viral immunization. Symptoms typically develop a week or two after the antecedent infection and include evidence of diffuse brain involvement with headache, lethargy, and coma rather than focal findings, as seen in MS.

Symptoms progress rapidly, with a fatal outcome in as many as 20% of cases; in the remaining patients there is complete recovery.
84. Morphology of ADEM
Macroscopic exam of the brain shows only *grayish discoloration around white matter vessels. On microscopic exam, myelin loss with relative preservation of axons can be found throughout the white matter. In the early stages of the disease, polymorphonuclear leukocytes can be found within the lesions; later, mononuclear infiltrates predominate.

The breakdown of myelin is associated w/the accumulation of lipid laden macrophages.
85. What is acute necrotizing hemorrhagic encephalomyelitis (ANHE)?
ANHE is a fulminant syndrome of CNS demyelination, typically affecting young adults and children.

*The illness is almost invariably preceded by a recent episode of upper respiratory infection.*

Sometimes, it is due to Mycoplasma pneumoniae, but often it is of indeterminate cause.

The disease is fatal in many patients, but some have survived with minimal residual symptoms.
86. Morphology of ANHE
Shows histologic similarities with ADEM, including perivenular distribution of demyelination and widespread dissemination throughout the CNS.

*However, the lesions are much more devastating than those of ADEM and include destruction of small blood vessels, disseminated necrosis of white and gray matter with acute hemorrhage, fibrin deposition, abundant neutrophils, and scattered lymphocytes recognizable in less severely damaged areas and in foci of demyelination.*
87. ADEM vs. ANHE
ADEM may represent an acute autoimmune reaction to myelin and ANHE may represent a hyperacute variant, although no inciting antigens have been identified.
88. What is central pontine myelinolysis?
Characterized by loss of myelin (with relative preservation of axons and neuronal cell bodies) in a roughly symmetric pattern involving the basis pontis and portions of the pontine tegmentum but sparing the periventricular and subpial regions.

Lesions may be found more rostrally; it is extremely rare for the process to extend below the pontomedulalry junction.

Extrapontine lesions occur in the supratentorial compartment, with similar appearance an apparent etiology.
89. What causes central pontine myelinolysis?
The condition is believed to be caused by rapid correction of hyponatremia; however, alternative pathogenetic hypotheses attribute the disorder to extreme serum hyperosmolarity or other metabolic imbalance.
90. Clinical presentation of central pontine myelinolysis
It is that of a rapidly evolving quadriplegia; radiologic imaging studies localize the lesion to the basis pontis.

It occurs in a variety of clinical settings, including alcoholism, severe electrolyte or osmolar imbalance, and orthotopic liver transplantation.
91. What is Marchiafava-Bignami disease?
This is a rare disorder of myelin characterized by relatively *symmetric damage* to the myelin of central fibers of the corpus callosum and anterior commissure.
92. What are degenerative diseases?
These are diseases of gray matter characterized principally by the progressive loss of neurons w/associated secondary changes in white matter tracts.
93. What are 2 general characteristics of degenerative diseases?
1. The pattern of neuronal loss is selective, affecting one or more groups of neurons while leaving others intact.

2. The diseases arise w/o any clear inciting event in pts w/o previous neurologic deficits.
94. What is a common theme among the neurodegenerative disorders?
A common theme is the development of protein aggregates that are resistant to normal cellular mechanisms of degradation thru the ubiquitin-proteasome system.

These aggregates can be recognized histologically as inclusions, which often form the diagnostic hallmarks of these different diseases.
95. What is Alzheimer disease?
AD is the most common cause of dementia in the elderly. The disease usually becomes clinically apparent as insidious impairment of higher intellectual function, w/alterations in mood and behavior.

Later, progressive disorientation, memory loss, and aphasia indicate severe cortical dysfunction, and eventually, in 5-10 years, the pt becomes profoundly disabled, mute, and immobile.
96. Who is most likely to get AD?
Pts rarely become symptomatic before 50, but the incidence of the disease rises w/age, and the prevalence doubles every 5 years.

Most cases are sporadic, although at least 5-10% of cases are familial. Pathologic changes identical to those observed in AD occur in almost all individuals w/trisomy 21 who survive beyond 45 years.
97. What does the gross exam of the brain look like in AD?
Macroscopic exam shows a variable degree of cortical atrophy w/widening of the cerebral sulci that is most pronounced in the frontal, temporal, and parietal loves.

With significant atrophy, there is compensatory ventricular enlargement (hydrocephalus ex vacuo) secondary to loss of parenchyma.
98. What are the 3 major microscopic abnormalities of AD?
1. Neuritic (senile) plaques
2. Neurofibrillary tangles
3. Amyloid angiopathy

All of these may be present to a lesser extent in the brains of elderly nondemented individuals.
99. What brain regions are affected first in AD?
Pathologic changes (specifically plaques, tangles, and the associated neuronal loss and glial reaction) are evident earliest in the entorhinal cortex, then spread thru the hippocampal formation and isocortex, and then extend into the neocortex.
100. What are neuritic plaques?
Senile or neuritic plaques are focal, spherical collections of dilated, tortuous, silver staining neuritic processes (dystrophic neurites) often around a central amyloid core, which ma be surrounded by a clear halo. Microglial cells and reactive astrocytes are present at their periphery.
101. Where are these neuritic plaques located?
They can be found in the hippocampus and amygdala as well as in the neocortex, although there is usually relative sparing of primary motor and sensory cortices (this also applies to neurofibrillary tangles).
102. What the hell are these plaques composed of?
The dystrophic neurites contain paired helical filaments as well as synaptic vesicles and abnormal mitochondria. The amyloid core, which can be stained by Congo red, contains several abnormal proteins.

The dominant component of the plaque core is Aβ, a peptide derived through specific processing events from a larger molecule, amyloid precursor protein (APP).
103. What are diffuse plaques in AD? What is their significance?
Immunostaining of Aβ demonstrates the existence in some pts of amyloid peptide deposits in lesions lacking the surrounding neuritic reaction.

These lesions are called diffuse plaques and they are found in superficial portions of cerebral cortex as well as in basal ganglia and cerebellar cortex.

*Diffuse plaques appear to represent an early stage of plaque development.
104. What is the main difference between neuritic and diffuse plaques?
While neuritic plaques contain both Aβ₄₀ and Aβ₄₂, diffuse plaques are predominantly made up of Aβ₄₂.
105. What are neurofibrillary tangles?
Neurofibrillary tangles are bundles of filaments in the cytoplasm of the neurons that displace or encircle the nucleus. In pyramidal neurons, they often have an elongated "flame" shape; in rounder cells, the basket weave of fibers around the nucleus takes on a rounded contour ("globose" tangles).

*Neurofibrillary tangles are visible as basophilic fibrillary structures but are clearly seen with silver staining.
106. Where are neurofibrillary tangles found?
They are commonly found in cortical neurons, esp in the entorhinal cortex, as well as in other sites such as pyramidal cells of the hippocampus, the amygdala, the basal forebrain, and the raphe nuclei.

*They are insoluble and resistant to clearance, thus remaining visible in tissue sections as "ghost" or "tombstone" tangles long after the death of the parent neuron.
107. What the hell are neurofibrillary tangles made of?
These tangles and composed predominantly of paired helical filaments along w/some straight filaments that appear to have a comparable composition.

A major component of paired helical filaments is abnormally hyperphosphorylated forms of the protein tau, an axonal microtubule-associated protein that enhances microtubule assembly.

Other antigens include MAP2, and ubiquitin.
108. What are neuropil threads?
Paired helical filaments are also found in the dystrophic neurites that form the outer portions of neuritic plaques and in axons coursing through the affected gray matter as neuropil threads.
109. What is central amyloid angiopathy (CAA)?
CAA is an almost invariable accompaniment of Alzheimer disease; however, ti can also be found in brains of individuals w/o AD.

Vascular amyloid is predominantly Aβ₄₀, as is also true when CAA occurs w/o AD.
110. What is granulovaculoar degeneration?
Granulovaculoar degeneration is the formation of small, clear intraneuronal cytoplasmic vacuoles, each of which contains an argyrophilic granule. While it occurs w/normal aging, it is most commonly found in great abundance in hippocampus and olfactor bulb in AD.
111. What are Hirano bodies? Where are they most commonly found?
Hirano bodies, found especially in AD, are elongated, glassy, eosinophilic bodies consisting of paracrystalline arrays of beaded filaments, with actin as their major component.

They are found most commonly w/in hippocampal pyramidal cells.
112. What correlates better w/the degree of dementia in AD - neurofibrillary tangles or neuritic plaques?
Neurofibrillary tangles
113. What is the best correlation of the severity of dementia in AD?
The loss of synapses.

Aβ is a critical molecule in the pathogenesis of this dementia.
114. How does Aβ cause AD?
Aβ aggregates readily, forming β-pleated sheets, and binds Congo red. It is relatively resistant to degradation, elicits a response from astrocytes and microglia, and can be directly neurotoxic.
115. Where does Aβ come from?
The Aβ peptides are derived through processing of APP. APP is a protein of uncertain cellular function that is expressed on the cell surface. A soluble form of APP can be released from the cell surface by proteolytic cleavage, by an enzymatic activity termed α-secretase. Molecules of APP that have undergone this cleavage cannot give rise to the Aβ fragment.

*However, surface APP can also be endocytosed and may then undergo processing to generate Aβ peptides that are less soluble and tend to aggregate into amyloid fibrils. These are generated through cleavage by β-secretase and cleavage w/in the transmembrane domain by γ-secretase. It is suggested that this γ-secretase gives rise to Aβ.
116. What are the four genes involved in AD?
1. The gene for APP on chromosome 21. Mutations in the APP gene all result in increased generation of Aβ.
2. Presenilin-1 (PS1) on chromsome 14
3. Presenilin-2 (PS2) on chromosome 1
4. Apolipoprotein E (ApoE) on chromosome 19
117. What is the significance of the presenilin gene mutations?
The presenilin gene mutations are linked to early-onset familial Alzheimer disease and probably account for the majority of early onset familial AD pedigrees.

***The cellular phenotype of these mutations was an increased level of Aβ generation, particularly Aβ₄₂.
118. How does presenilin affect Aβ generation?
The presenilins are a component of γ-secretase and possibly are the portion of a multiprotein complex containing the active proteolytic site.
119. What is the importance of Apolipoprotein E (ApoE) mutations?
ApoE mutations increases the risk of AD and lowers the age at onset of the disease.

People w/these mutations have a greater Aβ burden in their brains, as ApoE can bind Aβ and is present in plaques.
120. How, exactly, do aggregates of Aβ and larger fibrils damage neurons?
They are directly neurotoxic and can elicit various cellular responses, including oxidative damage and alterations in calcium homeostasis.

There is evidence that the inflammatory response that accompanies Aβ deposition may have both protective effects (through assisting clearance of the aggregated peptide) and injurious effects.
121. What are the clinical features of AD?
The progression of AD is slow but relentless, w/a symptomatic course often running more than 10 years. Initial symptoms are forgetfulness and other memory disturbances; with progression of the disease, other symptoms emerge, including language deficits, loss of mathematical skills, and loss of learned motor skills.

In the final stages of AD, pts may become incontinent, mute, and unable to walk. Intercurrent disease, often pneumonia, is usually the terminal event in these pts.
122. What is frontotemporal dementia w/Parkinsonism linked to chromosome 17 (FTD(P)-17)?
(FTD(P)-17) is a genetically determined disorder in which the clinical syndrome of a frontotemporal dementia is often accompanied by parkinsonian symptoms. In these families, the disease has been mapped to chromosome 17; in particular, it has been linked to a variety of mutations in the *tau gene.
123. What is the significance of tau?
Tau is a microtubule binding protein that has numerous sites of potential phosphorylation and exists in 6 splice forms as the result of alternative splicing of exons 2, 3, and 10.

The protein contains either 3 or 4 copies of the microtubule binding domain depending on whether exon 10 is included (4 repeat tau) or not (3 repeat tau).
124. What is the morphology of (FTD(P)-17)?
There is evidence of atrophy of frontal and temporal lobes in various combos and to various degrees. The pattern of atrophy can often be predicted in part by the clinical symptoms. The atrophic regions of cortex are marked by neuronal loss and gliosis as well as the presence of tau-contaning neurofibrillary tangles. These tangles may contain either 4 repeat tau or a mixture of 3 and 4 repeat tau, depending on the underlying genetic basis for the disease.

Nigral degeneration may also occur. Inclusions can also be found in glial cells in some forms of the disease.
125. What causes (FTD(P)-17)?
In some but not all pedigrees, there is linkage to mutations in the tau gene. The mutations fall into several categories; coding region mutations and intronic mutations that affect the splicing of exon 10.

The intronic mutations result in increased production of 4 repeat forms of tau. Coding region mutations appear to have several different consequences, including alterations in the interaction of tau w/microtubules (mutations in exon 10 will change this interaction only for 4 repeat tau) and altering the intrinsic tendency to aggregate.
126. What is Pick disease (lobar atrophy)?
Pick disease is a rare, distinct, progressive dementia characterized clinically by early onset of behavioral changes together w/alterations in personality (frontal lobe signs) and language disturbances (temporal lobe signs).

While most cases are sporadic, some familial forms identified are linked to mutations in tau.
127. What is the morphology of the brain in a gross exam in Pick disease?
The brain shows a pronounced, frequently asymmetric, atrophy of the frontal and temporal lobes w/conspicuous sparing of the posterior 2/3s of the superior temporal gyrus and only rare involvement of either the parietal or occipiatal lobe.

The atrophy can be severe, reducing the gyri to a thin wafer ("knife-edge" appearance).
128. How can one distinguish the atrophy of a brain in AD from Pick disease?
The pattern of lobar atrophy is often prominent enough to distinguish Pick disease from AD on macroscopic exam.

In addition to the localized cortical atrophy, there may also be bilateral atrophy of the caudate nucleus and putamen.
129. What are the morphologic features of Pick disease on microscopic exam?
On microscopic exam, neuronal loss is most severe in the outer 3 layers of the cortex.

***Some of the surviving neurons show a characteristic swelling (Pick cells) or contain Pick bodies, which are cytoplasmic, round to oval, filamentous inclusions that are only weakly basophilic but stain strongly with silver methods.
130. What are these Pick bodies made of?
Pick bodies are composed of straight filaments, vesiculated endoplasmic reticulum, and paired helical filaments that are immunocytochemically similar to those found in AD and contain 3 repeat tau.

Unlike the neurofibrillary tangles of AD, Pick bodies do not survive the death of their host neuron and do not remain as markers of the disease.
131. What is progressive supranuclear palsy (PSP)?
PSP is an illness characterized clinically by truncal rigidity w/dysequilibrium and nuchal dystonia; pseudobulbar palsy and abnormal speed; ocular disturbances, including vertical gaze palsy progressing to difficulty w/all eye movements; and mild progressive dementia in most pts.

The onset of the disease is usually btwn the 5ht and 7th decades, and males are affected approx 2x as freq as females. The disease is often fatal w/in 5-7 years of onset.
132. What is the morphology of progressive supranuclear palsy?
There is widespread neuronal loss in the globus pallidus, subthalamic nucleus, substantia nigra, colliculi, periaqueductal gray matter, and dentate nucleus of the cerebellum. Globose neurofibrillary tangles are found in these affected regions, in neurons as well as in glia.

*Ultrastructural analysis reveals 15-nm straight filaments that are composed of 4 repeat tau.
133. Are mutations in tau responsible for PSP?
Mutations in tau have not been found in PSP.

Analysis of the tau gene has shown that there is an extended haplotype. Of the two haplotypes, one of them is strongly overrepresented in PSP patients.
134. What is corticobasal degeneration (CBD)?
CBD is a disease of the elderly, and it is characterized by extrapyramidal rigidity, asymmetric motor disturbances (jerking movements of limbs: "alien hand"), and sensory cortical dysfunction (apraxias, disorders of language); cognitive decline occurs, and may be prominent in some cases.

*The same extended tau haplotype is linked to CBD as to PSP.
135. What is the morphology of CBD?
On macroscopic exam, there is cortical atrophy, mainly of the motor, premotor and anterior parietal lobes. *The regions of cortex show severe loss of neurons, gliosis, and "ballooned" neurons (neuronal achromasia).

Tau immunoreactivity has been found in astrocytes ("tufted astrocytes"), oligodendrocytes ("coiled bodies"), basal ganglionic neurons, and cortical neurons.
136. What are the most specific pathologic findings of CBD?
Clusters of tau-positive processes around an astrocytes ("astrocytic plaques") and the presence of tau-positive threads in gray and white matter may be the most specific findings of CBD.

Similar to PSP, the tau deposits in CBD contain predominantly 4 repeat tau.
137. What is motor neuron disease inclusion dementia?
Tau-negative, ubiquitin-positive inclusions can be found in superficial cortical layers in temporal and frontal lobe and in the dentate gyrus.

This pattern of path is termed motor neuron disease inclusion dementia. It has also been ascribed in the absence of ALS-like pathology.
138. What types of vascular injury to the brain can result in dementia?
Small areas of infarction (granular atrophy from cortical microinfarcts, multiple lacunar infarcts, cortical laminar necrosis associated w/reduced perfusion/oxygenation) and diffuse white matter injury (Binswanger disease, CADASIL).

Additional, dementia has been associated w/so called strategic infarcts, which are usually embolic and involve brain regions such as the hippocampus, dorsomedial thalamus, or frontal cortex including cingulate gyrus.
139. What is the interaction between vascular injury and AD?
It has been found that pts with vascular changes above a certain threshold have a lower burden of plaques and tangles for their level of cognitive impairment than do those w/o vascular base cerebral pathology.
140. What is Parkinsonism?
Parkinsonism is a clinical syndrome characterized by diminished facial expression, stooped posture, slowness of voluntary movement, festinating gait, rigidity, and a "pill-rolling" tremor.

This type of motor disturbance is seen in conditions that have in common damage to the nigrostriatal dopaminergic system.
141. What are four main diseases that involve the nigrastriatal system?
1. Parkinson disease
2. Multiple system atrophy
3. Postencephalitic parkonsonism
4. Progressive supranuclear palsy and corticobasal degeneration
142. What are the typical morphologic findings in parkinsons?
The typical macroscopic findings are pallor of the substantia nigra and locus ceruleus.

On microscopic exam, there is loss of the pigmented, catecholaminergic neurons in these regions associated w/gliosis. *Lewy bodies may be found in some of the remaining neurons.
143. What are Lewy bodies?
***Lewy bodies are single or multiple, cytoplasmic, eosinophilic, round to elongated inclusions that often have a dense core surrounded by a pale halo***
144. What are Lewy bodies made of?
Lewy bodies are composed of fine filaments, densely packed in the core but loose at the rim.

These filaments are composed of α-synuclein; neurofilament antigens, parkin, and ubiquitin are also present.
145. What is the main malfunction in Parkinsons?
The dopaminergic neurons of the substantia nigra project to the striatum, and their degeneration in Parkinson disease is associated w/a reduction int he striatal dopamine content. The severity of the motor syndrome is proportional to the dopamine deficiency, which can at least in part be corrected by replacement therapy with L-dopa.

Treatment does not, however, reverse the morphologic changes or arrest the progress of the disease; and w/progression, drug therapy tends to become less effective.
146. What can cause an acute parkinsonian syndrome and destruction of neurons in the substantia nigra?
Following exposure to MPTP, a contaminant in the illicit synthesis of psychoactive meperidine analogs. Action by MAO B is required for the toxicity of MPTP.

Also, pesticide exposure may increase the risk of PD, while caffeine and nicotine may be protective.
147. A gene encoding _______ was identified as the basis for inherited autosomal dominant for of PD.
α-synuclein, an abundant lipid-binding protein associated w/synapses.

However, only rare cases of PD have mutations in α-synuclein. Families w/autosomal dominant PD and genomic triplication of the region containing the gene for α-synuclein have been found.

***This suggests that gene dosage may also be related to PD, similar to the relationship btwn AD and trisomy 21.
148. What about the gene encoding the protein parkin?
A second gene, encoding the protein parkin, was linked w/a juvenile autosomal recessive form of PD. Alterations including deletions and nonsense and missense mutations resulting in loss of parkin functions have been found in various families.

***These mutations are most prevalent in the population of young-onset PD pts.
149. What is the role of parkin?
Parkin functions as an E3 ubiquitin ligase, with α-synuclein as one of its substrates.
150. What are the clinical features of PD?
About 10-15% of pts with PD develop dementia, w/increasing incidence w/advancing age.

Characteristic features of this disorder include a fluctuating course, hallucinations, and prominent frontal signs.

While many affected individuals also have pathologic evidence of AD, the dementia in others is attributed to widely disseminated Lewy bodies that are less distinct but still demonstrable by staining for ubiquitin and α-synuclein, particularly in the cerebral cortex but also involving the amygdala and brainstem neurons.
151. What is dementia with Lewy bodies (DLB)?
Similar pathology, with this distribution of cortical Lewy bodies, can also be found in individuals w/symptoms of dementia as their primary complaint 0 this is the disorder recognized as dementia with Lewy bodies (DLB).
152. What is the treatment for PD?
Sympatomatic resposne to L-dopa therapy is one of the features, in addition to clinincal sign and symptoms, that support a Dx of PD.

While L-dopa is often extremely effective in symptomatic treatment, it does not significantly alter the intrinsically progressive nature of the disease. Over time, L-dopa becomes less able to help the pt through symptomatic relief and begins to lead to fluctuations in motor function on its own.
153. What is multiple system atrophy?
MSA is now used to describe a group of disorders characterized by the presence of glial cytoplasmic inclusions (GCIs), typically w/in the cytoplasm of oligodendrocytes.

With recognition of GCIs, three entities of striatonigral degeneration, Shy-Drager syndrome, and olivopontocerebellar atrophy, were gathered into a single patholggic category.

*The identification of α-synuclein as the major component of the inclusions has resulted in this disorder being considered as a synucleinopathy. Unlike PD, no mutation sin the gene for this synaptic protein have been found in cases of MSA.
154. What is the morphology of MSA?
On macroscopic exam, there is typically atrophy of the cerebellum, including the cerebellar peduncles, pons, (especially the basis pontis), medulla (especially the inferior olive), substantia nigra, and striatum (especially putamen).

These brain regions show evidence of neuronal loss as well as variable numbers of neuronal cytoplasmic and nuclear inclusions.
155. What are the diagnostic features of MSA?
The cytoplasmic inclusion of MSA were originally demonstrated with silver impregnation methods and by immunostaining which stain α-synuclein as well as ubiquitin and αB-crystallin

***The inclusions are ultrastructurally distinct from those found in other neurodegenerative diseases and are composed primarily of 20-40 nm tubules. It appears the glial cytoplasmic inclusions can occur in the absence of neuronal loss, suggesting that they may represent a primary pathologic event.
156. What are the clinical features of MSA?
The two principal symptoms of MSA are parkinsonism and autonomic dysfunction, particularly orthostatic hypotension.

Parkinsonism can be related to the degree of cell loss from the substantia nigra and striatum; ataxia w/changes in the circuits involving the pons, cerebellum, and inferior olive; and autonomic symptoms with cell loss from the cetecholaminergic ncueli of the medulla and the IML column of the spinal cord.
157. What is Huntington disease?
HD is an inherited autosomal dominant disease characterized clinically by progressive movement disorders and dementia and histologically degeneration of striatal neurons.

The movement disorder chorea consists of jerky, hyperkinetic, sometimes dystonic movements affecting all parts of the body; pts may later develop parkinsonism w/bradykinesia and rigidity. The disease is relentlessly progressive, w/an average course of about 15 years to death.
158. What are the macroscopic characteristics of the brain in HD?
On macroscopic exam, the brain is small and shows ***striking atrophy of the caudate nucleus***, and less dramatically, the putamen. The globus pallidus may be atrophied secondarily, and the lateral and third ventricles are dilated.

Atrophy is freq also seen in the frontal lobe, less often in the parietal lobe, and occasionally in the entire cortex.
159. What are the microscopic characteristics of the brain in HD?
On microscopic exam, there is ***severe loss of striatal neurons; the most dramatic changes are found in the caudate nucleus, especially in the tail and portions nearer the ventricle.***

Pathologic abnormalities develop in a medial-to-lateral direction in the caudate and from dorsal to ventral in the putamen. The nucleus accumbens is the best preserved structure.
160. Which neurons are affected first in HD?
Both the large and small neurons are affected, but loss of the small neurons generally precedes that of the large. The medium-sized, spiny neurons that use GABA as their neurotransmitter, along w/enkephalin, dynorphin, and substance P, are especially affected.
161. Which two populations of neurons are relatively spared in HD?
1. The diaphorase-positive neurons that contain nitric oxide synthase
2. The large cholinesterase-positive neurons
162. What is the functional significance of the loss of medium spiny striatal neurons in HD?
The significance of the loss is to dysregulate the basal ganglia circuitry that modulates motor output.

The loss of the striatal inhibitory output to the external portion of the globus pallidus results in increased inhibitory input to the subthalamic nucleus. This inhibition of the subthalamic nucleus prevents it from exerting its regulatory effects on motor activity and thus leads to choreathetosis.
163. What gene is involved in HD?
The HD gene (genius!). It encodes a protein called huntintin. The coding region of the gene contains a polymorphic CAG trinucelotide repeat encoding a polyglutamine region of the protein.

Normal HD genes contain 6-35 copies of the repeat; in disease causing genes, the number of repeats is increased. Also, the larger the number of repeats, the earlier the onset of the disease.
164. So, what's the significance of the CAG repeat in HD?
The expanded polyglutamine repeat results in protein aggregation and formation of intranuclear inclusions.
165. What are the clinical features of HD?
The age at onset is most commonly in the 4th and 5th decades and is related to the length of the CAG repeat in the HD gene. Motor symptoms often precede the cognitive impairment. The movement disorder of HD is choreiform, w/increased and involuntary jerky movements of all parts of the body; writhing movements of the extremities are typical.

Early symptoms of higher cortical dysfunction include forgetfulness and thought and affective disorder, but there is progression to a severe dementia. HD pts have an increased risk of suicide, w/intercurrent infection being the most common natural cause of death.
166. What are spinocerebellar ataxias?
This is a group of genetically distinct diseases characterized by signs and symptoms referable to the cerebellum (progressive ataxia), brainstem, spinal cord, and peripheral nerves, as well as other brain regions in different subtypes.

Pathologically, they are characterized by neuronal loss from the affected areas w/secondary degeneration of white matter tracts.
167. What is a common genetic feature in the spinocerebellar ataxias?
Among dominantly inherited forms, there are many that are associated w/trinucleotide repeat expansion. In those forms in which there is expansion of a CAG repeat encoding polyglutamine tract (SCA1-3,6,7,17), neuronal intranuclear inclusions containing the abnormal protein can be found, similar to HD.
168. What is the significance of SCA6?
All but SCA6 show anticipation, w/expansion of the repeat length during gametogenesis. Curiously, SCA6 shares its genetic locus w/another cerebellar disorder - episodic ataxia type 2, which is associated w/point mutations.
169. What is Friedreich ataxia?
Friedreich ataxia is an autosomal recessive progressive illness, generally beginning in the first decade of life w/gait ataxia, followed by hand clumsiness and dysarthria.

DTRs are depressed or absent, but an extensor plantar reflex is typically present. Joint position and vibratory sense are impaired, and there is sometimes loss of pain and temp sensation and light tough.

Most pts develop pes cavus and kyphosciolosis. There is a high incence of cardiac disease w/arrhythmias and CHF. Most pts become wheelchair bound w/in 5 years of onset; the cause of death is intercurrent pulmonary infections and cardiac disease.
170. What is the gene involved in Friedreich ataxia?
The gene for Friedreich ataxia has been mapped to chromosome 9q13, and in most cases, there is a GAA trinucleotide repeat expansion in the first intron of a gene encoding a protein named frataxin.

*Affected individuals inherit abnormal forms of the frataxin gene from both parents and have extrememly low levels of the protein.*
171. What is the role of frataxin?
Frataxin undergoes processing and ends up in the inner mitochondrial membrane, where it has been suggested to play a role in regulation of iron levels.

B/c of the need for this metal in many of the complexes of the oxidative phosphorylation chain, mutations in frataxin have been suggested to result in generalized mitochondrial dysfunction.
172. What is the morphology of Friedreich ataxia?
The spinal cord shows loss of axons and gliosis in the posterior columns, the distal portions of the corticospinal tracts, and the spinocerebellar tracts. There is degeneration of neurons in the spinal cord, the brainstem (CN 8, 10, and 12), the cerebellum, and to some extent the Betz cells of the motor cortex.

Large dorsal root ganglion neurons are also decreased in number; their large myelinated axons, traveling first in the dorsal roots and then in dorsal columns, therefore undergo secondary degeneration. The heart is enlarged and may have pericardial adhesions.
173. What is ataxia-telangiectasia?
Ataxia-telangiectasia is an autosomal-recessive disorder characterized by an ataxic-dyskinetic syndrome beginning in early childhood, caused by neuronal degeneration predominantly in the cerebellum, the subsequent development of telangiectasias in the conjunctiva and skin, and immunodeficiency.
174. What are the immune characteristics of ataxia-telangiectasia?
Cells from pts w/the disease show increased sensitivity to x-ray induced chromosome abnormalities; these cells continue to replicate damaged DNA rather than stopping to allow repair or apoptosis.

Pts first come to medical attention b/c of recurrent sinopulmonary infections and unsteadiness in walking. Many affected individuals develops lymphoid malignant disease (T-cell leukemia, T-cell lymphoma); gliomas and CAs have been reported in some.
175. What is the gene responsible for ataxia-telangiectasia?
The ataxia-telangiectasia locus on chromosome 11q22-23 has been identified as a large gene, ATM, that encodes a protein w/a kinase domain. The protein orchestrates the cellular response to double-stranded DNA brakes.

As a result, the mutated ataxia-telangiectasia allele may underlie an increased risk of CA, specifically breast CA.
176. What is the morphology of ataxia-telangiectasia?
***The abnormalities are predominantly in the cerebellum, w/loss of Purkinje and granule cells; there is also degeneration of the dorsal columns, spinocerebellar tracts, and anterior horn cells and a peripheral neuropathy.***

The nuclei of cells in many organs show a bizarre enlargement of the cell nucleus to 2-5x normal size and are referred to as amphicytes. The lymph nodes, thymus, and gonads are hypoplastic.
177. What are the clinical features of ataxia-telangiectasia?
The disease relentlessly progresses to death early in teh second decade. Pts first come to medical attention b/c of recurrent sinopulmonary infectiosn and unsteadiness in walking.

Later on, speech is noted to become dysarthric, and eye movement abnormalities develop. Many affected individuals develop lymphoid malignant disease (T-cell leukemia, T-cell lymphoma); gliomas and CAs have been reported in some.
178. What is amyotrophic lateral sclerosis (ALS)?
ALS is characterized by neuronal muscle atrophy (amyotrophy) and hyperreflexia due to loss of lower motor neurons in the anterior horns of the spinal cord and upper motor neurons that project in corticospinal tracts, respectively.

The disease affects men slightly more freq than women and becomes clinically manifest in the 5th decade or later. 5-10% of cases are familial, mostly with autosomal dominant inheritance.
179. What are the genes responsible for ALS?
Fora subset of the familial cases, the genetic locus has been mapped to the copper-zinc superoxide dismutase gene (SOD1) on chromosome 21.

Also, an adverse gain of function phenotype caused by missense mutations have been identified. Among the mutations, the A4V mutation is the most common (50% of cases), and is associated w/a rapid course, and rarely has upper motor neuron signs.
180. What are the morphologic findings in ALS?

1/2
On macroscopic exam, the anterior roots of the spinal cord are thin; the precentral gyrus may be atrophic in especially severe cases. Microscopic exam demonstrates a reduction in the number of anterior horn neurons throughout the length of the spinal cord w/associated reactive gliosis and loss of anterior root myelinated fibers.

Remaining neurons often contain Bunina bodies: PAS positive cytoplasmic inclusions that appear to be remnant of autophagic vacuoles.
181. What are the morphologic findings in ALS?

2/2
Skeletal muscles innervated by the degenerated lower motor neurons show neurogenic atrophy.

Destruction of the upper motor neurons leads to degeneration of myelin the corticospinal tracts, resulting in pale staining that is particularly evident at the lower segmental levels but traceable throughout the corticospinal system w/special studies.
182. What are the clinical features of ALS?
Early symptoms include asymmetric weakness of the hands, manifested as dropping objects and difficulty in performing fine motor tasks, and cramping and spasticity of the arms and legs. As the disease progresses, muslce strength and bulk diminish, and involuntary contractions of individual motor units, called fasciculations, occur.

The disease eventually involves the respiratory muscles, leading to recurrent bouts of pulmonary infection.
183. What is progressive musclular atrophy?
The severity of involvement of the upper and lower motor neurons is variable; the term progressive muscular atrophy applies to those relatively uncommon cases in which lower motor neuron involvement predominates.
184. What is progressive bulbar palsy or bulbar amyotrophic lateral sclerosis?
In some pts, degeneration of the lower brainstem cranial motor nuclei occurs early and progresses rapidly, a pattern referred to as progressive bulbar palsy or bulbar amyotrophic lateral sclerosis.

**In these individuals, abnormalities of deglutition and phonation dominate, and the clinical course is relentless during a 1-2 year period.
185. What is bulbospinal atrophy (Kennedy syndrome)?
This X-linked adult onset disease is characterized by distal limb amyotrophy and bulbar signs such as atrophy and fasciculations of the tongue and dysphagia.

Affected individuals manifest androgen insensitivity w/gynecomastia, testicular atrophy, and oligospermia.
186. Tell me more about Kennedy syndrome...
On microscopic exam, there is degeneration of the lower motor neurons in the spinal cord and brainstem. The gene defect is expansion of a CAG/polyglutamine repeat in the androgen receptor (40-60 for affected individuals as opposed to 11-33 for the normal allele); nuclear inclusions containing aggregated androgen receptor can be found.
187. What are neuronal storage diseases?
Neuronal storage diseases are caused by a deficiency of a specific enzyme involved in the catabolism of sphingolipids, muopolysaccharides, or mucolipids.

They are often characterized by the accumulation of the substrate of the enzyme w/in the lysosomes of neurons, leading to neuronal death.
188. What are leukodystrophies?
Leukodystrophies show a selective involvement of myelin and generally exhibit no neuronal storage defects. Some of these disorders involve lysosomal enzymes; other affect peroxisomal enzymes.

Diffuse involvement of white matter leads to deterioration of motor skills, spasticity, hypotonia, or ataxia.
189. What is Krabbe disease?
Krabbe disease is an autosomal recessive leukodystrophy resulting from a deficiency of galactocerebroside β-galactosidase, the enzyme required for the catabolism of galactocerebroside to ceramide and galactose.
190. Is it the accumulation of galactocerebroside that causes toxicity in Krabbe disease?
While some accumulation of galactocerebroside may occur, this is not the direct toxic agent in Krabbe disease.

*Instead, it appears that an alternative catabolic pathway removes a fatty acid from this molecules, generating galactosylspingosine, which is a cytotoxic compound that could cause oligodendrocyte injury.
191. What are the clinical features of Krabbe disease?
The clinical course is rapidly progressive, with onset of symptoms often btwn the ages of 3 and 6 mos. Survival beyond 2 years is uncommon.

The clinical features are dominated by motor signs, including stiffness and weakness, w/gradually worsening difficulties in feeding. The brain shows loss of myelin and oligodendrocytes in the CNS and a similar process in peripheral nerves. Neurons and axons are relatively spared.
192. What is one unique feature of Krabbe disease?
The aggregation of macrophages filled w/cerebroside, forming multinucleated cells (globoid cells), around blood vessels.
193. What is metachromatic leukodystrophy?
Metachromatic leukodystrophy is an autosomal recessive disease that results from a deficiency of the lysosomal enzyme arylsulfatase A. This enzyme, present in a variety of tissues, cleaves the sulfate from sulfate-containing lipids, the first step in their degradation.

*Enzyme deficiency leads to an accumulation of the sulfatides, especially cerebroside sulfate which somehow breaks down myelin.
194. What are the different forms of metachromatic leukodystrophy?
Recognized clinical subtypes of the disorder include a late infantile form (the most common), a juvenile form, and an adult form.

The two forms w/childhood onset often present w/motor symptoms and progress gradually, leading to death w/in 5-10 years. In the adult form, psychiatric or cognitive symptoms are the usual initial complaint, w/motor symptoms coming later, and the disease has a slower course than in the infantile form.
195. What is the most striking histologic finding in metachromic leukodystrophy?
The most striking finding is demyelination w/resulting gliosis. Macrophages w/vacuolated cytoplasm are scattered throughout the white matter. The membrane bound vacuoles contain complex crystalloid structures composed of sulfatides. ***These sulfatides, when stained, shift the absorbance spectrum of the dye, a property called metachromasia.

The detection of metachromatic material in the urine is also a sensitive method for establishing Dx.
196. What is adrenoleukodystrophy?
Adrenoleukodystrophy is a progressive disease w/symptoms referable to myelin loss from the CNS and peripheral nerves as well as adrenal insufficiency. In general, forms w/early onset have a more rapid course. The X-linked form usually presents in the early school years w/neurologic symptoms and adrenal insufficiency and is rapidly progressive and fatal.

When it develops in adults, it is usually a slowly progressive disorder w/predominantly peripheral nerve involvement developing over a period of decades.
197. What gene is responsible for adrenoleukodystrophy? If mutated, what is the consequence?
The disease is associated w/mutations in the ALD gene, which encodes a member of the ATP-binding cassette transporter family of proteins.

*The disease is characterized by the inability to properly catabolize very-long chain fatty acids within peroxisomes, w/elevated levels of the long chain fatty acids in the serum.
198. What is Pelizaeus-Merzbacher disease?
Pelizaeus-Merzbacher disease is a fatal leukodystrophy beginning either early in childhood or just after birth, and is characterized by slowly progressive signs and symptoms resulting from widespread white matter dysfunction.

Pts present w/peduncular eye movements, hypotonia, choreoathetosis, and pyramidal signs early in the disease, followed later by spasticity, dementia, and ataxia.
199. What gene is defective in Pelizaeus-Merzbacher disease?
Pelizaeus-Merzbacher disease has been shown to arise in most cases from defects in a gene on the X chromosome that encodes proteolipid protein (PLP), a major protein of CNS myelin.

**Although myelin is nearly completely lost in the cerebral hemispheres, patches may remain, giving a "trigoid" appearance to tissue sections stained for myelin.
200. What is Canavan disease?
Canavan disease is characterized by megalocephaly, severe mental deficits, blindness, and signs and symptoms of white matter injury beginning in early infancy and relentlessly progressing to death by 18 mos of age.

Autopsy studies have shown spongy degeneration of the white matter, particularly affecting subcortical U fibers and Alzheimer type II astrocytes in the gray matter.

***Aspartoacylase activity is deficient in these pts, and point mutations and deletions in the gene for aspartylacylase underlie this autosomal-recessive disease.
201. What is Leigh syndrome (subacute necrotizing encephalopathy)?
Leigh syndrome is characterized in early childhood by lactic acidemia, arrest of psychomotor development, feeding problems, seizures, extraocular palsies, and weakness w/hypotonia. Death usually occurs w/in 1-2 years.

*Most cases show decreased activity of complex 4 (cytochrome c oxidase) of mitochondrial oxidative phosphorylation.
202. What are the morphologic features of the brain in Leigh syndrome?
On histologic exam, there are multifocal, moderately symmetric regions of destruction of brain tissue with a spongiform appearance and proliferation of blood vessels.

The areas that are most commonly affected include the periventricular gray matter of the midbrain, the tegmentum of the pons, and the periventricular regions of the thalamus and hypothalamus.
203. What is myoclonic epilepsy and ragged red fibers (MERRF)?
MERRF is a maternally transmitted disease in which pts have myoclonus, a seizure disorder, and evidence of a myopathy. Ataxia, associated w/evidence of neuronal loss from the cerebellar system, is also a common component.

MERRF is caused by mutations in mitochondrial tRNA, affecting protein synthesis w/in mitochondria.
204. What is mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS)?
MELAS is a mitochondrial disease in which children have acute episodes of neurologic dysfunction, cognitive changes, and evidence of muscle involvement w/weakness and lactic acidosis.

*Pathologically, areas of infarction are observed, often w/vascular proliferation and focal calcification. This syndrome is associated with mutations involvement a different mitochondrial tRNA.
205. What is Kearns-Sayre syndrome (KSS)?
KSS AKA "ophthalmoplegia plus" is a sporadic disorder, associated w/a large mtDNA deletion/rearrangement.

This disorder may present w/cerebellar ataxia in addition to the progressive external ophthalmoplegia, pigmentary retinopathy, and cardiac conduction defects. Pathologically, there is spongiform change in gray and white matter, w/neuronal loss most evident in the cerebellum.
206. What is Wernicke encephalopathy?
Wernicke encephalopathy is due to thiamine deficiency which can lead to development of psychotic symptoms or ophthalmoplegia that begins abruptly.

Treatment with vitamin B1 may reverse manifestations of Wernicke syndrome.
207. What is the morphology of the brain in Wernicke encephalopathy?
Wernicke encephalopathy is characterized by foci of hemorrhage and necrosis, particularly in the mammillary bodies but also adjacent to the ventricle, especially the 3rd and 4th ventricles.

Early lesions show dilated capillaries w/prominent endothelial cells. Subsequently, the capillaries leak red cells into the interstitium, producing hemorrhagic areas that are easily detectable macroscopically.
208. What is Korsakoff syndrome?
The acute stages of Wernicke encephalopathy, if left untreated, may be followed by a prolonged and largely irreversible condition known as Korsakoff syndrome. This syndrome is characterized clinically by memory disturbances and confabulation.

The syndrome is particularly common in the setting of chronic alcoholism but it may also be encountered in pts w/thiamine deficiency resulting from gastric disorders.
209. What are the morphologic findings in Korsakoff syndrome?
The capillaries that leak red cells into the interstitium, producing hemorrhagic areas in Wernicke encephalopathy, can over time, develop into cystic space w/hemosiderin-laden macrophages which are permanent.

These chronic hemosiderin-laden lesions predominate in pts with Korsakoff syndrome.
210. Which area of the brain appear to be best correlated with the neurologic symptoms in Korsakoff syndrome?
Lesions in the medial dorsal nucleus of the thalamus appear to be the best correlate of the memory disturbance and confabulation in Korsakoff syndrome.
211. Glucose deprivation in the brain initially leads to selective injury to which areas?
Glucose deprivation in the brain initially leads to selective injury to large pyramidal neurons of the cerebral cortex, which if it is severely involved, may result in pseudolaminar necrosis of the cortex, *predominantly involving layers 3-5.
212. What other areas of the brain are susceptible to hypoglycemic damage?
The hippocampus is also vulnerable to glucose deprivation, as it is to hypoxia, and may show a dramatic loss of pyramidal neurons in Sommer sector (CA1 of the hippocampus).

Purkinje cells of the cerebellum are also vulnerable to hypoglycemia, although to a lesser extent than to hypoxia.
213. What about carbon monoxide damage in the brain?
In CO poisoning, selective injury of the neurons of layers 3 and 5 of the cerebral cortex, Sommer sector of the hippocampus, and Purkinje cells is a common consequence.

Bilateral necrosis of the globus pallidus may also occur, and is more common in CO-induced hypoxia than in hypoxia from other causes.
214. What about methanol toxicity?
The pathologic findings of methanol toxicity are seen in the retina, where degeneration of the retinal ganglion cells may cause blindness.

Selective bilateral putamenal necrosis and focal white matter necrosis also occur when the exposure is severe.
215. Ethanol damage???
Cerebellar dysfunction occurs in about 1% of chronic alcoholics, associated w/a clinical syndrome of truncal ataxia, unsteady gait, and nystagmus.

**The histologic changes are atrophy and loss of granule cells predominantly in the anterior vermis.

In advanced cases, there is loss of Purkinje cells and proliferation of the adjacent astrocytes (Bergmann gliosis) btwn the depleted granular cell layer and the molecular layer of the cerebellum.
216. What are the CNS effects of radiation?
Delayed effects of radiation present w/rapidly evolving symptoms of an intracranial mass, including headaches, nausea, vomiting, and papilledema that may develop months to years after irradiation.

The pathologic findings consist of large areas of coagulative necrosis w/adjacent edema. The typical lesion is restricted to white matter, and all elements w/in the area undergo necrosis, including astrocytes, axons, oligodendrocytes, and blood vessels.

Also, proteinaceous spheroids may be identified adjacent to the area of coagulative necrosis.
217. What about MTX + radiation?
MTX toxicity most commonly develops when the drugs has been administered in associated w/radiotherapy, either together or at separate times. Symptoms often begin w/drowsiness, ataxia, and confusion, and may progress rapidly. Other may become comatose.

The pathologic basis of the symptoms are focal areas of coagulative necrosis w/in white matter, often adjacent to the lateral ventricles but at times distributed throughout the white matter or in the brainstem. Surrounding axons are often dilated and form axonal spheroids.
218. What are the two main dopamine receptors?
D1 and D2

Activation of D1 class receptors leads to increased cAMP, while activation of D2 class receptors inhibits cAMP generation.
219. What are the receptors in the D1 family?
D1: Straitum and neocortex

D5: Hippocampus and hypothalamus
220. What are the receptors in the D2 family?
D2: Striatum, substantia nigra, and pituitary

D3: Olfactory tubercle, nucleus accumbens, hypothalamus

D4: Frontal cortex, medulla, midbrain
221. What are the three main dopamine pathways in the brain?
1. Nigrostriatal pathway
2. Ventral tegmental area
3. Tuber-infundibular pathway
222. What is the nigrostriatal pathway?
Nigrostriatal pathway contains about 80% of the brains dopamine. This tract projects rostrally from cell bodies in the pars compacta of the substantia nigra to terminals that richly innervate the caudate and putamen (aka the striatum).

Dopaminergic neurons the nigrostriatal system are involved in the stimulation of purposeful movement.
223. What is the ventral tegemental area (VTA)?
Medial to the substantia nigra is an area of dopaminergic cell bodies in the midbrain called the VTA. The VTA has widely divergent projections that innervate many forebrain areas, most notably the cerebral cortex, the nucleus accumbens, and other limbic structures.

These systems play an important role in motivation, goal-directed thinking, regulation of affect, and positive reinforcement (reward). Derangement of these pathways may be involved in the development of schizophrenia.
224. What is the tubero-infundibular pathway?
Dopamine containing cell bodies i the arcuate and periventricular nuclei of the hypothalamus project axons to the median eminence of the hypothalamus.

This system is known as the tubero-infundibular pathway. Dopamine is released by these neurons into the portal circulation connecting the median eminence w/the anterior pituitary gland, and tonically inhibits the release of prolactin by pituitary lactotrophs.
225. Increased levels of dopamine in the straitum do what?
Increased levels of dopamine in the striatum tend to activate the D1-expressing neurons the direct pathway, while inhibiting the D2 expressing neurons of the indirect pathway.
226. What is levodopa?
Levodopa is still the most effective treatment for Parkinsons. Dopamine itself is not suitable b/c it cannot cross the BBB. However, dopamine's immediate precursor, L-dopa, is readily transported across the BBB by the neutral AA transporter.. Once in the CNS, L-dopa is converted to dopamine by the enzyme aromatic AA decarboxylase (AADC).

Thus, L-dopa must compete w/other neutral AAs for transport across the BBB, and its availability in the CNS may be compromised by recent protein meals.
227. Levodopa
MOA: Provide substrate for increased dopamine synthesis; levodopa is transported across the BBB by the neutral AA transporter and then decarboxylated to dopamine by AADC.

PURPOSE: Parkinsons disease

ADVERSE: Dyskinesia, heart disease, orthostatic hypotension, psychotic disorder, loss of appetite, vomiting

CONTRA: History of melanoma, narrow-angle glaucoma, concomitant use of MAOI
228. What are 3 therapeutic considerations for levodopa?
1. Levodopa, when administered alone, has low availability in the CNS. ***Therefore, it is almost always administered in combo with carbidopa, and inhibitor of DPA decarboxylase***
2. Continued use results in both tolerance and sensitization; pts develop periods of increased rigidity alternating with periods of normal or dyskinetic movement.
3. Dyskinesias are nearly ubiquitous in pts w/in 5 years of stating levodopa. As the disease progresses, continued levodopa therapy leads to worsening of both the dyskinesias and the "on/off" phenomenon.
229. What are the dopamine receptor agonists?
Another strategy for enhancing dopaminergic enurotransmission is to target the postsynaptic dopamine receptor directly thru the use of agonists.

These include:
1. Bromocriptine (D2 agonist)
2. Pergolide (D1 and D2)
3. Pramipexole (D3>D2)
4. Ropinirole (D3>D2)

This class has several advantages in that they do not compete with levodopa or toher neutral AAs for transport across the BBB. Furthermore, they do not require enzymatic conversion by AADC, and thus they remain effective later in the course of Parkinsons. Also, they have a longer half life.
230. Bromocriptine (MOA, PURPOSE, and ADVERSE)
MOA: A synthetic dopamine receptor agonist (D2 agonist) that inhibits lactotroph cells growth (decreases prolactin levels)

PURPOSE: Amenorrhea and galactorrhea from hyperprolactinemia, acromegaly, Parkinson's disease, premenstrual syndrome, Cushing syndrome, hepatic encepaholopathy, neuroleptic malignant syndrome related to neuroleptic drug therapy

ADVERSE: Cerebral vascular accident, seizure, acute myocardial infarction, dizziness, hypotension, abdominal cramps, nausea
231. Bromocriptine (CONTRA and NOTES)
Bromocriptine

CONTRA: Hypersensitivity to ergot derivatives, uncontrolled hypertension, toxemia of pregnancy

NOTES:
1. Ergot alkaloid; dose 2x/day
2. Intravaginal administration may reduce GI side effects
3. Alcohol intolerance may occur
4. First dose phenomenon occurs in 1% of patients and may result in syncope
5. Coadministration w/amitriptyline, butryophenones, imipramine, methyldopa, phenothiazines, or reserpine causes increased prolactin levels
6. Coadministration w/antihypertensives potentiates hypotension
232. Pergolide
MOA: Dopamine receptor agonist at D1 and D2 that also decreases prolactin levels

PURPOSE: Parkinson's disease, hyperprolactinemia

ADVERSE: Arrhythmias, MI, heart failure, pulmonary fibrosis, nausea, dizziness, dyskinesia, dystonia, hallucinations, somnolence, orthostatic hypotension, rhinitis

CONTRA: Hypersensitivity to ergot derivatives and uncontrolled hypertension

NOTES:
1. Use cautiously in patients prone to arrhythmias and underlying psychiatric disorders
2. CNS depressants have additive effects
233. Pramipexole and Ropinirole
MOA: Non ergot agonists at dopamine receptors (D3>D2), bind to and activate postsynaptic dopamine receptors directly

PURPOSE: Parkinson's disease, Restless leg syndrome (ropinirole)

ADVERSE: Dyskinesia, otherostatic hypotension, extrapyramidal movements, somnolence, dizziness, hallucinations, dream disorder, asthenia, amnesia

CONTRA: Concomitant use of other sedating meds

NOTES: These non-ergot dopamine agonists have fewer adverse effects than the ergo derivatives
234. Therapeutic considerations for dopamine agonists
1. Dopamine agonists have longer half lives than that of levodopa, which allows for less freq dosing.
2. The non ergot agonists have fewer adverse effects that ergot agonists
3. Cognitive effects can include excessive sedation, vivid dreams, and hallucinations
4. Some studies suggest that use of dopamine agonists rather than levodopa as initial treatment for Parkinson's disease delays the onset of "off" periods and dyskinesias, esp in younger people.
235. What are the inhibitors of dopamine metabolism?
Rasagiline, selegiline, tocapone, and entacapone
236. Rasagiline and Selegiline
MOA: Inhibit breakdown of dopamine in the CNS by inhibiting MAO-B

PURPOSE: Parkinson's

ADVERSE: BBB, GI hemorrhage, Orthostatic hypotension, dyskinesia, rash, dyspepsia, arthralgia, headache, weight loss, insomnia, (selegiline), confusion (selegiline)

CONTRA: Concomitant use of: cyclobenzaprine, mirtazapine, St. John's wort, DXM, meperidine, methadone, propoxyphene, tramadol, MOAI's, cocaine; elective surgery requiring general anesthesia; pheochromocytoma
237. Therapeutic considerations for rasagiline and selegiline
1. Selegiline in low doses is selective for MAO-B, which predominates in the striatum; higher doses inhibit MOA-A as well as MAO-B, w/associated risks of toxicity
2. *Selegiline forms the potentially toxic metabolite amphetamine, which may lead to insomnia and confusion esp in the elderly
3. Rasagiline does not form toxic metabolites
4. Both rasagiline and selegiline improve motor function when used alone and can augment the effectiveness of levodopa
238. Tolcapone and Entacapone
MOA: Inhibit breakdown of dopamine in the CNS by inhibiting COMP (tolcapone); also inhibit the breakdown of levodopa by COMT in the periphery (entacapone and tolcapone)

PURPOSE: Parkinson's

ADVERSE: Dyskinesia, dystonia, hallucinations, orthostatic hypotension (tolcapone), hyperpyrexia (tolcapone), fulminant hepatic failure (tolcapone), rhabdomyolysis (tolcapone), dyspepsia, dream disorder, sleep disorder

CONTRA: History of rhabdomyolysis or hyperpyrexia related to tolcapone; liver disease
239. Therapeutic considerations for tolcapone and entacapone
1. Tolcapone is a highly lipid-soluble agent that can cross the BBB, while entacapone distributes only to the periphery
2. ***COMT inhibitors can be used in combo w/carbidopa to further enhance the plasma half-life of levodopa; COMT inhibitors have been shown in some trials to reduce the "off" periods that are associated w/decreasing plasma levodopa levels
3. Rare but fatal hepatic toxicity has been reported w/tolcapone use
4. Entacapone is the more widely used COMT inhibitor
240. What about amantadine?
MOA: Antagonism of excitatory NMDA receptors

PURPOSE: Parkinson's; influenza A

ADVERSE:* Neuroleptic malignant syndrome*, exacerbation of mental disorder, insomnia, dizziness, hallucinations, agitation, orthostatic hypotension, peripheral edema, dyspepsia, livedo reticularis

CONTRA: Hypersensitivity

NOTES: Amantadine is used to treat levodopa-induced dyskinesias that develop late in the course of the disease; may exacerbate mental illness in pts with psychiatric illness or substance abuse problems
241. Trihexyphenidyl and Benztropine
MOA: Muscarinic receptor antagonists that reduce cholinergic tone in the CNS by modifying the actions of striatal cholinergic interneurons

PURPOSE: Parkinson's

ADVERSE: Angle-closure glaucoma, increased intraocular pressure, psychosis, hyperpyrexia (bentropine), paralytic ileus (benztropine); dizziness, blurred vision, nervousness, nausea, xerostomia, urinary retention

CONTRA: Narrow angle glaucoma, younger than 3 years, tradice dyskinesias (trihexyphenidyl)

NOTES: Trihexyphenidyl and Benztropine reduce tremor more than bradykinesia and are therefore effective in treating pts for whom tremor is the major symptom; may worsen dementia and cognitive impairment int he elderly
242. What does neuroleptic mean?
"Neuroleptic" emphasizes the drugs neurological actions that are commonly manifested as adverse effects of treatment.

These include extrapyramidal effects, resulting from DA receptor blockage in the basal ganglia, and include the parkinsonian symptoms of slowness, stiffness, and tremor.
243. What does antipsychotic mean?
"Antipsychotic" means the ability of these drugs to abrogate psychosis and alleviate disordered thinking in schizophrenic pts. The antipsycotics may be further divided into typical and atypical antipsychotics.
244. What are the 2 main categories of typical antipsychotics?

Give an example of the prototypical drug in each category.
1. Phenothiazines (Chlorpromazine)

2. Butyrophenones (Haloperidol)
245. What are the names of the phenothiazines?
1. Chropromazine
2. Thioridazine
3. Mesoridazine
4. Perphenazine
5. Fluphenazine
6. Thiothixene
7. Trifluoperzine
8. Chlorprothixene
256. Phenothiazines
MOA: Antagonize mesolimbic, and possibly mesocortical, D2 receptors; adverse effects are likely mediated by binding to D2 receptors in basal ganglia and pituitary

PURPOSE: Psychotic disorder, nausea and vomiting (chlorpromazine, perphenazine)

ADVERSE: Parkinsonian symptoms, *neuroleptic malignant syndrome*, tardive dyskinesia, anticholinergic symptoms

CONTRA: Myelosuppression, severe toxic CNS depression or comatose states, concomitant admin of drugs that prolong QT interval (thioridazine and mesoridazine), Parkinson's disease

NOTES: Fluphenazine is available as decanoate ester, deliver IM every 3-4 weeks
257. What are the therapeutic considerations for the phenothiazines?
In general, aliphatic phenothiazines are less potent antagonists at D2 receptors than butyrophenones, thioxanthenes, or phenothiazines functionalized w/a piperazine derivatives
3. Administration of typical antipsychotics to pts with Parkinson's disease often leads to marked worsening of parkinsonian symptoms
4. Typical antipsychotics potentiate the sedative effects of benzos and antihistamines
258. What is neuroleptic malignant syndrome?
NMS is a rare, but life-threatening syndrome characterized by catatonia, stupor, fever, and autonomic instability; myoglobinemia and death occur in about 10% of these cases.

NMS is most commonly associated w/the typical antipsychotic drugs that have a high affinity for D2 receptors, such as haloperidol.

NMS is thought to arise at least in part from the actions of the antipsychotics on the dopaminergic system in the hypothalamus, which are essential for the body's ability to control temperature.
259. What is the relationship between potency and adverse effects with the typical antipsychotics?
High-potency drugs tend to have fewer sedative adverse effects and cause less postural hypotension than drugs with lower potency (i.e., drugs that require high doses to achieve a therapeutic effect).

On the other hand, lower-potency typical antipsychotics tend to cause fewer extrapyramidal adverse effects.
260. What are the butyrophenones?
Haloperidol and droperidol.

These decanoate esters are highly lipophilic drugs and are injected via IM, where they are slowly hydrolyzed and released. The decanoate ester dosage forms provide a long-acting formulation that can be administered every 3-4 weeks.

These formulations are particularly useful for treating poorly compliant pts.
261. Haloperidol and Droperidol
MOA: Antagonize mesolimbic, and possibly mesocortical, D2 receptors; adverse effects are likely mediated by binding to D2 receptors in basal ganglia and pituitary gland

PURPOSE: Psychoses (haloperidol), Tourette's syndrome (haloperidol), Nausea and vomiting; anesthesia adjunct (droperidol)

ADVERSE: Same as phenothiazines

CONTRA: Parkinson's disease, severe toxic CNS depression or comatose states

NOTES: *Haloperidol is the most widely used butyrophenone. Decanoate ester formulation is great for treating poorly compliant pts due to its long acting delivery
262. What are three other typical antipsychotics?
1. Loxapine
2. Molindone
3. Pimozide
263.

Loxapine
Molindone
Pimozide
MOA: Antagonize D2 receptors.
Molindone exerts it antipsychotic effects on the ascending reticular activating system in the absence of muscle relaxation and incoordination effects; Pimozide has more specific dopamine receptor antagonism and less alpha adrenergic receptor blocking activity, which results in less potential for inducing sedation and hypotension

PURPOSE: Psychotic disorders, Tourette's syndrome (pimozide)

ADVERSE: Parkisonian symptoms, NMS, tardive dyskinesia, prolonged QT interval (pimozide), anticholinergic symptoms, sedation

CONTRA: Comatose or severe drug induced depressed states; Parkinson's; See next card!!!
234. What are the contraindications unique to pimozide?
1. Concomitant pemoline, methylphenidate, or amphetamines that may cause motor and phonic tics
2. Concomitant dofetilide, sotalol, and other class Ia and class III anti-arrhythmics, mesoridazine, thoridazine, chlorpromazine, or droperidol
3. Concomitant use of fluoroquinolone antibiotics, mefloquine, pentamidine, arsenic trioxide, levomethadyl acetate, dolasetron, mesylate, probucol, tacrolimus, ziprasidone, sertraline, or macrolide antibiotics
4. Concurrent administration w/drugs that have demonstrated QT prolongation and inhibitors of P450 3A4 (zileuton, fluvoxamine)
5. History of cardiac arrhythmias
235. What are the atypical antipsychotics?
1. Risperidone
2. Clozapine
3. Olanzapine
4. Quetiapine
5. Ziprasidone
6. Aripiprazole

*These atypical antipsychotics have efficacy and adverse effect profiles that differ from those of the typical ones. All of these drugs are more effective than typical antipsychotics at treating the "negative symptoms" of schizophrenia. In addition, risperidone is more effective at combating the positive symptoms of schizophrenia. These cause milder extrapyramidal effects as well.
236. Risperidone
MOA: Risperidone binds to and antagonizes D2, 5HT2, α1. α2. H1 receptors

PURPOSE: Psychotic disorders and bipolar disorder

ADVERSE: Mild extrapyramidal symptoms, QT prolongation, anticholinergic symptoms (dry mouth, constipation, urinary retention), sedation, weight gain

CONTRA: Hypersensitivity

NOTES: Atypicals are more effective than typicals at treating the negative symptoms of schizophrenia.
237. Clozapine
MOA: Clozapine binds to and antagonizes D1-D5, 5-HT2, α1, H1, muscarinic receptors

PURPOSE: Schizophrenia refractory to other antipsychotics

ADVERSE: Mild extrapyramidal symptoms, *agranulocytosis, anticholinergic symptoms (dry mouth, constipation, urinary retention), sedation, weight gain

CONTRA: History of clozapine-induced agranulocytosis or severe granulocytopenia; myeloproliferative disorders

NOTES: *Clozapine has not been used as a first-line agent b/c of a small but significant risk of agranulocytosis
238. Olanzapine
MOA: Olanzapine binds to and antagonizes D1-D4, 5-HT2, α1, H1, M1-M5 receptors

PURPOSE: Psychotic disorders; bipolar disorder

ADVERSE: Mild extrapyramidal symptoms, anticholinergic symptoms (dry mouth, constipation, urinary retention), sedation, weight gain

CONTRA: Hypersensitivity
239. Quetiapine
MOA: Quetiapine binds to and antagonizes D1, D2, 5-HT1, 5-HT2, α1, α2, H1 receptors

PURPOSE: Psychotic disorders, bipolar disorder

ADVERSE: Mild extrapyramidal symptoms, anticholinergic symptoms (dry mouth, constipation, urinary retention), sedation, weight gain

CONTRA: Hypersensitivity
240. Ziprasidone
MOA: Ziprasidone binds to and antagonizes D2, 5-HT1, 5-HT2, α1, H1 receptors

PURPOSE: Psychotic disorder, bipolar disorder

ADVERSE: Mild extrapyramidal symptoms, QT prolongation, anticholinergic symptoms (dry mouth, constipation, urinary retention), sedation, weight gain

CONTRA: Concomitant arsenic trioxide, chlorpromazine, class Ia and III antiarrhythmics , or other drugs that cause QT prolongation; QT prolongation history including congenital long QT syndrome; cardiac arrhythmias, recent acute MI, uncompensated heart failure
241. Aripiprazole
MOA: Aripiprazole is a D2 and 5HT1A partial agonist and a 5-HT2A antagonist

PURPOSE: Psychotic disorders and bipolar disorder

ADVERSE: Same as risperidone

CONTRA: Hypersensitivity