• 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

1/693

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;

693 Cards in this Set

  • Front
  • Back
What is responsible for central chromatolysis of neurons?
Axonal trans-section – in central chromatolysis, the nucleus moves to the periphery and nucleolus becomes more prominent (in an attempt to produce more RNA to regenerate more axonal proteins)
What is an example of an inclusion body in oligodendrocytes?
PML viral infection (JC virus) --> leads to intra-nuclear inclusions + glassy appearance of oligodendrocytes
What is the most commonly seen cell in white matter?
Oligodendrocytes: make myelin – if injured, result in demyelination
What are astrocytic responses to injury?
Gliosis (proliferation of astrocytes in response to injury (fibrosis-like), seen w/ any pathologic process)
Rosenthal fibers: intracytoplasmic inclusions in astrocytes
Corpora amylacea: non-pathologic (seen with aging), look like Lafora bodies of neurons
Alzheimer type 2 astrocytes: seen in metabolic diseases (not Alzheimer’s!)
What are the different types of astrocytes?
Gray-matter astrocytes (Protoplasmic astrocytes);
White-matter astrocytes (Fibrillary astrocytes) --> involved most commonly in disease processes of astrocytes – astrocytes in white matter are not obvious on microscopy, have role in formation of BBB
What are examples of intra-cytoplasmic inclusions in neurons?
Pathologic: Rabies infection (Negri bodies), neurofibrillary tangle (pathologic, seen in aging and Alzheimer’s disease), Lewy bodies (seen in pigmented neurons affected by Parkinson’s), Lafora bodies
Non-pathologic: Lipofuscin (non-pathologic, golden-brown material, seen with wear-and-tear of neurons), Colloid inclusion (non-pathologic)
What are examples of intranuclear inclusions in neurons?
Pathologic: CMV infection
Non-pathologic: Marinesco body (seen with aging, positioned next to nucleolus of neuron)
What characterizes neuron cells?
Large nucleus with prominent nucleolus is usually seen in neurons – variations exist
What changes are seen in the cell morphology of neurons at death?
Nucleus becomes more condensed and less well-defined, and cytoplasm becomes more eosinophilic (“red and dead”) – in gross brain, atrophy shows as narrowing of gyri and deepening of sulci
What are neuronal reactions to injury?
Acute neuronal injury, atrophy, central chromatolysis/axonal reaction, neuronal inclusions
What characterizes cerebral edema?
Increased brain volume due to increased water content, may complicate any pathologic process, types include: vasogenic, cytotoxic, interstitial/hydrocephalic
What is subfalcine herniation?
Herniation of cingulate gyrus underneath the falx, most commonly caused by asymmetric space-occupying lesion above the tentorium (supratentorial) --> brain herniation thru falx causes compression of adjacent ACA, leading to 2’ infarct
What are the major types of herniation in brain?
Cerebellar tonsillar herniation, subfalcine (cingulate) herniation, transtentorial herniation (types: lateral/uncal, central), “upward” herniation
What is seen grossly in all types of cerebral edema?
Flattening of gyri with narrowing of sulci – cerebral edema is a gross diagnosis (e.g. by imaging the entire brain), rather than a microscopic diagnosis (however, if do microscopic analysis, may see vacuolation of parenchyma, cell swelling, enlarged extracellular spaces, fluid around blood vessels)
What characterizes interstitial/hydrocephalic edema?
CSF accumulates in extracellular spaces of periventricular white matter --> reduces volume of periventricular white matter; usually due to obstruction of CSF flow, may be reversible surgically by shunting CSF out of the ventricular space
What characterizes cytotoxic edema?
Intra-cellular swelling due to increased cellular concentration of osmotically active solutes, gray and white matter affected equally, usually a complication of hypoxia/ischemia-induced damage to Na/K pump in affected cells
What is the most common type of cerebral edema?
Vasogenic edema: results from increased permeability of blood vessels across BBB, white matter is most severely affected, fluids mainly in extracellular spaces
What occurs in an injury to ependymal cells and choroid plexus?
Little regenerative capacity of the single epithelial layer of the ependyma --> (instead, will get) proliferation of glial cells beneath the ependyma, leading to bumps on the ependyma known as “granular ependymitis”
What are the various types of response to injury in microglial cells?
Become elongated --> “Rod cells” --> when rod cells proliferate, may cause “Microglial nodules”
Acquire foamy cytoplasm --> “Glitter cells”; Neuronophagia
What are the macrophages/histiocytes of the brain?
Microglia (BM derived like other histiocytes)
What characterizes lateral (uncal) transtentorial herniation?
Displacement of uncus and parahippocampal gyrus over the free edge of the tentorium --> can lead to PCA compression (supplies visual cortex --> may see blindness), can lead to Kernohan’s notch
What lesions are most commonly responsible for obstructive hydrocephalus?
Obstructive lesions in 3rd ventricle or cerebral aqueduct
What characterizes hydrocephalus?
Increase in water content of brain within the ventricular system (leading to dilation of ventricular system), can be obstructive (communicative or non-communicative), non-obstructive (increased CSF production, impaired absorption), or Hydrocephalus ex vacuo (loss of white matter volume leading to compensatory dilation of ventricular system)
What characterizes tonsillar herniation?
Herniation of cerebellum thru the foramen magnum, most commonly seen with posterior fossa or cerebellar space-occupying lesions, leads to respiratory arrest by medullary compression
What characterizes central transtentorial herniation?
Downward displacement of brainstem (e.g. hypothalamus and rostral brainstem) through the incisura --> can lead to “Duret hemorrhage” in the middle of brainstem (immediately fatal)
What is Kernohan’s notch?
Infarction of cerebral peduncle (opposite to brain lesion) as well as compression of CNIII (same side as brain lesion) --> leading to unopposed sympathetic stimulation of pupils causing pupillary dilation on same side as brain lesion + paralysis on same side as brain lesion (e.g. right hemisphere uncal lesion --> left cerebral peduncle infarct --> decussation of fibers --> right-sided paralysis)
What is dementia?
Progressive loss of cognitive function independent of the state of attention
What is seen microscopically in Alzheimer’s brain?
Senile plaques (due to β Amyloid deposition either in diffuse or neuritic fashion)
Neurofibrillary tangles (made up from Tau proteins – shows as elongated, flame-shaped intra-cytoplasmic Tau protein inclusions)
Granulovacuolar degeneration (“dot and halo”) + Hirano bodies (only in hippocampus), Amyloid angiopathy
What is seen pathologically in Alzheimer’s brain?
Diffuse cortical atrophy with disproportionately prominent atrophy of hippocampi (leading to small hippocampi), loss of white matter leading to hydrocephalus ex vacuo – vision and ability to move is lost very late in AD
What characterizes clinical features of Alzheimer’s?
Initial symptoms: forgetfulness and memory problems (AD initially affects hippocampus)
Later symptoms: language problems, loss of math skills, loss of learned motor skills
Final symptoms: incontinence, mutism, inability to walk
Death: due to other diseases (esp. pneumonia), Progression is slow and relentless
What is associated with genetic forms of Alzheimer’s?
Mutations affecting the β Amyloid (Aβ) production/processing;
APP gene (chr. 21) --> Down syndrome (Trisomy 21) can lead to Alzheimer’s due to having 3 copies of Amyloid Precursor Protein (APP); PS1 gene (chr. 14), PS2 gene (chr. 1)
What is the pathogenesis of Alzheimer’s?
Involves a combination of β-amyloid protein (Aβ) and Tau protein abnormalities
What is the most common cause of dementia in the elderly?
Alzheimer’s – most important risk factor is increasing age (40% in 85-89yo), 10% of cases are familial, true diagnosis made only on autopsy of brain, F > M
How are neurodegenerative disorders defined?
Progressive loss of neurons that typically arise without any inciting events – classified based on site of involvement and/or type of protein aggregates present
What are the various types of protein aggregates present in neurodegenerative disorders?
Amyloid (Aβ): seen in Alzheimer’s, Tau proteins (seen in tauopathies), α-synuclein (seen in synucleinopathies), ubiquitin (seen in almost all of neurodegenerative diseases)
What are the various types of neurodegenerative disorders based on site of involvement?
Cerebral cortex --> dementing disorders, subcortical areas (basal ganglia, brainstem) --> movement disorders, cerebellum and spinal cord --> spinocerebellar ataxias, motor neurons in spinal cord --> motor neuron diseases
What characterizes Progressive Supranuclear Palsy?
Hybrid disease (dementing and movement disorder) – mild progressive dementia + movement disorder (truncal rigidity, vertical gaze palsy), M > F, fatal within 5-7 years;
What characterizes Frontotemporal Dementia with Parkinsonism linked to Chromosome 17?
Familial disease with frontotemporal dementia and Parkinsonism, due to mutation in Tau gene (Tauopathy) – will have only neurofibrillary tangles (similar to AD) without any plaques found in AD
What characterizes Motor Neuron Disease Inclusion Dementia?
May or may not be associated with motor neuron disease; shows Tau-negative, silver-negative, ubiquitin-positive inclusions – most commonly found in the dentate fascia of the hippocampus
What characterizes pathology of Corticobasal Degeneration?
Cortical atrophy (less prominent than AD), ballooned neurons
Tauopathy in gray and white matter: Tau-positive inclusions in neurons (tangle-like) as well as astrocytes (astrocytic plaques and tufted astrocytes) and oligodendrocytes (coiled bodies)
Note: know that this disease extensively involves white matter as well as gray matter
What is Corticobasal Degeneration?
Cognitive decline with sensory cortical dysfunction (cortical) + movement disorder such as rigidity and asymmetric motor disturbances (basal); affects elderly
How does Progressive Supranuclear Palsy differ from Alzheimer’s?
M > F; Neurofibrillary tangles are globose (rounded) as opposed to elongated, flamed-shaped in AD, no plaques present --> pure Tauopathy; brain atrophy is less prominent and is more focused in deep gray structures (globus pallidus)
What characterizes pathology of Progressive Supranuclear Palsy?
Neuronal loss is more in deep gray structures (globus pallidus, substantia nigra, inferior/superior colliculus, etc), shows Globose (rounded) neurofibrillary tangles (Tau protein), no plaques (no amyloid role), Tauopathy
What are different types of frontotemporal dementias?
Pick disease, Progressive supranuclear palsy, Corticobasal degeneration, Motor neuron disease inclusion dementia, Frontotemporal dementia with Parkinsonism linked to chromosome 17, Dementia lacking distinctive histology
What are the pathologic findings in Pick Disease?
Lobar atrophy (mainly in frontal and temporal lobes, sparing the posterior 2/3rd of superior temporal gyrus --> “knife-edge atrophy”), deeper atrophy (caudate and putamen), Pick bodies (mainly seen in dentate fascia of the hippocampus), Tauopathy (stains positive for Tau proteins)
What characterizes Pick Disease?
Early on affects temporal lobes (not hippocampus) mainly --> characterized by early onset of behavioral changes (e.g. aggression) together with alterations in personality and language disturbances (no memory loss)
What characterizes the pathology of Parkinson’s disease?
Pallor of substantia nigra and locus ceruleus, loss of pigmented neurons with associated gliosis and pigment incontinence, Lewy bodies (!)
Lewy bodies = if see inclusion in cytoplasm in pigmented neurons --> think about PD
What is Friedrich Ataxia?
Spinocerebellar ataxia due to GAA trinucleotide repeat; hereditary (AR) due to changes on frataxin gene on 9q12; begins in 1st decade with gait ataxia, high incidence of CV disease, and concominant diabetes; death is due to pulmonary infection or CV disease
What are spinocerebellar ataxias?
Diseases affecting cerebellum and spinal cord – most are trinucleotide repeat disorders (CAG or GAA); includes the hereditary form of olivopontocerebellar atrophy
Note: if see atrophy of olive, pons, and cerebellum in young patient AND familial --> think Spinocerebellar ataxias
Note: if see the same in older patient with NO family history --> think Multiple System Atrophy
What is Multiple System Atrophy?
A group of movement disorders characterized by presence of glial cytoplasmic inclusions; include:
Sporadic form of olivopontocerebellar atrophy: ataxia, cerebellar dysfunction
Striatonigral degeneration: Parkinsonism without Lewy bodies (instead have glial cytoplasmic inclusions)
Shy-Drager syndrome (Parkinsonism with autonomic dysfunction)
Inclusions are positive for alpha-synuclein, ubiquitin, and alphaB-crystallin (hence, a synucleinopathy) and are present in oligodendrocytes and astrocytes (rather than neurons as in Parkinson’s disease)
What characterizes the pathology of Huntington’s disease?
Atrophy of caudate and putamen with loss of medium-sized spiny (GABAergic) neurons with corresponding gliosis
What movement disorder is seen with Huntington’s disease?
Chorea: jerky, hyperkinetic movements affecting all parts of the body
May also have dementia and depression (increased risk of suicide)
What characterizes Huntington’s disease?
Hereditary (AD) due to CAG trinucleotide repeat expansion in the gene (chromosome 4) encoding huntingtin; Paternal transmission is associated with anticipation (earlier onset in succeeding generations); larger repeats are also associated with earlier onset
What is a likely diagnosis in a presentation of dementia with “fluctuating course” and hallucinations?
Dementia with Lewy Bodies – dementia is fluctuating and associated with hallucinations and prominent frontal signs – important to ID because some drugs to treat AD (cholinesterase inhibitors) will worsen Dementia with Lewy Bodies – can see Lewy bodies (ubiquitin-positive) in brainstem as well as cortex, is a synucleinopathy (stains positive with synuclein)
What characterizes Parkinson’s disease?
Synucleinopathy: rare cases of juvenile AR Parkinson’s disease are due to mutations in the α-synuclein and parkin genes – patients present with Parkinsonism, 10-15% with dementia; treatment includes L-dopa and neurosurgery
What is Parkinsonism?
Clinical syndrome characterized by TRAP: Tremor (“pill-rolling”), Rigidity, Akinesia (diminished facial expression, slowness of voluntary movement), Posture and gait abnormalities (“festinating” gait: progressively shortened, accelerated steps) – all symptoms are due to damage to the nigrostriatal DA neurons (if damage is due to neurodegenerative disease --> Parkinson’s disease; other cause of damage include infectious, meperidine, progressive supranuclear palsy, corticobasal degeneration, multiple system atrophy)
What are the diseases affecting motor neurons?
ALS (progressive muscular atrophy and bulbar palsy)
Hereditary: Bulbospinal atrophy (Kennedy syndrome), Spinal muscular atrophy
What is a hereditary form of motor neuron diseases?
Bulbospinal atrophy (Kennedy syndrome) – X-linked due to CAG repeat in androgen receptor gene; presents as young adults with LMN disease as well as androgen problems (gynecomastia, testicular atrophy, low sperm count)
What microscopic findings are seen in ALS?
Bunina bodies in anterior horn neurons of spinal cord, loss of anterior horn cells, motor neurons in CN nuclei, myelinated axons in corticospinal tracts; neurogenic atrophy of skeletal muscle
What gross findings are seen in ALS?
Thinning of anterior roots of spinal cord, atrophy of precentral gyrus (UMN located here)
How does a patient with ALS present?
Early signs: asymmetric weakness of the hands, cramping and spasticity of the arms and legs
Later: fasciculations, Death: involvement of pulmonary muscles leading to pulmonary infections
What characterizes ALS?
Loss of LMN and UMN; M > F; 10% cases are familial with mutations in Cu-Zn SOD1 gene on chromosome 21
What is the general presentation of diseases affecting motor neurons?
Signs of LMN disease: Denervation of muscles (atrophy, weakness)
Sings of UMN disease: hyperreflexia, Babinski sign (positive if toes upgoing on scratch)
What is the pathology involved in Friedrich Ataxia?
Loss of neurons in cerebellar vermis (Purkinje cells) and dentate nucleus, brainstem (CN VII, X, XII), and in Clarke column of spinal cord; also, loss of axons and gliosis in posterior tracts, corticospinal, and spinocerebellar tracts in spinal cord
How is the pathology of AT different from FA?
In addition to spinocerebellar degeneration (e.g. loss of cerebellar Purkinje cells), will see telangiectasias and amphicytes (bizarre, enlarged nuclei due to inability to repair DNA) in many organs
How is Ataxia-Telangiectasia (AT) different from Friedrich Ataxia (FA)?
AR and childhood onset but due to mutation of ATM gene on chromosome 11; also in addition to cerebellar ataxia, patients have increased risk of x-ray-induced chromosome abnormalities; patients have telangiectasias in conjunctiva and skin, increased risk of lymphoid malignancies, and immunodeficiency
What is spinal muscular atrophy?
Group of AR diseases due to Survival Motor Neuron (SMN1) gene on chromosome 5; infantile form is the most significant with lowest survival (Werding-Hoffman disease)
Which drugs are best for 1st, 2nd, and neuropathic pain, respectively?
2nd: NSAIDs; 1st/2nd: Opioids; Neuropathic: Na+ channel antagonists and Gabapentin (Neurontin)
What is the effect of Substance P at the spinal nociceptive synapse?
It works synergistically with the NMDA receptor
What is the effect of Glutamate at the spinal nociceptive synapse?
Acts on AMPA receptor to generate APs; Acts on NMDA receptor to cause an influx of Ca2+ which subsequently leads to increased protein phosphorylation, NO production, and prostaglandin synthesis (can be blocked by NSAIDs), all of which lower the threshold to electrical excitability
How does Acetaminophen work?
It inhibits central prostaglandin formation (but not peripheral)
What noxious chemicals cause electrical activation of primary afferents (e.g. C-fibers)?
5-HT, Histamine, H+, Bradykinin
How are NSAIDs effective against inflammatory or 2nd pain?
They block molecular alterations that lead to the sensitization of nociceptive neurons (i.e. they block the peripheral sensitization of C-fibers to noxious chemicals by inhibiting the formation of prostaglandins), sensitization in the CNS may also be attenuated (by inhibiting central prostaglandin formation)
What happens in the primary afferents and in the neurons of the spinal cord in response to acute injury?
Primary afferents: immediately --> massive burst of action potentials, later on --> peripheral sensitization (regularly occurring APs); Neurons in spinal cord: immediately --> same as primary afferent, later on --> wind-up (progressively increasing frequency of APs) followed by central sensitization (maintenance of hyperexcitable state)
What is meant by endogenous pain inhibition?
Neuronal processes that arise primarily in the midbrain/brainstem periaqueductal gray (PAG), travel downwards to the spinal cord, and inhibit transmission of pain at the spinal synapses;
they contain NE and 5-HT neurons – the system may be activated consciously in response to learned reactions, emotionally, by noxious input or by drugs
What types of pain are associated with neuropathic pain (e.g. with diabetic neuropathy)?
Inflammatory pain as well as an intense, episodic, lightning-like pain with sensitivity to touch
What is meant by 1st pain and 2nd pain?
1st pain is pain in the acute phase: intense/sharp; 2nd pain is pain in the tonic/chronic phase: occurs as tissues become inflamed, aching/burning/throbbing/sensitivity to touch (allodynia)
Which α2-adrenergic agonist has a greater effect on the nervous system than on the cardiovascular system?
Dexmedetomidine
How does the anti-depressant Amitriptyline block chronic pain?
It alters the “affective reaction” to pain, and it blocks reuptake of monoamines (e.g. 5-HT) thus affecting noxious input
What are the effects of general anesthetics at safe doses?
They block the perception of pain by inhibiting synapses in route to the cortex to create unconsciousness; at these safe doses most agents attenuate, but do not eliminate transmission of impulses at synapses in the CNS – because of this the patient will be unaware, but reflex movements and autonomic responses will still occur (supplementation with opioids is often required to block pain reflex actions)
How do local anesthetics work?
Prevent conduction of pain impulses by blocking Na+ flux into nerves; applied peripherally or spinally
What characterizes Capsaicin?
Extract from hot red peppers, applied topically; Depletes Substance P from pain sensory afferents (attenuates peripheral/central sensitization phenomenon); Takes a few weeks to become maximally effective
What adverse effects are associated with Tramadol?
Dizziness, vertigo, constipation, n/v, pruritus, xerostomia
What characterizes Tramadol (Ultram)?
MOA: weak affinity for opioid receptors, and blocks reuptake of NE/5-HT;
Metabolized to O-desmethyltramadol (M1, has a much greater affinity for the opioid receptor);
Tramadol can act as an opioid antagonist when the patient is taking a full opioid agonist like Morphine, partially reversing the analgesia generated by the Morphine, as well as initiating mild withdrawal actions in chronic Morphine users
What are the actions of Opioids?
They block the release of Glutamate and Substance P from nociceptive presynaptic terminals of primary afferents (which are upregulated during inflammation); They hyperpolarize postsynaptic neurons; They change the affective reaction to pain
What is the mechanism of action of α2-adrenergic agonists (e.g. Clonidine) on neurons?
They mimic the action of endogenous NE released from descending pain inhibitory pathways (blocks Glutamate/Substance P release from primary afferents), require spinal/epidural delivery
What are the effects of Opioids on spinopetal neurons (from PAG)?
They block the inhibitory actions of the GABAergic neurons, thus leading to disinhibition and a subsequent increase in NE/5-HT release (since NE/5-HT are inhibitory to pain input, this will decrease the perception of pain)
What can be said about the treatment of neuropathic pain?
Does not respond well to NSAIDs or Opioids; Responds to anticonvulsants (e.g. Carbamazepine or local anesthetics like Mexiletine) and Gabapentin/Pregabalin (inhibit Ca2+ flux which may potentiate GABA, though not via direct interaction with the GABA receptor) – best response is found with a combination of drugs directed at different parts of the problem
What is the most common congenital malformation of the brain in human fetus?
Anencephaly: F >M, develops at gestation day 28, with folate (!) deficiency being a possible risk factor *** KNOW
What is polymicrogyria?
Excessive folds of “micro-gyri” (small folds) that may be fused together or piled on top of each other, resembling cobblestones or Moroccan leather, resulting in an abnormally thin, laminated, excessively folded cortical ribbon – may be due to intrauterine infections (CMV), intrauterine hypoperfusion, or metabolic disorders (Zellweger syndrome)
What are disorders of cell migration?
Too many gyri: Polymicrogyria
Too few gyri, smooth brain: Lissencephaly (agyria, type I: failure in normal cell migration, type II: over-migration of neurons)
Neuronal heterotopias
What organs are affected in Meckel-Gruber syndrome?
Occipital encephalocele, polydactyly, polycystic kidneys, hepatic fibrosis with bile duct proliferation
What is encephalocele?
Diverticulum of malformed CNS tissue through a defect in the cranium – mostly in occipital region
What are the pathological findings in anencephaly?
Area cerebrovasculosa (flattened remnant of disorganized brain tissue with admixed ependyma, choroid plexus, and meningothelial cells), shallow orbits with eye protrusion, calvarium is hypoplastic or absent, descending fiber tracts are absent, brainstem and cerebellum are often spared
How is anencephaly detected prenatally?
Detected prenatally by screening maternal blood for elevated alpha-fetoprotein (!) *** KNOW
What are neural tube defects caused by failure of neural tube closure?
Anencephaly (cranial end closure defect), Myelomeningocele (caudal end closure defect), Craniorachischisis, Exencephaly
What are neural tube defects caused by herniation of neural tissue/meninges thru a defect in skull or vertebtral column?
Encephalocele (cranial end protruding out), Meningocele (spinal cord protruding out)
What is myelomeningocele?
Neural tissue and meninges herniate through a vertebral defect – mostly in lumosacral region  mostly affect lower extremities (motor, sensory) and bowel/bladder control; may be associated with Chiari II syndrome; the protrusion may be covered by skin or thin membrane, and central spinal canal may open and blend into skin
What are midline patterning defects?
Problems with separation of brain hemispheres; include Holoprosencephaly (incomplete or faulty separation of cerebral hemispheres), Agenesis of the corpus callosum, Septo-optic dysplasia
What are different types of Chiari malformations? (types III and IV not on exam)
Chiari I = cerebellar tonsils extend into the upper cervical canal, not due to increased intracranial pressure – not serious and surgically treatable
Chiari II = Arnold-Chiari = displacement of vermis into the upper cervical canal, always associated with a myelomeningocele, main risk factor is maternal Vitamin A deficiency
What are cerebellar malformations?
Chiari malformations
Too big posterior fossa: Dandy Walker malformations
What characterizes agenesis of corpus callosum?
Incidental finding, may be seen with other malformations; patients have higher incidence of mental retardation and seizures; will show slightly dilated ventricles with an irregular “batwing” contour
What conditions often accompany holoprosencephaly in the affected individual?
Craniofacial anomalies: single eye (cyclopia), proboscis (instead of nose) in severe cases; hypotelorism, cleft lip/palate in moderate cases; single midline incisor in mild cases – note: craniofacial abnormalities are often a clue to the severity of underlying holoprosencephaly
Brain abnormalities: single ventricle (instead of two lateral ventricles), fusion of deep gray structures in midline (basal ganglia, thalami), absent corpus callosum/olfactory bulbs/tracts
Note: least severe abnormalities: absence of one olfactory bulb/tract (cannot smell on one side), single midline incisor
Mutation in what gene has been associated with holoprosencephaly?
Mutations in the Sonic hedgehog gene have been identified in AD cases
What conditions are associated with holoprosencephaly?
Trisomy 13 and maternal diabetes
What is the most common type of Lissencephaly?
Type I: inherited; due to under-migration of neurons - cortex is too thick for gestational age, and only contains 4 layers of cells; in localized form, it is referred to as “pachygyria”
What is cerebral heterotopia?
Misplaced brain tissue; a lesser form of migration abnormality than lissencephaly and polymicrogyria; include cortical dysplasia (heterotopic neurons in layer 1); are associated with focal epilepsy (treatable by surgery)
What is Type II Lissencephaly (cobblestone lissencephaly)?
Due to over-migration of neurons – brains shows highly disordered cerebral cortical structure; may lead to cerebellar and ocular abnormalities & congenital muscular dystrophy; is due to defect in O-mannosylation
What pathologic findings are seen in Chiari II malformation?
Lumbosacral myelomeningocele, displacement of vermis into the upper cervical canal, 2’ Hydrocephalus (due to obstruction from vermis displacement)
4th ventricle, pons, and medulla are elongated, Kinked (“S”-shaped) lower medulla, Fusion (“beaking”) of the inferior tectum/colliculi
Note: any time have a baby with lumbosacral myelomeningocele --> scan for Chiari II
What are spinal cord malformations?
Hydromyelia (dilation of central canal lined with normal ependymal cell)
Syringomyelia (dilation of central canal not lined with ependymal cells – instead, have glial lining)
Note: syringomyelia has association with neoplasms of spinal cord
Duplication of spinal cord: diplomyelia
Partial duplication of spinal cord: diastematomyelia
What drug use is associated with Dandy Walker malformation?
Isotretinoin use during pregnancy
What is Dandy Walker malformation?
4th ventricle dilation causing hypoplasia or absence of cerebellar vermis --> causing enlargement of posterior fossa --> causing elevation of tentorium (between cerebrum and cerebellum) and transverse sinus; also can see hydrocephalus
What is seen grossly in brain with diffuse hypoxic/ischemic encephalopathy?
Diffuse cerebral edema with dusky-colored gray matter lesions, pseudolaminar necrosis of neocortex (infarct of layers III and V), “watershed” infarcts in regions with blood supply overlap
What are the gross and microscopic findings in subacute infarct?
Gross: gelatinous, friable tissue (“cracking artifact”), more distinct boundary between normal/abnormal
Microscopic: macrophage predominance, reactive astrocytes --> gliosis
What are the gross and microscopic findings in acute infarct?
Gross: soft, edematous tissue; corticomedullary junction becomes indistinct
Microscopic: pale tissue, “red and dead” neurons, neutrophils
What are the sources of emboli in cerebral vessels?
Cardiac mural thrombi, plaques within carotid arteries, paradoxical emboli (congenital heart disease), tumor/fat/air emboli – most commonly involves MCA region
What are the most common sites for thrombosis in cerebral vessels?
Carotid bifurcation, origin of the MCA, ends of the basilar artery; major contributing cause for thrombosis is cerebral atherosclerosis (risk factors: HTN, diabetes)
How are infarcts categorized based on time?
Acute: up to 48hrs – Subacute: 2-3 days to several weeks – Remote: >1 month
What are various types of infarcts?
Non-hemorrhagic (pale, bland, anemic): due to arterial thrombosis (usually from atherosclerosis), can be treated with anticoagualants/fibrinolytics, may become hemorrhagic
Hemorrhagic (red): due to embolism or venous thrombosis, anticoagulant is CI
Note: do imaging studies to determine if hemorrhagic or non-hemorrhagic --> determine treatment
Note: ischemic infarct may become hemorrhagic 2’ to anti-coagulant therapy
What is stroke?
Abrupt onset of focal or global neurological symptoms caused by hypoxia/ischemia or hemorrhage lasting more than 24 hours – if symptoms resolve in <24hrs, it is called “transient ischemic attack” (TIA)
Note: hypoxia is low pO2 supply to tissue for any reason (low inhaled O2, CO poisoning, inhibition of oxygen use by tissue); ischemia is due to reduction in perfusion pressure (hypotension) or obstruction of blood vessels
What are pathologic findings in diffuse hypoxic/ischemic encephalopathy?
Selective vulnerability: death of gray matter more than white matter (neurons more sensitive to hypoxia than glial cells) – within gray matter, CA1 region of hippocampus, Purkinje cells of cerebellum, and pyramidal neurons in neocortex (layers III and V) are the most susceptible
What types of ischemic damage can occur in brain?
Focal cerebral ischemia (infarct) due to thrombosis or embolism
Global cerebral ischemia (diffuse hypoxic/ischemic encephalopathy) due to cardiac arrest, shock, severe hypotension --> Mild cases: leads to transient confusion with recovery;
Severe cases: PVS, brain death, role for mechanical ventilation to save the brain (“respirator brain”)
What are different types of aneurysms?
Saccular (berry) aneurysms – “congenital” (not present at birth, but predisposing factors present at birth)
Atherosclerotic (fusiform) aneurysms; mycotic (infectious) aneurysms; traumatic, dissecting aneurysms
What are the most common sites for hypertensive hemorrhage?
Basal ganglia (putamen), thalamus, pons, cerebellum (HTN hemorrhage occurs deep in the brain)
What is the most common cause of intracerebral hemorrhage?
Hypertension – accounts for 50% of clinically significant hemorrhages; thought to cause weakening of arterial vessels or small microaneurysms (Charcot-Bouchard)
What are various hypertensive cerebrovascular diseases?
Hypertensive intracerebral hemorrhage, lacunar infarcts, hypertensive encephalopathy
What is the classic presentation of saccular aneurysm rupture?
“Worst headache I’ve ever had” with collapse – associated with increased intracranial pressure (straining at stool, sexual orgasm) – 25-50% of patients die with first rupture, rebleeding common in survivors, rupture may lead to vasospasm of adjacent vessels leading to ischemic injury/infarcts, reorganized blood in CSF space can cause obstructive hydrocephalus
What are the predisposing factors for saccular (berry) aneurysms?
Most important: cigarettes, HTN, AD polycystic kidney disease, CT-related problems (Ehlers-Danlos, Marfan, neurofibromatosis, fibromuscular dysplasia)
What characterizes saccular (berry) aneurysms?
2% of post-mortem brains, mostly sporadic, due to underlying defect in tunica media
Most common locations: anterior circulation (ACA-Acomm junction), bifurcation of MCA, MCA-internal carotid junction, tip of basilar artery where PCA comes off
What are the gross and microscopic findings in remote infarct?
Gross: cystic cavity
Microscopic: cavity (clear space) surrounded by gliosis
What are the two most common settings causing subarachnoid hemorrhage?
Trauma (#1 answer on exam), rupture of a saccular (berry) aneurysm
How does hemorrhagic acute/subacute/remote infarct differ from non-hemorrhagic?
Hemorrhagic will have extravasated blood cells in an otherwise similar-looking lesion
What is the disease that causes amyloid deposition in cerebral vessels?
Cerebral amyloid angiopathy: Amyloidogenic proteins (usually Aβ40) deposit in the walls of medium- and small-caliber meningeal and cortical vessels – microscopically, vessels will have apple-green birefringence under polarized light with congo-red stain due to amyloid deposition
Note: if see hemorrhage in a place that’s uncommon for HTN hemorrhage, think about amyloid angiopathy
What is suspected in a young patient presenting with recurrent strokes, free of any risk factors for stroke, and a strong FH of stroke?
CADASIL: rare, hereditary form of stroke due to mutations in Notch3 gene; Characterized clinically by recurrent strokes (usually subcortical infarcts, sometimes hemorrhages); diagnosed by finding Notch3 mutations or by biopsying vessels of skin/muscles (looking for concentric thickening of media and adventitia, as well as basophilic, PAS-positive deposits)
What is primary angiitis of CNS?
Inflammation of blood vessels only in CNS (aka granulomatous angiitis of CNS); some cases may be due to VZV reactivation; treatment is steroids/immunosuppressants
Note: granulomatous inflammation of blood vessels only in CNS --> think primary angiitis
How can vasculitis lead to cerebral hemorrhage?
Infectious vasculitis, primary angiitis of CNS, systemic vasculitis (most common cause is polyarteritis nodosa)
How is cavernous angioma different from AV malformations?
Distended, thin-walled vessels without intervening brain tissue - bleed less often than AV malformations, but can cause clinically insignificant bleeds forming old hemorrhages/calcifications
What characterizes AV malformations?
M>F, more in 10-30yo men; can cause seizures, intracerebral hemorrhage, or subarachnoid hemorrhage; is seen as tangled mass of abnormal vessels separated by gliotic brain tissue
What vascular malformations in brain tend to bleed (clinically significant)?
AV malformations (#1 type), cavernous angiomas (capillary telangiectasia and venous angioma are incidental findings that tend not to bleed)
What changes are seen in hypertensive hemorrhage?
Thickening of arterioles (hyaline change), extravasation of blood with compression of surrounding tissue, hypoxic/ischemic change and edema in adjacent tissue;
Old lesions: hemosiderin-laden macrophages and gliosis
What is hypertensive encephalopathy?
Clinical syndrome – if due to acute HTN, patient can come in with diffuse cerebral dysfunction (headaches, confusion, vomiting, coma); if due to chronic HTN, patient can have “multi-infarct dementia” (dementia, gait abnormalities, focal neurological deficits)
Besides intracerebral hemorrhage, what can occur in the brain as a result of HTN?
Single or multiple small cavitary infarcts called “lacunar infarcts”, <15mm in size, occur in same locations as HTN hemorrhage – cavitary lesions contain macrophages and gliosis; microscopically, appear as vessels with widened perivascular spaces (État crible)
What characterizes peripheral neuropathy?
Most common cause of weakness; starts in feet/hands/stocking and glove principal b/c longest nerves are affected first; length-dependent in origin/nature; can affect any/all parts of nerve; knowing which components of nerve are affected helps narrow DD
Is ALS a central or peripheral disease?
Both; involves UMN & LMN; affects corticospinal tract so get brisk reflexes; ONLY EMG
What characterizes ALS?
Combination of UMN signs (brisk reflexes) with LMN signs (atrophy, fasciculations); EMG helps support dx when shows fasciculations (but NO sensory loss); no other dx tests; must r/o others
What are examples of anterior horn cell/motor neuron diseases?
ALS, SMA, polio, radiation, lymphoma
What characterizes anterior horn cell diseases?
Motor neuron diseases; motor only, NO sensory; can start anywhere even speech/swallowing
What tests are used in peripheral neuropathy?
EMG/nerve conduction studies, blood tests, spinal tap, biopsy
What are the causes of peripheral neuropathy?
Hereditary (CMT), metabolic (DM, thyroid, vit B12), infectious (leprosy, HIV), entrapment (CTS), inflammatory (Guillan-Barre, CIDP, vasculitis), toxic, tumors
What are the steps in determining the type of neuromuscular problem you have?
1) central vs peripheral, 2) component/localization of the central or peripheral, 3) timing/history
Working from the top down, what are the possibilities as sources of weakness?
Motor cortex, anterior horn cell, peripheral nerves, NMJ, muscle
What are the possible peripheral components (prox to distal)?
Anterior horn cell (ALS, SMA), peripheral nerve, which could include roots, plexus, nerves, etc (CMT, DM), NMJ (Myasthenia Gravis, Lambert Eaton), muscle (Duchenne’s, polymyositis)
What characterizes Lambert-Eaton?
Also involves ABs; often from small cell lung CA; ABs bind to Ca ch’s in presynaptic portion of nerve; prevents Ca from entering, prevents ACh release, presynaptic dz; weakness but briefly improves with exercise, then worse again; reflexes reduced but briefly improve with exercise
What is the tx for NMJ defects?
Immunosuppression (prednisone, azathioprine, cellcept), AChE inhibitors (mestinon), thymectomy, CA removal, plasma exchange/IVIG
How do you evaluate NMJ defects?
Tensilon test (inject IV, inhibits AChE so ACh lasts longer; must have crash cart and atropine nearby); NCS/EMG with repetitive stimulation to fatigue NMJ; ABs
What are the examination findings in NMJ defects?
FATIGABLE weakness anywhere (bulbar, ocular motility, appendicular), so, when m testing, must hold arms up for awhile first then test; have them look up to set off ptosis; reflexes NL except in LEMS, dysarthria, SOB, must take seriously all the time/may need urgent admission
What are examples of pre and postsynaptic NMJ dz’s?
Pre: botulism, Lambert-Eaton, congenital myasthenia; post: myasthenia gravis, congenital myasthenia
What are common NMJ defects?
More common in adults; fatigable weakness; diplopia, ptosis; more proximal than distal; painless/no sensory change; may be insidious; M. gravis most common.
What is typical of presentation of myasthenia gravis?
Affects young women and old men; see fatigable weakness
How is ALS treated?
LE is 3-5 years, can be extended 3 mos with riluzole, a glutamate antagonist; supportive care
What characterizes myasthenia gravis?
Body directs ABs against ACh rec’s & destroys them → when ACh is released, can’t bind to rec’s → weakness, a postsynaptic disease; weakness worsens with ex, reflexes NL, associated with other AI diseases
What are the two main diseases of the NMJ?
Myasthenia gravis & Lambert-Eaton S.
What characterizes myopathies?
Will see motor deficits only, NOT sensory loss; proximal is worse than distal weakness (opposite of neural problems which are length dependent); see atrophy, no sensory abNLs
How are myopathies treated?
Depends on dx – treat underlying problem or immunosuppression; supportive, counseling.
How are myopathies evaluated?
NCS/EMG with NL NCS and abNL motor unit APs; lab studies (CK), genetic testing, heart/lung testing, muscle bx.
What are the clinical findings in myopathies?
Proximal greater than distal weakness; pattern of weakness may help ID type of myopathy; can see associated skin rash, early respiratory problems, cardiomyopathy, or arrythmias; may first present to pulmonologist or after anesthesia trouble.
What are the types of myopathies?
Inflammatory (polymyositis, dermatomyositis, inclusion body myositis), metabolic (thyroid), toxic (cholesterol lowering agents, prednisone), mitochondrial, infectious, hereditary (Duchenne, Beckers, Limb-Girdle, Myotonic Dystrophy), congenital (central core, nemaline); the most common are inflammatory and hereditary.
What is the risk of using Isotretinoin (Accutane) for cystic acne treatment?
HIGHLY teratogenic, causing craniofacial and cardiovascular defects
What is a premalignant precursor for Squamous Cell Carcinoma?
Actinic keratosis
What does “pearly papule” describe?
Basal Cell Carcinoma, Malignant cells from the basal layer of the epidermis
What are ABCDE’s of Malignant Melanoma?
Asymmetry, Border irregularity, Color variegation, Diameter, Evolving
What is the number one prognostic factor in Malignant Melanoma?
Thickness
Hepatitis C virus infection is associated with what type of skin disorder?
Lichen Planus
What characterizes anaplastic astrocytomas?
aka “malignant” or “high-grade” astrocytoma, may arise de novo or from low-grade astrocytoma
What is histologically seen in pilocytic astrocytomas?
Pilocytic: cells appear “hair-like” and elongated – are biphasic in pattern (dense + loose patterns) – show Rosenthal fibers (one of the astrocytic responses to injury)
What are the signs and symptoms of pilocytic astrocytoma?
Optic nerve/chiasm --> visual defects; Hypothalamus --> diabetes insipidus; Cerebellum --> ataxia; Brainstem --> increased intracranial pressure (all occur in children, first 20 years of life)
What characterizes pilocytic astrocytomas?
WHO grade I, #1 glioma in children, may be seen with neurofibramotisis I
Common sites: cerebellum, optic nerve/chiasm, hypothalamus, brainstem (all below tentorium)
Present as discrete, contrast-enhancing cysts with mural nodule - can be excised (excellent prognosis)
What are the genetic alterations implicated in astrocytomas?
Primary glioblastoma: EGF-receptor amplication, loss of PTEN gene on chromosome 10
Secondary gliobastoma: p53 mutation
Note: Primary: 50yo comes in with ring-enhancing lesion of glioblastoma, no history of low-grade lesions
Note: Secondary: 30yo comes in with seizure, diagnosed as diffuse astrocytoma, excised as much as possible, comes back 4 years later with ring-enhancing lesion diagnosed as glioblastom
What are the prognostic factors for various astrocytomas?
Age (worse grade at higher age), proliferation markers (mitotic activity correlates with worse prognosis), extent of surgical resectability – all are non-curable
What characterizes Glioblastoma?
May arise de novo (primary glioblastoma) or from low-grade astrocytoma (secondary glioblastoma), short clinical history unless developed from low-grader astrocytoma, poorest prognosis (death in 1-2 years), histologic variants include gliosarcoma and giant cell glioblastoma
What are different astrocytic tumors?
Grade I: pilocytic astrocytoma (#1 glioma in children – cystic lesion with mural nodule - curable);
Grade II: diffuse (low-grade) astrocytoma (30-40yo – no contrast enhancement on CT);
Grade III: anaplastic astrocytoma (40+ yo – show enhancement on CT);
Grade IV: Glioblastoma (50+ yo – ring-like zone of contrast enhancement, #1 primary brain tumor)
Note: grade I, in all CNS tumors, means curable by neurosurgery (!)
Note: allastrocytomas are more common in males, higher grade lesions tend to occur in older patients (!)
Note: adult astrocytomas (Grade II, III, IV) occur supratentorially and are non-curable – children astrocytoma (pilocytic astrocytoma) occur below tentorium and is curable;
Grade I: subependymal giant cell astrocytoma (assc. tuberous sclerosis – located in lateral ventricle);
Grade II-III: pleomorphic xanthoastrocytoma (superficial location – cystic lesion with mural nodule);
What characterizes diffuse (low-grade) astrocytoma?
Represent 25% of tumors of cerebral hemispheres, are diffuse, spread progressively, and are difficult to excise due to diffuseness – mean survival 6-8 years, most are supratentorial (e.g. occur in hemispheres, not common in cerebellum), manifest often with new onset of seizures
What is seen histologically with astrocytomas?
Diffuse (low-grade): increase in astrocytic cells --> diffuse expansion of white matter
Anaplastic: same as above + mitotic activity (can stain with proliferation markers to detect)
Glioblastoma: same as above + mitotic activity + vascular (endothelial) proliferation + necrosis
Pleomorphic Xanthoastrocytoma: like glioblastoma but no mitotis, vascular proliferation, or necrosis
What characterizes oligodendrogliomas?
(chicken tumor)
WHO grade II, 5-18% of all intracranial gliomas, peak 40-50yo (3-5 year post-operative survival), presents with longer history of neurological s/s than astrocytomas, hallmark feature is “calcifications on CT”
Histology: “fried-egg” cells with perincular halo, braching blood vessels called “chicken-wire”
Note: Chicken tumor: egg-shell calcifications, fried-egg cells, chicken-wire blood vessels
Note: oligodendrogliomas are sensitive to chemotherapy, astrocytomas are not (!)
What characterizes anaplastic ependymoma?
WHO grade III - will show mitotis, vascular proliferation, necrosis; higher grade does not mean poorer prognosis than low-grade ependymoma (!)
How can one distinguish between clear cell ependymoma (clear cells) and oligodendrogliomas?
Both have clear cytoplasms around central nucles (fried-egg like), but clear cell ependymomas occur in the ventricles (!)
What are various types of rossetes in CNS tumors?
True (ependymal) rosettes: ependymal cells line up to form a true lumen – diagnostic of ependymomas
Perivascular pseudorosettes: ependymal cells line up againt central blood vessel – common in ependymoma
Homer-Wright rosettes: no true or pseudo lumen exists, fibrillary material fill up the center
Note: if see perivascular psuedorosettes --> think ependymomas
What characterizes (low-grade) ependymoma?
WHO grade II, 3-9% of all neuroepithelial tumors, #1 primary tumor of spinal cord – tumor of ependymal cells predominantly intraventricularly (with increased intracranial pressure); can also occur in ependymal cells of spinal cord central canal (more common in adults)
What genetics is involved in oligodendrogliomas?
Loss of chromosome 1p and 19q (except for anaplastic types: 9p loss --> worse prognosis)
Note: depending on which loss, treatment is chosen (1p, 19q --> better prognosis, 9p --> worse prognosis)
What characterizes anaplastic oligodendrogliomas?
Same as above, except will have mitosis, vascular proliferation, and necrosis, and loss of chromosome 9p (worse prognosis)
What characteriezes pleomorphic xanthoastrocytoma?
WHO grade II-III, less than 1% of astrocytomas, superficially located with easy excision – may look like glioblastomas (peak age 50+) but occur in young patients with long history of seizures (not new onset as in diffuse astrocytoma) – appear as cystic lesion with mural nodule – thought to originate from subpial astrocytes
Note: if hear cystic lesion with mural nodule --> think either pilocytic astrocytoma or this (!)
What characterizes subependymal giant cell astrocytoma?
WHO grade I, occur in children (0-20yo), associated with tuberous sclerosis – tumor is found in lateral ventricles (intraventricular tumor), when tumor develops in tuberous sclerosis patients (have epilepsy), the epilepsy gets much worse or intracranial pressure increases (due to obstruction in ventricles)
Note: tuberous sclerosis setting + astrocytic lesion --> think subependymal giant cell astrocytoma
What characterizes the histology of pleomorphic xanthaastrocytomas?
As ugly as glioblastoma but no mitosis, vascular proliferation, or necrosis – unique feature is “chronic inflammatory infiltrate” due to slow-growing of tumor (long history of seizures)
What genetics are associated with ependymomas?
Monosomy 22, associated with neurofibromatosis II (which also has mutation in chromosome 22)
What should be suspected in a children with metastatic tumor of CNS origin?
Choroid plexus carcinoma
Is metastasis significant in CNS tumors?
No – since the CNS effect of these tumors kill the patient before any significant metastasis presents
Exception: choroid plexus carcinomas in children metastasize and disseminate via meninges (!)
What location is associated with choroid plexus tumors?
Intraventricular: adults (4th ventricle), children (lateral ventricles), M>F, can cause obstructive hydrocephalus
What are the various choroid plexus tumors?
Most common are papillomas (5:1) - Choroid plexua papilloma (WHO grade I - curable), Choroid plexus carcinoma (WHO grade III – 80% occur in children – can metastasize)
What is a curable form of ependymoma?
Myxopapillary ependymoma (WHO grade I): occur in “filum terminale”, peak age 35yo, M>F
Subependymoma (WHO grade I): occur “intraventricular”, #1 site is 4th ventricle, incidental finding, M>F
What characterizes Medulloblastoma/sPNET?
(malignant)
Small, round, blue cells tumor of childhood (embryonal tumor) – can have neuronal and/or glial differentiation – uniquely have Homer-Wright pseudorosettes (no true lumen, fibrillar in center)
Medulloblastoma (always in cerebellum): good prognosis with chemotherapy, isochromosome 17q
sPNET (always in hemispheres): bad prognosis, incurable (s = supratentorial)
What characterizes Hemangioblastoma?
Sporadic or associated with VHL disease – #1 site is cerebellum – have cyst with mural nodule (--> Grade I)
Histology: composed of vessels (varying sizes) + neoplastic cells (lipidized/foamy stromal cells)
What is the classification for a Meningioma that shows mitotic figures?
Atypical meningioma (Grade II lesion)
What is the only CNS tumor that is more common in females? ***********
Meningioma: usually attached to dura, mostly benign + resectable (Grade I), show diffuse contrast enhancement – associated with chromosome 22 loss, various histologies possible – associated with NFII
Note: neurofibromatosis II is associated w/ chromosome 22 loss --> may have multiple meningiomas
Note: know meningiomas are F>M, associated with NFII, many histologic patterns
What is the most common tumor of the pineal gland?*********
**Germ cell tumor of pineal – pineocytoma (benign, in adults, composed of round/regular pineal ells + pineocytoma rosettes), pineoblastoma (malignant, in children - small, round, blue cell tumor)
Note: if show small, round, blue cell tumor in cerebellum --> medulloblastoma (curable)
Note: if show small, round, blue cell tumor in hemispheres --> sPNET (incurable)
Note: if show small, round, blue cell tumor in pineal --> pineoblastoma
What CNS tumors are seen that resemble dysgerminoma/seminoma of genitals?
Germ cell tumors: germinoma, teratoma, yolk sac tumor, embryonal carcinoma, choriocarcinoma, mixed germ cell tumors
In what patients Primary CNS Lymphomas are seen?
(most are diffuse B-cell lymphomas)
Immunosuppressed patients (HIV, transplant) – most are EBV-associated – most cases are diagnosed by CSF evaluation, poor prognosis despite therapy
What characterizes Dysembryoplastic Neuroepithelial Tumors (DNT)?
(mixed tumor)
Occur in young patient with medically intractable epilepsy and temporal lobe lesions, commonly associated with cortical dysplasia (also causes seizures); lesions are intracortical and superficial, mimic oligodendroglioma but have neurons – probably not true neoplasma (hamartomatous)
Histology: oligodendroglioma-like cells (perincular halo) + neuronal cells “floating” in the background of glioma
Case: 20yo patient with long history of seizures, unresponsive to all AEDs, scan shows superficial temporal lobe lesion, biopsy shows oligodendroglioma-like cells but also shows neurons
What characterizes Central Neurocytoma?
Occur in adults - composed exclusively of neuronal cells, resembles oligodendroglioma but neuronal markers (synaptophysin) show the exclusively neuronal composition – occurs in lateral ventricle, near foramen of Monro – good prognosis, surgically curable (Grade I)
What characterizes Ganglioglioma?
(mixed tumor)
Occur in young patient with medically intractable epilepsy, presents as cyst with mural nodule, contains glial + “dysplatic” neuronal components, surgically curable (Grade I)
Histology: shows perivascular chronic inflammation, neuronal cells in background of glioma
Note: when hear youn patient with medically intractable epilepsy --> usually low-grade, curable lesion
Note: when hear cyst with mural nodule --> usually low-grade, curable lesion
Case: 20yo patient with long history of medically intractable seizures, lesion is cystic with mural nodule
What is von Hippel Lindau disease?
AD, loss of tumor suppressor on 3p25 --> can lead to hemangioblastoma of brain/retina – at risk for renal cell carcinoma, pheochromocytoma
Case: child with hemangioblastoma --> think VHL and 3p25 loss --> check for RCC, pheochromocytoma
Note: most CNS tumor syndromes are AD and due to loss of tumor suppressor genes (!)
What is the most common cause of excess production of anterior pituitary hormones?
Pituitary adenomas – may get bitemporal hemianopsia, pituitary apoplexy (acute hemorrhage into adenoma) - microadenoma < 1cm, macroadenoma > 1cm – look like adenomas --> need evidence of metastasis to make a diagnosis of carcinoma
What is the malignant counterpart of neurofibroma?
(Schwanommas do not become malignant)
Malignant peripheral nerve sheath tumor – are sarcomas (spindle shaped cells with mitotis and necrosis), tend to be in patients with *** NF type I (why? the more neurofibromas, the more chance of developing malignancy) – cells may show divergent differentiation (e.g. peripheral nerve + skeletal muscle), poor prognosis
Note: Triton tumor = MPNST with rhabdomyoblastic differentiation
What characterizes Neurofibromatosis type I?
(aka von Recklinghausen (17 letters) disease) = benign
AD, loss of tumor suppressor gene 17q11.2 – have multiple neurofibromas (“plexiform” neurofibromas), café-au-lait spots, Lisch nodules (eye lesion), may have glioma of optic nerve/hypothalamus or pheochromocytomas
What characterizes neurofibroma, in general?
= benign
Occur in skin (SC tissue) at distal ends of peripheral nerves: closely associated with and expand the peripheral nerve – composed of mixed cells (Schwann cells, perineural cells, fibroblasts) – may be de novo or in association with NF type I
Note: Schwanomma hangs off the peripheral nerve – Neurofibroma is intimately associated with the peripheral nerve and diffusely expands it (!) ****
What can cause bilateral CN8 Schwanommas?
= benign
Neurofibromatosis type II – AD, loss of tumor suppressor 22q12, develops bilateral CN8 schwanommas, ependymomas, meningiomas (!)
Note: patient with bilateral CN8 schwanommas --> definitely NFII --> ID because at increased risk for other brain tumors (meningiomas, ependymomas)
What is Schwannoma?
= benign
Can occur centrally (cranial cavity) --> on the CN8 (vestibular branch = vestibular schwanomma) or on peripheral nerves – are well-separated from the nerve tissue – show Verocay bodies
Occur in two patterns: Antoni A (dense, compact), Antoni B (loose, myxoid)
Note: Schwanommas do not undergo malignant degeneration (!)
What portion of CNS tumors are due to metastatis?
50% - tend to be multiple, well-demarcated, located at gray-white junctions, usually ring-enhancing
Note: ring-enhancing lesion in brain: glioblastoma (old patients), metastasis (old patient) (!)*******
Note: when see multiple lesions --> think metastasis
Above = CNS tumors
Now = PNS tumors
What are the opioids 4 major sites of action?
Peripheral pain receptors and nerves (during inflammation); Spinal, midbrain, thalamic and cortical nociceptive synapses (pain relays from periphery to cortex); Limbic brain (affective reaction); Endogenous pain inhibitory processes descending from brainstem to spinal cord
What are the Endogenous Opioid Peptides that Opioids act by mimicking?
Enkephalins, beta-endorphin, Dynorphins, Endomorphins
What are two ways Opioids regulate Ca2+ Flow?
(1) direct effect of G protein on the Ca2+ channel (causes inhibition of Ca2+ entry on a nerve terminal membrane, Ca2+ doesn’t move in, & neurotransmitter release will↓);
(2) indirect effect through the underlying biochemical process through cAMP (opioids alter adenylate cyclase & cAMP is ↓, so phosphoproteins aren't formed to extent they normally would be, & these are one of the things that keeps the Ca2+ channel open, if ↓, the Ca2+ channel becomes a little ineffective)
What type of opioid receptor produces analgesia, sedation (drowsy), euphoria & respiratory depression?
Mu receptor (vs Kappa (): analgesia, sedation (sleep), dysphoria & limited respiratory depression)
What is characteristic of the Kappa-receptors?
Selective agonists for kappa & also interact as antagonists at the --> receptor** (Mixed receptor function --> mixed agonist/antagonist)
What substitution and where must occur to provide optimal receptor interaction of opioids and opioid receptors?
Simple alkyl substitution on 17-N and a non- substituted 3-OH (Bulky substitutions at 17 position create partial agonists or antagonists at Mu opioid receptor)
What type of substitutions can improve the bioavailability of opioids?
Alkyl groups on OH at positions 3 & 6 decreases charge and improves oral bioavailability. Also, reduces first pass metabolism (i.e. Codiene (OCH3 & OH in positions 3 & 6 respectively); Oxycodone & Hydrocodone (OCH3 & =O at 3 & 6 respectively); Heroin (OCOCH3 at both positions))
How is the combination product of Pentazocine (agonist) & Naloxone (antagonist) effective?
Helpful in pts who may abuse drugs, admin PO b/c Naloxone wont go across GI tract & Pentazocine does, get analgesic effect of Pentazocine not blocked by the antagonistic effect of Naloxone, but if injected, Pentazocine analgesic/rush effect won’t happen b/c blocked by Naloxone
What are Orally active Opioids, Orally active, but w/ decreased bioavailability, and Poor oral bioavailability opioid drugs?
Orally active: Hydromorphone (Dilaudid)*, Pentazocine, Naltrexone;
Orally active, but w/ decreased bioavailability: Morphine, Meperidine (Demerol);
Poor oral bioavailability: Naloxone (Narcan)
What are different ways opioids can be administered to avoid the first pass effect?
Buccal (fentanyl lollipop (esp. children));
Sublingual (buprenorphine);
Intranasal (butorphanol);
Rectal suppositories;
Dermal patch (fentanyl, esp. cancer)
What is the effect of glucuronidation of 6-OH in morphine (not involved in receptor binding)?
Creates a compound morphine 6-glucuronide (M6G) which is an effective & potent Opioid agonist, M6G accumulates slowly in CNS, & may play a role when morphine is taken chronically (act as an active metabolite)
What is the effect of glucuronidation of 3-OH in morphine?
Morphine & other phenathrenes can’t be substituted at 3 OH position or it wont fit into receptor, M3G, the metabolite formed is a CNS excitant and can cause seizures
What are the commonly used opioid drugs?
Morphine (classic compound remains the gold standard); Heroin (popular drug of abuse in the US ); Nalbuphine (sedation always occurs with analgesia, kappa-receptor drug); Meperidine (used in dentistry & medicine); Fentanyl (highly potent, used in cancer and anesthesia)
What are the moderately effective opioid drugs used to block pain?
Codiene, oxycodone, hydrocodone;
Partial Agonist (Mu receptor): Buprenorphine;
Mixed agonists (agonists at kappa, antagonists at Mu receptor): Pentazocine, Nalbuphine, Butorphanol
What are the opioid antagonists?
Naloxone, Naltrexone
What are the highly effective opioid drugs used to block pain?
Morphine, meperidine, methadone, hydromorphone, oxymorphone heroin, levorphanol, fentanyl & congeners,
What is the Triad of Symptoms seen in an opioid overdose?
Miosis, Respiratory depression, Unconsciousness
How do opioids cause constipation & how do you treat it?
They cause decrease in propulsive movement (muscle is contracted rather than being rhythmical) & sphincters are closed & decreased secretions,; Chronic opioid use requires treatment, Stool softeners Docusate Na+ (Colace), Milk of magnesia, Stimulants Bisacodyl (Dulcolax)
What are Diphenoxylate, Loperamide, & Paregoric used for?
Used to treat diarrhea; Diphenoxylate & loperamide synthetic opioids with low abuse potential & Paregoric – and alcohol extract (tincture) of opium
What is the order of sensitivity of the opioids for inducing biliary colic?
Morphine > Meperidine > Codiene
What is the metabolite of Meperidine and what characterizes it?
Normeperidine: chronic use complicated by accumulation of CNS excitatory and proconvulsant metabolite
How is the prominent itching in the facial area that occurs w/ opioids treated?
Titration using an opioid antagonist (Naloxone or Naltrexone), antihistamines, minimally effective because the itch is opioid-R regulated
What class of opioid drugs causes skeletal muscle rigidity?
Phenylpiperidines in high doses, Basal ganglia involved, causes chest wall rigidity during anesthesia requires reversal (Opioid antagonist or skeletal muscle relaxant)
What characterizes Dextromethorphan?
Compound currently seen in OTC cough preparations, it is the d-isomer of an opioid & does not produce analgesia (l-isomers)
What class of opioid drugs has the lower incidence of histamine release?
Phenylpiperidines
By what two mechanisms does nausea occur with opioid use?
Chemoreceptor trigger zone and vestibular effect
What is the phenomena called when a patent who takes opioids for an extended period of time & the body adapts so that stopping the drug may result in physiological disruption?
Dependence
What is the phenomenon that is classified by craving and drug-seeking behavior, perhaps to continue the rewarding (e.g. euphoria) aspects of the drug?
Psychological Dependence
What drug is used for opioid abuse that reduces action of illicit opioids?
Buprenorphine: partial Mu opioid agonist
What drug can be given to reduces sympathetically-mediated opioid withdrawal symptoms?
Clonidine
What type of receptors have the highest abuse potential?
Mu opioid agonists (due to euphoric effect and development of psychological dependence) vs Partial Mu and Kappa agonists which have lower abuse potential
(Kappa agonist’s dysphoric effect)
What is the difference between intravenous and inhalational general anesthetics?
Intravenous: rapid onset (<1min), short duration (due to redistribution), can be given by continuous infusion to extend anesthesia (recovery is slower and depends on metabolism, excretion, degree of tissue accumulation, and duration of infusion --> context sensitive half-time);
Inhalational: slow onset (>4min), duration dependent upon tissue solubility, used for maintenance of anesthesia
What characterizes Sufentanil (and other fentanyls)?
Induction: <1min; Anesthesia: incomplete amnesia; Eliminate reflex reaction to pain: yes;
Muscle paralysis: no; BP: minimal change (cardiac contractility is mostly maintained!); Ventilation: decrease; Note: anesthesia supplemented due to concern for partial awareness
What characterizes Midazolam?
Same as Thiopental except that there is only a minimal change in BP/ventilation, it is a water-soluble benzodiazepine, and it is most commonly employed for conscious sedation or preoperative anxiety
What characterizes Ketamine?
Induction: <1min; Anesthesia: dissociative (!); Eliminate reflex reaction to pain: yes (!);
Muscle paralysis: no; BP: slight decrease; Ventilation: minimal change;
Note: it blocks the NMDA receptor (MOA), may produce emergence delirium (patients wakes up crying, having delusions/bad dreams), it is a PCP derivative
What characterizes Etomidate?
Same as Thiopental except that there is only a minimal change in BP/ventilation, and there is a decrease in adrenocortical response to stress (patients may need to receive glucocorticoids post-anesthesia in order to maintain their stress response; especially the elderly)
What characterizes Propofol?
Same as Thiopental except that with its use there is a decrease in the opioid requirement, it is an antiemetic (reduces overall feeling of nausea upon recovery), there is rapid recovery with extrahepatic metabolism, and pain on injection (because the formulation is a viscous solution)
What characterizes Thiopental?
Induction: <1min; Anesthesia: yes; Eliminate reflex reaction to pain: no; Muscle paralysis: no;
BP: transient decrease; Ventilation: depressed; Note: given by IV bolus, travels rapidly to brain to cause unconsciousness, used for induction of anesthesia
What defines general anesthesia?
A drug-induced loss of consciousness where patients cannot be aroused, even by painful stimulation (excludes reflexive reactions to pain as these are not considered a purposeful reaction initiated by conscious perception)
What is a common sequence of balanced anesthesia?
Propofol (provides rapid anesthesia) --> Succinylcholine (neuromuscular blocker, for placing ventilation tube) --> N2O and halogenated hydrocarbon (general anesthetic) --> narcotic (block reflexive pain) --> non-depolarizing neuromuscular blocker (maintain skeletal muscle paralysis)
What are the aspects of an ideal anesthetic management?
Hypnosis and amnesia, block reflex reactions to pain, provide skeletal muscle paralysis, maintain vital functions, rapid induction of anesthesia, physical safety
What characterizes N2O?
Induction rate: 3-5min; Anesthesia: incomplete; Eliminate reflex reaction to pain: yes;
Muscle paralysis: no; BP/ventilation: no change; Note: laughing gas, MAC only achieved at hyperbaric conditions, at 20-50% N2O in inspired air analgesia is excellent but memory/response to commands may not be completely suppressed, at >50% dysphoria and nausea may occur with a risk of aspiration and subsequent pneumonitis (!)
What characterizes Halothane?
Rare liver dysfunction
What characterizes Isoflurane?
Commonly used, good cardiovascular safety, increased sympathetic outflow, maintains cardiac output (!)
What characterizes Desflurane?
Bad odor, airway irritation, increased sympathetic outflow, maintains cardiac output (!)
What characterizes Sevoflurane?
Pleasant smell, nephrotoxic by-product (not prominent), combative behavior, disorientation
Which inhalational volatile liquid anesthetics are not used anymore and why?
Methoxyflurane: causes excessive fluoride production which is toxic to the kidney;
Ether: flammable and explosive
What characteristics do most inhalational volatile liquid anesthetics have in common?
Induction: 3-5min (Sevoflurane/Desflurane), 10-30min (Isoflurane, Halothane, Enflurane), >60min (Methoxyflurane); Anesthesia: yes; Eliminate reflex reaction to pain: some;
Muscle paralysis: minimal-some; BP: decrease; Ventilation: decrease (marked, use ventilator)
What characterizes the inhalational anesthetics?
There are two physical forms: gases (--> supplied in gas tanks, currently only N2O) and volatile liquids (--> delivered by vaporizers, several halogenated hydrocarbon anesthetics exist) –
Minimum Alveolar Concentration (MAC): a measure of dose/potency of inhalational anesthetics
What are the MAC values associated with Desflurane and Halothane?
Desflurane: 6.0; Halothane: 0.75 (this means that 0.75% of inspired air will consist of Halothane when 50% of patients are not responding to surgical incision)
How is the minimum alveolar concentration (MAC) measured?
As a percentage of lung gases that are anesthetic gas, when 50% of patients are not responding to surgical incision
What are some special characteristics of the shorter-acting general anesthetics?
Propofol: extrahepatic metabolism; Sevoflurane/Desflurane: low tissue solubility; Short-acting narcotic, Remifentanil: metabolized by RBC/tissue esterases
How does alveolar tension increase with each breathing cycle as new gases are delivered?
At an exponential rate, up to the inspired tension
What characterizes the uptake/distribution of inhalation anesthetics?
Partial pressure gradients control equilibria of gases between various tissue compartments;
Alveolar partial pressure of the anesthetics is the driving for to establish brain partial pressure (i.e. the partial pressure in the brain can never be higher than that in the alveoli);
The rate at which the alveolar tension of an anesthetic gas approaches the inspired tension is influenced by the rate of gas delivery to the lung and tissue solubility of the drug
Which general anesthetics have the most beneficial context sensitive half-times?
Ketamine, Propofol, Etomidate (Midazolam less so; Thiopental/Diazepam are bad)
What characterizes the distribution of an IV bolus of an IV anesthetic over time?
Brain/heart/liver/kidney --> skeletal muscle/skin --> connective tissue/adipose tissue/bone
What characterizes the pharmacokinetics of IV anesthetics when given as an IV infusion?
Done primarily with drugs that are rapidly metabolized and/or excreted; Computer-assisted delivery for longer procedures
What characterizes the pharmacokinetics of IV anesthetics when given as an IV bolus?
Rapid initial distribution to the brain due to proportionally higher CO to tissues like brain, as well as lipid solubility of drug; Short action of single IV bolus since it is rapidly redistributed from brain to skeletal muscle and skin – Bolus is ideal for induction, very short procedures, and as a supplement to other anesthetic procedures
What characterizes Enflurane?
Myoclonus (contraindicated in patients with a history of seizures)
What is the purpose of “conscious sedation” for minor operative procedures?
Minimizes drug-induced alterations of consciousness and vital functions
What is the logic in using a combination of N2O with a halogenated hydrocarbon?
Less of each drug will be needed, thus lowering the toxicity of the halogenated drug while preserving good anesthesia and analgesia
Why does an inhaled drug with a greater solubility take longer to reach inspired tension?
Because there is greater uptake into blood and tissues (large reservoir), thus reducing alveolar tension
In what ways can the speed at which we attain an anesthetic concentration in the brain be increased, when agents of high solubility are being used?
Increase MAC initially, then drop it down; Increase breathing rate
What is the relationship between tissue solubility and anesthesia induction rate?
The lower the tissue solubility, the faster the induction rate (i.e. if the drug is less absorbed into tissues, it will be more readily available in the plasma to cause induction of anesthesia in the brain)
What does the Ostwald Solubility Coefficient express?
The blood/gas partition coefficient (i.e. the concentration of the drug in blood divided by the concentration of the drug in the alveoli) – the higher the coefficient, the more soluble it is
What head finding does indicate fracture of petrous portion of temporal bone?
Mastoid ecchymosis (Battle sign): discoloration over mastoid area behind ear
Where does contusion occur from a moving head that strikes a surface (e.g. falling to the floor)?
At frontal and temporal regions of the brain: specifically at bony prominences of anterior and middle cranial fossae where brain moves against interior bones: called “contrecoup contusion” **** - location of contusion on the brain is stereotypical and does not indicate the location of blow to the head ****
Where does contusion occur from a blow when head is stationary (e.g. beating of restrained head)?
Directly beneath the site of impact: called “coup contusion”**
If on autopsy, brain is shown to have hemorrhage on the superficial cortical layer, how can one justify a determination of contusion vs. hemorrhagic stroke?
Superficial cortical layer receives some blood supply from meningeal vessels, and hence, is not affected with a cerebral vessel stroke; Contusion causes microscopic hemorrhage at the most superficial layer of cortex due to torn vessels
What characterizes contusions?
Wedge-shaped bruises of cortical surface due to hemorrhages from torn vessels – sub-pial layer is affected --> usually superficial, but can extend to subcortical white matter (“contusion hemorrhage”)
What are various types of basilar fractures?
Ring fracture: fall from great height causing spinal column moving up into foramen magnum
Hinge fracture
What characterizes skull fractures?
Linear fracture: simple fracture line that originates from point of impact – from broad-based forces
Basilar fracture: fracture at base of skull (thick) – from significant forces;
Depressed fracture: bone pushed inward into brain – from heavy force striking over a small surface area
Diastatic fracture (only in children): fracture that opens up a suture
Comminuted fracture: bone is fragmented; Compound fracture: overlying scalp is lacerated
What characterizes CNS trauma?
Head injury is #1 cause of death due to trauma, traffic/transport accidents are #1 cause of head injuries
What head finding does indicate fracture of the orbital plate?
Periorbital ecchymosis (“raccoon eyes”): swelling/discoloration of upper/lower eyelids – does not necessarily mean injury to the eye
What are various types of blunt head trauma?
Abrasion: superficial injury with scraping of skin; Contusion: bruise from trauma due to underlying vessel breakage; Laceration: splitting open/tearing of skin
How is severity of diffuse axonal injury graded?
Grade I: microscopic diffuse axonal damage in corpus callosum, white matter of hemispheres, brainstem
Grade II: focal hemorrhage in corpus callosum
Grade III: tissue tear hemorrhage in rostral brainstem
Note: survivors of diffuse axonal injury usually become PVS
What can tearing of vertebral artery lead to?
Traumatic basilar subarachnoid hemorrhage at the base of the brain
What characterizes subarachnoid hemorrhage?
Blood beneath the arachnoid membrane – may be non-traumatic (berry aneurysms), #1 cause is trauma – almost any type of brain injury can result in subarachnoid hemorrhage (e.g. all contusions, torn axons, etc) --> non-specific indicator of trauma – usually not at base of injury
How are subdural hemorrhages dated?
Acute, subacute, and remote – further out it gets, the harder it is to pinpoint the date - after 1 year, can get calcification/ossification of dura from subdural hemorrhage
What characteriezes subdural hemorrhage?
Bleeding between dura and arachnoid – venous in origin (#1: cortical bridging veins), no need for skull fractures – crescent-shaped, mostly in frontoparietal region; prognosis depends on rate of evolution and age
Note: tearing of bridging vein most common in the elderly with very minor degrees of trauma (no fractures needed) – subdural hematoma does not indicate foul play (!)
Case: dilated ventricle due to minor hydrocephalus, older patient, minor trauma --> minor subdural hemorrhage --> probably not clinically significant
Case: subdural hemorrhage --> brain pushed over on one side --> herniation --> blocking of foramen of Monro --> obstructive hydrocephalus --> dilated lateral ventricle on imaging --> clinically significant
What characterizes epidural hemorrhage?
Bleeding between skull and dura – arterial in origin (#1: middle meningeal artery), mostly associated with skull fractures – lens-shaped on imaging; prognosis depends on rate of evolution and volume
What type of injury is seen when hitting hard in the face, causing twisting of head from the force?
Brainstem avulsion: tearing of pons, cerebellum
What contusion occurs when bone is driven into the brain?
Fracture contusion
What type of injury is seen in angular acceleration (e.g. sudden stopping of moving vehicle)?
Diffuse axonal injury: shearing of axons – also occurs with shaken infants; can immediately lead to unconsciousness and death, but brain usually looks unremarkable on gross section
Note: if suspect shaken baby injury --> must look at microscopic sections of brain (gross is unremarkable)
Note: usually unremarkable on gross, but if see a lesion, they are hemorrhagic and involve the deep white matter such as corpus callosum, quadrants of rostral brainstem (as opposed to contusion, which involved superficial gray matter)
What contusion occurs when a blow to the head causes rapid expansion of intracranial contents?
Herination contusion – bullet shot to the head causing rapid expansion due to its high kinetic energy
21. What are complications of CNS trauma – things to avoid from happening?
Brain swelling and increased intracranial pressure --> may cause blockage of cerebral vessels --> may lead to hypoxic/ischemic brain damage
Note: small white matter lesions are associated with diffuse axonal injury
Note: intraventricular hemorrhage can be an extension of hypertensive hemorrhage or due to trauma (e.g. if see intraventricular hemorrhage and subdral hemorrhage --> trauma is the most likely cause)
What are common types of peripheral nerve injuries?
Segmental demyelination (10%), axonal degeneration (90%)*****
What are inflammatory neuropathies?
Guillain-Barre syndrome: “acute demyelinating polyradiculoneuropathy” --> acute demyelinating signs
CIDP: “chronic inflammatory demyelinating polyradiculoneuropathy” --> chronic demyelinating sign = “onion bulb”
What characterizes the regeneration of axons after Wallerian degeneration due to transection?
Multiple axonal sprouts will regenerate within the Schwann cell “tube”
What is the response to axonal transaction in the nerve cell and in the axons?
In nerve cells: central chromatolysis – In axons: Wallerian degeneration (degeneration of nerve fiber distal to site of transection) (!) ***
What characterizes axonal degeneration?
Axon destruction with 2’ demyelination --> if neuronal cell body and Schwann cells survive, axonal regeneration occurs; if motor nerve destroyed --> may get 2’ neurogenic atrophy of skeletal muscle
What does repeated episodes of remyelination and demyelination result in?
Onion bulb: concentric layers of Schwann cell cytoplasm and BM surround the thinly myelinated axon****
Note: onion bulb (know the EM picture) indicates a “chronic demyelinating disease” (!) ***************
What characterizes segmental demyelination?
Damage to Schwann cell or myelin sheath (axons spared), remyelination occurs but with shorter internodes/thinner myelin --> never as good as pre-injury; if repeated --> “onion bulb”
What is the composition of normal peripheral nerves?
Axon, its myelin sheath, and CT surrounding the entire nerve (epineurium), CT surrounding each fascicle and forming blood-nerve barrier (perineurium), and CT surrounding each nerve fiber (endoneurium) – thickness of myelin sheath and distance between nodes of Ranvier are directly proportional to the diameter and speed of conduction of axons
What is the most common nerve biopsied?
Sural nerve (pure sensory)
What is used to visualize peripheral nerves?
Myelinated: toluidine blue, Unmyelinated: electron microscopy (not used much anymore)
What are causes of infectious neuropathy?
Leprosy: due to Mycobacterium leprae, two forms: Tuberculoid (immune response, few M. leprae, granuloma formation), Lepromatous (no immune response, many M. leprae in Schwann cells)
Diphtheria (toxin-mediated demyelination), VZV (inflammation in ganglia), CMV (neuropathy in immunosuppressed)
What can Lead poisoning do to peripheral nerves?
Demyelination
What characterizes malignancy-associated peripheral neuropathy?
(less commonly) due to direct infiltration or compression from tumor
(more commonly) Paraneoplastic syndromes: small cell carcinoma of lung (anti-Hu antibodies againt CNS and PNS), plasma cell dyscrasias (e.g. multiple myeloma) --> MAG antibodies against myelin, light-chain amyloid deposition on peripheral nerves
What can cause amyloid neuropathy?
Plasma cell dyscrasias (e.g. multiple myeloma)
What type of neuropathy is Diabetes Mellitus peripheral neuropathy?
Axonal neuropathy (degenerating/regeneration axons) – may see thickening/hyalinization of endoneurial arterioles with BM reduplication (e.g. thickened blood vessels in biopsy)
What pattern of neuropathy is seen in long-standing Diabetes Mellitus?
“Stock-and-glove” pattern: distal symmetric sensori-motor neuropathy
Autonomic dysfunction (esp. vagus) --> can cause masking of angina in CV disease --> sudden death
What is the most common hereditary neuropathy?
Charcot-Marie-Tooth disease (HMSN I) – AD (17p duplication at Peripheral Myelin Protein 22 gene)****
Present in childhood/early adulthood with progressive atrophy of calf muscle (peroneal);
Is a progressive/chronic demyelinating disease --> “onion bulbs” seen
Note: most hereditary enzyme disorder --> AR since both gene copies of enzyme must be absent for disease
Note: most hereditary structural protein disorders --> AD since one fauly copy can cause disease
Note: know protein and gene for Charcot-Marie-Tooth disease ***********************************
Why is nerve biopsy not helpful in diagnosis of GBS?
Sural nerve is pure sensory – GBS is motor nerve disease (causes motor weakness)
What characterizes CIDP?
= chronic demyelination
Relapsing and remitting immune-mediated neuropathy --> “onion bulb” formation
What is seen histologically in GBS?
Segmental demyelination; inflammation around venules and endoneurium in spinal and cranial motor (anterior) nerve roots
What characterizes Guillain-Barre syndrome?
= acute demyelination - motor illness
Preceding flu-like illness --> distal weakness --> “ascending paralysis” to proximal weakness --> most recover, some die from respiratory failure; thought to be due to immune cross-reaction with myelin --> plasmapheresis may help
What are traumatic neuropathies?
Traumatic neuromas: composed of peripheral nerve + collage – is painful, may form a bump at amputation site (remove tumor instead of reamputation)
Compression neuropathies: carpal tunnel syndrome, “Saturday night palsy”, Morton neuroma (interdigital nerve foot, from wearing high heels)
What characterizes Myasthenia Gravis pathology?
Initially: extraocular muscle weakness (ptosis, diplopia), improve with anticholinesterases, associated with thymoma/thymic hyperplasia, on EM will see simplification of post-synaptic membrane with AchR loss
What are the diseases of NMJ?
Myasthenia Gravis – Ab againt Ach receptors, associated with thymomas/thymic hyperplasia
Lambert-Eaton myasthenic syndrome – Ab againt presynaptic Ca2+ channels, paraneoplastic process associated with small cell carcinoma of lung
What is classically seen in neurogenic atrophy of skeletal muscles?******
Angular, atrophic, esterase-positive myofibers
Fiber type grouping: type I fibers group together in one location, type II fibers group together elsewhere (instead of the alternating fiber distribution seen in normal muscle)
Target fiber (hole in muscle fiber): stained with NADH
Note: fiber type grouping is indicative of chronic denervation (!)
To what form of the Na+ receptor do local anesthetics bind?
Open/active state, and the inactive state (but not the channel at rest); this means that the block forms more quickly in rapidly conducting nerve (i.e. it is use dependent) – the binding site for the local anesthetic is located on the cytosolic surface of the Na+ channel
What is responsible for hypersensitivity reactions with Ester-linked local anesthetics?
They are derivatives of para-aminobenzoic acid (PABA); metabolism creates PABA or a PABA-like compound which can be allergenic in sensitive patients – formulations of either esters or amides may contain methylparaben or sulfite as preservatives which may also be allergenic
What characterizes the clearance of local anesthetics?
Most esters are metabolized by plasma pseudocholinesterase (rapidly hydrolyzed, caution in atypical pseudocholinesterase); Amides and Cocaine (an ester) are metabolized in the liver, most undergo N-dealkylation followed by hydrolysis, the Prilocaine metabolite o-toluidine causes methemoglobinemia; A slow rate of absorption into blood prolongs action (Lidocaine is a potent vasodilator, Epinephrine can be included in solution to retard absorption)
How are Cocaine and Benzocaine different from other local anesthetics?
Cocaine: vasoconstrictor (blocks the reuptake of NE), reduces bleeding when applied to thin mucosal membranes such as nasal mucosa, chronic use leads to ischemic necrosis of tissue, abuse leads to cardiovascular toxicity; Benzocaine: uncharged, possess solubility characteristics that allow it to penetrate the skin, used in ointment for sunburn and skin irritation
What are the classes of local anesthetics?
Divided up by the linkage between their lipophilic and hydrophilic portions: amide or ester linkage; Amide-linked: Lidocaine, Mepivacaine, Bupivacaine, Etidocaine, Prilocaine, Ropivacaine; Ester-linked: Procaine, Chloroprocaine, Tetracaine, Cocaine, Benzocaine
Besides their anesthetics effects, what is the effect of local anesthetics on the heart?
Blockade of Na+ channels, providing an antiarrhythmic effect; at higher doses they can cause a conduction block (toxic effect) – drugs incl. Lidocaine, Tocainide, Mexiletine
By what mechanism of action does the non-ionized local anesthetic, Benzocaine, act?
Dissolves into the membrane causing conformational changes in the membrane: lipid bilayer expands causing pressure that deforms the ion channels causing failure of ion conduction
By what mechanism of action do local anesthetics work?
They inhibit Na+ influx in neural tissue
When local anesthetics are injected into inflamed tissue, what will happen to the potency of the drug?
It will decrease due to an increase in acidity, causing more of the anesthetic to be in the ionized (less lipophilic) form
What characterizes local anesthetics?
They are weak bases with a pKa from 7..7-9.0; at physiological pH they exist largely in ionized form in equilibrium with a smaller non-ionized portion; the non-ionized portion is essential for diffusion to the site of action, whereas the ionized form is essential for channel binding
What are the adverse cardiovascular effects from absorption of local anesthetics?
Systemic vasodilation (leading to severe hypotension), depression of cardiac conduction leading to AV/ventricular conduction blocks (cardiac arrest may result)
Why may anatomical position of fibers in a large nerve trunk create exceptions to the rules regarding differential nerve block?
Since the local anesthetic diffuses from the outside of the large nerve trunk to the inside, the order of nerve block will be skeletal muscle innervation first, followed by a block of proximal sensory innervation, and finally a block of distal sensory innervation
What is the general order of sensitivity regarding nerve fiber type and local anesthetics?
First: C-fiber (dull pain, temp.), pre-/post-ganglionic autonomic fiber, Aδ-fiber (sharp pain, temp);
Second: Aγ-fiber (muscle tone), Aβ-fiber (pressure, touch); Third: Aα-fiber (proprioception, motor activity) – factors promoting high sensitivity to local anesthetic: small fibers, high firing frequency, longer AP duration (i.e. pain fibers), short internodal distance (2-3 nodes need to be blocked)
What should be a concern regarding a spinal conduction block?
Spinal headache due to loss of CSF and inflammation after puncturing the dura; should also be concerned about infection, and avoid the upward spread towards T1-4 (going to the heart) or brain
What is meant by infiltration regarding local anesthetics?
Any injection in which a specific cutaneous nerve is not targeted: subcutaneously (e.g. removal of warts and suturing), intravenously (Bier’s block, done with tourniquet applied, allows full block of an entire extremity)
What local anesthetic is applied to the cornea?
Proparacaine (ester-linked), has low antigenicity in eye since the amine group is in the meta rather than the para position on the benzene ring (i.e. won’t form PABA)
What are the most common topical applications of local anesthetics?
Benzocaine, Pramoxine, EMLA cream (eutectic mixture of lidocaine and prilocaine with adequate solubility for skin/mucosa)
What characterizes the CNS toxicity of local anesthetics?
Low plasma level: talkative, disoriented, drowsy; Higher level: restlessness, tremors, clonic seizures (administer oxygen, benzodiazepines, and perhaps succinylcholine); Higher yet: medullary paralysis leading to respiratory and cardiovascular collapse (supportive therapy)
Which neuromuscular blockers have the fastest speed of onset?
Succinylcholine<1min; Rocuronium<2min
What is the association between the neuromuscular blockers and their effects on cardiac muscarinic receptors?
None: Curare, Atracurium, cis-Atracurium, Vecuronium;
Slight: Rocuronium; Moderate: Pancuronium; Stimulation: Succinylcholine
What is the association between the neuromuscular blockers and their effects on autonomic ganglia?
None: Atracurium, cis-Atracurium, Pancuronium, Rocuronium, Vecuronium;
Weak block: Curare; Stimulation: Succinylcholine
What characterizes the metabolism of steroidal neuromuscular blockers?
Pancuronium, Vecuronium, and Rocuronium are partially metabolized to pharmacologically active compounds (i.e. their 3-OH, 17-OH, and 3,17-diOH metabolites): these accumulate in renal insufficiency, so with prolonged use caution since paralysis may persist
Which neuromuscular blocking drug should be used in patients with liver/kidney disease?
Atracurium – however, its metabolite Laudanosine may cause seizures and is inactivated in the liver (i.e. use low doses of Atracurium in liver disease or with prolonged use); less Laudanosine is formed from cis-Atracurium
What drug is used for the pharmacological reversal of non-depolarizing blockers?
Neostigmine
What characterizes the elimination of neuromuscular blockers?
Succinylcholine: eliminated in 5-10 min by butyrylcholinesterase (pseudocholinesterase); Atracurium: eliminated in 40 min by Hoffmann degradation and plasma pseudocholinesterase;
Others: eliminated by liver/kidney (generally slower and subject to alterations with diseased state)
In what ways do neuromuscular blocking drugs produce skeletal muscle paralysis?
By depolarizing the neuromuscular junction: Succinylcholine; By competitively antagonizing nicotinic receptors in the junction: isoquinoline derivatives (Curare, Atracurium, cis-Atracurium), steroid derivatives (Pancuronium, Vecuronium, Rocuronium)
What structural characteristic do neuromuscular blockers have in common?
They are quarternary ammonium compounds which act with anionic groups on skeletal muscle cholinergic receptors – they do not readily penetrate the BBB
What are the therapeutic uses of neuromuscular blockers?
Promote access to the operative field by removing muscle tone; Facilitates intubation by relaxing the jaw and by preventing reflex movement when tube touches trachea; Reduces the dosage of anesthetics required to eliminate movement (must be used with anesthetics!)
What is used as a premedication to prevent bradycardia in a patient who is given Succinylcholine?
Atropine
How is the degree of neuromuscular blockade monitored (two methods)?
Via a functional analysis based on the occurrence of a progressive dose-related paralysis: eyes/fingers > head/neck > limbs > respiratory musculature (recovery is in reverse order);
Via the use of a nerve stimulator to test the effectiveness of the neuromuscular blockade
What are pharmacogenomic concerns with Succinylcholine?
Plasma pseudocholinesterase deficiencies prolong action, so assisted ventilation may be required; Malignant hyperthermia can be initiated by Succinylcholine (antidote = Dantrolene: inhibits Ca2+ release from sarcoplasmic reticulum, can be used prophylactically in a hereditary condition)
What are the adverse effects of Succinylcholine?
Fasciculations cause muscle aches, particularly of the tongue; Stimulation of muscarinic receptor in the sinoatrial node may lead to arrhythmias with therapeutic doses; Stimulation of autonomic ganglia, particularly with high doses; Seriously injured patients may experience arrhythmias from K+ release from traumatized tissue
What characterizes the pharmacology of Succinylcholine?
Rapid onset, short duration; Two phases of blockade: Phase I --> persistent depolarization of end plate from repetitive agonistic interaction with receptors (initial muscle stimulation, sarcolemmal action potentials rapidly decline since Na+ entry is voltage gated and requires cyclical end plate potential), Phase II --> desensitization of cholinergic receptors when Succinylcholine persists within the neuromuscular junction – neither phase is reversible with Neostigmine
By what mechanism can the pharmacological reversal of non-depolarizing neuromuscular blockers occur?
By increasing ACh levels with Neostigmine (acetylcholinesterase inhibitor); must co-administer with Atropine in order to control the adverse effect of Neostigmine on the respiratory system, heart, GI tract, etc. without effecting skeletal muscle cholinergic receptors – concern for “recurarization” since Neostigmine has a shorter duration than most neuromuscular blockers
What drug interactions are seen with the non-depolarizing neuromuscular blockers?
Aminoglycosides enhance paralysis by reducing ACh release (blockade is not reversible with neostigmine, but may be partially reversed with calcium salts);
Calcium channel blockers potentiate the blockade; Synergistic interaction is seen with halogenated hydrocarbon anesthetics
What is the association between the neuromuscular blockers and their effects on histamine release?
None: cis-Atracurium, Pancuronium, Rocuronium, Vecuronium;
Slight: Atracurium, Succinylcholine; Moderate: Curare
What will occur when Pancuronium interacts with its cardiac muscarinic receptor?
Causes tachycardia and increased BP
What is beta amyloid derived from?
Amyloid precursor protein
What off-label treatment strategies have been used for AD?
Memantine, Statins (may be effective in reducing AD severity), antioxidants (Vit. E/C, Selegiline)
What characterizes other NMDA antagonists?
MK-801, Phencyclidine, Ketamine: they have slow off rates and are associated with coma, hallucinations, and anesthesia
What characterizes Memantine?
NMDA receptor antagonist: uncompetitive, channel must be active for Memantine to bind, the more active the channel the greater the blockade, results in some neuroprotection;
AEs: constipation, dizziness, HA, pain
What are the AChE inhibitors and what are their associated adverse effects?
Donepezil: nausea, diarrhea, HA, insomnia, generalized pain, dizziness;
Rivastigmine: n/v, diarrhea, HA, loss of appetite, abd. pain, accidental injury, tremor, dizziness;
Galantamine: n/v, diarrhea, HA, loss of appetite, UTI, dizziness
What describes the formation and breakdown of ACh?
Acetyl-CoA + Choline --> (via ChAT) ACh --> (via AChE) Choline + Acetate
What differentiates between the non-amyloidogenic and amyloidogenic pathways?
Non-amyloidogenic: α-secretase cleaves the pre-cursor leaving behind a water soluble molecule (α-sAPP) and another particle that is subsequently cleaved by γ-secretase to p3;
Amyloidogenic: β-secretase cleaves the precursor leaving behind β-sAPP and another particle that is subsequently cleaved by γ-secretase to β-amyloid
What are possible causes of neuronal loss in neurodegenerative diseases?
Apoptosis, necrosis, oxidative stress, inflammation
What are risk factors for AD?
High BP, high cholesterol, diabetes, smoking, family history (slight increase in risk
Of the top 5 causes of death, which is the only one that is increasing in incidence?
Alzheimer’s disease (AD)
What describes the use of Lithium in ALS?
When used in combination with Riluzole, a small study showed no progression of symptoms – Lithium promotes autophagy which induced neuroprotection; it may also suppress glial cell activation
What neurons are lost in Huntington’s disease?
GABA/enkephalin containing projection neurons from the striatum (specifically the D2 receptors) --> direct pathway becomes prominent causing excess movement
What describes the indirect pathway of the basal ganglia?
Glutamate is released from the neocortex onto the striatum --> GABA is released from the striatum, causing inhibition of the external pallidum --> less GABA is released from the external pallidum, meaning less inhibition of the subthalamic nucleus --> more glutamate is released by the subthalamic nucleus, causing stimulation of the internal pallidum --> more GABA is released from the internal pallidum, meaning inhibition of the thalamus --> less glutamate is released from the thalamus --> decreased stimulation of the neocortex – DA inhibits the excitatory effects of glutamate on the striatum (via D2), thus somewhat increasing thalamic output
What describes the direct pathway of the basal ganglia?
Glutamate is released from the neocortex onto the striatum --> GABA is released from the striatum, causing inhibition of the internal pallidum --> less GABA is released from the internal pallidum, meaning less inhibition of the thalamus --> more glutamate is released from the thalamus to the neocortex --> enhanced stimulation of the neocortex – DA enhances the excitatory effects of glutamate on the striatum (via D1), thus increasing thalamic output
What is the end-result of activation of the direct and indirect pathways, respectively?
Direct: increases movement (i.e. increases thalamic output);
Indirect: decrease movement (i.e. decreases thalamic output)
What differentiates between D1 and D2 receptors in the striatum?
D1: excitatory, GPCR, activates adenylyl cyclase which increases cAMP to induce APs;
D2: inhibitory, GPCR, inhibits adenylyl cyclase which reduces cAMP thus reducing excitability
What substance may be involved with increased incidence of ALS?
β-N-methylamino-L-alanine (found in Guam flying foxes and cyanobacteria)
What describes the use of anti-psychotics for the treatment of psychiatric symptoms in AD?
Short-term benefit, long-term may increase mortality – should be used to control violence, not for warehousing
What drugs are used in the symptomatic treatment of ALS?
Muscle cramping/twitching: Quinine, Carbamazepine, Phenytoin; Spasticity: Baclofen, Tizanidine, Memantine, Tetrazepam; Salivation: Amitriptyline, Glycopyrrolate, Atropine;
Laughing/crying: Amitriptyline, Fluvoxamine, Lithium, Levodopa
What drug is used in the treatment of ALS?
Riluzole: inhibits glutamate release, blocks postsynaptic glutamate receptors, inhibits voltage-dependent sodium channels – prolongs patient’s life, but does not provide symptomatic improvement
What is the effect on the direct and indirect pathways with Parkinson’s disease (PD)?
Direct pathway: lack of DA leads to a decreased inhibition of the internal pallidum which leads to an increase in thalamic inhibition, thus decreasing thalamic output to the cortex;
Indirect pathway: lack of DA leads to an increased inhibition of the external pallidum which via the subthalamic nucleus leads to an increased activation of the internal pallidum, thus causing an increase in thalamic inhibition, decreasing thalamic output to the cortex
What are the Dopamine agonists used in PD?
Bromocriptine mesylate (associated with heart valve damage), Pergolide mesylate (same as Bromocriptine), Pramipexole, Ropinirole, Rotigotine (transdermal), Amantadine
What is the purpose of adding Carbidopa to a regimen of Levodopa?
As a dopa decarboxylase inhibitor that does not cross the BBB, it reduces conversion of Levodopa to DA in the periphery, thus increasing availability of Levodopa in the brain and reducing peripheral side effects of DA
What are the advantages and disadvantages of Levodopa?
Advantages: most potent drug for PD, effective in early and advanced disease, well tolerated, low risk of cognitive disturbances; Disadvantages: short half-life, multiple dosing, wearing off, dyskinesias
What are the advantages and disadvantages of Dopamine agonists?
Advantages: effective in early and advanced disease, long half-life, low dosing frequency, well tolerated, low risk of wearing off or dyskinesia; Disadvantages: not as potent as Levodopa, risk of cognitive/behavioral problems, cardiovascular side effects for ergot agonists
What are the advantages and disadvantages of MAO-B inhibitors?
Advantages: effective in early and advanced disease, once-daily dosing, well tolerated, possible neuroprotection; Disadvantages: mild benefit, Selegiline metabolizes to amphetamine
What drugs should be used depending on the severity of motor disability with PD?
Mild: MAO-B inhibitor; Mild/moderate: Dopamine agonist; Moderate/Severe: Levodopa
What drugs can be used in the treatment of Huntington’s disease?
Dopamine receptor blockers: Haloperidol, Fluphenazine; Dopamine transporter blocker: Tetrabenazine; SSRIs: Fluoxetine, Paroxetine, Sertraline (used for psychiatric symptoms)
What describes the metabolic pathway for Dopamine?
Tyrosine --> (via tyrosine hydroxylase) DOPA --> (via aromatic L-amino acid decarboxylase) DA --> (via COMT) 3MT --> (via MAO) HVA; And, DA --> (via MAO) DOPAC --> (via COMT) HVA
What does the treatment of Parkinson’s disease focus on?
Neuroprotection: antioxidants, anti-inflammatory, block glutamate toxicity; Dopamine agonists: Ropinirole, Pramipexole; Levodopa
What are the MAO-B inhibitors?
Selegiline, Rasagiline
What are the COMT inhibitors?
Entacapone, Tolcapone (associated with hepatotoxicity)
What is seen in the presentation of tonic-clonic seizures?
An initial sequence of maximal tonic muscle spasms, followed by a period of synchronous clonic jerking of the limbs, followed by a period of prolonged depression (“postictal depression”)
What is the treatment for complex partial seizures?
Carbamazepine or Phenytoin (1st choice), Primidone, Valproic acid – partial seizures may be the most difficult to control
What characterizes partial seizures?
Psychomotor epilepsy, temporal lobe epilepsy; most difficult to treat effectively; Can see simple or focal cortical seizures with various manifestations, but without loss of consciousness; May involve only a single limb; May involve only a specific sensory system
What is the treatment for absence seizures?
Valproic acid, Ethosuximide, Clonazepam (sometimes useful, but tolerance develops to anticonvulsant effects) – seizures are made worse by Phenytoin or Carbamazepine
What is the problem with seizures lasting longer than 5min?
They can result in neurological damage
What characterizes absence seizures?
No warning (“aura”); Will see a brief loss of consciousness, a characteristic EEG pattern (3/sec spike and dome pattern), some clonic muscle movement (blinking of eyelid, jerking of entire body), no depression following seizure; Patient will remember little of seizure
What is the treatment of tonic-clonic seizures?
Phenytoin (first choice), Primidone, Carbamazepine, Valproic acid (effective in some patients)
What characterizes the epilepsies?
A collective designation for a group of CNS disorders; all have in common the spontaneous occurrence of brief involuntary dysfunction; disturbances are all associated with abnormal and excessive EEG discharges
What characterizes tonic-clonic (grand mal) seizures?
Often preceded by a “prodrome” (feeling of tenseness/depression, may precede seizure by several hours), an “aura” will immediately precede seizure, patient will remember little of actual seizure
What are the major classifications of epilepsy?
Generalized: tonic-clonic (grand mal), absence (petit mal); Partial: complex partial, simple partial
What epilepsy drugs potentiate the effects of GABA at the GABA receptor?
Benzodiazepines: limited use due to marked sedation and tolerance development (Diazepam is the drug of choice for status epilepticus); Barbiturates: Phenobarbital (too many adverse effects);
Deoxybarbiturates: Primidone (similar to Phenobarbital, used some in partial complex seizures);
What drug interactions are seen with Phenytoin?
Displaces other drugs from plasma protein binding sites: oral anticoagulants, salicylates, valproic acid, benzodiazepines; Can alter the metabolism of other drugs
What toxicities are associated with Phenytoin?
Dose dependent; When administered IV: cardiac arrhythmias, hypotension, CNS depression;
With acute oral overdose: cerebellar-vestibular disturbances; With chronic administration: gingival hyperplasia, hirsutism (can be minimized by reduction in dose)
What characterizes Phenytoin?
Oral absorption is slow and variable; rapidly distributed in total body water; extensively bound to plasma proteins; half-life is dose dependent; no active metabolites
What epilepsy drugs block the Na+ channel?
Hydantoins: Phenytoin, limits repetitive firing of action potentials by producing a sustained depolarization (slows rate of recovery of voltage-gated Na+ channels, effect is fairly selective at therapeutic doses); Carbamazepine: similar MOA to Phenytoin
What epilepsy drugs increase GABA at the synapse?
Tiagabine: blocks GABA uptake, adjunct therapy for partial complex seizures, adverse effects incl. confusion/dizziness/difficulty concentrating; Vigabatrin: irreversible inhibitor of GABA-transaminase, role in therapy uncertain, not approved in US
What characterizes Primidone?
Well absorbed orally, 60% of administered dose is metabolized (--> PEMA, Phenobarbital), half-life is 5-15hrs; Half-life of PEMA is 16hrs; Has similar MOA to Phenobarbital
What characterizes febrile seizures?
The more they occur in childhood, the more likely the child is to become epileptic later in life;
Start treatment after the second seizure: treat during times of elevated body temperature with phenobarbital
What are the problems with therapy for epilepsy?
Long-term treatment: costs, P450 induction, protein binding, minor toxicities (nausea, sedation)
What characterizes status epilepticus?
A series of recurrent seizures; represents a life-threatening emergency; treated with IV Diazepam, followed by administration of Phenytoin (longer duration of action)
What characterizes the pharmacokinetics of Carbamazepine?
Oral absorption is slow and erratic, widely distributed in total body water, induces P450 system, half-life following acute administration is ~36hrs (with chronic administration: 10-20hrs, in combination with phenobarbital: 10-12hrs); It has an active metabolite (the 10, 11-epoxide)
What characterizes Valproic acid?
Good oral absorption, over 90% plasma protein bound, distributed in extracellular water, half-life is 9-18hrs, no active metabolites
What are epileptic drugs with an unknown or multiple mechanisms of action?
Valproic acid: no effect on GABA systems, small reduction in low-threshold calcium currents, limits sustained repetitive firing and reduced T currents (may explain broader activity)
What toxicities are associated with Ethosuximide?
GI disturbances, fatigue, HA, dizziness – no significant drug interactions
What characterizes Ethosuximide?
Complete oral absorption, distributed in total body water, half-life of 40-50hrs in adults (30hrs in children), no active metabolites
What are epilepsy drugs that block calcium T-currents?
Succinimides: Ethosuximide, reduced low-threshold calcium currents in thalamic neurons, no effect on GABA systems, effective in absence epilepsy
What characterizes Oxcarbazepine?
Converted to active metabolite; very similar to Carbamazepine
What toxicities are associated with Carbamazepine?
Acute overdose: stupor, coma, hyperirritability, convulsions, respiratory depression;
Chronic administration: drowsiness, vertigo, ataxia, diplopia, blurred vision, water retention, hematological disorders (aplastic anemia, agranulocytosis) – can also see P450 induction
What is the clinical use of Fosphenytoin?
Pro-drug of Phenytoin that is rapidly converted to Phenytoin in the body – used parenterally in status epilepticus
What are the uses of Carbamazepine?
Tonic-clonic seizures and partial complex seizures – contraindicated in absence seizures
What are the clinical indications for Valproic acid?
Mainly used in absence seizures, but useful in all seizure disorders as adjunctive therapy; also effective in acute mania and migraine headaches
What are the general principles in the treatment of epilepsy?
Correct diagnosis is the key, may take weeks to establish correct dose, when at all possible use one drug, when seizure control is not adequate check compliance and consider an alternate drug, when altering the concentration of a drug taper it slowly, teratogenic potential in pregnancy is a significant concern (with Phenytoin, Carbamazepine, Valproic acid, Phenobarbital)
What adverse effects are associated with Valproic acid?
GI disturbances, and blah-blah-blah (sedation, ataxia, tremor, hepatic toxicity, acute pancreatitis, hyperammonemia, increased appetite, weight gain, rash, hair loss, thrombocytopenia) – drug interactions are related to high protein binding and competitive inhibition of drug metabolism with Phenobarbital
What monitors are used during general anesthesia?
Hypnosis: EEG analyzers, vapor concentrations; Analgesia: HR, BP; Amnesia: none;
Muscle relaxation: neuromuscular stimulators; Physiology: temp., O2 sat., CO2, ventilation, cardiac output, blood loss, urine output, evoked responses, ECG
What are characteristics of non-depolarizing neuromuscular blockers?
Decrease in twitch height (train-of-four fade), post-tetanic potentiation, absence of fasciculations, augmentation of other non-depolarizing neuromuscular blockers
What drugs are used for the reversal of neuromuscular blockers?
Acetylcholinesterase inhibitors: Neostigmine, Pyridostigmine, Edrophonium (minimize muscarinic stimulation with glycopyrrolate/atropine); Drug binding agents: Sugammadex
(binds Rocuronium, not yet approved)
What side effects are associated with neuromuscular blockers?
All: can mask awareness by preventing muscular response to stimulation; Succinylcholine: prolonged effect if pseudocholinesterase deficient, myalgia, hyperkalemia, bradycardia, increased IOP; Cisatracurium: histamine release, slow onset, ma require reversal; Rocuronium: slow onset, renal excretion, requires reversal, not associated with malignant hyperthermia
Which neuromuscular blocker is used for quick intubation?
Succinylcholine (depolarizing)
What drugs are considered general anesthesia adjuvants?
Lidocaine (anesthetized veins), Glycopyrrolate (decrease secretions), Phenylephrine/Ephedrine (sympathetic stimulation), Metoprolol/Labetalol (control cardiac function)
What preoperative medications are used prior to general anesthesia?
Anxiety relief: Diazepam; Sedation: Pentobarbital; Amnesia: Midazolam, Scopolamine; Analgesia: Morphine, Fentanyl; Anti-sialogogue effect: Glycopyrrolate;
Prevent ANS reflexes: Atropine, Esmolol
What basic considerations is every anesthetic plan based on?
Planned surgery, medical condition of patient, anesthetic history of patient, current drugs/allergies, patient requests/approvals – other consideration incl. patient’s physiologic status, post-op pain management, and skills of the anesthesiologist
What are the steps in general anesthesia?
Induction: Propofol, Thiopental, Etomidate, Ketamine; Maintenance: N2O, Desflurane, Sevoflurane; Emergence: reversal agents, oxygenation, pain relief; Recovery
What are the major components of general anesthesia?
Hypnosis: Propofol, Thiopental, inhalational anesthetics; Analgesia: opioids;
Amnesia: benzodiazepines; Muscle relaxation: Rocuronium, Succinylcholine –
halogenated inhalational anesthetics have all four properties
What anesthetics can be used in a patient with major depression and HTN for electroconvulsive therapy?
Requires general anesthesia for seizure induction (so that the patient doesn’t hurt himself): Labetalol (or Esmolol for HTN prophylaxis), Thiopental, Remifentanil, Succinylcholine, Ketorolac (NSIAD for symptoms after the procedure)
What anesthetics can be used with a 30yo for appendectomy?
Premedication: Fentanyl, Midazolam; Induction: Lidocaine (decreases consciousness), Propofol (induction agent), Rocuronium (paralyze vocal cords); Maintenance: Desflurane, Fentanyl; Adjuvants: Ondansetron (prophylaxis); Emergence: Neostigmine, Glycopyrrolate
What anesthetics can be used with a 5yo for tonsillectomy?
Midazolam syrup, Sevoflurane induction, Fentanyl, Ondansetron/Dexamethasone
What anesthetics can be used in a patient for hysterectomy with a history of post-operative nausea/vomiting (PONV)?
General anesthetic (long-lasting), muscle relaxant for abdominal surgery, infection and PONV prophylaxis: Cefazolin (abx), Ondansetron/Droperidol/Dexamethasone (n/v), Midazolam, Lidocaine, Propofol (induction and anti-nausea), Rocuronium, Fentanyl, Desflurane, Neostigmine, Glycopyrrolate
What EEG pattern is seen with juvenile myoclonic epilepsy?
4-6Hz spike and wave pattern
What type of seizures can be seen in chronic alcoholics who suddenly stop drinking?
ETOH withdrawal seizures – caused by the downregulation of GABA-mediated inhibition and the upregulation of NMDA receptors
What is the drug of choice for temporal lobe seizures?
1st choice: Carbamazepine, Oxcarbazepine, Phenytoin;
2nd choice: Lamotrigine, Valproate, Topiramate
What type of seizures should be suspected in a patient who experiences strange smells and feelings prior to the onset of the seizure?
Temporal lobe seizures
Which seizures medications cause induction of CYP3A4?
Carbamazepine, Phenytoin, Phenobarbital – they will decrease contraceptive effectiveness
What describes the drug interaction between Lamotrigine and oral contraceptives?
Lamotrigine is cleared by glucuronidation, and estrogens can induce glucuronosyltransferase causing a reduction in the plasma levels of Lamotrigine
What is the drug of choice for juvenile myoclonic epilepsy?
Valproic acid (can control all three types of seizures); watch for side effects: liver toxicity, weight gain; contraindicated in pregnancy – Lamotrigine may be effective to treat all seizure types as well (not FDA approved yet) – treatment is lifelong
What are 3Hz spike and wave patterns on EEG indicative of?
Absence seizures
Which drug can make absence seizures worse?
Carbamazepine (or phenytoin)
What is the drug of choice for absence seizures?
Ethosuximide (does not control tonic-clonic seizures); can also use Valproic acid (can cause hepatotoxicity and pancreatitis), Lamotrigine (can cause rash) – Valproic acid and Lamotrigine can treat absence and tonic-clonic seizures
What should alcohol withdrawal seizures be treated with?
Phenytoin and benzodiazepines – no need for long-term treatment
What seizure state can be seen in patients undergoing benzodiazepine withdrawal?
Nonconvulsive status epilepticus – treat accordingly
With what AEDs is gingival hyperplasia seen?
Phenytoin
Which antiepileptic drugs (AED) are more commonly associated with Stevens-Johnson syndrome?
Lamotrigine patients receiving valproate and/or aggressively titrated
What are the two main types of seizures?
Partial seizures and generalized seizures; seizures can progress simple partial → complex partial → generalized
What are the causes of seizures?
Idiopathic (68.7%), stroke, developmental, head trauma, brain tumor, infection, degenerative; most causes unknown; see a lot of mesotemporal sclerosis; a high fever can cause a seizure, basically ANYTHING that affects the brain can cause a seizure.
What are more classifications of seizures?
Partial: simple partial, complex partial, secondarily generalized; generalized: absence, myoclonic, tonic-clonic, tonic (stiffening), clonic (pattern of contraction/relaxation), atonic (lose all m tone)
What characterizes generalized seizures?
Initial involvement of BOTH hemispheres; almost always produces alteration/LOC and/or BL motor activity. Ex: fall to floor, stiffen, jerk
What characterizes complex partial seizures?
A partial seizure that involves some impairment of consciousness. Ex: impairment of consciousness with lip smacking, fiddling with hands
What characterizes simple partial seizures?
A partial seizure where there is NO impairment of consciousness. Ex: arm jerking, flashing lights, funny smells; may get auras.
What characterizes partial seizures?
It starts clinically/electrographically on ONE SIDE of the brain; usually the temporal lobe but can be anywhere; partial b/c starts in “part” of brain. There are two types of partial seizures, simple partial and complex partial.
What is the definition of a seizure?
Episode of sudden, relatively brief disturbance of mental, motor, sensory, or autonomic activity caused by anNL paroxysmal, electrical, or cerebral activity (gray matter)
What percent of people have a seizure in their lifetime?
1%; (epilepsy: 2 million Americans, up to 3% cumulative incidence by age 75)
What is epilepsy?
State of chronic, recurrent (2 or more) unprovoked seizures. Provoked seizures (such as via alcohol withdrawal, hyponatremia, etc) are NOT the cause of epilepsy b/c these can be easily prevented if the cause is reversed.
What characterizes West Syndrome?
Affects infants; triad of hypsarrythmia (chaotic EEG), infantile spasms, MR; poor px
What is the WV law regarding driving after a seizure?
Anyone who has a seizure with an impairment of consciousness cannot operate a motorized vehicle for ONE YEAR. But NOT required to report them by law. (Varies state to state.)
What is the general tx for seizures?
Time is the best test; after 1st unprovoked sz, chance of having 2nd unprovoked sz in 5 years is 35-50%, therefore, tx depends on pt, EEG/MRI findings, family hx, physical exam. But after 2nd unprovoked sz, the chance of a 3rd is 75% so everyone must be treated. Can depend some on patient after 1st, must discuss it.
What characterizes Juvenile Myoclonic Epilepsy?
Teenagers and young adults; see 4-6 Hz generalized spike and wave; pt’s often have morning jerks, generalized seizure; responds very well to meds, but need to always take meds or sz’s return
What characterizes absence?
School age children; see “staring spells”, 3Hz spike and wave; responds to ethosuximide
What characterizes centrotemporal epilepsy?
Affects kids 5-10 y/o; see centrotemporal spikes, drooling face, slurring words; often outgrow this
What characterizes Lennox-Gestaut?
Affects infants- young children; see slow spike and wave on EEG, multiple sz types, MR; poor px but not as bad as West S.
What is the most important aspect of evaluating seizures?
History. When, how often, how long, circumstances, aura, bowel/bladder, tongue biting, postictal state, sleep deprivation, video games, head trauma, meningitis/encephalitis, family hx, prenatal or postnatal problems, hx of sz’s/febrile sz’s as child; often confused, tired, stupified afterwards.
What is epileptic syndrome?
Term that describes certain seizure disorders, defined by a cluster of symptoms, signs, and findings on EEG and imaging studies. Depends on age, cause and type of sz’s, prognosis. Two particularly bad ones are West S and Lennox-Gestaut; others include centrotemporal epilepsy, absence, and juvenile myoclonic epilepsy
What kind of tests are used to evaluate seizures?
EEG: looking for spikes, sharp waves, spike and wave patterns; MRI especially checking the temporal lobe; also can look at spinal fluid, blood tests, genetic tests; MRI is like a picture, EEG tells you how it is “running”; if see squiggly’s all over at same time – it’s a generalized sz
What are the principles of meds for seizures?
There are many different seizure meds with different SEs and different efficacies against the different sz types. There is no single best drug.
What is the tx for status epilepticus?
ABCs, ativan .1mg/kg, dilantin 20mg/kg, head scan, AED levels.
What are the causes of status epilepticus?
Kids: fevers, CNS infections, electrolyte disturbances, hypoxia, previous epilepsy, low AEDs; adults: fevers, CNS infections, tumors, trauma, previous epilepsy, low AEDs.
What is status epilepticus?
Recurrence of sz episodes at intervals too short to allow recovery to pre-seizure condition, or any seizure lasting longer than 5-10 minutes. (CAN be in this state for months!)
What other treatments are used to treat seizures?
Ketogenic diet, vagal nerve stimulator, surgery for “lesional epilepsy” (invasive)
What are some of the common meds and related SEs used to tx seizures?
Dilantin (rash, purple glove syndrome, nystagmus), tegretol (hyponatremia), depakote (wt gain), lamictal (rash), keppra (cognitive), topamax (wt loss). The first 3 are the old standards and are cheaper; last 3 are newer. Difference is in SEs.
What are the rules for efficacy?
1st drug: 60%, 2nd drug: 75%, 3rd drug 80%, and so on. No drug will stop ALL seizures.
What nerve mediates migraine pain?
Trigeminal nerve which inn’s bld vessels and dura
What stx appears to be involved in cluster h/a?
Hypothalamus; seems to be triggered by REM sleep. There is ↑ parasympathetic outflow. Signal is sent from hypothalamus down to trigeminal nerve then to superior salivatory nucleus which regulates pain and sympathetic outflow.
What characterizes cluster h/a?
Very severe; can lead to suicide; occurs in groups for weeks-mos then may disappear for mos/years; often around the eye; occurs at night and can wake person, M > F; associated with eye redness, tearing, rhinorrhea, Horner’s S. alcohol and cigg’s can be a trigger.
What is a tension h/a?
Can occur daily; can last hours or all day; BL; squeezing or pressure-like; no aura and no significant neuro sx’s; can be triggered by stress; take Tylenol/aspirin and it goes away.
What is a complicated migraine?
Symptoms last longer than 24 hours, h/a may be secondary. Looks like a stroke.
What is a migraine equivalent?
When have aura but not followed by a h/a; could have scotomas, confusion
What is an aura?
Occurs in 25% of migraines; area of brain becomes hyper then hypoexcitable; “spreading corticodepression” that does NOT occur in vascular distribution, rather, thought to be a neuronal thing. Pain is the response of blood vessels TO that neuronal event.
What important things need to be asked in history?
Date of onset, duration, timing of attacks, frequency, severity, triggers, quality, factors that ↑ or ↓ h/a, meds including OTC, previous/current, trauma, drug abuse.
What characterizes a migraine h/a?
Usually <3x/wk, begins UL, may alternate sides, throbbing/pulsating, +/- photophobia, N/V. Can be triggered by stress, lack of sleep or food, menses, a food trigger. May have + family hx.
What are the most common type of h/a?
Tension h/a, but most commonly seen in clinic is migraine b/c people seek help for migraines
What are CIs for performing a LP?
Anticoagulants, platelets <50,000, signs of ↑ ICP; don’t want to cause herniation!
Besides meningitis/encephalitis, what other condition commonly has neck pain?
Carotid or vertebral artery dissection
What characterizes carotid or vertebral artery dissection?
May happen spontaneously or following trauma; h/a with some neck pain, neuro deficits that may wax/wane, may see BS signs with vertebral A dissection. Need neuroimaging.
What characterizes meningitis/encephalitis?
H/a common, associated fever, neuro deficits esp MS change, loss of smell (HSV), may/may not have stiff neck, sz’s may occur, neuroimaging if focal deficit, then LP. CT can look NL – if suspicious STILL do LP!
What condition predisposes to aneurysms?
CN palsies: MUST check CNs and LOOK for aneurysms. 20% of pt’s have multiple aneurysms.
What is xanthochromia?
Helps distinguish true bleeds from trauma from tap; if see xanthochromia (yellowish discoloration), you know bleed has been there a/l 12 hours (RBCs have had chance to break down bilirubin)
What characterizes an aneurysm/hemorrhage?
Pt describes as “worse h/a of my life”; acute, explosive onset, meningeal irritation/stiff neck if SAH, may/may not have neuro deficit/HTN; need stat CT scan without contrast and if no bleed seen, then do LP; if LP +, then need angiogram
What are some causes of secondary headaches?
Aneurysm/hemorrhage, meningitis/encephalitis, carotid or vertebral A dissection, cerebral venous thrombosis (DVT of head)
What is CT/MRI better at dectecting?
CT: blood and bone, so better if suspect acute bleed, fx, or sinus problems. MRI: more sensitive for little lesions in parenchyma like posterior fossa lesions, ischemia, subdural and epidural hematomas, meningeal disease. So MRI is good if suspect stroke. NOT every h/a pt needs neuroimaging.
What additional testing may be needed?
In primary h/a, usually none. But in secondary h/a: neuroimaging (CT or MRI), blood work (SED rate if suspect temporal arteritis), LP (esp if personality changes present), angiography (stenosis, aneurysms)
What characterizes Giant Cell Arteritis?
55 and older, pain in temple or around eye, pain brushing hair, tender scalp, jaw claudication, episodes of vision loss. Anorexia, fever, joint pains. ↑ SED rate (esp if over 40). Must start steroids NOW to avoid blindness THEN get temporal artery bx.
What characterizes cough/exertional h/a?
Might also reflect aneurysm leak, Arnold-Chiari malformation type 1. Usually benign but must be SURE they are benign.
What drugs can lead to medication overuse h/a?
Narcotics/opioids, NSAIDS, triptans, acetaminophen, butalbital, caffeine; duration of use also important. May need steroids or medrol dose packs
What is the mechanism for h/a to be worse in am?
After laying down at night and CO2 levels ↑, get dilation of blood vessels making h/a worse.
What characterizes AVM, tumor, or intracranial mass?
H/a with cough/exertion, esp if posterior fossa lesion, may be worse in am, neuro s/s reflect location of mass, may have N/V; if fever, suspect abscess, if antecedent head trauma, suspect hematoma. Neuroimaging: CT with and without contrast or MRI. Note that NOT everyone with a brain tumor has a h/a.
What drugs are associated with pseudotumor cerebri?
Tetracyclines, Vit A stuff, nitrofurantoin, sulfonamides, bactrim. Also hypercapnia.
What characterizes pseudotumor cerebri?
Diffuse h/a, blurred vision or seconds of obscured vision; most seen in obese F, NL neuro exam except DO have papilledema (disc margins look funny), +/- CN6 nerve palsy and visual field deficits. CT and MRI NL. LP also NL but ↑ opening pressure. (anything over 20). Rarely seen in men, but if do, usually have sleep apnea/hypercapnea.
What characterizes CVT?
H/a as first symptom, neuro deficit depending on location of thrombosis, sz’s may occur, predisposing conditions: pregnancy, postpartum, dehydration, CA, coagulopathy, trauma (hypercoagulable conditions). Neuroimaging including CT with contrast, MRV and arteriogram.
What are some causes of recurrent secondary h/a?
Giant cell arteritis (temporal arteritis), pseudotumor cerebri (benign intracranial HTN), AVM, tumor, intracranial mass, medication overuse, cough/exertional, CNS vasculitis.
What happens if a CVT affects the brain around the superior sagittal sinus?
BL LE weakness. (the part of the brain taking care of the legs is over medial aspect)
What characterizes CNS vasculitis?
Neuro deficits represent different sites of ischemia; MRI abNL.
What is the mechanism of the Valsalva?
Cough → ↑ pressure around SC/CSF → transmitted up → causes descent of cerebellum thru f. magnum → h/a
What Serotonin receptor family is not a GPCR, but rather a 5-HT-gated ion channel?
The 5-HT3 family
What Serotonin drug class is used in the treatment of GI disorders (IBS, GERD)?
5-HT4 agonists: increase gut contractile activity, increase colonic transit/emptying;
5-HT3 antagonists: decrease visceral perception, slow colonic transit
What Serotonin drug class is used in the treatment of emesis?
5-HT3 antagonists: Ondansetron, Granisetron, Dolasetron (and other –setrons); MOA: block the effects of Serotonin released from enterochromaffin cells by cytotoxic agents (e.g. those used in the treatment of cancer); AE: HA, enhanced effects of alcohol, cardiovascular effects (esp. with Dola-, Palono-, and Tropisetron); DI: all are metabolized by P450 (all drugs, except Granisetron, are metabolized by a CYP isoform that exhibits polymorphisms, significantly affecting activity)
What characterizes Serotonin syndrome?
Can be caused by many different drugs; produces mental status changes, autonomic hyperactivity, and neuromuscular abnormalities – difficult to diagnose; in severe cases, can see delirium, neuromuscular rigidity, hyperthermia
What agents are associated with the pharmacological modulation of Serotonin?
Tryptophan/tryptophan hydroxylase, MAOIs, SRIs (selective/non-selective), releasing agents (amphetamine), depleting agents (reserpine), receptor agonists/antagonists
What diseased states are associated with Serotonin?
Carcinoid tumors, anxiety/depression/psychosis, GI disorders, emesis, migraine, obesity
What are the physiological roles of endogenous Serotonin?
CNS neurotransmitter, stimulant of pain/itch, activates chemoreceptor reflex, smooth muscle constriction (direct action), smooth muscle dilation (via NO), modulation of GI function (!), modulation of platelet function, role in cardiovascular function, probable role in bone metabolism, possible role in glucose regulation
What describes the synthesis of Serotonin?
L-tryptophan --> (via tryptophan hydroxylase --> rate-limiting step) L-5-hydroxy-tryptophan -->
(via dopa decarboxylase) 5-hydroxy-tryptamine (5-HT, Serotonin)
What characterizes the metabolism of Serotonin?
5-HT --> (via MAO) 5-hydroxyindole acetaldehyde --> (via aldehyde dehydrogenase) 5-HIAA and (via aldehyde reductase) 5-hydroxy-tryptophol
What are examples of derivatives of Serotonin?
Melatonin, Dimethyltryptamine, LSD
What characterizes Cilansetron?
Being tested for use in diarrhea-predominant IBS
What are the most important Serotonin receptors, drugs, and their associated functions?
5-HT1D agonists: Sumatriptan (and others), used in the treatment of migraines;
5-HT3 antagonists: Ondansetron (and others), used for chemotherapy induced emesis;
5-HT4 agonists: Cisapride, used for GI disorders
Which Serotonin receptor subtype is involved in the modulation of satiety, and what drugs affect this receptor?
5-HT2C: Sibutramine (controlled substance, inhibits reuptake of 5-HT, NE, DA),
Fenfluramine/Dexfenfluramine (stimulates release/inhibits uptake, associated with heart valve hypertrophy via 5-HT2A/2B and pulmonary side effects)
What drugs are used in the prophylaxis of migraine?
Beta blockers (first choice) and other hypertensives (CCBs, ATII blockers);
Tricyclic antidepressants (amitriptyline, nortriptyline – not safe from cardiovascular standpoint); antiepileptic drugs (valproate, topiramate, gabapentin – reduce migraine frequency)
What Serotonin drug classes are used in the treatment of migraine?
5-HT1D/1B agonists: Sumatriptan, Naratriptan, Rizatriptan (and other –triptans); used for acute migraine and cluster headaches (not useful for prophylaxis); CI: cardiovascular disease;
PK: metabolized by MAO – Serotonin syndrome can develop with concurrent use of SSRIs/SNRIs
What are the major regions of drug action associated with various Serotonin modulators?
5-HT3 antagonists: act at the vagus nerve to inhibit emesis; SSRIs: stimulate intestinal motility and cause nausea; Alosetron: slows colonic propulsion and might reduce intestinal sensation;
Sumatriptan/Buspirone: relax gastric fundus; Metoclopramide/Tegaserod: increase GI propulsion
What characterizes Sumatriptan?
5-HT1B agonist: useful in dyspepsia; decreases GI tone with NO release from neuron in the gut
What characterizes Tegaserod?
5-HT4 partial agonist: used for constipation predominant IBS; withdrawn due to increased risk of cardiovascular events (still available through age/gender restricted IND protocol)
What characterizes Alosetron and Cisapride?
5-HT3 antagonists (Cisapride also has 5-HT4 agonist activity): restricted usage due to ischemic colitis (Alosetron) and cardiovascular events (Cisapride)
What are common myths about opioids?
Respiratory depression is a common side effect;
Opioid addiction is a common problem (not addiction, but dependence);
Opioid tolerance develops rapidly;
Adequate dosing causes unmanageable constipation;
A majority of patients require prophylactic anti-emetics (not much of an issue when given PO);
Short-acting opioids are preferred for moderate/severe pain;
Unacceptable sedation and confusion occur frequently;
Controlled release opioids should only be used for cancer pain;
Dosage titration is difficult to achieve with controlled release opioids;
Severe cancer pain requires parenteral opioids;
Plasma opioid concentrations correlate directly with analgesia
What is the benefit of pain control in a patient suffering from extreme pain, who also has a high risk of suffering an MI?
Extreme pain causes stress which produces an elevation in NE; NE increases cardiac function which can lead to MI
What is the relationship between hydrocodone and hydromorphone?
Hydromorphone is a metabolite of hydrocodone
What is given to treat neuropathic pain?
Tricyclic antidepressants: Amitriptyline – can also give an SNRI, or Pregabalin/Gabapentin (potency = 6:1)
What can occur in a patient taking chronic oxycodone, when nalbuphine is administered?
Iatrogenic withdrawal (since nalbuphine is a mixed agonist/antagonist) – treat by giving morphine
What are the acquired NMJ disorders?
Botulism: cleaves ACh release-needed proteins; Lambert-Eaton syndrome: antibodies against presynaptic calcium channels; Myasthenia Gravis: antibodies against postsynaptic ACh receptor or MuSK (receptor tyrosine kinase); Carbamates/organophosphates: postsynaptic AChE inhibition
What is the difference between Edrophonium and Pyridostigmine?
Edrophonium has a very short duration of action whereas Pyridostigmine lasts longer
What will be seen with repetitive nerve stimulation in a patient with MG?
A decrement in neuromuscular transmission (with 2Hz nerve stimulation)
What tests are available for MG?
Tensilon test: give patient Edrophonium (AChE inhibitor) and watch weakness disappear; can also do repetitive nerve stimulation, look for abnormal antibodies to AChR/MuSK, or do a chest CT (some patients will have a tumor of the thymus gland) – MuSK antibodies are more so associated with bulbar weakness: tongue, swallowing, facial muscles, muscles of the eyes (this subclass is highly sensitive to immunosuppressive therapy)
What is the hallmark of Myasthenia Gravis (MG)?
History of fatigable weakness: mainly affects cranial nerves (ptosis, tongue weakness, problems swallowing) and proximal musculature – respiratory function should be monitored since disease can affect respiratory musculature
What are the treatment options for MG?
Anticholinesterase drugs (Mestinon = Pyridostigmine); Airway/respiratory support (essential); Thymectomy; Immunosuppressive therapy (Prednisone, Azathioprine); Plasmapheresis and IVIG
What characterizes the use of plasmapheresis and IVIG with MG?
Used with non-compliant patients in the ICU in order to rapidly remove the offending antibodies – treatment is very expensive and doesn’t last very long
What are the major side effects of Azathioprine?
Bone marrow and liver involvement – must monitor closely
What is the major issue with Prednisone treatment for MG?
It has a terrible side effect profile – but it is very effective
What are the rules for doing a thymectomy in a patient with MG?
Teens/twenties: must clear out entire anterior mediastinum to remove all fragments of the thymus;
30+: still perform thymectomy, but need double-blind study to determine whether appropriate;
50+: no thymectomy needed – only major side effect of thymectomy is the surgery itself
What side effect is associated with Pyridostigmine?
Diarrhea, treated with Atropine
In what type of autopsy must the family request an autopsy?
Hospital Autopsy
What is involved in the external examination?
Identification, Identifying characteristics (body type, clothing, age, weight), Head to Toe Examination (skin, hair color, chest, abdomen, back, extremities), Toxicology
Under what circumstances do cases full under the medical examiner authority?
Criminal Violence, Suicide, Accident, Sudden death when in good health, Death unattended by practicing physician, Death under suspicious or unusual circumstances, Abortion, Poisoning, Disease constituting a threat to public health, Disease, injury or toxic agent resulting from employment, associated w/ diagnostic or therapeutic procedures, any prison or penal institution, in legal custody, cremation, dissected or buried at sea, Unclaimed bodies, Body brought into a new medicolegal jurisdiction w/o proper medical certification
What are the components of an autopsy?
External Examination, Internal Examination, Inspection
What are the characteristics of Livor Mortis?
Lividity, postmortem hypostasis, Reddish purple coloration, Accumulation of blood in the small vessels, Evident 30 min-2 hrs after death, Can appear antemortem, Maximum coloration at 8-12 hrs --> Fixed
What are the characteristics of Rigor Mortis?
Stiffening of the body after death, Cessation of ATP generation, Develops in all muscles at the same time, Cold-delay onset and prolong presence, Heat-speed up onset, Disappears with decomposition
What are Rigor Mortis and Livor Mortis used for?
Not very important in determining the time of death. It is important in determining whether the body has been moved
What is the most method for determining time of death?
When was the person last seen? (i.e. 10:00 pm), When was the person found? (i.e. 9:00 am); The person died sometime between 10:00 pm and 9:00 am
What are the components of toxicology?
Blood (most important), Vitreous Humor, Urine, Bile, Gastric contents
What is involved in the external examination?
Identification, Identifying characteristics (body type, clothing, age, weight), Head to Toe Examination (skin, hair color, chest, abdomen, back, extremities), Toxicology
What are the types of blunt force wounds?
Contusion (Bruise), Abrasions, Laceration
What is the difference between a recent and old bruise?
Recent is purple or red; Old is yellow or brown
How can you tell a laceration from a stab wound?
Skin bridging, there is tissue that is still connected where the skin was town, laceration has rough edges (stab wound is smooth)
What is seen in a Basilar Skull Fx?
Have a battle sign (bruising behind the ears) & raccoon eyes
What are the types of sharp force wounds and what differentiates them?
Incised wounds (Length > depth, No Abrasion, No Contusion, No Bridging) and Stab wounds (Pointed instruments, Homicidal, Depth exceeds length, Sharp edges)
What is the most important thing about GSW?
Determining if it is an entrance or exit wound
How can you tell if a GSW is an Entrance wound & what types of entrance wound exist?
If there is injury to the skin present, reddening & abrasions of the superficial skin, there is tissue lost;
Contact Entrance Wound (muzzle imprint);
Intermediate Entrance Wounds (Powder Tattooing: abrasions created by powder);
Distance Entrance Wounds: no muzzle imprint, no powder tattooing, just skin abrasion & tissue lost of skin around it
What is included in the Internal Examination?
Organs in anatomical Position, Gross examination of each organ, Documentation of injury to organs, Microscopic sections if needed
What are the most common natural disease processes that cause death?
Heart disease, Lung Disease (COPD, asthma), Cancer, Liver Disease
What organism can affect normal valves and cause Acute IE?
Staphylococcus aureus
What is the most important thing for a pathologist to figure out for MVA?
Who was driving?
What is dicing?
The side paneling on glass windows breaks into cubes and causes this characteristic abrasions and lacerations to the arm in MVAs
What is the protocol for thermal injuries?
Complete X-Rays due to their Pugilistic position (Arms and legs will be contracted (i.e. like a boxer), Toxicology
What are the characteristic findings in thermal injuries?
Skin splitting (expose muscle); Ruptured muscle; Unburned skin (seared leathery appearance); Burned bone (grey/white with fine superficial fractures (curved))
When is it protocol to perform complete X-Rays?
Gunshot wounds, Sharp force deaths, Fire deaths
What is a defensive type wound?
Shows whether the person tried to fight back or not, usually on the fingers (person grabs knife), back of the hands, back of the arm
What is important in Infants and Childhood Deaths?
Complete X-Rays (Battered babies and abused children often have old, healing fractures), Possible cultures, Heart very important, CPS reports (old Hx of abuse), SUID (Sudden Undetermined Infant Death), Doll reenactment
What classifies Drowning?
Dx of exclusion, wrinkled skin, possible pulmonary edema
What is a Nasal Plume?
Due to pulmonary edema, seen w/ asphyxiated death (i.e. drowning, drug overdose)
What are the types of Asphyxia deaths?
Hanging, Smothering, Strangulation, Chemical Asphyxia
What classifies how autoerotic asphyxia?
Asphyxial Deaths, Anoxia induced to enhance sexual arousement produced by masturbation
What is the primary lesion seen in Multiple Sclerosis?
Plaques of demyelination: periventricular white matter (classical site), gray-white matter junctions, optic nerves/chiasm/tracts, brainstem ascending/descending tracts, subpial region of spinal cord
Note: on actual gross sections, areas of white matter afflicted with MS look discolored and grayish
What disease is similar to ADEM in pathology but is usually fatal?
Acute Hemorrhagic Leuko-Encephalitis (AHLE) aka Weston-Hurst disease: acute, monophasic (following a viral illness or immunization), causes perivenous demyelination (like ADEM) as well as petechial hemorrhage in the demyelinated white matter, and possibly neutrophilic infiltrate
Note: AHLE is usually fatal due to petechial hemorrhages, whereas ADEM patients recover (!)
What is found grossly and microscopically in ADEM?
Grossly: normal brain – Microscopically: perivascular pattern of demyelination (no hemorrhage)
What characterizes Acute Disseminated Encephalo-Myelitis (ADEM)?
Perivenous encephalomyelitis: acute, monophasic demyelination in children (following a viral illness or immunization), usually recovers with supportive therapy
What are clinical variants of Multiple Sclerosis?
Classic or Charcot type: relapsing/remitting form
Acute or Marburg type: first lesion can rapidly progress, cause extensive demyelination, and be fatal acutely (!)
Devic disease: affects only optic pathway and/or spinal cord (aka neuromyelitis optica)
Balo concentric sclerosis: only concentric areas of demyelination
Schilder disease: all MS variants that do not fit elsewhere
What morphological variant of macrophages are seen in demyelinating diseases?
Crutzfeldt cells: macrophages with mitotic figures (not unique to MS, may be seen in all demyelinating disease)
What is seen microscopically with Multiple Sclerosis?
Acitve plaques (demyelination): loss of myelin, macrophages (sometimes Crutzfeldt cells), **relative sparing of axons and neurons, **perivascular mononuclear T cell infiltrates, reactive astrocytosis, edema
Remyelination: “Shadow plaques” showing some remyelination, Schwann cell migration to spinal cord --> remyelination from Schwann cells in spinal cord
Inactive plaques: demarcated/acellular/inactive areas of previous demyelination with little inflammatory infiltrate
Note: axons are less involved than the white matter in Multiple Sclerosis ****
Note: remyelination never produces equally good myelin as original (!)
What characterizes Multiple Sclerosis?
F > M, linkage to HLA-DR2, 15-fold greater risk when 1st relative has MS, 25% concordance rate in monozygotic twins, increased incidence with distance away from the equator – pathogenesis is thought to involve genetic predisposition, immune factors, and possibly environmental antigens (virus?)
What is a screening test for Multiple Sclerosis?
CSF evaluation: looking for oligoclonal bands in CSF
How does Multiple Sclerosis present clinically?
Most common presentation: relapsing/remitting neurological deficits
Other signs: optic neuritis, brainstem involvement (CN signs, ataxia, nystagmus, ophthalmoplegia), spinal cord involvement (motor/sensory abnormalities, bladder dysfunction), oligoclonal bands in CSF
What characterizes Krabbe Disease (Globoid Cell Leukodystrophy)?
Galactocerebroside b-galactosidase deficiency (AR) --> psychosine accumulation, demyelination with multinucleated globoid cells, curved tubular inclusions in macrophages/Schwann cells (EM)
Note: globoid cells are multinucleated marcophages (!)
What leukodystrophies can cause demyelination of PNS?
Metachromatic leukodystrophy, Krabbe disease
What are some demyelinating diseases of PNS?
Guillain-Barre syndrome, Chronic Inflammatory Demyelinative Polyradiculoneuropahy (CIDP)
What characterizes Canavan Disease?
Aspartoacylase enzyme deficiency (AR) --> “spongy” degeneration of white matter, Alzheimer type 2 astrocytes
Note: Alzheimer type 2 astrocytes are seen in Canavan disease and Hepatic Encephalopathy (!)
What characterizes Pelizaeus-Merzbacher Disease?
Proteolipid protein synthesis defect (X-linked, M>F) --> widespread demyelination + perivascular myelin sparing (“tigroid” demyelination)
Note: proteolipid protein is the precursor for CNS myelin
What characterizes Alexander Disease?
Sporadic pattern of inheritance (most patients have new mutations), patients present with macrocephaly, demyelination, and large numbers of Rosenthal fibers (astrocytic inclusions)
Note: demyelination + lots of Rosenthal fibers --> think Alexander Disease
What characterizes Adrenoleukodystrophy?
ATP-binding cassette transporter deficiency in Perixosomes (X-linked, M>F) --> very long chain fatty acid accumulation (from peroxisomal dysfunction), adrenal dysfunction (enzyme has role in adrenals)
Demyelination is more prominent posteriorly, with **perivascular lymphocytes (mimicking MS) and cleftlike inclusions in macrophages
Note: case wording may be: adrenal cortex thinning, posterior brain demyelination, demyelination with macrophages + perivascular lymphocytes, cleftlike inclusions in macrophages
What are the infectious demyelinating diseases?
Progressive multifocal leukoencephalopathy (PML): due to JC virus, primarily affects oligodendrocytes (leading to demyelination), oligodendrocytes appear with intranuclear viral inclusion (glassy appearance, chromatin pushed to the side), JC viruses (under EM) look like spaghetti and meatballs (!)
Others include subactue sclerosing panencephalitis, Rubella, HIV leukoencephalopathy
What characterizes Metachromatic Leukodystrophy?
Arylsulfutase A deficiency (AR) --> metachromatic accumulation in macrophages/Schwann cells, prismatic/tuffstone inclusions (EM)
Note: metachromasia refers to staining that produces a different color from what’s expected of the stain (!)
What are leukodystrophies?
Hereditary disorders of myelin – most are due to enzyme deficiency (AR) – in all, demyelination is seen as “confluent areas of discolored, gelatinous white matter” - diseases are listed below …
Note: leukodystrophies spare the subcortical arcuate fibers, whereas in MS, they are affected (!)
What are vascular disorders that can cause demyelination of white matter?
Binswanger disease, Chronic edema
What can Thiamine deficiency lead to in the CNS?
Wernicke encephalopathy + Korsakoff psychosis + (possible) beriberi (involvement of PNS, CV)
Wernicke encephalopathy: eye problems + nystagmus/ataxia + global confusion + disorientation
Korsakaoff psychosis: cannot form new memories (retrograde amnesia), confabulation
What characterizes methanol poisoning?
Metabolic acidosis (systemically), blindness, respiratory depression – causes **** bilateral necrosis of putamen, degeneration of retinal ganglions – treated with ethanol
Note: if see “bilaterally symmetric” lesion of putamen --> think methanol poisoning ****
What lesions are seen in CO poisoning?
Acute lesion: cherry-red discoloration
Chronic lesion: **** bilateral necrosis of globus pallidus, demyelination of white matter tracts
Note: most hypoxic, stroke-type damages are not bilaterally symmetric (!)
Note: if see “bilaterally symmetric” lesion of globus pallidus --> think CO poisoning (Slide 21 – KNOW)
What characterizes CO poisoning?
Acutely leads to hypoxic damage to brain --> in surviving patients, leads to delayed encephalopathy and Parkinsonism – seen in accidental or suicidal settings
What characterizes Marchiafava-Bignami disease?
Demyelination/necrosis of Corpus Callosum --> symptoms of interhemispheric disconnection (Neuropsychiatric disorders), dysarthia, impaired consciousness – 1st described in Italian red wine drinkers (but can occur in anyone, with any type of drink)
What characterizes Central Pontine Myelinolysis? ****
Iatrogenic disorder seen most commonly in setting of alcoholism: too rapid correction of hyponatremia --> diamond-shaped demyelination in basis pontis --> rapidly evolving quadriplegia
What lesions are seen in Wernicke-Korsakoff syndrome?
Mamillary body lesions (acute: edema, hemorrhage, capillary proliferation – chronic: shrunken, brown mamillary bodies with gliosis) – other periventricular area lesions may be seen (!)
What are CNS lesions in alcoholism?
Trauma, cerebellar degeneration, Wernicke-Korsakoff syndrome, Central Pontine Myelinolysis, Marchiafava-Bignami disease, Fetal Alcohol Syndrome
What are other causes of cerebellar vermis atrophy?
Ethanol, phenytoin, heavy metals (Mercury), spinocerebellar ataxias
What characterizes Alcoholic Cerebellar Degeneration?
Atrophy of anterior, superior vermis (midline cerebellum) --> truncal ataxia, unsteady gait, nystagmus – affects 1% of chronic alcoholics – histology shows loss of Purkinje cells with Bergmann gliosis
What chemotherapeutic agent can cause brain damage?
Methotrexate --> white matter damage with swollen axons
What are lysosomal storage diseases of CNS?
Sphingolipidoses: enzyme deficiency (AR), affect either gray or white matter
Mucopolysaccharidoses
Neuronal ceroid lipofuscinosis
What characterizes the CNS pathology of Vitamin B12 deficiency?
Subacute combined degeneration of the spinal cord (combined posterior + lateral column degeneration)***
What characterizes the pathology of Vitamin B12 deficiency in CNS?
(all insidious, progressive)
Spinal cord problems: spastic paraparesis, sensory ataxia, severe paresthesias in lower limbs
Megaloblastic anemia, increased methylmalonic acid in urine, improvement with IV B12
What are the etiologies for Vitamin B12 deficiency in CNS?
#1 cause is Intrinsic Factor deficiency (from pernicious anemia or gastrectomy), other causes include dietary deficiency, malabsorption (Crohn’s disease), fish tapeworm infection, or increased metabolism of Vitamin B12 (pregnancy, hyperthyroidism, cancer)
What is seen microscopically with Solvent Abuse Leukoencephalopathy?
Brain atrophy with discoloration of white matter (more subtle than leukodystrophies)
Loss of myelin, ** characteristic PAS-positive macrophages around blood vessels (!)
Note: all changes are extremely subtle in this disease, except PAS-positive macrophages around vessels (!)
What occurs with inhalant abuse of toluene-containing solvents (e.g. glue sniffing)?
Solvent Abuse Leukoencephalopathy: volume loss of white matter, loss of gray-white matter demarcation, increased white matter signal on MRI - Mimic leukodystrophies, but have more subtle discoloration of white matter
What characterizes Lead poisoning?
Mimic of meningitis (encephalopathy, increased intracranial pressure, nuchal rigidity), characteristic peripheral neuropathy (demyelinating, with wrist/foot drop), epigastric pain/vomiting/constipation, anemia, chronic tubulointerstitial nephritis
What occurs to the brain as a result of cranial irradiation?
Acute: patchy edema of white matter – Delayed: white matter damage, fibrinoid changes in blood vessels
Secondary tumors can appear (#1: meningioma)
Histologically: typical change is appearance of atypical cells with big nucleus AND big cytoplasm
Given the major function and G-protein class name the receptor

↑ H2O permeability and reabsorption in the collecting tubules of the kidney

What mechanims does it use?
V2

Receptor --(Gs)--> adenylyl cyclase...

Lipids --(adenylyl cyclase)--> ↑ cAMP --> protein kinase A
How is Gaucher disease diagnosed?
CNS findings are subtle: Gaucher cells in perivascular spaces – not usually tested for diagnosis
Diagnosis is done by looking for Gaucher cells in liver or BM
What findings are seen in CLN disorders?
CLN1 (infantile) – Granular bodies on EM;
CLN2 (late-infantile) – Curvilinear bodies on EM;
CLN3 (juvenile) – Fingerprint profiles on EM
What disease causes accumulation of Lipofuscin-like material in neurons?
Neuronal Ceroid Lipofuscinosis (CLN) aka Batten disease – unknown enzyme defect --> progressive neurologic illness with blindness
Which Mucopolysaccharidoses is X-linked?
Hunter syndrome
What are the most common findings in Mucopolysaccharidoses?
Common findings: involvement of soft tissue/bone of face --> coarse facial features, corneal clouding (cataracts), joint stiffness, mental retardation
What characterizes Mucopolysaccharidoses?
Glycosaminoglycan degradative enzyme deficiency (most are AR) --> systemic involvement – storage material in cells are called “zebra bodies” - can be diagnosed by urine monitoring for accumulated mucopolysaccharides
What characterizes Niemann-Pick disease?
Sphingomyelinase deficiency (AR) --> visceral storage mainly in macrophages (hepatosplenomegaly, lymphadenopathy), CNS storage mainly in neurons and macrophages (progressive psychomotor deterioration)
What are lysosomal storage diseases of CNS that predominantly affect the white matter?
Leukodystrophies: Metachromatic leukodystrophy (Arylsulfatase A), Krabbe disease (Galactocerebroside -->-galactosidase, Globoid cells)
What characterizes Gaucher disease?
Glucocerebroside deficiency (AR) --> systemic disease --> distended, PAS-positive phagocytes (Gaucher cells) systemically, in CNS found in perivascular spaces
Note: know that Gaucher disease is a systemic sphingolipidoses and enzyme replacement therapy is available for it (!)
Note: Gaucher cells are supposed to look like “wrinkled tissue paper” (!)
What characterizes Tay-Sachs disease (GM2 Gangliosidoses)?
(more common in Ashkenazi Jews)
Hexosaminidase A deficiency (AR) --> progressive motor/mental deterioration beginning in infancy
Characteristic but not pathognomonic finding is “macular cherry-red spots”
Storage material accumulates exclusively in the gray matter (neurons) – not a systemic disease (!)
Case: 6-month old child with cherry-red spot on retinal exam, know what enzyme is missing (!)
Note: this is on the test (99% chance)
What peroxisomal disorder presents as a combination of leukodystrophy and abnormal neuronal migration? ********
Zellweger syndrome: is a generalized peroxisomal dysfunction --> accumulation of very long chain fatty acids --> abnormal neuronal migration and/or myelination disturbances
Which mitochondrial disease is due to large deletions in mitochondrial DNA?
Kearns-Sayre syndrome (sporadic)
What mitochondrial encephalomyopathies are due to mutations in mitochondrial tRNA (and maternally transmitted)?
MERRF: myoclonus, myopathy, ataxia – neuronal loss in inferior olives, cerebellar cortex, cerebellar outflow nuclei
MELAS: neurologic dysfunction, weakness, lactic acidosis – patients will have strokes in white/gray matter
Note: see a child with stroke-like lesion --> think MELAS (!) *********
What characterizes Leigh syndrome?
Due to decreased cytochrome c oxidase activity - lesions look like Wernicke-Korsakoff, but occurs in children: spongiform degeneration and proliferation of blood vessels in periventricular gray matter
What characterizes Mitochondrial Encephalomyopathies?
Mutations can be either in the mitochondrial DNA (maternal transmission) or in nuclear DNA – common finding in all disorders are “ragged red fibers”, lactic acidosis, weakness with hypotonia
Which Glycogen Storage Disease involves CNS?
Pompe disease due to Acid Maltase deficiency --> systemic involvement of muscle, liver, CNS (!) --> MORE SEVERE than McArdle’s
What are common findings in Glycogen Storage Disorders?
Hepatic enlargement, hypoglycemia, muscle cramps after exercise – only know the following …
McArdle’s disease due to Myophosphorylase deficiency --> only involves muscles (!) --> LESS SEVERE
What are the routes of entry for CNS infections?
Hematogenous spread (most common);
Direct implantation (traumatic, iatrogenic);
Local extension (sinuses, cranial bones);
Via the peripheral nervous system (rabies)
What are the causative organisms for community acquired acute pyogenic meningitis?
Streptococcus pneumoniae (most common), Neisseria meningitidis (i.e. outbreak on college campus, military groups), group B streptococcus, Listeria monocytogenes
What are the causative organisms for acquired acute pyogenic meningitis?
S. pneumoniae, Staphylococcus species, gram-negative bacilli
What are the causative organisms for immunocompromised patients w/ acute pyogenic meningitis?
L. monocytogenes, gram-negative bacilli
What are the symptoms associated with acute pyogenic meningitis?
headache, photophobia, irritability, clouding of consciousness, neck stiffness
What is the pathology & what found in the CSF of a pt w/ acute pyogenic meningitis?
Path: Prominent meningeal vessels, Neutrophils in subarachnoid space, Phlebitis with hemorrhagic infarction, Chronic adhesive arachnoiditis;
CSF: Cloudy/purulent, increased pressure, increased neutrophils, increased protein, decreased glucose
What are the Predisposing conditions for brain abscess?
Bacterial endocarditis, cyanotic congenital heart disease, & chronic, pulmonary sepsis
What are the primary causative organisms for brain abscess & how do patients present?
Streptococci & staphylococci;
Present with focal deficits & increased intracranial pressure
What types of CNS lesions are discrete with central necrosis surrounded by a fibrous collagen capsule & edema and can be treated with surgery and antibiotics?
Brain Abscess
What are the causes of chronic bacterial meningoencephalitis?
Tuberculosis, Neurosyphilis, Lyme disease (neuroborreliosis)
What are describes Tuberculous Meningitis?
Symptoms: headache, malaise, mental confusion, vomiting;
CSF: mononuclear cells, increased protein, normal or moderately decreased glucose (less severe extent that in acute bacterial meningitis);
What is a Tuberculoma?
Well-circumscribed intraparenchymal mass (mass lesion)
How does TB meningitis present pathologically?
Gross: Gelatinous or fibrinous, basally located exudate
Histo: Granulomatous inflammation (like it is w/ TB in other organs) with caseous necrosis & obliterative endarteritis
What is vertebral involvement in TB meningitis called?
Pott disease (can cause the bones to collapse and compress the spinal cord)
What are the 3 types of neurosyphilis?
Meningovascular neurosyphilis - chronic meningitis with obliterative endarteritis & a perivascular plasma cell infiltrate;
Paretic neurosyphilis - invasion of the brain by treponemal organisms with brain atrophy & resultant severe dementia;
Tabes dorsalis - damage to dorsal root sensory nerves with impaired sensation & absence of deep tendon reflexes
What causes Lyme disease and what are the neurologic findings?
Spirochete Borrelia burgdorferi; Neurologic findings are non-specific & include aseptic meningitis, facial nerve palsies, mild encephalopathy, & polyneuropathies
What is the most common cause of aseptic meningitis?
Enteroviruses, can be viral or chemical
What is seen in the CSF of Aseptic meningitis?
Lymphocytic pleocytosis with moderate protein elevation & normal glucose (vs Bacteria meningitis which has decreased glucose)
What is the difference between Aseptic meningitis and Bacterial meningitis?
Aseptic meningitis is usually self-limited (vs bacterial which needs surgery or ABX treatment)
What is viral meningoencephalitis?
Parenchymal infection of the brain associated with meningeal inflammation
What histological features are always seen in all viral CNS infections?
Perivascular & parenchymal mononuclear cell infiltrates, glial nodules, Neuronophagia
What are responsible for epidemic viral encephalitis (Arthropod-Borne viral encephalitis)?
Include Eastern, Western, & Venezuelan equine, St. Louis, Japanese B, California, Murray Valley, West Nile, and tick-borne encephalitis viruses; Animal hosts & mosquito or tick vectors, they present with Generalized or focal neurologic deficits
What is the most common cause of sporadic encephalitis?
HSV-1 (hemorrhagic encephalitis involving the temporal lobes & orbital gyri of the frontal lobes) (vs HSV-2: severe generalized encephalitis in neonates & immunocompromised patients)
What CNS virus are Cowdry A nuclear inclusions present in?
Herpes Simplex Virus
What type of CNS manifestation can VZV cause in immunocompromised patients?
Acute meningoencephalitis
What characterizes CMV in the CNS?
Infections occur in fetuses & immunocompromised patients; Periventricular necrosis & calcification; Prominent cytomegalic cells with intranuclear & intracytoplasmic inclusions
What classified the poliovirus?
Enterovirus that has been controlled by immunization; Specifically attacks lower motor neurons producing a flaccid paralysis with muscle wasting & hyporeflexia
What is syndrome presents as late neurologic syndrome characterized by progressive weakness associated with decreased muscle bulk & pain?
Postpolio syndrome
What are Negri bodies?
Cytoplasmic eosinophilic inclusions in pyramidal neurons of the hippocampus & cerebellar Purkinje cells, found in Rabies
What is characteristic presentation of rabies?
Patients exhibit CNS excitability (sensitivity of light and fear of water)
How does Rabies enter the CNS?
Virus enters CNS by ascending along the peripheral nerves from the wound site
What are the CNS manifestations of HIV?
Aseptic meningitis (early HIV); Meningoencephalitis with microglial nodules & multinucleated giant cells (AIDS); Vacuolar myelopathy (myelin damage); Cranial & peripheral neuropathies; Myopathies (inflammatory and drug-related); Opportunistic infections
What does the JC virus cause?
Progressive Multifocal Leukoencephalopathy, infects & kills oligodendrocytes, Demyelination with lipid-laden macrophages, Nuclear viral inclusions in oligodendrocytes, Bizarre giant astrocytes
What can a defective form of the measles virus cause?
Subacute Sclerosing Panencephalitis: Cognitive decline, spasticity of the limbs, and seizures; Loss of myelin and gliosis (predominantly a white matter disorder); Nuclear and cytoplasmic viral inclusions
What are the manifestations of the CNS of fungal infections?
Cause chronic meningitis, vasculitis, & parenchymal invasion; Usually result from hematogenous dissemination from a primary infection elsewhere; Encountered primarily in immunocompromised patients
What Encapsulated yeast causes meningitis or parenchymal lesions with Sparse inflammatory cell infiltrate?
Cryptococcus neoformans causes Cryptococcus (5-10 micrometer yeasts with wide capsular halo; narrow-based unequal budding)
What fungus produces microabscesses in the CNS?
Candidiasis (Candida albicans, Hyphal and yeast forms, Nonbranching pseudohyphae + Blastocondia)
What fungus has a predilection for vessel walls leading to hemorrhagic abscesses in the CNS?
Aspergillosis (Aspergillus fumigatus or A. flavus most common; Septate, branching hyphae (45 degree angle; Acute angle = Aspergillosis), rare fruiting bodies)
What fungus causes Rhinocerebral infection in patients with diabetes and ketoacidosis and is angioinvasive?
Mucormycosis (Mucor, Rhizopus, and Absidia, Irregular, Irregular broad (empty-looking) non-septate hyphae that branch at right angles [wide-angle])
mandón/mandona
bossy
What characterizes histoplasmosis, coccidioidomycosis, and blastomycosis?
Histoplasmosis (Tiny (2-5 micrometers) yeasts, occasional unequal budding;
Coccidioidomycosis (20-60 micrometer spheres with endospores);
Blastomycosis (Larger (up to 25 micrometers) yeasts, double-contoured, broad-based budding)
What parasitic infection must be clinically distinguished from primary CNS lymphoma?
Toxoplasmosis
What characterizes Toxoplasmosis?
Toxoplasma gondii, Infects immunosuppressed patients and fetuses, Necrotic lesions surrounded by inflammation; Free tachyzoites and encysted bradyzoites
What are the types of Amebias that can infect the CNS?
Naegleria: Rapidly fatal meningoencephalitis;
Acanthamoeba and Balamuthia: Chronic granulomatous encephalitis, Keratitis in contact lens wearers
What classifies cysts containing larval forms of the organism that are commonly found in the CNS?
Cysticercosis (Taenia solium (pork tapeworm))
What are the different types of Spongiform Encephalopathies?
Creutzfeldt-Jakob disease; Kuru; Gerstmann-Straüssler-Scheinker syndrome; Fatal familial insomnia; Variant Creutzfeldt-Jakob disease
What is the cause of spongiform encephalopathies & what are the manifestations?
Caused by prion proteins: normal cellular prion protein (PrP) undergoes a conformational change to an abnormal beta-pleated sheet isoform which resists digestion and accumulates in neural tissue; Rapidly progressive dementia, Spongiform change, neuronal loss, & reactive astrocytosis
What type of spongiform encephalopathy can be iatrogenic transmission?
Creutzfeldt-Jakob disease (Rapidly progressive dementia with myoclonus; Uniformly fatal; Spongiform change, neuronal loss, and gliosis)
What spongiform encephalopathy is an inherited disorder that causes cerebellar ataxia & is characterized by Large amyloid plaques in cerebellum?
Gerstman-Straussler-Scheinker Syndrome
What classifies Fatal Familial Insomnia?
Spongiform encephalopathy, Inherited, Sleep disturbances, ataxia, and autonomic problems, Pathologic changes in thalamus
What spongiform encephalopathy has characteristic florid plaques pathologically?
Variant CJD (United Kingdom, Younger age of onset, more prominent behavioral disturbances, and a longer clinical course than typical CJD cases, Linked to bovine spongiform encephalopathy)
What are the two main types of Infections that bacteria can cause?
(1) Inflammation of the Meninges (Meningitis)
(2) Localized infections due to bacteria (Abscesses)