• 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

1/999

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;

999 Cards in this Set

  • Front
  • Back
What characterizes mucoepidermoid carcinomas?
Malignant tumor composed of a mixture of squamous cells, mucus-secreting cells, and intermediate cells; occurs mainly in the parotids, but accounts for a large fraction of tumors in other salivary glands (esp. minor salivary glands)
What characterizes Warthin tumors?
2nd most common salivary gland neoplasm; almost always in parotid gland (sometimes bilateral); M>F, usually in 5th-7th decades of life; smoking increases the risk (x8); very low recurrence rate after complete resection
When a pleomorphic adenoma undergoes malignant transformation (rare), what malignancies can arise?
Carcinoma, Sarcoma (chondrosarcoma), True malignant mixed tumor
What characterizes a pleomorphic adenoma?
Most commonly seen in parotid gland (60%); contains a mixture of ductal and epithelial cells, with mixed mesenchymal and epithelial elements; painless, benign, slow-growing tumor – recurrence is rare after complete excision
What are the malignant salivary gland tumors?
Mucoepidermoid carcinoma (most common), adenocarcinoma NOS, acinic cell carcinoma, adenoid cystic carcinoma, and others
What syndrome is xerostomia (dry mouth) associated with?
Sjögren syndrome
(autoimmune destruction of exocrine glands); also seen as a complication of radiation therapy
What characterizes mucoceles?
Most common salivary gland lesion; result from ductal blockage or rupture with saliva leakage into surrounding stroma; most often found on the lower lip, typically result from trauma – complete excision is necessary
What causes nonspecific (bacterial) sialoadenitis?
Ductal obstruction by stone (sialolithiasis) is associated with bacterial overgrowth (usually Staph. aureus or Strep. viridans) which induces painful enlargement and purulent discharge
What characterizes adenoid cystic carcinomas?
Most often found in minor salivary glands; slow-growing by relentless/recurrent; perineural invasion is common; disseminate widely after time; early survival is good, but long-term is not
What characterizes acinic cell carcinomas?
Resemble normal serous acinar cells; most often in the parotid; recurrence after resection uncommon; survival rates are good
What characterizes an olfactory neuroblastoma (esthesioneuroblastoma)?
Neuroendocrine tumors resembling a neuroblastoma; it is a primitive neuroectodermal tumor but it does not have the 11;22 translocation of Ewing sarcoma – tumor of small cells, metastasize widely
What characterizes an extramedullary plasmacytoma?
Malignant plasma cells that arise in lymphoid structures adjacent to nose/sinuses – rarely progress to multiple myeloma
What characterizes sinonasal (Schneiderian) papillomas?
Benign neoplasms composed of squamous or columnar mucosa; caused by HPV 6 and 11 – can be septal, inverted (not exophytic), and cylindrical
What concept is thought to be responsible for the fact that many people with oral cancer also often get laryngeal cancer?
Field cancerization
In what age group does otitis media most commonly occur and what are the causative agents of acute and chronic disease?
Infants and young children – Acute: S. pneumoniae, H. influenza, M. catarrhalis;
Chronic: P. aeruginosa, S. aureus, fungus or mixed flora
What characterizes laryngeal squamous papillomas?
Benign neoplasms, usually on the true vocal folds; small, soft, raspberry-like excrescences that have slender, finger-like projections with fibrovascular cores and squamous epithelium; usually single in adults and multiple in children (juvenile laryngeal papillomatosis); multiple recurring papillomas are associated with HPV 6/11 (not malignant, often regress at puberty)
What is the prognosis for a patient with laryngeal carcinoma?
At presentation 60% are confined to the larynx; they generally spread to lymph nodes; cancer may recur at primary site or adjacent sites (field cancerization); about 1/3 die of disease (usually due to local spread of the cancer)
Where do laryngeal cancers first metastasize to?
Cervical lymph nodes (just like Head/Neck cancer)
What is the treatment for laryngeal carcinomas?
Depends on the stage: can include radiation/surgery (laryngectomy with/without ipsilateral radical neck dissection), chemotherapy/radiotherapy, or a combination of surgery/radiation/chemo
What characterizes laryngeal carcinomas (site of origin, type)?
Their site of origin can include the vocal cords or surrounding structure (epiglottis, aryepiglottic folds, piriform sinuses); 95% are squamous cell carcinomas; they are ulcerating/fungating lesions
What are the most common causes of laryngitis and what are the possible consequences?
Infection or heavy tobacco smoke exposure – β-hemolytic strep and Haemophilus influenzae can cause a lethal laryngoepiglottitis in infants/young children (may require intubation/tracheostomy); inflammation of the larynx can cause croup in children (laryngotracheobronchitis with inspiratory stridor); Laryngitis in smokers may have pre-malignant changes
What are the major risk factors for laryngeal carcinoma?
Cigarette smoking, alcohol exposure – minor factors: nutritional causes, asbestos, radiation, HPV
What characterizes reactive nodules on the vocal cords?
They most commonly occur in smokers or in those with strain on the vocal cords (singers); they occur more frequently in males than females and cause hoarseness; morphologically they are smooth rounded excrescences composed of myxoid connective tissue, covered by squamous epithelium – surgical excision is curative
What are the most common types of ear tumor?
Those affecting the external canal: squamous cell and basal cell
What do carotid body tumors consist of?
Zellballen: nests of polygonal neuroendocrine cells enclosed by fibrous trabeculae and elongated sustentacular cells
Where are the most common paragangliomas located?
Carotid body paraganglion, vagal paraganglia, jugulotympanic paraganglia
What characterizes paragangliomas?
Tumors arising in extra-adrenal paraganglia (paravertebral or associated with the great vessels of the head); often arise singly/sporadically in the sixth decade of life; may be familial, associated with autosomal dominant transmission of MEN II syndrome – they frequently recur after incomplete resection; many metastasize to local/distant sites; about half are fatal
What characterizes a thyroglossal duct cyst?
Midline remnants of the thyroid tract between the foramen cecum of the tongue and the thyroid;
they are 1-4 cm cysts with stratified squamous/pseudostratified columnar epithelial lining; contain lymphoid aggregates or remnants of thyroid tissue – excision with recurrence if incomplete, malignant transformation is rare
What characterizes a branchial cleft cyst?
Lateral remnants of the branchial arches or salivary gland inclusions in cervical lymph nodes;
they are 2-5 cm circumscribed cysts with fibrous walls, composed of stratified squamous/ pseudostratified columnar epithelial lining; they have clear, watery-to-mucinous contents or desquamated, granular cellular debris – they enlarge slowly and rarely transform to malignant
What are the most common neck masses caused by?
Reactive lymphadenopathy; Cysts: branchial cleft (lateral), thyroglossal duct (medial); Metastatic tumors; Lymphoproliferative disorders
What can result from severe otitis media?
Eardrum perforation;, Spread within ear, temporal cerebritis, abscess; Destructive necrotizing otitis media in diabetics (P. aeruginosa)
What characterizes otosclerosis?
Abnormal bone deposition in the middle ear, hampering stapes footplate mobility (hearing loss);
starts in young adults and is slowly developing; has an autosomal dominant pattern with variable penetrance – can be surgically corrected with a prosthesis
What characterizes cholesteatoma?
A non-neoplastic mass of keratinized squamous epithelium and cholesterol in the middle ear, seen with tympanic membrane perforation or chronic otitis media; complications include erosion into surrounding structures or production of a neck mass
What are the benign salivary gland tumors?
Pleomorphic adenoma (most common), Warthin tumor, oncocytoma, other adenomas, ductal papillomas
What is the prognosis with mucoepidermoid carcinomas?
Low-grade: invade locally, rarely metastasize, 5-year survival > 90%; High-grade: invasive, difficult to excise, higher recurrence rate, many disseminate, 5-year survival rate is 50%
What characterizes periodontitis?
Inflammatory process affecting supporting structures of the tooth (due to a shift in bacterial flora and poor oral hygiene) – can result in loss of teeth, infective endocarditis, pulmonary/brain abscesses, and adverse pregnancy outcomes (premature labor)
What characterizes gingivitis?
Inflammation of soft tissues around the teeth (due to a lack of oral hygiene) leading to the accumulation of dental plaque (microbes) and calculus (mineralized plaque) – most common in adolescents (but reversible)
What characterizes hairy leukoplakia?
White, confluent patches of fluffy (“hairy”) hyperkeratotic thickening and squamous hyperplasia found on the lateral border of the tongue – mostly associated with AID
What pattern of metastasis is usually associated with HNSCC?
Invades locally first (cervical lymph nodes), and then to distant sites (mediastinal lymph nodes, lungs, liver, bones)
What are common sites for HNSCC?
Floor of mouth, ventral surface of tongue, soft palate/gingiva, base of tongue
What pathogenesis is associated with head and neck squamous cell carcinoma (HNSCC)?
Most common in middle aged men who smoke or abuse alcohol; some are HPV+
What is meant by field cancerization?
After a primary cancer has been discovered, second primaries will often present later on – this is due to the fact that cancer causing agents are effective over a large field of distribution
What is the most common type of head/neck cancer (95%)?
Squamous cell carcinoma (most common in the oral cavity) – long-term survival is < 50%
What distinguishes leukoplakia from erythroplakia?
Leukoplakia is a white lesion without specific diagnosis (5-25% premalignant) whereas erythroplakia is a red, velvety area (level/slightly depressed) that is associated with an atypical epithelium 90% of the time (dysplasia, CIS, cancer)
What viruses are associated with oral pathology?
HSV-1, HSV-2, VZV, EBV, CMV, Rubeola (measles)
What are examples of deep fungal infections?
Zygomycosis, Aspergillosis, Coccidiomycosis, Cryptococcosis, Histoplasmosis, Blastomycosis
In what patients is oral Candidiasis often seen?
Immunosuppressed, diabetics, transplant patients, AIDS, patient taking chemotherapy
What characterizes an ameloblastoma?
Cystic, slow-growing, locally invasive, benign growth – can also occur in the paranasal sinuses
What are mucormycosis and how are they treated?
Spreading fungal infections (especially in diabetics) – can be treated by surgical debridement
What can chronic tonsillitis lead to?
Obstruction of breathing, sleep apnea
What can an infection with β-hemolytic streptococci lead to?
Rheumatic fever and glomerulonephritis
What are nasal polyps?
Edematous mucosa with a loose stroma and inflammatory cells – can occlude the nasal canal
Which viruses are responsible for the common cold?
Adeno-, echo-, and rhinoviruses
What characterizes an odontoma?
Arises from epithelium and shows extensive deposition of enamel/dentin – probably hamartomatous
What is the most common odontogenic cyst and what characterizes it?
Periapical cyst – inflammatory cyst found at the apex of the tooth
What characterizes an odontoma?
Arises from epithelium and shows extensive deposition of enamel/dentin – probably hamartomatous
What is the most aggressive odontogenic cyst and what characterizes it?
Odontogenic keratocyst; most common in the posterior mandible of males age 10-40;
It has well-defined rediolucencies and contains a layer of parakeratinized/orthokeratinized (“basket weave pattern”) epithelium – needs to be aggressively and completely removed
What characterizes a dentigerous cyst?
Unilocular cyst around the crown of an unerupted tooth; contains a thin layer of stratified squamous epithelium – complete resection is curative
What characterizes a nasopharyngeal angiofibroma?
Vascular neoplasm of adolescent males, can bleed profusely
What characterizes the nasopharyngeal carcinoma?
Associated with EBV, seen in African children and adults in S. China; EBV is associated with the undifferentiated form (lymphoepithelioma); they tend to grow silently and are very radiosensitive
What characterizes an irritation fibroma?
Caused by a accidental bite in the buccal mucosa leading to a nodular mass of fibrous tissue covered by squamous mucosa
What characterizes herpes simplex virus?
Usually HSV-1 (increase in HSV-2); primary infection occurs in children, and reactivation occurs with triggers (e.g. stress, infection); cold sores consist of vesicles with molding, intranuclear inclusion (identified by Tzanck test)
What characterizes glossitis?
Inflammation of the tongue, often associated with vitamin deficiencies, iron-deficiency anemia, and esophageal dysphagia
What characterizes apthous ulcers?
Very common shallow ulcer with this exudate and narrow erythematous border – most common in the first two decades and within families; causation is obscure
What characterizes a peripheral giant cell granuloma?
Multinucleate, foreign body-like giant cells in a fibrovascular stroma; similar to a pyogenic granuloma but more bluish-purple
What characterizes a peripheral ossifying fibroma?
Red, ulcerated, nodular, reactive growth of the gingiva (etiology unknown) – common in young and teenage females
What characterizes a pyogenic granuloma?
Ulcerated, red-purple vascular proliferation (form of capillary hemangioma) – common in children, young adults, and pregnant women
What is the most common cause of tooth loss below age 35?
Caries (focal degradation of tooth structure by bacterial acid metabolic end products)
What is the purpose of hyphae?
They serve to penetrate substance, translocate nutrients via coordinated cytoplasmic streaming, and produce aerial spores
What are some nutritional characteristics of fungi?
Grow over wide pH range (1-11); filamentous fungi are mainly aerobic, whereas yeasts are often facultative anaerobes (fungi can also be obligate anaerobes); they can grow at very low moisture levels (>50%)
What is meant by germination of fungi?
Hyphal growth from a spore
What effects do the death cap/death angel and fly agaric have on the body, respectively?
Death cap/death angel: destroys the liver; Fly agaric: muscarinic agonist (hallucinations)
How does sexual reproduction of fungi occur?
Via sexual spores --> fusion of nuclei from 2 haploid mating strains --> zygote --> meiosis --> recombinant haploid spores
What fungus is commonly found in a grain silo, and causes sick building syndrome?
Aflatoxicosis – infection can lead to liver cancer
How are fungi identified in the lab?
By looking at their spores (conidia)
What are the two methods by which fungi grow and reproduce?
Yeast: budding ; Hypha: tubules/spores (sexual/asexual)
What substance is found in the hyphal cell walls?
Chitin (β-1,4 N-acetyl glucosamine); may contain septa (cross-walls) or may be coenocytic (no cross-walls)
What is meant by a fungus that is dimorphic?
It can grow as yeast in its infective/pathogenic stage, but is capable of hyphal growth saprophytically (in lab culture at room temperature)
By what mechanism, other than meiosis, can recombination of fungi occur?
Mitotic recombination in multinucleated cells after hyphal anastomosis (the spindle apparatuses of two cells undergoing mitosis become confused and chromosomal material is exchanged)
What are the various types of fungal infections (i.e. what locations/organ systems can they be associated with)?
Systemic mycoses, CNS mycoses, opportunistic mycoses, subcutaneous mycoses (wound infections), dermatomycoses (caused by keratinophilic fungi), superficial mycoses (fungal infections of the cutaneous integument – not associated with serious complications)
How do opportunistic mycoses relate to nosocomial infections (hospital acquired)?
They are the third most common, but cause 50% of nosocomial mortality – also, 90% of AIDS patients are afflicted by opportunistic mycoses
What does the Gomori Methenamine Silver (GMS) stain attach to?
Cell wall of fungi: chitin
What key differences are found between fungi and bacteria?
Cell walls: chitin vs. peptidoglycan; Staining: PAS/GMS vs. Gram+/-; Cell membranes: ergosterol vs. no sterol; Spore functions: resist desiccation vs. resist heat; Secondary metabolites: FK506 vs. none – also, fungi are larger, have organelles, undergo meiosis, have longer regeneration times, have infectious airborne propagules, and more commonly form spores
What are some examples of recombinant drugs that are developed using fungi (yeast fermentation)?
Human insulin (humulin) and hepatitis B vaccine
What are the most common species of Candida seen clinically?
C. albicans > C. glabrata > C. tropicalis > C. parapsilosis > C. krusei > C. lusitaniae
What acute and chronic adverse effects are associated with Amphotericin B?
Acute: fever, chills, rigors, hypotension, arrhythmias, thrombophlebitis, HA;
Chronic: nephrotoxicity (!), hypokalemia, hypomagnesemia, sodium wasting, anemia
What is the mechanism of action of Amphotericin B?
Binds to ergosterol in fungal cell membranes and alters permeability, leading to leakage of vital cell components
Why is Amphotericin B given with deoxycholate in systemic formulations?
Amphotericin B is not water soluble and requires a water-soluble ionic detergent (deoxycholate) and must be given in dextrose 5% water in order to form a micellular dispersion so that the drug is equally spread out in the formulation
What is the “gold standard” for fungal infections?
Amphotericin B
What is the most common life-threatening fungal infection in HIV-positive patients?
Cryptococcus
What characterizes Aspergillus clinically?
Most common in immunodeficient/immunosuppressed patients; expensive to treat; high mortality
Which common fungi are classified as yeasts, moulds, and dimorphic (endemic), respectively?
Yeasts: Candida, Cryptococcus; Moulds: Aspergillus, Mucormycosis, Fusarium;
Dimorphic: Histoplasma capsulatum, Blastomyces dermatitidis, Coccidioides immitis
What endemic areas are associated with the Dimorphic fungi?
Histoplasma capsulatum: Ohio river valley, Mississippi river valley; Blastomyces dermatitidis: Mississippi river valley, Southern US; Coccidioides immitis: Western US (causes valley fever)
What are the most common fungal pathogens seen clinically?
Yeast: Candida; Mould: Aspergillus
What is the mechanism of action of Flucytosine (5-FC)?
It is a pro-drug that is converted into 5-FU; this is a pyrimidine analog which disrupts DNA synthesis by inhibiting thymidylate synthetase
Which patients should be given Amphotericin B rather than Fluconazole?
Neutropenic patients, since Fluconazole is fungistatic, and not fungicidal like Amphotericin B
What adverse effects are associated with the older azoles?
Although they have a good therapeutic index, there is the chance of a drug-induced hepatotoxicity and they have an inhibitory effect on the CYP450 enzymes
How do Fluconazole and Itraconazole differ?
Fluconazole: narrower spectrum, yeast infections; Itraconazole: broader, yeast and mould activity
What are the older azoles and what is their mechanism of action?
Imidazoles: Miconazole, Ketoconazole; Triazoles: Itraconazole, Fluconazole – they function by inhibiting 14-α-lanosterol demethylase (CYP51A1, normally converts lanosterol to ergosterol), leading to a more fragile cell membrane; they are fungistatic
What adverse effects are associated with 5-FC?
Marrow toxicity (neutropenia); also, hepatic and GI effects (same as with 5-FU)
How and for what is 5-FC used?
In combination with other antifungal drugs since resistance develops rapidly; its main use is in Cryptococcal infections
What specific effect does Amphotericin B have on the kidneys?
It damages the renal tubules, thus inhibiting the tubule’s capacity for electrolyte reabsorption; this can lead to hypokalemia, hypomagnesemia, sodium wasting, and anemia – saline-loading is done to prevent renal insufficiency
What are the three lipid formulations of Amphotericin B and how do they differ?
ABLC (ribbon complex): very large size; L-AMB (liposome): very small size, mildest infusion-related reactions, highest cost; ABCD (disc-like complex): intermediate size, high rate of infusion-related reactions, lowest cost – all three formulation are less nephrotoxic, but none have superior efficacy nor a survival benefit associated with them
What drugs are given to lessen the infusion-related adverse effects of Amphotericin B, and what is given if a patient develops rigors?
Infusion-related side effects: Acetaminophen, anti-histamines – Rigors: Meperidine
What is the mechanism of action of the echinocandins?
They inhibit β-D-glucan synthase which causes disruption of the enzyme complex that is responsible for the formation of glucan fibrils; a disturbance of these fibrils will cause the fungal cell wall to break down (in a similar fashion by which Penicillin disrupts bacterial cell walls)
What is special about Posaconazole?
It is an oral Triazole with broad-spectrum activity: approved for Candida/Aspergillus and used off-label for Zygomycosis infections (!)
What pharmacokinetics are associated with Voriconazole?
Oral and IV formulations; good bioavailability; food can decrease absorption; IV formulations use a carrier; large Vd and penetrates CSF (!); inter-individual pharmacokinetic variability is high (due to CYP450 variability); since it is metabolized by CYP450, drug interactions are common;
dosing is difficult
What adverse effects are associated with Voriconazole?
Elevated liver enzymes (has greater hepatotoxicity than Fluconazole); Skin rash;
Visual disturbances (! does mean that the drug is effective for fungal infections of the eye)
What is characteristic about Voriconazole as compared to Fluconazole?
Voriconazole has been engineered to be more broad-spectrum: it has enhanced activity against all Candida species and is effective against Aspergillus and some other moulds
What issues are associated with the echinocandins?
Although they have minimal adverse effects and drug interactions, they are very expensive and only available in IV formulation
What fungi are affected by the echinocandins?
Candida and Aspergillus – but NOT Cryptococcus
Although all common forms of Candida are susceptible to Amphotericin B, which species are susceptible to Fluconazole?
Fluconazole is effective in C. albicans/parapsilosis/tropicalis, but not in C. glabrata/krusei (in reality C. glabrata susceptibility is dose dependent whereas C. krusei is always resistant)
What are the echinocandins?
Caspofungin, Micafungin, Anidulafungin
What issues are associated with Itraconazole?
Takes a long time to get to steady-state concentrations; no good oral absorption (especially when taken with antacids); significantly more drug interactions than Fluconazole
How does treatment of Candida differ when a patient is septic/immunocompromised vs. when they are not?
Septic/immunocompromised: Amphotericin B, echinocandins; Not: Fluconazole, echinocandins
What drugs are used for endemic fungal infections?
Itraconazole and Amphotericin B
What is the first-line treatment for Cryptococcal meningitis?
Amphotericin B with Flucytosine, followed by Fluconazole maintenance (in HIV patients, Fluconazole treatment is continued prohylactically)
What is the first-line treatment for invasive Aspergillosis?
Amphotericin B or Voriconazole (second-line is Caspofungin or high-dose AmphotericinB)
What are the Serotonin reuptake inhibitors (SRIs)?
Fluoxetine, Fluvoxamine, Paroxetine, Sertraline, Citalopram, Escitalopram
What is the treatment for Serotonin syndrome?
Discontinue SRI; Treat with benzodiazepines or 5-HT2A antagonists (cyproheptadine); Use short-acting drugs to control BP, and a non-depolarizing neuromuscular blocker with intubation for severe hyperthermia
What drug interactions are seen with SRIs?
Never use with MAOIs: SRI must be withdrawn for a least two weeks before MAOI use (and vice versa), combined use can result in Serotonin syndrome (autonomic instability, rigidity, fever, stupor, death) – there are also concerns with other drugs that enhance serotonergic transmission
What characterizes the pharmacokinetics of SRIs?
Good PO absorption, long half-life (more than a day), some produce longer-lived/active metabolites, oxidized by hepatic enzymes; some have complex pharmacokinetics (CYP inhibition)
What adverse effects are associated with SRIs?
Initial agitation/excitation, nausea, sexual dysfunction – fewer side effects relative to other antidepressants (low incidence of cardiovascular/anticholinergic effects); little risk of overdose
What is the primary method of inactivation of Serotonin?
Reuptake – thus the use of SRIs
What characterizes the SRIs?
First-line therapy in patients diagnosed with major depression; also used to treat OCD and some eating disorders – failure of one SRI does not signal failure on another; similar efficacy to other antidepressants (may be better compliance)
What characteristic do all antidepressants have in common?
They show a therapeutic lag: it takes a couple of weeks for their effects to become evident, and even longer to become maximal (~8wks) – side effects are immediate
What black box warning is associated with most antidepressants?
Increased “suicidality” – especially in children/adolescents; more prevalent in early treatment
What does the delay in the effects of antidepressants suggest?
That the patient undergoes a delayed response (regulation of receptors, transporters; neurogenesis) – however, acute effects appear essential
What characterizes Venlafaxine?
SNRI with non-CYP metabolism; First-line agent for depression, also used for anxiety disorders;
AEs: HTN, tachycardia, nausea, anorexia, drowsiness, dry mouth, sexual effects like SRIs;
CI: HTN (not absolute); DI: Serotonin syndrome (like SRIs)
What characterizes the pharmacokinetics of TCAs?
Good PO absorption, long half-life (more than a day), some produce longer-lived/active
What adverse effects are associated with TCAs?
Orthostatic hypotension: due to antagonism of α-adrenergic receptors; Anticholinergic effects: blurred vision, worsening of narrow-angle glaucoma, dry mouth, constipation, urinary retention, tachycardia, confusion; Sedation: antagonism of histamine and α-adrenergic receptors;
Cardiovascular effects: seen with overdose, arrhythmias, direct myocardial depression, worsening of CHF, can be lethal, limit access to drug; With bipolar illness: precipitation of manic episode
What is the mechanism of action of TCAs?
Inhibit reuptake of Serotonin/NE; potentiate and prolong the actions of these neurotransmitters; receptor and transporter regulation with repeated treatment
What characterizes the TCAs?
Similar efficacy to SRIs; have been supplanted in most cases by SRIs as first-line therapy;
Generally cause weight gain; Also used to treat certain types of pain, panic disorder, ADHD
What are the tricyclic antidepressants (TCAs)?
Amitriptyline, Clomipramine, Desipramine, Doxepin, Imipramine, Nortriptyline, Protriptyline, Trimipramine
What characterizes Duloxetine?
SNRI with CYP metabolism; Used in treatment of depression itself and for the treatment of physical pain associated with depression (useful in treatment of pain from diabetic neuropathy);
AEs: nausea, anorexia, drowsiness, constipation, dry mouth, rare hepatic injury;
CI: liver disease, alcoholism; DI: Serotonin syndrome and CYP interactions
What are some characteristics of the individual SRIs?
Fluoxetine: potent CYP inhibitor, insomnia/anxiety more common, long T½ , active metabolites;
Sertraline: diarrhea more common; Paroxetine: potent CYP inhibitor, anticholinergic (sedation, constipation), weight gain, sexual dysfunction, ↑ risk of cardiac birth defects in 1st trimester;
Fluvoxamine: used for OCD, potent CYP inhibitor; Citalopram: faster onset of action;
Escitalopram: active stereoisomer of citalopram, more potent/more selective/faster onset
What are the Serotonin-Norepinephrine reuptake inhibitors (SNRIs)?
Venlafaxine, Duloxetine
What are the effects of SRIs on the CYP enzymes?
Strong inhibitors: Fluoxetine, Fluvoxamine, Paroxetine;
Weak inhibitors: Sertraline, Citalopram, Escitalopram
What contraindications are associated with MAOIs?
Alcohol abuse, cardiovascular disease, HTN, cerebrovascular disease/bleeding, renal failure, children – use with extreme caution
What characterizes Bupropion?
Weak blocker of DA, Serotonin, NE uptake; Can cause agitation/anxiety/restlessness; Associated with a risk of seizure (administered as divided doses or slow release formulation); Also used as aid in quitting smoking – used in patients who can’t tolerate the sexual side effects of more common antidepressants
What are the atypical antipsychotics?
Bupropion, Mirtazapine, Trazodone, Nefazodone
What characterizes the reversible inhibitors of MAO (RIMAs)?
MAO-A inhibitors: more effective antidepressants; MAO-B: used with Parkinson’s (Selegiline) – not used clinically in the US, fewer drug/food interactions
What drug interactions are seen with MAOIs?
Food containing high amounts of tyramine, sympathomimetic drugs (cold remedies, diet aids, stimulants), SRIs, Meperidine and some other analgesics
What characterizes the pharmacokinetics of MAOIs?
Good PO absorption, daily dosing required (in spite of irreversible enzyme inhibition), inactivated by acetylation (caution in slow acetylators)
What adverse effects are associated with MAOIs?
Orthostatic hypotension, restlessness, insomnia, sometimes fatal hypertensive crisis (when combined with drugs like sympathomimetics, and with foods containing tyramine – strict monitoring required)
What drug interactions are seen with TCAs?
Interactions with alcohol and other sedatives, antiparkinson drugs, antipsychotic drugs, biogenic amines – also via competition for plasma protein binding
What characterizes the use of MAOIs?
Only used in patients who have failed courses of treatment with other antidepressants and for whom electroconvulsive therapy is not suitable – MAO levels remain low for at least two weeks after withdrawal of an MAOI (MAO inhibition is irreversible)
What are the MAOIs?
Phenelzine, Tranylcypromine
What adverse effects are seen with Trazodone, Nefazodone?
Drowsiness, dizziness, lightheadedness, dry mouth, priapism, Serotonin syndrome with serotonergic drugs
What atypical antipsychotics are used for the treatment of acute mania and long-term management?
Olanzapine, Risperidone, Quetiapine, Ziprasidone, Aripiprazole
What adverse effects are associated with Lithium?
Mild AEs: HA, fatigue, confusion, weakness, thirst, polyuria; Severe AEs: myoclonic jerks, seizures, impaired consciousness, coma, death (levels about 3.5 mEq/L are often fatal);
Cardiac effects: ECG irregularities, cardiac arrest; Fetal/lactational effects: Ebstein anomaly, fetal goiter, floppy baby syndrome, arrhythmias in nursing infants
What characterizes Lithium?
Mechanism of action poorly understood: inhibits inositol phosphate signalling, inhibits neurotransmitter-stimulated adenylyl cyclase; Low therapeutic index: monitor plasma concentrations; Renal clearance: sensitive to diuretics and anti-inflammatory drugs
What characterizes the pharmacotherapy of bipolar depression?
Mood stabilizers: Lithium, Carbamazepine, Valproate, other anticonvulsants, atypical antipsychotics; Acute anti-mania effects: benzodiazepines, antipsychotics
What are the stages of treatment of MDD?
Acute treatment (1-3 months): decrease symptoms; Continuation treatment (4-9 months): prevent relapse; Maintenance treatment (12+ months): used for high risk patients
What contraindications are associated with Bupropion?
Patient with seizure disorders, during lactation – can see Serotonin syndrome with serotonergic drugs
What characterizes Trazodone, Nefazodone?
Moderate inhibition of Serotonin reuptake, Serotonin-2 receptor antagonist – useful in the treatment of depression characterized by anxiety and sleep disturbances
What adverse effects are seen with Mirtazapine?
Drowsiness/fatigue (H1 receptor antagonist), appetite stimulation, weight gain, elevated blood lipids, rare blood dyscrasia; CI in severe liver disease and blood dyscrasia – do not use with MAOIs, watch for signs of neutropenia, caution due to sedation
What characterizes Mirtazapine?
Enhances the release of Serotonin and NE by antagonizing presynaptic α2-adrenergic receptors; also antagonizes 5-HT2/3 receptors – relative to SRIs, there is a reduced incidence of GI and sexual side effects
What characterizes the phases of treatment with bipolar illness?
Acute phase: control symptoms of mania (with an antipsychotic); Continuation phase: remission (mood stabilization with Lithium, anticonvulsants, atypical antipsychotics) and maintenance (lifetime therapy may be necessary)
What are the different ways in which the viral genome can be protected?
Via an icosahedral capsid or a helical capsid; and with or without a membrane present
What are the stages of the virus life cycle?
Initiation of infection: adsorption and penetration; Synthesis of viral proteins and genetic material: genome replication and transcription/translation of viral proteins; Maturation: assembly and release
What is the key difference between a virus containing +RNA and one containing –RNA?
The virus containing +RNA can immediately be translated by host ribosomes, whereas the virus containing only –RNA must first be transcribed to a +RNA by an RNA-dependent RNA polymerase before viral proteins can be synthesized
How does a virus containing a single strand of +RNA create many more of that same +RNA?
It uses an RNA-dependent RNA polymerase to create a –RNA (i.e. it is in 3` to 5` orientation instead of the normal 5` to 3` orientation); this –RNA serves as a template to create a large number of +RNA
What are the parameters for viral classification?
Nucleic acid structure and composition (e.g. RNA vs. DNA, ss vs. ds); Nucleocapsid symmetry (e.g. icosahedral vs. pleomorphic vs. filamentous); Presence/absence of an envelope
Which viral enzyme is an attractive drug target since the enzyme is not produced by humans?
RNA-dependent RNA polymerase (creates an mRNA transcript from RNA)
What structures are associated with viral membranes and what is their clinical significance?
Membrane-spanning glycoproteins that are specific to a particular virus and function in their adhesion to host cells; these glycoproteins can be the target of antibodies, thus preventing adhesion (e.g. hemoglutinin of influenza)
What determines the specificity of a virus?
The organ that contains the protein that is recognized by a particular virus
What are the functions of the
viral capsid?
Packaging/condensation; Protection of nucleic acid; Transport of nucleic acid from cell to cell; Provide specificity for attachment
What is a virion?
Viruses released from cells after the initial infection
What occurs during the eclipse period of the viral life cycle?
It replicates its genome
What characterizes the picornaviruses?
Icosahedral, no envelope present, ss-linear, nonsegmented, +RNA; includes poliovirus (gut/brain), rhinovirus (upper respiratory tract), Hepatitis A (liver), meningitis, hand-foot-mouth disease
Which type of RNA does not bring in the RNA-dependent RNA polymerase and why?
+RNA, since it can immediately be translated and start producing RNA-dependent RNA polymerase; –RNA has to bring the enzyme with it since host ribosomes cannot read the –RNA, so the initial +RNA will have to made by the virus itself
What are plaque assays used for?
To quantify the number of viral particles in a particular sample
What are the three main host responses in regards to viral particles?
Humoral response: antibodies; Cell mediated response: T-cells; Interferons: sense double-stranded RNA and block viral replication
What happens in certain viruses when the host cell reaches yield?
It bursts and releases all intracellular virions
What occurs at the end of the latent period of the viral life cycle?
Intracellular virions are released into the extracellular environment
By what two mechanisms can viral particles gain entry into the cell?
It can directly fuse with the host cell membrane (e.g. HIV); It can gain entry through a host endocytic vesicle (e.g. West Nile)
What is a mechanism by which virus assembly and release occurs?
Viral glycoproteins insert themselves into the host cell membrane --> viral matrix proteins associated with the intracellular portion of these glycoproteins --> when the viral genome comes in contact with the matrix proteins, it induces folding and budding of the viral particle (some host cell proteins may become part of the virus)
Why do viruses have to regulate what genes are transcribed when?
So that they produce their replication machinery first, before producing the capsid
What characterizes the flaviviruses?
Identical morphology to alphaviruses (icosahedral, envelope present, ss-linear, nonsegmented, +RNA), but different replication strategy; includes West Nile virus (transmitted by mosquitoes), Yellow fever (mosquitoes, liver), Dengue fever (hemorrhagic-type fever), Hepatitis C
What characterizes paramyxovirus?
Helical, envelope present, ss-linear, nonsegmented, –RNA; includes measles (very infectious, encephalitis), mumps (parotid gland, encephalitis, sterility), parainfluenza (croup), respiratory syncitial disease
What characterizes orthomyxovirus?
Helical, envelope present, ss-linear, segmented (8 strands), –RNA; includes influenza, common cold (influenza C)
What characterizes the retroviruses?
Icosahedral, envelope present, 2 strands of ss-linear +RNA with a reverse transcriptase attached; includes HIV, HTLV-I, HTLV-II
What characterizes the coronaviruses?
Helical, envelope present, ss-linear, nonsegmented, +RNA; includes SARS, common cold
What makes Hepatitis C a difficult virus to treat?
It has a RNA-dependent RNA polymerase that makes many mistakes, which means it has a high rate of mutation
What characterizes the calciviruses?
Icosahedral, no envelope preset, ss-linear, nonsegmented, +RNA; includes Norwalk virus (“cruise ship diarrhea, self-limting/acute), Hepatitis E
What characterizes the alphaviruses (arboviruses/togaviruses)?
Icosahedral, envelope present, ss-linear, nonsegmented, +RNA; includes Western equine encephalitis (also Venezuelan equine encephalitis and Eastern equine encephalitis, all are transmitted by mosquitoes), and Rubella virus (especially harmful in first trimester)
What characterizes the reoviruses?
Icosahedral, double-shelled capsid, no envelope present, ds-linear, segmented; includes reovirus, rotavirus (diarrhea), Colorado Tick Fever
Which class of viruses is very similar to the calciviruses and usually affects children?
Astroviruses
What characterizes the arenaviruses?
Helical, envelope present, 2 strands of circular –RNA, ambisense (contains both – and + RNA characteristics); includes Lassa fever (hemorrhagic), Lymphocytic choriomeningitis (hamsters)
What characterizes herpesviruses?
Icosahedral, envelope present, ds-linear, complex genome; includes Herpes I (cold sores), Herpes II (genital herpes), Herpes III (chicken pox, shingles), Herpes IV (Epstein-Barr, mononucleosis), Herpes V (roseola, child with fever/rash), Herpes VI-VIII (less characterized, although VIII is associated with Kaposi’s sarcoma)
What characterizes adenovirus?
Icosahedral with pentons, no envelope present, ds-linear; includes common cold
What characterizes papovaviruses?
Icosahedral, no envelope present, ds-circular DNA, supercoiled; includes papillomavirus (chondylomas, warts, cervical cancer)
What characterizes hepadnaviruses?
Icosahedral, envelope present, partially ds-circular DNA, spherical to filamentous, contain reverse transcriptase; includes Hepatitis B
What characterizes parvoviruses?
Icosahedral, no envelope present, ss-linear, +DNA; includes B19 (“slapped cheek disease”), Fifth disease
What characterizes bunyaviruses?
Helical, envelope present, 3 strands of circular –RNA, ambisense; includes Lacrosse fever (epilepsy-like symptoms), Rift valley fever, Hantavirus (Hanta pulmonary syndrome, upper respiratory tract)
What characterizes rhabdoviruses?
Helical, envelope present, ss-linear, nonsegmented, –RNA; includes rabies virus (slow virus)
What characterizes the filoviruses?
Helical, envelope present, ss-linear, nonsegmented, –RNA, filamentous virus; includes Ebola, Marburg virus (these are hemorrhagic)
What characterizes poxviruses?
Complex capsid structure, envelop present, ds-linear; includes smallpox, vaccinia, monkeypox
How is acute MI diagnosed?
With a diagnostic ECG (about half of patients) or if the patient has a non-diagnostic ECG, through the use of cardiac markers
What changes in cardiac Troponin are seen with unstable angina?
A small increase (peaks in the gray zone: 0.050-0499) that is not sufficient for a diagnosis of AMI
How is BNP metabolized?
It is rapidly removed from the blood through clearance receptors on the endothelial cells and degraded by neutral endopeptidases
What are the important natriuretic polypeptides and where are they produced?
Atrial natriuretic peptide (ANP): produced and stored in atria granules; B-type natriuretic peptide (BNP): produced in ventricles, but not stored – ANP/BNP secretion increases when the ventricular myocardium is overloaded (BNP is secreted in much greater quantities)
What is the purpose of natriuretic polypeptides?
They counter the effects of the renin-angiotensin-aldosterone system by acting on guanylyl cyclase A receptors and NPR (natriuretic polypeptide receptor)
What are the early and definitive markers for MI?
Early: myoglobin, CK-MB; Definitive: cTnT, cTnI
What are causes of elevations in Troponin levels?
Cardiac contusions, trauma (e.g. steering-wheel injuries), acute heart failure, pulmonary embolism, inflammatory diseases, extreme exertion
What can cause a false elevation of cTnI?
Heterophile antibodies; Rheumatoid factor; Human anti-mouse antibodies (as seen with some anti-cancer and anti-viral mouse monoclonal antibodies); and other animal antibodies (e.g. in people who work with animals)
How is myoglobin used to diagnose MI?
It is a small molecule that leaks easily out of damaged cells; this makes it very sensitive for MI, but not very specific – 1-3 hours: rise; 6-9 hours: peak; 12-24 hours: normal
What levels of cTnI are expected post-MI?
3-4 hours: rise; 24-48 hours: first peak (due to cytosolic pool); 72-100 hours: second peak (due to structural pool); 4-7 days: normal (10-14 days for cTnT)
What is the standard cardiac marker used for the diagnosis of MI?
Troponin: Troponin T and Troponin I have specific cardiac forms (cTnT, cTnI) whereas Troponin C is structurally identical in cardiac and skeletal muscle
What is hs-CRP?
High sensitivity C-reactive protein; it is an acute phase reactant released during acute inflammation (i.e. it is a marker of low-grade inflammation seen in unstable atherosclerotic plaques) – it is useful to help assess risk of an initial acute coronary event in apparently healthy individuals
What are the classes and levels defined in the NACB practice guidelines?
Class I: treatment/procedure should be used; Class II(a/b): reasonable/may be considered;
Class III: should not be used – each treatment/procedure also has a level associated with it:
Level A: recommendation based on multiple randomized trials/meta-analyses;
Level B: based on one randomized trial or non-randomized studies;
Level C: based on expert opinion, case studies
What is Myeloperoxidase?
An ischemia marker involved in the inflammatory process (released from neutrophils); may identify Troponin-negative patients at risk for MI
What does the Albumin Cobalt Binding Test detect?
Cobalt binding to Albumin, which is decreased in ischemia (due to Ischemia Modified Albumin – IMA); IMA rises within minutes of an ischemic event and returns to baseline within 6 hours; very effective when used in conjunction with Troponin
What is the clinical use of LP(a)?
Higher levels of LP(a) correlate with an increased risk of coronary heart disease; Niacin can be used to lower LP(a) levels
What is Lipoprotein(a) (LP(a))?
Acute phase reactant that promotes atherogenesis and thrombogenesis (via inhibition of fibrinolysis and by increasing cholesterol deposition in arterial walls, amongst other effects)
What drugs have an effect on hs-CRP?
Estrogen therapy increases hs-CRP, whereas low-dose aspirin and “statin” drugs decrease it
What is the precursor of BNP?
ProBNP; it is cleaved into NT-proBNP and BNP
What are the differences between BNP and NT-proBNP?
BNP has a shorter half-life, can detect a treatment response rapidly, is biologically active, and cannot be used to monitor response to Nesiritide therapy; both compounds will be increased in renal failure (due to increased secretion and slower removal)
What is the most important use of BNP, clinically?
It is a marker for the diagnosis and assessment of severity associated with CHF (NT-proBNP is similarly useful); It can be used in risk stratification for future cardiovascular events; It is a screening tool in patients with known risk factors; And, it can be used to monitor the effectiveness of drug therapy
What happens to CK-MB levels when a MI is reperfused?
A wash-out effect takes place where CK-MB levels rise rapidly, and return to normal more rapidly – in a patient who underwent reperfusion, infarct size cannot be assessed using CK-MB levels
What does the MB index elicit?
Whether an elevated CK is due to skeletal muscle injury (if value is < 2.0) or due to MI/necrosis (if value is > 5.0)
What are the causes of an elevated CK and what causes an elevated CK-MB, specifically?
Elevated CK: acute MI, heart surgery/trauma, skeletal muscle breakdown (for various reasons);
Elevated CK-MB: acute MI, cardiac ischemia, chronic renal failure, heart surgery/trauma, electrical injury, extreme exercise, muscle disease (polymyositis, Duchenne’s MD)
What levels of CK are expected post-MI?
4-6 hours: rise; 22-24 hours: peak; 48-72 hours: normal
How is Creatinine Kinase (CK) measured?
By immunoassay (called mass CK-MB)
How soon should thrombolytic treatment be administered for maximal benefit?
Within the first 2 hours post-MI (since myocardial necrosis begins within 30 minutes)
What is acute coronary syndrome (ACS) and what are the two associated categories?
Symptoms due to acute myocardial ischemia – STEMI: ST elevation MI; NSTEACS: non-ST elevation ACS, consists of NSTEMI (MI with myocyte necrosis) and UA (unstable angina, no necrosis)
What ECG changes are seen with MI?
ST segment elevation (hours after MI), inverted T waves (hours to days), enlarged Q waves (hours to days) – in days to weeks, ST segment returns to near normal; in weeks to months, the T waves becomes upright again, but the large Q wave may remain (and the R wave may disappear)
What describes the “textbook” MI?
Male, >65, chest pain
What treatments/procedures are considered Class I in regards to the diagnosis of MI?
In conjunction with the clinical history and ECG, cardiac Troponin is the preferred marker (although CK-MB may be used when Troponin is not available); Troponin should be measured on presentation and during the 6-9 hour period (need >99th percentile = >0.500)
With what patient are natriuretic peptides considered Class I?
The patient who present with SOB of unclear cause
What treatments/procedures are considered Class I/IIa/III in early risk stratification?
Class I: cardiac Troponin by serial sampling; Class IIa: hs-CRP/BNP;
Class III: the use of biomarkers in patients with a low clinical probability of ACS
What treatments/procedures are considered Class III in regards to the diagnosis of MI?
Total CK, AST, LDH; also in patients with a diagnostic ECG, they should be treated immediately instead of waiting for cardiac marker tests to come back
What treatments/procedures are considered Class IIa/b in regards to the diagnosis of MI?
Class IIa: Myoglobin; Class IIb: frequent early sampling of markers (e.g. every 2-3 hours)
What pathological mechanisms are associated with parasites?
Toxic products: hydrolytic enzymes, proteinases, collagenases, endotoxins; Mechanical tissue damage: blockage of internal organs, pressure atrophy, migration through tissue (larval stage);
Immunopathology (damage done by our immune system)
Which amoebae can be pathologic to humans?
Parasitic: Entamoeba histolytica; Free-living: Acanthamoeba, Naegleria (all have pseudopodia)
What are the four medically-important protozoa?
Amoebae, Flagellates, Ciliates, Sporozoans/Apicomplexans
What characterizes the protozoa?
Unicellular eukaryotes; Have organelles which serve in locomotion (used for categorization); Glucose is their chief source of energy and ATP (glycolysis, Krebs cycle)
What are the two main groups that parasites can be divided
up in?
Protozoa, Helminths
What evasion mechanisms of host defenses are employed by parasites?
Antigenic variation; Molecular mimicry; Concealment of antigenic site (masking); Intracellular location; Immunosuppression
What immunopathologic responses are associated with parasites?
Anaphylactic shock, local inflammation; Lysis of cell bearing microbial antigens (e.g. RBCs); Immune complex deposition; Chronic inflammation, mononuclear accumulation, macrophage activation
What occurs in the definitive host and what in the intermediate host?
Definitive host (human): the parasite passes its adult existence and/or sexual reproductive phase;
Intermediate host: animal in which a parasite passes its larval stage or asexual reproductive phase
What are the ports of entry by which parasites gain access to their host?
Ingestion by host; Direct penetration: arthropod bite, transplacental penetration, organism-directed penetration (e.g. penetration through intact skin)
What is the difference between the direct and indirect life cycle of parasites?
Direct: involves one host that passes cysts in feces which then contaminate food/water sources;
Indirect: involves multiple hosts, in which the definitive host passes eggs in feces which produce larvae, which are subsequently eaten by the intermediate host (this intermediate host can then be eaten by other intermediate hosts), which is then eaten by the host
What are reservoirs associated with amoebae and what symptoms are often seen with an infection of amoebae?
Reservoirs: humans and animals; Symptoms: usually diarrhea
What are the stages associated with Leishmania?
Promastigote stage (infective stage): flagellated, transmitted from insect to human; Amastigote stage (diagnostic): non-flagellated, found in tissue
What stage can certain types of Trypanosoma be found in when they enter the tissue?
Amastigote stage (diagnostic stage): non-flagellated
What are the stages associated with Trypanosoma?
Trypomastigote (infective and diagnostic stage): flagellated, found in blood/lymph, transmitted from insect to human, back to insect
How are hemoflagellates transmitted into their host?
Via an arthropod vector (specific arthropod varies between genera)
What stages are associated with the intestinal/urogenital flagellates?
Trophozoite and cyst stages; although Trichomonas vaginalis only has a trophozoite stage
What characterizes the flagellates and what main categories are they separated in to?
They have flagella – Intestinal/urogenital flagellates: Giardia lamblia, Trichomonas vaginalis;
Hemoflagellates: Trypanosoma brucei, Trypanosoma cruzi, Leishmania species
What are the stages of amoebae?
Trophozoite (troph): vegetative state of organism (active/feeding/motile), pseudopodia are temporary cytoplasmic extensions, very fragile organisms; Cyst: nonfeeding/nonmotile, stage infective for humans, passed from host in feces and can remain active for long periods of time
How do the free-living amoebae gain access to their host?
Through direct penetration: Naegleria in a host with meningeal encephalitis, and Acanthamoeba in a host with keratitis
What happens when an amoeba in the cyst stage is consumed by an appropriate host?
It becomes a trophozoite – the trophozoite will once again convert to a cyst when the required signals are present (i.e. it is close to exiting the intestinal tract)
What characterizes sporozoans/apicomplexans and what are some examples of associated species?
They are intracellular parasites; Contain an apical complex for penetration into the host cell;
Life cycle consists of a sexual phase (sporogony) and an asexual phase (schizogony);
Examples include Plasmodium species and Cryptosporidium species
What reservoirs are associated with Sporozoans?
Plasmodium: humans, mosquitoes; Cryptosporidium: humans, animals
What symptoms are associated with a Cryptosporidium infection?
In healthy individuals, usually little to none (possible self-limiting diarrhea); but in immunocompromised patients they can have a life-threatening diarrhea
What describes the life cycle of Cryptosporidium?
It is direct: fecal contamination and ingestion of oocyst --> sexual part of life cycle in intestinal wall --> asexual part of life cycle in lumen --> oocyst elimination in fecal material
What are the diagnostic stages associated with Plasmodium?
In blood: trophozoite, schizont (merozoites), gametocyte; Exoerythrocytic: schizont in liver (merozoite) – merozoites reproduce in schizonts and are released when the schizont bursts (this is what causes the symptoms of fever, chills, sweating in malaria)
What are the names of the stages associated with Plasmodium that transmits from insect to human, and vice versa?
Insect to human: sporozoite; Human to insect: gametocyte
What vector is associated with Plasmodium and what characterizes its metabolism?
Vector: female Anopheles mosquito; Metabolism: facultative anaerobe, degradation of glucose to lactate, usually no pentose phosphate pathway, use host hemoglobin as a source of nutrition, cannot assimilate folic acid
What reservoirs are associated with flagellates/hemoflagellates?
Humans, animals, arthropods
What is the only ciliate that is pathological to humans and what characterizes it?
Balantidium coli: possess cilia for a pulsing locomotion, goes through trophozoite and cyst stages, associated with fecal contamination, associated with pigs
What modes of transmission are associated with Nematodes?
Ingestion of eggs: Ascaris, pinworm, whipworm; Penetration of larvae: Hook worm, Strongyloides; Ingestion of larvae: Trichinella
What is the first intermediate host in the life cycle of trematodes?
Snails (a miracidium is released from the host and taken up by the snail --> inside the snail it is known as a sporocyst/redia --> once eliminated from the snail, it is known as a cercaria --> the cercaria can live on vegetation or enter a second intermediate host before returning to the definitive host) – in schistosomes, the cercaria do not require vegetation or a second intermediate host, but can penetrate the definitive host directly
What differentiates the hermaphroditic trematodes from the schistosomes?
Hermaphroditic: associated with a single organ system (intestine, liver, lung), complex life cycle;
Schistosomes: blood flukes, associated with the veins of a particular organ (intestine, bladder), complex life cycle
What characterizes the flukes?
Dorsoventrally flattened, non-segmented, leaf-/lancet-shaped, smaller in size; They have 2 suckers: oral sucker and ventral sucker (acetabulum, used to attach itself to the host);
Outer covering is called tegument: very absorptive for nutrient acquisition
What pathology/symptoms are associated with filarial worms and what is the treatment?
Blockage of the lymphatics (elephantiasis when chronic); Treatment: Diethylcarbamazine and Ivermectin (microfilariae), prevention – there is no treatment for adult worms
What characterizes the filarial worms?
Arthropod borne (mosquito, fly, midge): varies from worm to worm; Stages found in human: adult and microfilariae (larvae found in blood, produced after sexual reproduction); Often found in tropical locations (geographic distribution can be very diagnostic)
What characterizes the roundworms?
Direct life cycle; Widespread geographical distribution; Pathology/symptoms are associated with adult and larval stages; Prevention is stressed
What are the three different classes of Helminths
Nematodes: roundworms and filarial worms; Trematodes: flukes; Cestodes: tapeworms
What are the three stage associated with the Nematodes?
Adult stage; Egg stage (contains larvae); Larvae stage (once they hatch out of the egg)
Which are the most complex class of Helminths and what describes them?
Nematodes: round in cross-section, complete digestive tract, have separate sexes with male/female reproductive organs
What modes of transmission are associated with Nematodes?
Ingestion of eggs: Ascaris, pinworm, whipworm; Penetration of larvae: Hook worm, Strongyloides; Ingestion of larvae: Trichinella
What is the first intermediate host in the life cycle of trematodes?
Snails (a miracidium is released from the host and taken up by the snail --> inside the snail it is known as a sporocyst/redia --> once eliminated from the snail, it is known as a cercaria --> the cercaria can live on vegetation or enter a second intermediate host before returning to the definitive host) – in schistosomes, the cercaria do not require vegetation or a second intermediate host, but can penetrate the definitive host directly
What differentiates the hermaphroditic trematodes from the schistosomes?
Hermaphroditic: associated with a single organ system (intestine, liver, lung), complex life cycle;
Schistosomes: blood flukes, associated with the veins of a particular organ (intestine, bladder), complex life cycle
What characterizes the flukes?
Dorsoventrally flattened, non-segmented, leaf-/lancet-shaped, smaller in size; They have 2 suckers: oral sucker and ventral sucker (acetabulum, used to attach itself to the host);
Outer covering is called tegument: very absorptive for nutrient acquisition
What pathology/symptoms are associated with filarial worms and what is the treatment?
Blockage of the lymphatics (elephantiasis when chronic); Treatment: Diethylcarbamazine and Ivermectin (microfilariae), prevention – there is no treatment for adult worms
What characterizes the filarial worms?
Arthropod borne (mosquito, fly, midge): varies from worm to worm; Stages found in human: adult and microfilariae (larvae found in blood, produced after sexual reproduction); Often found in tropical locations (geographic distribution can be very diagnostic)
What characterizes the roundworms?
Direct life cycle; Widespread geographical distribution; Pathology/symptoms are associated with adult and larval stages; Prevention is stressed
What are the three different classes of Helminths
Nematodes: roundworms and filarial worms; Trematodes: flukes; Cestodes: tapeworms
What are the three stage associated with the Nematodes?
Adult stage; Egg stage (contains larvae); Larvae stage (once they hatch out of the egg)
Which are the most complex class of Helminths and what describes them?
Nematodes: round in cross-section, complete digestive tract, have separate sexes with male/female reproductive organs
What is the geographical distribution, pathology/symptoms, and treatment of cestodes?
Geographical distribution is widespread; Pathology/symptoms: blockage, diarrhea, constipation, pernicious anemia (with Diphyllobothrium latum, since it absorbs Vitamin B12);
Treatment: praziquantel, niclosamide, prevention (cooking meat thoroughly)
What are virulence factors associated with adherence/colonization?
Pili; Motility/chemotaxis (flagella); Outer membrane proteins (for adhesion/invasion)
What are some examples of tissue damaging/cytolytic exotoxins?
Tissue damaging: enzymes (proteases, DNases); Phospholipase C (with C. perfringens);
Surfactant (with S. aureus); Cholesterol-dependent cytolysins (with Streptolysin O)
What are diseases caused by exotoxins?
Diphtheria, Tetanus, Botulism, Anthrax, Cholera, Pertussis (“whooping cough”), Gas gangrene, Toxic Shock Syndrome, Enterohemorrhagic E. coli, Necrotizing fasciitis (“flesh-eating bacteria”)
How does the cell wall of Gram– organisms activate the immune systems?
Via LPS which functions as an endotoxin (PAMP interacting with a TLR)
How does the cell wall of Gram+ organisms activate the immune system?
The lipoteichoic acid found on the cell wall functions as a pathogen-associated molecular pattern (PAMP) which activates toll-like receptors (TLRs) of the innate immune system
What are virulence factors associated with the infectious process?
Cell wall/endotoxin; Exotoxins; Secretion systems (type III); Intracellular growth
What are the three types of bacterial-host interactions?
Symbiotic (both benefit), commensal (association that causes no harm), parasitic (association where one organism gains benefit from the other without compensation)
What are the types of disease caused by bacteria?
Localized infection produces toxins that are release systemically (e.g. Clostridium tetani);
Disseminated infection in which organisms themselves invade the blood stream (Staph aureus);
Intoxication in which spores multiply and produce toxin outside of the body, which is subsequently ingested (e.g. Clostridium botulinum)
By what two mechanisms can bacteria cause disease?
Release of factors (cell wall/endotoxins), direct host cell manipulation (intracellular/extracellular)
What is the measurement used to check virulence?
ID50: number of bacteria necessary to infect 50% of animals exposed to bacterium
What are the Type II secretion effectors in Gram– bacteria?
AB toxins, proteases
How is the function of microbial communities in biofilm analyzed?
Via metagenomics: analyze complete genetic information of community using pyrosequencing
What technique is used in research to determine to composition of a particular biofilm?
Bacterial 16S RNA sequences
What are examples of biofilm infections?)
Chronic infections, catheter-related infections, soft contact lens infections, dental carries/ periodontal disease (most common disease in humans
What is a biofilm composed of?
Living, 3-D community of microorganisms, adherent to a surface and enclosed within a polysaccharide matrix (highly resistant to antibiotics)
What are some examples of ways that bacteria protect against host defenses?
Antigenic variation (e.g. of pili), capsule (highly antigenic protective shell – used in vaccines), biofilm formation
What molecule is often used by bacteria to get into host cell and sometime to move around inside the cell?
Actin
What are some examples of cell receptor interaction toxins?
Heat-stable toxins (e.g. from E. coli): activate guanylate cyclase which causes an increase in cGMP  this creates an ion flux which causes diarrhea; Superantigens (e.g. Toxic Shock Syndrome)
How do the Type III secretion effectors differ from the AB toxins?
They do not have a leader sequence – they do contain a GAP protein which is a GTPase-activating protein (can turn host G-proteins on and off)
What is the largest group of exotoxins and how do they function?
AB toxins: consist of an A subunit (contains a leader sequence which moves the protein outside of the bacterial cell, a particular enzyme, and a signal sequence that traffics the protein to a location inside the target cell) and a B subunit (contains a receptor binding region and internalization domain) – both subunits are required for toxicity to occur
What are examples of transmissible spongiform encephalopathies in animals?
Scrapie, Bovine spongiform encephalopathy (mad cow disease), Chronic wasting disease of deer and elk
What genetic characteristic do people who have developed vCJD in common?
They have all been homozygous for the methionine amino acid (pos. 129) of PrPc due to polymorphisms in the PRNP gene (found in 40% of population)
How does brain tissue differ between patients with sCJD and vCJD?
In sCJD there is fine spongiform degeneration that is evenly distributed, whereas in vCJD the spongiform degeneration is clustered around a plaque surrounded by a vacuole (“florid”/”daisy” plaques)
How can vCJD be transmitted?
Via blood and blood products (which is why people who have visited the UK can’t donate blood), whereas conventional/sporadic CJD (sCJD) cannot
By what mechanisms can iatrogenic CJD come about?
Improperly sterilized equipment, corneal/dura matter transplant, contaminated human growth hormone (not an issue anymore)
What are examples of transmissible spongiform encephalopathies in humans?
Kuru (ceremonial cannibalism), Creutzfeldt-Jakob disease (CJD), variant CJD (vCJD, human form of Mad cow disease)
What is the pathogenesis of transmissible spongiform encephalopathy?
Uniformly fatal disease of CNS with a long incubation period, characterized by development of vacuolization of neurons and accumulation of amyloid-like plaques
How are transmissible spongiform encephalopathies most commonly diagnosed?
On autopsy, by finding the characteristic “spongy” brain appearance
What symptoms are associated with transmissible spongiform encephalopathy?
Loss of muscle control, shivering, dementia, tremors
What are some key differences between sCJD and vCJD?
vCJD is found in young adults (vs. older people in sCJD), the median duration of the illness is much longer, and it can be found in the lymphatics and blood (not seen in sCJD)
What are some key differences between sCJD and vCJD?
vCJD is found in young adults (vs. older people in sCJD), the median duration of the illness is much longer, and it can be found in the lymphatics and blood (not seen in sCJD)
What are some key differences between prions and viruses?
Prions do not contain nucleic acid and they do not have a defined morphology on EM; they are also more difficult to kill in sterilization procedures (i.e. they are not sensitive to ionizing radiation, formaldehyde, and steam heat)
What is the concern regarding chronic wasting disease in deer?
The possibility of transmitting this prion disease across species barriers to infect humans who hunt and eat deer
What is the proposed mechanism by which PrPsc can infect the brain after ingestion?
They travel to Peyer’s patches in the intestine where the follicular dendritic cells contain PrPc; here they convert those normal prions to PrPsc at which point they propagate via nerve fibers in order to enter the brain
What is the proposed mechanism for prior replication?
PrPsc has enzymatic activity by which it converts normal prions into PrPsc; these PrPscs then accumulate in the neuronal cytoplasm and cause neuronal death
What are some key differences between normal and aberrant prion proteins?
Aberrant prion proteins (PrPsc)have a globular structure (with characteristic β-pleated sheets) that is resistant to protease degradation, are found in the cytoplasm (vs. plasma membrane) and turn over in days (vs. hours for normal prions)
How is the diagnosis of a Babesia microti infection made and what is the treatment?
By Giemsa-stained thin and thick smears (or by Wright stain) of blood;
Treatment: clindamycin/oral quinine or azithromycin/quinine (or atovaquone)
What geographical distribution is associated with Leishmania species and what are its reservoirs?
Mediterranean, SW Asia, central/NW Africa, central/S America – reservoirs: human, rodents, dogs, other animals
What class of parasites do Leishmania species belong to and what are its definitive/intermediate hosts?
Hemoflagellates – the intermediate host is an arthropod (e.g. sand fly) that injects the flagellated promastigote into the definitive host during a blood meal; once inside the human host, the promastigote is ingested by macrophages at the bite site and converted to an amastigote (loses its flagella)
How is the diagnosis of a Toxoplasma gondii infection made and what is the treatment?
↑ IgM titer to Toxoplasma gondii; also, appearance of neurological symptoms, multiple ring-enhancing lesions on CT/MRI, detection of tachyzoites in brain biopsy using fluorescently-labeled antitoxoplasma antibodies – Treatment: pyrimethamine with either sulfadiazine/clindamycin
What pathology/symptoms are associated with Toxoplasma gondii?
Immunocompetent: usually asymptomatic or mild mononucleosis-like syndrome; Immunocompromised: encephalitis with severe tissue damage in brain, lesions in heart/lungs;
Developing fetus: stillbirth, hydrocephalus, chorioretinitis, liver damage, deafness, MR
By what three ways can humans become infected by Toxoplasma gondii?
Ingestion of oocysts in cat feces; Ingestion of bradyzoites in undercooked/rare meat; Tachyzoites cross placenta of primarily-infected mother during pregnancy (not an issue with bradyzoites)
What class of parasites does Toxoplasma gondii belong to and what are its definitive/intermediate hosts?
Coccidian (sporozoans) – Definitive host: feline, sexual life cycle in gut --> release of oocyst;
Intermediate host: other vertebrates (incl. humans) --> tachyzoite stage in tissue/fluids --> formation of cysts containing bradyzoites in tissue (after the immune system has walled it off)
What class of parasites does Babesia microti belong to and what are its definitive/intermediate hosts?
Sporozoans – Definitive host: Ixodes tick (deer tick), sexual life cycle;
Intermediate host: White-footed mouse, human (accidental), asexual life cycle
What pathology/symptoms are associated with Babesia microti?
In the immunocompetent: asymptomatic/mild flu-like illness, parasitemia <2%;
Splenectomized person: parasitemia = 10%, symptoms include HA/fatigue/fever/shaking chills/ joint and muscle aches/hemolytic anemia (pathology due to infection and lysis of RBCs)
What is the geographical distribution associated with Babesia microti?
Northeast, Pacific coast, Missouri, Georgia, Wisconsin
What class of parasites does Trypanosoma cruzi belong to and what are its definitive/intermediate hosts?
Flagellate – the intermediate host is the reduviid bug (Triatoma) that releases the trypomastigote in its feces while taking a blood meal on the definitive host (usually on the face around moist areas)
What class of parasites does Trypanosoma brucei belong to and what are its definitive/intermediate hosts?
Flagellate – the intermediate host is the tsetse fly (Glossina) that transmits trypomastigotes from its saliva as it takes its blood meal; the definitive hosts include humans, wild game, cattle
What is the treatment for Trypanosoma cruzi?
Early: nifurtimox or benznidazole; Chronic: treat symptoms (e.g. for heart condition, megacolon)
How is the diagnosis of Trypanosoma cruzi made?
Early: check for trypomastigotes in blood or through xenodiagnosis (let reduviid bug take a blood meal on a potentially infected patient, then observe the bug over the following weeks for changes);
Later: amastigotes in tissue or serological tests for antibodies to T. cruzi
What pathology/symptoms are associated with Trypanosoma cruzi?
Chagas’ disease --> Chagoma (chancre) at site of bite; Acute phase: fever, weakness, enlarged lymph nodes/liver/spleen with a Romaña’s sign (unilateral swelling of the face, if bitten on face, especially seen in children; Chronic phase: destruction of ganglia of the ANS (megacolon, megaesophagus, can be fatal, also possible to see chronic heart disease in young adults)
How are Trypanosoma cruzi and Trypanosoma brucei distinguished in a blood smear?
T. cruzi is small and will form C-shapes in the blood, whereas the larger T. brucei forms S-shapes
What is the geographical distribution of Trypanosoma cruzi?
Central/S America, maybe southern US
What are the two forms of Leishmaniasis?
Cutaneous Leishmaniasis: L. tropica, L. major, L. mexicana, L. brasiliensis;
Visceral Leishmaniasis: L. tropica, L. donovani (bone marrow)
How is the diagnosis of Leishmaniasis made and what is the treatment?
Biopsy of skin lesion/bone marrow/lymph node followed by Giemsa stain or PCR to look for amastigotes in macrophages – Treatment: antimony-containing compounds (sodium stibogluconate or meglumine antimonate)
What pathology/symptoms are associated with the two forms of Leishmaniasis?
Cutaneous: dry/long-lasting lesion (12-18 months), moist/short-term lesion (3-6 months);
Visceral: cutaneous lesion followed by chills, fever, sweating, diarrhea, weight loss, splenomegaly, hepatomegaly, hyperplasia of tissue/organs, anemia, death within 2 years
What are the causative agents of lymphatic filariasis and what is their geographical distribution?
Nematodes --> Wuchereria bancrofti: tropic/subtropics; Brugia malayi: Asia and South Pacific
What is the geographical distribution of Onchocerca volvulus?
Mostly in central Africa, but also some foci in central/South America
What class of parasites does Onchocerca volvulus belong to and what are its definitive/intermediate hosts?
Nematodes – Arthropod intermediate host (Simulium fly/black fly) transmits infective larvae into definitive host (human) during blood meal; Simulium fly prefers to breed near fast flowing water
How is the diagnosis of lymphatic filariasis made and what is the treatment?
Detection of microfilariae in blood smears using Giemsa/Wright stain (blood sample taken at night since this is when microfilariae are released) – Treatment: diethylcarbamazine/ivermectin (for microfilariae, not adults), vector control (kill mosquitoes)
What pathology/symptoms are associated with lymphatic filariasis?
Early: low grade fever and inflammation of lymphatics/lymph nodes due to adult worms;
Late: lymphatics become occluded, fluids leak into tissues, severe swelling --> elephantiasis
What distinguishes between Wuchereria bancrofti and Brugia malayi, morphologically?
Wuchereria bancrofti: nuclei end before the tail tip; Brugia malayi: nuclei extend into the tail tip
What intermediate host is associated with lymphatic filariasis?
Arthropods: Anopheles/Culex/Aedes mosquitoes (transmit the infective larvae during blood meal)
What is the geographical distribution of Trypanosoma brucei?
African continent --> western part: T.b. gambiense (less virulent); eastern part: T.b. rhodesiense (more virulent)
How is the diagnosis of Trypanosoma brucei made and what is the treatment?
Detection of trypomastigote in blood or CSF by Giemsa stain; Treatment --> blood: pentamidine isethionate or suramin; CSF: melarsoprol or eflornithine
What pathology/symptoms are associated with Trypanosoma brucei?
African sleeping sickness --> Initially, high fever and swollen postcervical lymph nodes (Winterbottom’s sign) every 10 days to 2 weeks (due to antigenic switching of variable surface glycoprotein); Once the parasite gets into the CNS, patients will suffer from meningitis, lethargy, motor changes, staggered gait, loss of balance, coma, and eventually death
What class of parasites does Schistosoma mansoni belong to and what are its definitive/intermediate hosts?
Trematode (blood fluke, schistosomes – not hermaphroditic); the intermediate host is the fresh water snail which releases free-swimming cercariae that then penetrate intact skin of the definitive host (human); once in the host, the flukes resides in the veins of the large intestine
What pathology/symptoms are associated with Schistosoma hematobium infection?
Onset of symptoms is delayed: painful urination, hematuria; chronic infection is correlated with bladder cancer
What is the geographic distribution of Schistosoma hematobium?
Africa and Middle East
What class of parasites does Schistosoma hematobium belong to and what are its definitive/intermediate hosts?
Trematode – same life cycle as Schistosoma mansoni, although S. hematobium has a predilection for the veins of the bladder, not the large intestine
How is the diagnosis of Schistosoma mansoni infection made and what is the treatment?
Finding eggs in stool exam or biopsied tissue (eggs are oval with large lateral spine);
Treatment: praziquantel
What pathology/symptoms are associated with a Schistosoma mansoni infection?
Initially: rash at the site of cercarial entry a few days after penetration (ground itch);
4-6 weeks later: Schistosomiasis, Katayama fever --> Typhoid fever-like symptoms (cough, malaise, myalgias), hepatosplenomegaly, cirrhosis of the liver (with chronic infection), bloody diarrhea, hypertension, toxic reactions due to granuloma formation around eggs (all of the symptoms are in response to the eggs, not the adults)
What is the geographical distribution of Schistosoma mansoni?
Africa, South/Central America, Caribbean area (incl. Puerto Rico and the West Indies)
Where in the human body are the adult worms and microfilariae of Onchocerca volvulus found?
Adults: subcutaneous tissue in nodules; Microfilariae: in skin
How is the diagnosis of Onchocerca volvulus infection made and what is the treatment?
Detection of microfilariae in skin snip; Treatment: remove skin nodules to kill adults, give ivermectin to kill microfilariae, also vector control
What pathology/symptoms are associated with Onchocerca volvulus?
Skin nodules where the adult reside; females travel through the skin causing chronic dermatitis, severe itching, and discoloration of the skin; Migrating microfilariae cause blindness (thus the name river blindness)
How is the diagnosis of Schistosoma hematobium infection made and what is the treatment?
Detection of eggs in sedimented urine or bladder biopsy (eggs are oval with a large terminal spine, peak egg excretion is noon to 3pm); Treatment: praziquantel
What is the rate-limiting step in the phosphorylation of Acyclovir?
The first phosphorylation, performed by herpes thymidine kinase (for HSV, Varicella Zoster Virus) or by phosphotransferase (CMV)
What adverse effects are associated with Ganciclovir?
Bone marrow suppression (!): neutropenia, anemia, thrombocytopenia; Renal insufficiency (uncommon); Contraindicated in pregnancy (teratogenic) – if used for more than 3 months, 10% of patients will excrete resistant strains; More toxic than Acyclovir
What is the “gold standard” for CMV?
Ganciclovir
What adverse effects are associated with Acyclovir?
n/v/d, nephrotoxicity (especially when given IV and in high doses, due to crystallization), HA, CNS effects/hallucinations/seizures – category B in pregnancy
If a patient has a strain of herpesvirus that is resistant to Acyclovir and Ganciclovir due to resistance in thymidine kinase, what alternatives can be given?
Foscarnet and Cidofovir
What are the inhibitory concentrations of Acyclovir regarding the different herpes viruses?
HSV-1 < HSV-2 < VZV < CMV (the dose needs to go up considerably to treat VZV and the dose to treat CMV would be too high to tolerate)
Although Acyclovir resolves symptoms and prevents shedding of the virus, what does it not do?
It does not terminate herpesvirus latency
What is the “gold standard” antiviral for herpes simplex viruses?
Acyclovir
What mechanism of action do most anti-herpes virus agents have in common?
They mimic natural deoxynucleosides (purines, esp. guanine) and in this fashion become phosphorylated and inhibit DNA polymerase, stopping viral DNA synthesis
What are the anti-herpes virus agents and which are effective
against CMV?
Acyclovir, Valacyclovir, Famciclovir, Ganciclovir, Foscarnet, Cidofovir, Valganciclovir (the latter 4 are effective against CMV) – Famciclovir has the greatest bioavailability (78%) and Acyclovir has a bioavailability of 20-30%
What is the mechanism of action of Foscarnet?
It is a pyrophosphate compound that functions as a non-competitive inhibitor of viral RNA and DNA polymerases
What viral surface proteins define the strain of the virus?
Neuraminidases, Hemagglutinins
What is the treatment for VZV?
Normal adult: Acyclovir, Valacyclovir; Immunocompromised: higher dose Acyclovir
What adverse effects are associated with Cidofovir?
Nephrotoxicity (administer with probenecid and saline hydration), neutropenia, uveitis, decreased intraocular pressure, neuropathy
Besides Foscarnet, what other drug can be used in the treatment of acyclovir/ganciclovir-resistant strains and what is its mechanism of action?
Cidofovir (nucleotide monophosphate): it is somewhat similar to acyclovir/ganciclovir but it already has the first phosphate (rate-limiting) attached, so it will function in resistant stains
What adverse effects are associated with Foscarnet?
Severe nephrotoxicity, hypocalcemia, hypo-/hyperphosphatemia, hypomagnesemia, hypokalemia, n/v, penile ulcers (unique)
What strains of herpesvirus are affected and why?
Since it does not have to be phosphorylated by thymidine kinase or phosphotransferase, it affects all herpesviruses (HSV, CMV, VZV) and even most acyclovir/ganciclovir-resistant strains
Why is oral Ganciclovir not used much anymore?
Its bioavailability is 5-10%, so the pro-drug Valganciclovir is used instead (bioavailability 60%, used in the treatment of several types of CMV)
What is special about the dosing of Ganciclovir?
There is an initiation dose (high) and a consolidation dose (lower) – also requires renal adjustment
What are the approved indications for Ganciclovir?
CMV retinitis, prevention of CMV in transplant patients
In what case would influenza antivirals not be recommended?
If a patient recently had a live flu virus vaccine; this is not an issue with killed vaccine
What are the selective neuraminidase inhibitors?
Oseltamivir, Zanamivir – both are indicated for the prophylaxis and treatment of influenza; they both have action on Type A and B influenza
What is the purpose of neuraminidase?
It allows the viral particles to break free from the host cell in which they were synthesized;
It prevents aggregation of newly formed viral particles by removing sialic acid on the viral particles and the infected cells; It allows for penetration of the respiratory tract
What class of drugs is used currently for influenza infections?
Neuraminidase inhibitors:
What adverse effects are associated with Amantadine and Rimantadine?
Amantadine: (90% excreted in urine) n/v, CNS (>5-10%), insomnia, altered concentration, seizures, suicidal ideation; Rimantadine: (20% excreted in urine), n/v, CNS (<3-6%), insomnia, altered concentration, seizures – contraindicated in pregnancy
What are the effects of giving someone an influenza antiviral?
Post-exposure: decrease symptom duration by 1-2 days (and decrease viral shedding);
Pre-exposure: 70% decrease in symptomatic influenza infection
What structures, found on the surface of cell membranes (especially RBCs and cell membranes of the upper respiratory tract), are recognized by the hemagglutinins?
Sialic acid
What is the mechanism of action of the influenza antivirals Amantadine and Rimantadine?
They inhibit the M2 ion channel functioning, thus blocking the uncoating of the viral genome and viral entry – active against Influenza Type A only (since M2 ion channel is not as important in Types B and C)
What are the different types of influenza viruses classified as?
Type A: potentially severe illness, cause of epidemics/pandemics, rapidly changing form;
Type B: usually less severe illness, has caused some epidemics, more uniform antigens;
Type C: Usually mild/asymptomatic illness, minimal public health impact
What is responsible for influenza’s year-to-year bypassing of human immunity?
Antigenic drift: makeup of the hemagglutinin changes every year so antibodies become ineffective
What is the difference in dosing regarding Oseltamivir and Zanamivir?
Oseltamivir is an oral dosing whereas Zanamivir is inhaled
Which influenza type has been responsible for the avian flu?
Type A, H5N1
When should neuraminidase inhibitors be given for the treatment of influenza?
Within 1-2 days after the onset of symptoms (if given at this time, there is a reduction in the duration of uncomplicated illness by 1-2 days)
What adverse effects are often seen with dry powder inhalers (i.e. Zanamivir)?
Cough and bronchospasm (watch in patients with COPD, asthma) – Zanamivir is also Category C
What adverse effects have been found with Oseltamivir?
n/v, neuropsychiatric events (self-injury, delirium) – Category C in pregnancy
What should use of the neuraminidase inhibitors be avoided with?
Patients who have recently taken the live flu vaccine
What characterizes a congenital pulmonary airway malformation?
Hamartomatous lesion of the lung;
Type 1: most common, large cysts, good prognosis;
Type 2: medium-sized cysts, poor prognosis
What conditions are associated with ARDS?
Infection: sepsis, diffuse pulmonary infections, gastric aspiration; Physical injury: mechanical trauma; less commonly: inhaled irritants, chemical injury, hematologic conditions, others
What characterizes ARDS?
Rapid onset of severe life-threatening respiratory insufficiency, cyanosis, and severe arterial hypoxemia that is refractory to oxygen therapy (must balance therapy carefully) – CXR will show diffuse alveolar infiltration
What are the various names for adult respiratory distress syndrome (ARDS)?
In the wards: ARDS; Internationally: acute interstitial pneumonia; Autopsy: diffuse alveolar damage (DAD) – other synonyms: shock lung, acute alveolar lung, acute lung injury
What are causes of pulmonary edema and what
characterizes each?
Hemodynamic: increased hydrostatic pressure, edema seen in basal portions of lower lobes (dependent edema), presence of frothy pink fluid in alveoli;
Injury to capillaries: leakage of fluids due to injury to vascular endothelium or alveolar epithelial cells, seen with pneumonia and adult respiratory distress syndrome
What are the three types of atelectasis (collapse)?
Resorption atelectasis: due to complete airway obstruction causing resorption of oxygen in alveoli (causes mediastinum to shift towards the affected lung); Compression atelectasis: pleural cavity become partially/completely filled with fluid, tumor, or air (e.g. pulmonary edema, pneumothorax) causing elevation of the diaphragm and a mediastinal shift away from the affected lung;
Contraction atelectasis: local/generalized fibrosis prevent full lung expansion (not reversible)
What characterizes pulmonary sequestration?
Lung tissue without any normal connection to the airway system; can be extralobar (external to lung in thorax/mediastinum) or intralobar (within lung substance – often presents with recurrent localized infection or bronchiectasis)
What distinguishes the bronchi from the bronchioles?
Bronchioles have no cartilage
What characterizes foregut cysts?
Abnormal detachment of primitive foregut; most commonly bronchogenic cysts lined by ciliated, pseudostratified epithelium; they are usually an incidental finding in children/young adults;
What characterizes pulmonary hypoplasia?
Defective development of both lungs (found in 10% of neonatal autopsies); can be caused by space-occupying lesions in the uterus, oligohydramnios, congenital diaphragmatic hernia, renal cystic disease, anencephaly – microscopically there will be a lack of alveolar development
What histologic features are seen with DAD?
Diffuse distribution, uniform temporal appearance, alveolar septal thickening, hyaline membranes! – pertinent negatives include lack of granulomas, lack of infectious agents, and negative cultures
What distinguishes centriacinar from panacinar emphysema?
Centriacinar: destruction of the respiratory bronchioles (mostly in upper lobes – apices), occurs in heavy smokers and often associated with chronic bronchitis;
Panacinar: destruction of the entire alveolus (mostly in lower lobes – bases)
What characterizes emphysema?
Abnormal enlargement of airspaces distal to the terminal bronchiole due to destruction of walls without obvious fibrosis; Types of emphysema are classified according to their anatomical distribution: centriacinar, panacinar, paraseptal, irregular
What is the pathogenesis of COPD?
Mild, chronic inflammation throughout the lung, characterized by increased macrophages T cells, and neutrophils as well as increased inflammatory mediators (IL-8, TNF, etc.)
What characterizes COPD?
Chronic bronchitis (disease of bronchi) and emphysema (disease of acinus); 4th leading cause of morbidity/mortality in US; 90% of patients with COPD smoke
How do lung test values differ between obstructive and restrictive lung disease?
Obstructive: decreased FEV1 (seen in chronic bronchitis, emphysema, asthma, brochiectasis);
Restrictive: decreased FEV1 and FVC leading to a normal or increased FEV1/FVC (seen in ARDS and chest wall disorders – polio, obesity)
What is the difference between obstructive and restrictive lung disease?
Obstructive: increased resistance to airflow due to partial/complete obstruction in airway;
Restrictive: reduced expansion of lung parenchyma causing a decreased total lung capacity
What is the pathogenesis associated with ARDS?
Damage to alveolar capillary walls leading to increased vascular permeability, alveolar flooding, loss of diffusion capacity, widespread surfactant abnormalities (due to type II pneumocyte damage), and scarring (may not cause any functional consequences) – during this process pro-inflammatory mediators are expressed (NF-κB and IL-8) and inflammatory cells (neutrophils, macrophages) cause damage and maintain the inflammatory cascade
What is the clinical course associated with ARDS?
Usually occurs in hospitalized patients; they will experience profound dyspnea/tachypnea although an early CXR is normal; there will be increasing cyanosis/hypoxemia, respiratory failure and diffuse bilateral infiltrates; hypoxemia can become unresponsive to oxygen and respiratory acidosis can develop – 60% mortality rate
What enzymatic deficiency can lead to emphysema?
PiZZ: α1-antitrypsin deficiency (protease inhibitor); patients are more likely to develop panacinar emphysema at a young age (especially when they smoke)
What characterizes irregular emphysema?
Acinus involvement is irregular (many parts are involved); associated with scarring and coal workers pneumoconiosis
What clinical features are associated with chronic bronchitis?
Persistent productive cough, dyspnea on exertion, eventual hypercapnia/hypoxemia/cyanosis;
often seen in conjunction with emphysema; can lead to cor pulmonale and cardiac failure;
death is often due to pulmonary infection
What is the pathogenesis of chronic bronchitis?
It is a protective metaplastic response to tobacco smoke/pollutants, leading to hypertrophy of the submucosal glands in the trachea/bronchi; will see an increase in goblet cells of the small airways and hypersecretion of mucus
What characterizes chronic bronchitis?
Associated with tobacco smoke and other substances (grain, cotton, silica dust); infections may trigger acute exacerbations; most frequent in middle-aged men
How is chronic bronchitis defined?
Persistent cough with sputum for >3 months in 2+ years, in the absence of other causes
What are some rare types of emphysema?
Compensatory hyperinflation: dilation of alveoli without septal destruction; Obstructive overinflation: due to airtrapping; Interstitial emphysema: air in connective tissue stroma
What characterizes bullous emphysema?
Any form of emphysema that produces large subpleural blebs; often seen at the apex of the lung, and rupture can cause pneumothorax
How does smoking affect the protease-antiprotease balance in the lungs?
Smoke attracts neutrophils and causes the formation of ROS; these two factors will cause an increase in lung degradation leading to centriacinar emphysema
What characterizes paraseptal (distal acinar) emphysema?
Preferential destruction of the distal airway structures (starting around the septae of the lungs and affecting the alveolar sacs/ducts first); often forms adjacent to fibrosis/scarring; more severe in the upper half of the lung; can cause spontaneous pneumothorax (seen especially in young adults)
What is the clinical course associated with emphysema?
Clinical manifestations occur once >1/3 of the pulmonary parenchyma is damaged; symptoms start with a progressive insidious dyspnea (may have cough/wheezing), followed by weight loss, cor pulmonale and eventual CHF – death due to respiratory acidosis, right heart failure, and pneumothorax
What is seen on CXR with COPD?
Increased vascular markings (with bronchitis); Hyperinflation (emphysema)
What is the clinical course associated with bronchiectasis?
Severe, persistent cough; foul-smelling expectoration (often bloody); dyspnea/orthopnea; occasional life-threatening hemoptysis;
What characterizes bronchiectasis?
Permanent dilation of bronchi/bronchioles due to destruction of the muscle and elastic tissue;
can result in chronic necrotizing infections; also associated with CF, intralobar sequestration, immunodeficiency, tumor, foreign body, mucus impaction, autoimmune disease
How does the appearance bronchitis patient differ from a patient with emphysema?
Bronchitis: blue bloater; Emphysema: pink puffer – *check table in slide show
What causes the rigor/crises of Malaria?
The RBCs are lysed in a synchronous fashion by the parasites; once this occurs Merozoites will be free in the blood, causing an immune response which will present as rigor/crises
Why are most African blacks not affected by P. vivax?
They lack the Duffy antigen on their RBCs
Which strain of Plasmodium is known to cause benign tertian malaria?
Plasmodium vivax (40% of world-wide malaria cases) – infects only young RBCs
What is Plasmodium falciparum very lethal?
It attacks RBCs of all ages, causing them to form knobs that block capillaries, especially those in the brain (cerebral malaria) and kidneys (black water fever)
Which strain of Malaria is responsible for 40-50% of the world’s Malaria cases and around 95% of total mortality?
Plasmodium falciparum (malignant tertian malaria); it also has the most drug resistance
What is the vector associated with Malaria and when does it feed?
Anopheles mosquito, feeds after dusk
What are the classic Malaria symptoms?
Prodrome: malaise, myalgia, HA, fever (also cough, diarrhea, vomiting, abdominal pain);
Abrupt paroxysms of rigor (violent chills) followed by high fever, HA, sweating;
Fever periodicity is often absent initially, but indicates severe illness when present;
Other symptoms: nausea, jaundice (hemolysis), delirium, low glucose, splenomegaly
What are the resistance zones associated with Malaria drugs?
A: sensitive to chloroquine, includes central America, Middle East, temperate China;
B: partially chloroquine resistant, includes India, Western Indonesia;
C: significant chloroquine resistance, includes Amazon, SE Asia, PNG, sub-Saharan Africa
What are Hypnozoites?
Dormant form of the Malaria life cycle which can cause relapses in the disease (only in Vivax/Ovale species)
What are the stages of the Malaria life cycle?
Sporozoite (injected into human host during blood meal) --> travel to the liver which functions as a nursery bed --> after leaving the liver the infective organisms are called Merozoites --> they will enter the RBCs to feed and reproduce at which point they are known as Trophozoites --> once they have multiplied enough, the RBC will lyse and release more Merozoites (since now they are infective again) --> some of these Merozoites can enter the reproductive stage and become Gametocytes --> the Gametocytes can then be taken up again by another intermediate host
Which type of malaria do humans share with primates?
P. malariae (benign quartan): oldest/least virulent form, 1% of world malaria, infects oldest RBCs, persists in blood without the use of Hypnozoites – associated with transfusions malaria and nephrotic syndrome
What adverse effects are associated with Chloroquine?
GI upset, itching (especially in African blacks since they have slower metabolism), psoriasis exacerbation, intradermal HDCRV (rabies vaccine) interference; safe for retina at prophylactic doses (avoid of diseased), but requires annual eye exam if life dose is exceeded – safe in pregnancy, avoid with epilepsy, pills taste very bitter
What is the mechanism of action of Chloroquine?
It inhibits the enzyme heme polymerase; this enzyme normally functions in the biocrystallization of hemozoin molecules (heme --> hemozoin), thus removing toxic heme from infected RBCs; however, Chloroquine caps the hemozoin molecules, preventing further biocrystallization, and in effect causing a lethal buildup of heme
What are the oral treatment options for malaria?
Chloroquine (only in central America); Quinine sulfate/doxycycline (cinchonism);
Mefloquine (neuropsych reactions); Pyrimethamine-sulfa (Fansidar, resistance problems); Atovaquone/proguanil (Malarone, expensive)
What are the ABCDs of malaria prevention?
be Aware of malaria risk at destination; avoid being Bitten by mosquitoes; take Chemoprohylaxis when appropriate; immediately seek Diagnosis/treatment if fever develops (within 1 year)
What are the four drugs used for the prophylaxis of malaria?
Mefloquine (weekly dosage); Chloroquine (weekly); Doxycycline (daily); Malarone (daily, expensive, shortest post-trip course)
What should be done if malaria is suspected?
Treat immediately; don’t wait for test results
Why must a P. vivax infection also be treated with Primaquine?
Because it infects the host’s liver as Hypnozoites, causing the infection to relapse (P. vivax adapted to this mechanism in order to survive the cold winters in temperate climates)
What form of malaria is P. ovale malaria most like?
P. vivax malaria – infects only young RBCs, makes Hypnozoites; <10% of world malaria, most common in W Africa; usually Chloroquine sensitive
Which antimalarial drugs are effective against the blood stage, and which against the liver stage (Hypnozoites)?
Blood: Quinine, Chloroquine, Mefloquine; Liver: Primaquine
What adverse effects are associated with Mefloquine?
Sleep disturbance, vivid dreams, GI upset (take with water), dizziness, panic attacks, cardiac conduction problems; rarely: hallucinations, seizures – OR for psych reactions is 3.56
What is Doxycycline usually taken with and why?
Quinine, since Doxycycline is slow-acting (effective in chloroquine-resistant malarias)
What is the mechanism of action of Doxycycline?
Ribosomal inhibition (in pre- and erythrocytic phases)
What contraindications and drug interactions are associated with Doxycycline?
Contraindications: pregnancy, children<9 yo, allergy, liver disease; Drug interactions: antagonized by seizure meds (barbiturates, phenytoin, carbamazepine), avoid with Pepto-Bismol/iron/antacids, oral contraceptive become less effective, monitor PT (INR) if on Coumadin;
Doxycycline is cheap and effective, but has poor adherence
What does Doxycycline also protect against (besides Malaria) and what adverse effects are associated with it?
Also protects against Leptospirosis – Adverse effects: photosensitivity, esophagitis (take with water and keep upright!), Monilia (yeast-like fungus)
What contraindications are associated with Mefloquine?
Seizure disorders, h/o psychosis/depression, cardiac conduction defects (!); avoid use with quinine, quinidine, halofantrine (due to conduction defects, but BB/CCB use is OK) –
relative contraindications: 1st trimester pregnancy, people who perform tasks involving fine motor coordination (airline pilots), infants (unless high risk case)
What can be problematic regarding the pharmacokinetics of Mefloquine?
It has a very long half-life (21 days), meaning that adverse effects continue for a long time
What dosage is associated with Chloroquine and Mefloquine?
Once weekly, starting one week prior to departure and continuing 4 weeks after return
How has Chloroquine resistance come about?
Plasmodium has developed the ability to pump Chloroquine out of its food vacuoles where hemozoin biocrystallization occurs
What is the purpose of Nivaquine?
It is a syrup of Chloroquine that does not taste as bitter (not sold in US since it can cause a lethal overdose in children)
How does Quinine affect blood glucose levels?
It can exacerbate the hypoglycemia already caused by P. falciparum (can mimic changes of cerebral malaria)
What adverse effects are associated with Primaquine?
n/v, HA, methemoglobinemia
What must be screened for before giving a person Primaquine?
G6PDH deficiency (could be fatal due to hemolysis)
What is the treatment of choice in the US for malaria?
Primaquine phosphate – can also be used prophylactically
What drug interactions are associated with Proguanil?
It interacts with Coumadin, Rifampin, Tetracycline
What adverse effects are associated with Atovaquone/Proguanil?
Very few: GI upset, HA, mouth sores – not yet approved for pregnancy (use Mefloquine instead)
What drug is the best choice for short-term effective Chloroquine-resistant P. falciparum prophylaxis?
Atovaquone/Proguanil (Malarone)
What is the most common adverse effect associated with Quinine?
Cinchonism (flushing, ringing of ears, hearing loss, dizziness), may necessitate discontinuation;
Where should Quinine never be administered?
In the buttocks of a child, since it can cause sciatic nerve paralysis (use thigh injection)
What is the mechanism of action of Quinine?
Inhibits hemozoin biocrystallization
Why should long-term treatment with Fansidar (Pyrimethamine/Sulfadoxine) be avoided?
Since it has a sulfa drug in it, long-term treatment can lead to Stevens-Johnson syndrome – also avoid in 1st trimester and in last 2 weeks of pregnancy (due to kernicterus)
What homeopathic preparations are not effective against malaria?
Demal 200, Papaya leaf tea, Sweet wormwood home-brewed tea, Thiamine, ultrasound devices, repellant-impregnated wristbands
How is Artemisinin best used?
In combination with a long-acting antimalarial (e.g. Lumefantrine, which is the safer alternative to Halofantrine = no QT prolongation)
What is the mechanism of action of Artemisinin?
As the parasite consumes hemoglobin, it frees up the heme which releases iron --> the iron can then reduce a peroxide bond in Artemisinin, generating high-valent iron-oxo species --> these species result in a cascade of reactions that produce oxygen radicals which kill the parasite
What new drug is reserved for severe cases of Malaria in the US?
Artemisinin (derived from Sweet Wormwood or Artemesia annua) – has a very short half-life
What is used in the US as an alternative to IV Quinine treatment?
IV Quinidine
What is the adverse effect associated with Halofantrine?
QT prolongation – also, avoid with Mefloquine
What distinguishes Tafenoquine from Primaquine?
It has a longer half-life
What does Maloprim (Deltaprim) consist of, where is it used, and what adverse effects are associated with it?
Pyrimethamine and Dapsone; used overseas as an affordable alternative for malaria prophylaxis; not used in US/Europe due to adverse effects: agranulocytosis, thrombocytopenia
What is the treatment for Babesia microti?
Clindamycin with quinine (same treatment as for malaria in pregnant women)
What is the treatment for Trichinella spiralis (Trichinosis)?
Albendazole and steroids
How does Trichinella spiralis cause disease?
Worms enter the body in undercooked meat (pork/bear meat), mature in the GI tract, and their progeny encyst in host muscle; It causes painful, tender muscles and fever, as well as eyelid edema, diarrhea, and high eosinophil counts
What characterizes Albendazole?
MOA: interferes with microtubule assembly and impairs glucose uptake; PK: part of the Benzimidazole class, it is metabolized in the liver to Albendazole sulfoxide which is the most active form;
What is the treatment for cutaneous larva migrans?
Albendazole or Ivermectin; can also use ethyl chloride or freezing ahead of the advancing margin
What is cutaneous larva migrans also known as and what characterizes it?
Creeping eruption: causes uncontrolled itching and a characteristic skin track (serpiginous)
What characterizes Clindamycin?
MOA: attaches to ribosomes; Uses: Gram+/anaerobes/anti-malarial/oral and bone infections;
AE: can cause C. difficile colitis (since it is resistant), as well as diarrhea and a rash
What are tissue helminth infections often associated with?
Eosinophilia – whereas protozoan infections usually do not develop eosinophilia
What characterizes Babesia microti?
It can appear as a malaria-like blood infection, but is distinguished by RBCs containing tetrads (or “Maltese cross”); it is spread by ticks or via transfusion, and is endemic to the NE and Midwest US (also globally)
What is the danger with Naegleria?
They live in warm springs and can attack the human brain by traveling through the cribriform plate - can be treated with Amphotericin B
What is the pathology of Onchocerciasis (river blindness)?
adasd
What characterizes Diethylcarbamazine (DEC)?
Used in the treatment of filarial infections (available only from CDC); MOA: damages surface of microfilariae, allowing the host immune system to kill it; AE: Mazzotti reaction = host immune reaction due to dying microfilarial antigens (itching, rash, fever, edema, conjunctivitis, encephalitis) – often given with corticosteroids to prevent these adverse effects
What is the pathology of Loa loa?
How is Loa loa diagnosed?
Take a blood sample during the day (since that’s when the vector is active) and look for microfilariaeIt is a filarial worm; the adults migrate through the tissue and may travel to eye (causing conjunctivitis); wherever the worms are, it can create an inflammatory swelling (Calabar swelling); the microfilariae live in the blood where it can be picked up by the deer fly during daylight hours
What is the treatment for Strongyloides?
Ivermectin (or Albendazole)
What is the pathology of Strongyloides?
It is a parasitic/free-living nematode that can invade humans, especially when immunocompromised (in this case it can cause a hyper-infection syndrome which is fatal in 80% of cases) – symptoms include a rash due to migrating larvae in the skin (larva currens)
What side effects are important for Ivermectin?
Although it kills the microfilariae, it does not kill the adult Oncocerca worms (it is curative for Strongyloides and Mansonella); most side effects are due to the dying microfilariae (itching, rash, edema, eye inflammation); there is a risk of fatal encephalitis if co-infected with Loa loa
What is the treatment for Onchocerciasis and how does it work?
Ivermectin – derived from Streptomyces avermitilis; MOA: opens chloride channels in invertebrates (but not in mammals); it is also used for Strongyloides, Mansonella, and scabies
What is the pathology of visceral larva migrans?
Dog/cat roundworm eggs are deposited in soil/sandbox; dirt can then be ingested (often by a child < 3yo) causing nausea, fever, hepatomegaly, edema, urticaria, and seizures
What is the treatment for visceral larva migrans?
Albendazole or Mebendazole
What parasites can be responsible for Filariasis/Elephantiasis, what is the vector, and what is the treatment?
Wuchereria bancrofti, Brugia malayi, Mansonella – vector: mosquitoes (feed at night); treatment: DEC
What is the treatment for Leishmaniasis?
Cutaneous lesions: infiltrate lesion with stibogluconate/liquid nitrogen, or give oral antimony/fluconazole; For L. braziliense or recalcitrant infections, pentavalent antimony is given IV/IM (i.e. sodium stibogluconate/meglumine antimonate)
What can Schistosomiasis lead to?
Liver cancer
What are the three species that cause Schistosomiasis and what is the treatment?
S. mansoni (inferior mesenteric vessels, terminal spine on side); S. haematobium (bladder vessels, terminal spine on end); S. japonicum (superior mesenteric vessels) – treatment is praziquantel
What can be a complication of visceral leishmaniasis?
Post Kala Azar Dermal Leishmaniasis – causes hypopigmented macules (usually occurs up to several years after treatment); may require pentavalent antimony treatment
What characterizes Miltefosine?
Used for visceral Leishmaniasis in India; Category X in pregnancy; investigational treatment in HIV
What adverse effects are associated with pentavalent antimonials?
Cardiac arrhythmias, chest pain, pancreatitis, very painful when given IM
What is the treatment for visceral Leishmaniasis?
Pentavalent antimonials, although resistance in India has required replacement by Miltefosine; in the US, treatment is L-AMB (less nephrotoxicity)
What is the treatment for Dracunculiasis (Guinea worm infection)?
Mechanical removal of worm (found in Southern Sudan and war-torn areas of Africa)
What form of Leishmaniasis is responsible for Espundia?
L. braziliense
What are the three types of Leishmaniasis and what is the vector?
Cutaneous, Mucosal (Espundia), Visceral (Kala Azar/black fever – common in India) – all are caused by the sand fly
What are the different types of Trypanosomal infections?
South American Trypanosomiasis: Chagas disease (caused by Reduviid bug);
African Trypanosomiasis: African sleeping sickness (W. African variety – T. brucei gambiense, slow course; E. African variety – T. brucei rhodesiense, fast course)
What is the usual treatment for PCP pneumonia?
Bactrim – however in AIDS patients, the adverse sulfa reaction can be too severe, so alternative treatments may have to be used
What adverse effects are associated with Pentamidine?
Hypoglycemia; later on, diabetes
What is the treatment for African sleeping sickness?
West African --> early: pentamidine (also used for PCP pneumonia); late: eflornithine
East African --> early: suramin; late: melarsoprol (arsenic)
What is an important symptom of African sleeping sickness?
Winterbottom’s sign (posterior cervical lymph node enlargement); also Tsetse bite chancre with lymphadenitis
What is the treatment of choice for Chagas disease?
Benznidazole
What is an important symptom of Chagas disease?
Romana’s sign (unilateral swelling of the face)
What is the treatment for non-intestinal flukes?
Praziquantel – does not work for F. hepatica (use Triclabendazole)
What characterizes Praziquantel?
Best treatment for most flukes and tapeworms; MOA: penetrates integument of worms causing tetanic contraction and paralysis; AE: dizziness (don’t drive), interacts with Rifampin, Category B in pregnancy
What can be a complication of ingestion a pork tapeworm eggs?
Neurocysticercosis (common cause of seizures in the developing world) – treatment is praziquantel or albendazole (sometimes seizure medication)
What is an unusual side effect of eflornithine?
Retards facial hair growth in women
How do people often get infected with Toxoplasma gondii?
By cleaning out the litter box and inhaling the parasite
What is the treatment for Toxoplasmosis and when is it used?
Pyrimethamine and sulfadiazine (work by folic acid synthesis inhibition) – only used in pregnant women, acute neonatal toxicity, and immunocompromised patients
What are the various ways in which Toxoplasmosis can present?
Congenital toxoplasmosis: seizures, intracerebral calcifications, MR, microcephaly, blindness;
Mono-like presentation: in adults; Meningoencephalitis: in AIDS patients – however, most patients will be asymptomatic
What are examples of antigen-specific mechanism for mucosal host defense?
Lymphoid elements associated with internal surfaces of the body; Exocrine secretory glands (e.g. salivary, lacrimal, pancreas, mammary glands)
What is the difference between monomeric and dimeric IgA?
Monomeric: produced mostly in bone marrow and found in the blood; Dimeric: produced mostly in lamina propria of mucosal tissues and found mainly in external secretions, also dimeric IgA is actively transported into external secretions via the polymeric immunoglobulin receptor (Pig-R)
By what mechanism does migration of mucosal-derived lymphoblasts to distant mucosal sites take place?
By the interaction of lymphocyte adhesion receptors (integrins) and vascular addressin
What can happen when an antigen-response is generated in the gut?
It can generalize to other mucosal surfaces (e.g. bronchial tract, mammary glands) – mediated by lymphoblast trafficking
How does the MALT differ from lymph nodes?
MALT is associated with T cells that preferentially support a B cell class switch to IgA
Where are M cells located and what is their function?
They are part of the specialized epithelium that overlies Peyer’s patches and function in the facilitation of antigen uptake
What are examples of mucosa-associated lymphoid tissue (MALT)?
Nasal-associated lymphoid tissue (NALT): tonsils, adenoids; Gut-associated lymphoid tissues (GALT): Peyer’s patches; Bronchus-associated lymphoid tissues
What are examples of non-antigen specific mechanisms for mucosal host defense?
Nasal function (e.g. being able to clear particulates), cough reflex, gag reflex, mucociliary clearance
What problems can lead to dysfunction of mucociliary clearance and what can this result in?
Improperly functioning cilia, abnormally thick mucus (with CF) --> stasis --> bronchiectasis (abnormal enlargement of conducting airways) due to recurrent infection
What can an impaired cough/gag reflex lead to?
Aspiration --> pneumonia, lung abscess
How does IgA2 differ from IgA1?
IgA2 is more resistant to bacterial proteases, thus more IgA2 is secreted than IgA1
What binds two IgA monomers together to form the dimeric IgA?
J chain – B cells can be either J chain-positive or J chain-negative; this will determine whether they secrete monomeric or dimeric IgA
What is the purpose of the secretory component of secretory IgA?
It provides the secretory IgA with some form of protection against enzymatic cleavage, thus prolonging its half-life
How does dimeric IgA cross the mucosal epithelial cell layer?
After being secreted from B cells, they bind to the poly-Ig receptor found on the basal surface of mucosal cells --> this causes them to be taken up via endocytosis, after which they are transported to the apical surface of the cell --> once they are released from the apical surface into the lumen, a secretory component from the poly-Ig receptor remains attached to the dimeric IgA
Why is IgA better suited for mucosal immunity than IgG?
It does not activate inflammatory systems to the same extent as IgG (i.e. it can block infection without inducing an inflammatory response that damages the surface)
By what mechanism can TH2 cells cause suppression of TH1 cells?
TH2 can secrete IL-10 which interferes with the co-stimulator function of APCs to TH1 cells
What is the purpose of oral tolerance to antigens and how is this thought to be achieved?
Its purpose is to prevent a harmful immune response to harmless antigens/foods – it is thought to be achieved through clonal deletion, clonal anergy, or active suppression by T cells
What is the significance of the presence of TCRγδ/CD8+ T cells as part of the intra-epithelial lymphocytes?
They are thought to play an important role in immunoregulation and epithelial renewal during infection or enteropathy
How do intra-epithelial lymphocytes differ from lamina propria lymphocytes?
Intra-epithelial lymphocytes contain more of the TCRγδ-type whereas those of the lamina propria are almost exclusively the TCRαβ-type – also, the intra-epithelial lymphocytes are mostly CD8+. Whereas those of the lamina propria are mostly CD4+
What type of infection is common in people who have low T cell counts (e.g. AIDS patients)?
Fungal infections (e.g. Candida, Pneumocystis)
What is the effect of having HIV on malarial infections?
Increased incidence and increased mortality
What describes the clinical symptoms of malaria?
Non-specific (must take epidemiological history)
How does Plasmodium falciparum affect RBCs and what are the consequences?
They make them less deformable and more sticky causing them to form knobs --> knobs block capillaries and small blood vessels in essential organs --> impaired oxygen/substrate delivery, focal ischemia, anaerobic glycolysis, lactic acidosis --> cellular injury
How do Plasmodia derive energy from their host?
By anaerobic glycolysis of glucose to lactic acid
What causes the symptoms of malaria?
Rupture of schizont in RBC with release of merozoites
What form of malaria has recently been found to transmit to humans (formerly only seen in primates)?
Plasmodium knowlesi
What are modes of malaria transmission besides via the Anopheles vector?
Sharing of needles (drug abuse), blood transfusion, mother to infant transmission, organ transplantation
What does it mean when a patient with malaria has crescent/banana-shaped gametocytes in their blood?
They are infected with Plasmodium falciparum
What is meant by airport malaria?
Mosquitoes around international airports pick up Plasmodium from travelers and spread it to other human hosts
What is seen in a blood smear from a patient infected with P. vivax/ovale?
Enlarged RBCs containing small, red dots
What are the quinolone derivatives used for malaria treatment?
Chloroquine, quinine, quinidine (IV), mefloquine, halofantrine, primaquine
What must be done before giving primaquine?
Check for G6PDH deficiency
What is the main treatment for P. vivax/ovale?
Chloroquine and primaquine; if patient from PNG/Indonesia, use quinine with doxycycline and primaquine
What is the main treatment for P. falciparum with chloroquine-resistance?
Quinine with doxycycline (if pregnant: quinine with clindamycin)
What type of treatment is given in patients with an extremely high parasitemia?
Exchange transfusion (replace plasma)
What are the ribosomal inhibitors used for malarial treatment?
Tetracycline, clindamycin – used in combination with other drugs (e.g. Quinine)
What are the antifolates used for malaria treatment?
Pyrimethamine, sulfonamides
How is malaria diagnosed?
By thick and thin smears; thick smear is more sensitive, thin smear allows for species identification – also useful for quantifying the degree of parasitemia
What is an example of a substance detected by rapid diagnostic tests for malaria?
Histidine-rich protein (from P. falciparum infected RBC)
What are the pros and cons of PCR testing for malaria?
Can find very small number of parasites, but cannot tell whether they are dead or alive
What antimalarial is used prophylactically when traveling to areas with no drug-resistance?
Chloroquine – treatment must be started earlier and continued longer
What is a problematic side effect associated with mefloquine prophylaxis?
Neuropsychiatric and cardiac issues – cheaper and weekly dosing
What is a problematic side effect associated with doxycycline prophylaxis?
Photosensitivity (easy to get burned in areas where malaria is endemic)
Why are chemoprophylaxis antimalarials started prior to travel?
Determine whether drugs are tolerated (also, to build up steady-state levels of drugs)
What antimalarial is used prophylactically when traveling to areas of drug-resistance?
Malarone (atovaquone/proguanil) – treatment is quite expensive, but has short post-trip dosage; other options include doxycycline or mefloquine
When is vaccination against Strep pneumo especially important?
In the elderly and pre-splenectomy
What characterizes community-acquired atypical pneumonia (CAAP)?
Pathy inflammatory changes in the lung, mostly interstitial; Little/no sputum; No physical signs of consolidation; Little/no elevation of WBCs; Extrapulmonary signs
Which bacteria often cause pneumonia in patients with CF?
Pseudomonas aeruginosa (also seen with nosocomial pneumonia)
What is the most common cause of Gram-negative pneumonia?
Klebsiella: causes formation of thick/gelatinous sputum; often seen in debilitated/malnourished people (e.g. chronic alcoholism)
What bacteria often cause pneumonia following a viral infection?
Staph aureus
What characterizes Moraxella catarrhalis?
Mostly seen in elderly; common cause of acute exacerbation of COPD; common cause of otitis media in children
What disease is associated with H. flu?
Children: lower respiratory tract infection, meningitis, epiglottitis, conjunctivitis (“pink eye”); Adults: CAP; Elderly: septicemia, endocarditis, pyelonephritis, cholecystitis, suppurative arthritis
What are the key distinctions between lobar pneumonia and bronchopneumonia?
Lobar pneumonia: consolidation of a large part of a lobe/the entire lobe (4 stages: congestion, red hepatization, gray hepatization, resolution); Bronchopneumonia: patchy consolidation, consolidated areas of acute suppurative infection, multilobar/bilateral/basal
What (soft) radiologic findings are associated with pneumonia?
Lobar distribution --> RLL (S. pneumo/H. flu), Upper lobe (Klebsiella); Effusions --> H. flu/streptococcal, not S. pneumo; Cavitation --> Klebsiella, not S. pneumo/H. flu
What physical exam findings are associated with pneumonia?
Dullness to percussion, crackles/rales, bronchial breath sounds, egophony
What is absolutely necessary for the diagnosis of CAAP?
CXR – since physical exam is not as helpful
What clinical signs are associated with a lung abscess?
Foul-smelling purulent/sanguineous sputum; Fever, chest pain, weight less; Clubbing
What characterizes SARS?
Coronavirus; First found in China; Lower respiratory tract syndrome; Diagnosis by PCR or detection of antibodies; 10% mortality rate; No treatment, supportive care only
What type of infection are immunosuppressed patients especially vulnerable to?
CMV(e.g. CMV pneumonia, CMV retinitis)
What extrapulmonary signs are associated with CAAP?
Ear/throat lesions: mycoplasma; Diarrhea: mycoplasma, legionella; Abdominal pain: legionella;
Skin rash: mycoplasma, psittacosis
What are disadvantages of combination therapy?
Cost, IV lines, drug interactions, adverse effects, difficulty in identifying single agent effects (both positive and negative effects), antagonism, superinfection (antibiotics kill natural flora, allowing opportunistic infection to take root)
What is meant by the terms inappropriate/inadequate therapy?
The drugs that were used initially to treat the infection are shown to be ineffective for the organisms later found to be causative – a change of therapy is required
What is the procedure for nosocomial pneumonia or intraabdominal infections?
Multi-drug therapy based on a variety of potential pathogens
Why do bacteria respond differently to gentamicin/tobramycin and streptomycin?
They are affected by different aminoglycoside-modifying enzymes
In what clinical setting are synergistic drug combinations a must?
Serious infections, like endocarditis
What combination of drug has been shown to be synergistic against enterococcal infections?
Aminoglycosides and ampicillin/vancomycin; this combination can make two bacteriostatic drugs bactericidal (ampicillin/vancomycin break up the cell wall so that aminoglycosides can get in and inhibit the 30S bacterial ribosomes)
What class of drugs are enterococcal infections inherently resistant to?
Cephalosporins
What is the major reason why combination therapy is used?
Empiric therapy to cover all most likely pathogens
How is synergism defined, in vitro?
As a greater than 2 log10 reduction in bacterial count when comparing combination treatment with a single agent (e.g. Enterococcus time-kill study)
What indications for combination therapy have not yet been established to a great extent?
Prevention of the emergence of resistance, disease severity/organisms with high/rapid mortality
How often does inappropriate therapy take place?
about 30% of the time – often due to antibiotic-resistant pathogens
How does outcome differ between combination therapy vs. appropriate monotherapy (i.e. bug is susceptible to monotherapy drug)?
It does not; there is no difference in mortality rates
What drug combinations have not been shown to be synergistic in the treatment of Pseudomonas aeruginosa?
Ciprofloxacin + aminoglycosides; Double β-lactams
What drug combinations have been shown effective in treating Pseudomonas aeruginosa?
Antipseudomonal β-lactam + aminoglycosides (gold standard); also antispseud β-lactam + ciprofloxacin or aztreonam; other options include carbapenems + aminoglycosides or ciprofloxacin, or also aztreonam + aminoglycosides
Why is combination therapy important in treating Pseudomonas aeruginosa?
It is a common nosocomial pathogen with a high mortality rate; it has multiple mechanisms for attaining drug-resistance
What is the significance of the SPACE organisms?
Two antibiotics should be used for Serratia, Pseudomonas, Acinetobacter, Citrobacter, Enterobacter – unless the exact species is known (monotherapy is sufficient)
What organisms are known to be resistant/difficult to treat?
MRSA, Pseudomonas aeruginosa, Acinetobacter, Klebsiella pneumoniae, Enterobacter, Enterococcus, Candida
What describes the activity of barbiturates?
Sedation to coma/death; Induces a state resembling normal sleep (decreases REM; at hypnotic doses can see distortion of mood, impairment of judgment, impairment of fine motor coordination); No analgesia; Anticonvulsant activity
What characterizes the clinical use of benzodiazepines in the treatment of anxiety?
Most effective in patients showing high levels of emotional and somatic symptoms, and low levels of depression; Some use in patients showing anxiety associated with other disease states; Improvement usually seen within the 1st week or two of treatment
What activity is seen with benzodiazepines?
Sedative/hypnotic, anxiolytic, anticonvulsant (not used prophylactically), muscle relaxant, amnestic
What advantages do benzodiazepines have over barbiturates?
Do not induce liver enzymes, have very little effect on REM sleep, have a much wider margin of safety, withdrawal is mild
When is giving a barbiturate contraindicated?
When the patient has a family or personal history of acute intermittent porphyria: overproduction and excretion of porphyrins and their metabolites, causes severe abdominal pain and neurological disturbances; barbiturates increase the production of porphyrins
What is the treatment for barbiturate overdose?
Maintain respiration, rule out narcotics (e.g. Naloxone), treat symptoms (maintain respiration and perfusion, gastric lavage [?], administer bicarbonate, hemodialysis)
What are adverse effects of barbiturates?
Symptoms of overdose incl. comatose state, marked respiratory depression (immediate cause of death in overdose), shortened REM sleep, marked hypotension, weak/rapid pulse; Induces P450; Can see physical/psychological dependence – At one time barbiturates were the most common drug class used in suicide
What is the mechanism of action of benzodiazepines?
Bind between the gamma and alpha subunits on the GABAA receptor, thus increasing the influx of Cl-, causing hyperpolarization of the cell
What are barbiturates used for clinically?
Induction in general anesthesia; Seizure disorders
What are examples of barbiturates and their half-life?
Long-acting: Phenobarbital; Intermediate-acting: Pentobarbital, Secobarbital;
Short-acting: Thiopental, Methohexital
What side effects and toxicities are seen with benzodiazepines?
Drug interactions: do not induce P450, combination with other CNS depressants can be lethal;
Adverse effects: very few, safe, toxic doses treated with supportive care, caution in 1st trimester;
At time of peak plasma levels: marked effects on driving, patients underestimate impairment;
Tolerance: develops to many effects, but not to antianxiety effects;
Physical dependence: abrupt discontinuation of chronic administration leads to withdrawal
What characterizes Temazepam?
Oral absorption slow and erratic; No active metabolites
What characterizes Lorazepam?
Absorbed slowly when administered orally; Well absorbed following IM administration; No active metabolites; Proper dose adjustment can give a good night’s sleep with minimal residual effects
What characterizes Flurazepam?
Metabolized to active metabolites; peak concentration of active metabolites reached in 1-3 hrs after oral intake; good BA; Proper dose can give 8hrs of sleep with minimal residual effects the next day
Which benzodiazepines are used as sedatives/hypnotics?
Flurazepam, Lorazepam, Temazepam, Triazolam
What characterizes Flumazenil?
It is a benzodiazepine antagonist: used for overdose to reverse respiratory depression; it will precipitate withdrawal so don’t give to patients with a history of seizures or to patients with barbiturate/tricyclic antidepressant overdose – variable effects with ethanol intoxication; it has a rapid onset and short duration
What is the best way to avoid side effect when withdrawing benzodiazepines?
Gradually taper the dose
Besides treating anxiety, what other uses do benzodiazepines have?
Sleeping disorders, convulsive disorders, neuromuscular disorders, preoperative medication, treatment of withdrawal from CNS depressants
What characterizes the metabolism of benzodiazepines?
Many are metabolized to active metabolites with longer half-lives and possibly more activity; Only Lorazepam, Oxazepam, Temazepam, and Triazolam have no active metabolites
What characterizes the absorption and distribution of benzodiazepines?
Oral absorption within 1-3 hours; Only Lorazepam is absorbed following IM injection; Apparent volume of distribution is 1-3L/kg; Typically have 80% of dose protein-bound
What characterizes Oxazepam?
Slow oral absorption; T½: 5-10hrs; No active metabolites
What describes the absorption, distribution, and metabolism of Buspirone?
Rapid and complete absorption following oral administration; Extensive first-pass effect;
T½: 2-3hrs, active metabolite half-life is twice as long
What characterizes Buspirone?
May act through serotonin systems; possibly has some effect on dopamine systems;
CNS activity: anxiolytic, delayed effects (4-6wks), not as effective in patients with prior history of benzodiazepine use, may be drug of choice in patients with history of sedative/hypnotic abuse;
Does NOT have sedative, anticonvulsant, or muscle relaxant properties (may be helpful in treating elderly patients) – minimal adverse effects, no dependence/cross-tolerance
What type of drug class are the azaspirodecanediones?
Non-benzodiazepine anxiolytic; prototypical drug is Buspirone
What are the advantages of benzodiazepines in sleep disorders?
Safety, do not disrupt REM, do not induce P450, milder withdrawal syndrome
What characterizes Alprazolam?
Oral absorption slower than Diazepam (like Chlordiazepoxide); T½: 10-14hrs; Metabolized to active metabolite (significant levels of active metabolite are not observed at therapeutic doses)
What characterizes Chlordiazepoxide?
Slower oral absorption than Diazepam; T½: 7-13hrs; Metabolized to Desmethyldiazepam and Oxazepam
What characterizes Triazolam?
Well absorbed orally; peak plasma levels 1 hr after oral administration; No active metabolites; May have higher abuse potential and more withdrawal with chronic use; Reported higher incidence of adverse behavioral reactions
What characterizes Diazepam?
Well absorbed orally; Elimination is biphasic and variable; Metabolized to Desmethyldiazepam which is further metabolized to Oxazepam (both metabolites are active)
Which benzodiazepines are used as anxiolytics?
Diazepam, Chlordiazepoxide, Oxazepam, Alprazolam
What is the mechanism of action of GABAA-α1 agonists?
Non-benzodiazepines; bind to with high affinity to α1 receptor subtype, but with much lower affinity to the other subtypes
What are the spasmolytic drugs?
GABA agonists (e.g. Diazepam and other benzodiazepines)
What drug interactions are seen with GABAA-α1 agonists?
Additive effects with ethanol and other CNS depressants; Inhibitors of CYP enzymes can increases serum concentrations of Eszopiclone
What can occur at high doses of Zaleplon?
Rebound insomnia
What adverse effects are seen with GABAA-α1 agonists?
Respiratory depression is NOT a major problem; Hypoxia/hypercarbia may be seen in patients with obstructive sleep apnea; All have some residual daytime sedation
What characterizes the absorption, distribution, and metabolism of GABAA-α1 agonists?
Rapid onset with short duration of action; well absorbed orally; T½: Zolpidem --> 2-2.5hrs, Zaleplon --> 1hr, Eszopiclone --> 6hrs; Metabolized by liver to inactive metabolites; Need to lower dose for elderly and patients with liver disease; All are metabolized by P450
What are the clinical uses of GABAA-α1 agonists?
Sedative/hypnotic: decrease sleep latency, little effect on sleep cycles at low doses; Zaleplon has a shorter duration of action than Zolpidem/Eszopiclone – compared to benzodiazepines they have less anti-anxiety effect, less anti-convulsant effects, and less muscle relaxant effects
What drug interaction is found with Buspirone?
binding sites
Displaces Digoxin but not Warfarin from plasma protein
What are the GABAA-α1 agonists?
Zolpidem, Eszopiclone, Zaleplon
What is the key difference between viral and bacterial sinusitis?
Viral sinusitis does not last as long as bacterial sinusitis (and it is not associated wit pus formation)
What is the most common cause of pneumonia?
Bacterial infection (Strep pneumo)
What characterizes the lower respiratory tract infections?
The etiologic agents for these infections vary widely
What causes the most severe type of epiglottitis?
Bacterial infection (mainly H. flu) – not seen much anymore due to H. flu vaccine
What is almost always the underlying cause of laryngitis?
Viral infection
What is the most important organism causing otitis media?
Strep pneumonia (also H. flu)
What type of bacterial infection can be a complication of a viral infection?
Infection due to bacteria that comprise the normal flora of the nasopharynx; the viral infection causes swelling and inflammation which prevents normal drainage, thus giving the normal flora an opportunity to cause overgrowth and infection
What are considered upper respiratory infections?
Any infection affecting the larynx or structures above it
What is the most important microbe responsible for pharyngitis?
Group A Strep (Strep pyogenes)
What is the most common cause of rhinitis?
Viral infection
What is the most common cause of bronchitis?
Viral infection (presents as a lingering cough/cold), although Mycoplasma is also common
Why does rhinovirus mostly affect the upper respiratory tract?
Because it is transmitted in larger particles (>10 microns)
What type of atelectasis can be caused by an empyema?
Compressive atelectasis
What can atelectasis (collapse of a segment of lung) lead to?
Pneumonia (since normal functioning is impaired)
Why are patients with COPD more likely to get pneumonia?
They have a reduced capacity to expel organisms from the lungs; they also often smoke, which further impairs host defense mechanisms
What host defense mechanisms are specific to the respiratory tract?
Clearance: mucous layer, cilia, macrophages, cough – these mechanisms can be impaired by smoking
What can result from aspiration of food?
Anaerobic bacteria are carried along with the food and can create a lung abscess (seen as a cavity on x-ray) – location of aspiration depends on position of patient while aspiration occurred (lower lobes are more common)
What is the most common cause of bronchiolitis?
Viral infection (most common in children, can be very serious)
How can a patient with an empyema present?
Patient being treated in the hospital for pneumonia appear to be getting better but is still febrile and CXR shows a pleural effusion
What does the term empyema mean?
A collection of pus in a potential space (e.g. located around the lung) – can be due to a complication of pneumonia, often requires surgical drainage
What is the classic virus that is considered intermediate size (2-10 microns)?
Influenza virus (can damage cilia thus increasing the chances for secondary bacterial pneumonia – this is a major reason why people die from influenza virus)
What can be a distinguishing factor between bacterial pharyngitis and viral pharyngitis?
If the patient has a concomitant rhinitis, the cause is most likely viral
What organisms are part of the differential diagnosis for pharyngitis?
Viral: rhinovirus, EBV (mono); Bacterial: group A strep, diphtheria; also, deep neck abscess
What organism can be responsible for pneumonia in the bronchioles and alveoli?
A very small organism (1-5 microns); e.g. Mycobacterium tuberculosis
What circadian-specific physiologic events occur during sleep?
GH secretion spikes during first bout of Delta sleep (stage III/IV); Cortisol secretion is maximal at dawn; Body temperature falls until 5AM; REM propensity is maximal between 3-5AM; Sleepiest times of the day are between 2-4PM and 3-5AM
What occurs during Stage II sleep?
Sleep spindles, K complexes; bodily movements continue; lasts 10-25 minutes during first sleep cycle; constitutes ~50% of sleep
What occurs during Stage I sleep?
Transition from Alpha to Theta waves; easily responsive to sounds; lasts 1-7 min.; Hypnic myoclonus may occur
During which states of sleep does arousal cause only a limited confusional state?
Stage I, II, REM
How long does it take on average to enter REM sleep?
Hour and a half (in narcolepsy, 20 min)
What results from a chronic lack of sleep?
Being tired/fatigued; Kids are hyperactive; Impaired performance at work/school/sports; Reduced alertness/attention; Mood swings, irritability; Impaired social interactions; Decreased creativity
What are the major chief complaints in sleep medicine?
Insomnia, excessive daytime sleepiness (EDS), unusual movements/behaviors/sensations while asleep or attempting to sleep
How much sleep is necessary?
Infants: 16-20 hours (half in REM), Adults: 8 hours
What percent of sleep is taken up by REM sleep?
~20%
What are reasons that sleep is important?
Restore wakefulness, tissue growth/repair/cellular division/protein synthesis
What are the three subtypes of insomnia?
Delayed sleep onset, impaired sleep continuity, early morning awakenings
What characterizes the Multiple sleep latency test?
Patients are given ~5 nap opportunities; time to sleep onset and REM is measured for each nap; can measure narcolepsy
What characterizes the Epworth Sleepiness Scale?
Consists of 8 questions that assess sleepiness under varying conditions; >10 is a significant score
What are tests that objectively measure sleepiness?
Multiple sleep latency test (MSLT), Maintenance of Wakefulness Test (MWT), Epworth Sleepiness Scale (ESS)
What characterizes narcolepsy?
REM sleep onset, cataplexy, hypnagogic/hypnopompic hallucinations, disturbed nocturnal sleep, excessive daytime sleepiness
What are the main causes of excessive daytime somnolence?
Insufficient sleep, sleep disordered breathing, restless leg syndrome overlapping with nocturnal myoclonus or PLMS, narcolepsy
In what groups of people is insomnia more commonly seen?
Women, elderly, thin; Chronic illness, pain; Anxiety, depression, stress; Drug/alcohol use/abuse
What occurs during Stage III/IV sleep?
High voltage Delta waves predominate; high stimulus threshold for arousal; important stages for feeling well-rested in the morning; skeletal muscles relaxed by still active
What characterizes REM Behavior Disorder?
REM sleep activity without
skeletal muscle atonia
What occurs during REM sleep?
Brain is active and EEG resembles awake state; Body is paralyzed except for diaphragm/eyes; REM is “dream sleep time”
What are physiologic consequences of sleep apnea?
Systemic/pulmonary hypertension, cardiac dysfunction, polycythemia
What should be considered in the patient with restless leg syndrome?
Renal failure, iron deficiency anemia
What are the main causes of central sleep apnea?
Stroke, CHF
How is OSA treated?
Positive airway pressure
What is a common cause of obstructive sleep apnea (OSA)?
Cranio-facial abnormalities
What are the main complications of sleep apnea?
MVA, CVA, MI, arrhythmias, airway obstruction/pulmonary edema after general anesthesia
What characterizes the Multiple sleep latency test?
Patients are given ~5 nap opportunities; time to sleep onset and REM is measured for each nap; can measure narcolepsy
What are risk factors for sleep apnea?
Small airway, Cushing’s, hypothyroidism, acromegaly, morbid obesity
What is sleep apnea?
Ability to maintain the patency of the upper airway during sleep
What is the treatment for narcolepsy?
CNS stimulants, SSRIs/antidepressants, scheduled naps
What sleep disorder sometimes precedes Parkinson’s disease?
REM behavior disorder
What are the most common sleep disorders in children?
Night terrors, nightmares, insomnia
What are examples of parasomnias?
REM behavior disorders, sleep walking, night terrors, PLMS, head banging
What is the treatment for AOM?
Analgesics/antipyretics, antibiotics, possible decongestants, possible drainage with myringotomy or ventilation tube
Besides middle ear atelectasis, what other problem can be caused by ETD?
Cholesteatoma – skin/keratin debris trapped behind the ear drum; usually arise at the top of the eardrum (pars flaccida)
What is the problem with middle ear atelectasis?
Collapse of the middle ear (severely retracted) – end-result of chronic and severe Eustachian tube dysfunction (ETD)
What treatment can be given for the treatment of otitis media with effusion?
Antibiotics, possible decongestants, possibly steroids, tincture of time (most important), myringotomy, ventilation tubes
What can be seen in otitis media with effusion?
A retracted TM, air-fluid levels (bubbles), discoloration, impaired mobility, hearing loss, not painful
What are the complications of AOM?
TM perforation (usually heal spontaneously), meningitis, brain abscess, facial nerve paralysis, labyrinthitis (otitis interna --> can predispose to meningitis and permanent hearing loss), otitis media with effusion
What antibiotics are used for AOM?
β-lactams (penicillins/cephalosporins), macrolides, sulfonamides, quinolones (in adults)
What are the two biggest clues to the condition of the middle ear?
Translucency and color of the TM
What are the most common bacteria seen in AOM?
Strep. pneumo.; H. flu.; M. cat.
What are the symptoms of acute otitis media (AOM)?
Fever, otalgia, hearing loss (!), systemic symptoms
What treatment is used with otitis externa?
Gentle debridement, ototopical medications (ear drops), wick placement if swelling prevents proper delivery of ear drops, occasionally oral antibiotics (ciprofloxacin)
What is the treatment of ABS?
Antibiotics (for Strep pneumo./H. flu./M. cat); decongestants, warm soaks to face, analgesics, rarely surgery (when ABS becomes complicated)
How does bacterial sinusitis compare to viral sinusitis?
The duration differs (viral lasts about 7-10 days, whereas bacterial lasts longer); there is more intense nasal obstruction with bacterial sinusitis as well as purulent discharge (in the nose)
What symptoms are associated with ABS?
Common symptoms: nasal obstruction, rhinorrhea (usually purulent), HA, facial pain;
Variable symptoms: odontalgia, fever, sore throat
What is the challenge in diagnosing acute bacterial sinusitis (ABS)?
It is very hard to distinguish
from allergic rhinitis, viral URI, or nasal masses
What is the treatment of malignant otitis externa?
Treat aggressively with systemic antibiotics against pseudomonas and possible surgical debridement
What is characteristic of malignant otitis externa?
Usually seen in diabetics; most commonly caused by pseudomonas; can lead to necrotizing osteitis of skull base and paralysis of cranial nerves (e.g. VII, XI); often fatal
What microbes are associated with otitis externa?
Staph, pseudomonas, fungi
What characterizes otitis externa?
Infection/inflammation of external auditory canal causing severe pain (especially on manipulation of pinna), drainage, swelling, and hearing loss (if canal is completely occluded)
What symptoms are seen with cholesteatoma?
Pain, hearing loss (due to the production of osteolytic enzymes), recurring otorrhea
What differentiates the early phase from the late phase reaction in allergic rhinitis?
Early: pruritus, urticaria, vasodilation/leakage, bronchoconstriction, mucous secretion;
Late: infiltration with eosinophils, fibrin deposition, infiltration with monocytes, tissue destruction
What hormonal imbalances can cause rhinitis?
Pregnancy, hypothyroidism
What is the problem with the use of nasal decongestant sprays?
Although they provide immediate relief to patients with nasal congestion (by causing vasoconstriction), once the effects wear off, they are followed by a rebound nasal vasodilation that will be worse than their initial problem (thus causing continuous use of the nasal spray)
What characterizes allergic shiners and Dennie lines?
Allergic shiners: dark discoloration under the eyes; Dennie lines: folds below the lower eyelids;
these signs are due to chronic obstruction of nasal venous return and hypoxia leading to spasm of striated muscles of Mueller
What characterizes the nasal salute?
Maneuver of scratching the nose by lifting the tip (temporarily improves congestion); leads to a nasal crease developing right above the nasal tip
What are the characteristics of an adenoid face?
Chronic mouth breathing which results in hypognathic jaw, open-mouthed facies, arched palate
What characterizes nasal polyps?
Benign growths arising from nasal or sinus mucosa; unclear etiology; treat underlying factors; if symptomatic, surgical extirpation may be necessary (send to pathologist)
What factors are important in diagnosing allergic rhinitis?
Symptoms: hypersecretion (rhinorrhea), itching, irritation (sneezing/burning), congestion;
Seasons/circumstances: spring (grasses/trees/molds), summer (grasses), fall (weeds/molds), winter (dust mite, animal dander) – molds can be year-round antigens;
Family history: there is a genetic predisposition (ask about allergies/asthma) --> 1 parent allergic: 1/3 chance for child; 2 parents allergic: 2/3 chance for child
What type of immunological reactions is associated with allergic rhinitis?
Type I (Gell and Coombs)
What is an antrochoanal polyp?
A polyp originating in the maxillary sinus protruding in the middle nasal meatus (may be visible behind the soft palate if very large)
What distinguishes acute tonsillitis from infectious mononucleosis?
In mono, the lymph nodes affected are mostly posterior whereas with Strep pyogenes they are mostly anterior
What causes biphasic stridor?
Usually a more severe obstruction at the level of the larynx or below (sign of impending airway obstruction)
What differentiates inspiratory vs. expiratory stridor?
Inspiratory stridor is caused by extrathoracic obstruction (vocal cords and above);
Expiratory stridor is caused by intrathoracic obstruction (in the lungs, upper trachea, subglottis)
What are some examples of laryngeal dysfunction?
Aspiration, hoarseness (usually first symptoms to present), cough, dyspnea/stridor
What is the main function of the larynx?
Airway protection (keep food/fluids out of the lungs); also important are respiration and speech
How is tonsil size graded?
Grade 1+: totally contained within the tonsillar fossa; Grade 2+: barely protruding out of the tonsillar fossa; Grade 3+: protrude to touch the uvula; Grade 4+: tonsils touch in midline
What are the indications for tonsillectomy?
Obstructive hypertrophy (interferes with breathing or swallowing); Recurrent infection (6 episodes in one year, or 3-4 per year for 2+ years)
What is the management of allergy?
Environmental control (most effective and difficult); Pharmacotherapy; Immunotherapy (when other options fail) – avoidance --> antihistamines --> corticosteroids --> immunotherapy
What symptoms are associated with acute tonsillitis?
Fever, sore throat, odynophagia – symptoms are frequently caused by Strep pyogenes
What is the usual treatment of acute tonsillitis and what complication can occur?
Antibiotics – if an abscess forms underneath the tonsil, it may have to be removed – peritonsillar abscess will cause the tonsil to become displaced and the patient will present with pain on opening the mouth (trismus)
What can a wart-like growth on the vocal cords indicate?
Laryngeal papilloma
What aspects are included in a neck mass evaluation?
History (duration, mode of onset, symptoms); Complete head and neck exam (size, location, tenderness, mobility, consistency); Fine needle aspiration biopsy (most important test);
CT, ultrasound, other radiological evaluations
What are the etiologies associated with a neck mass?
Infectious/inflammatory, neoplastic, congenital (cysts) – neck masses should be assumed to be malignant until proven otherwise (especially when asymptomatic and not present from birth)
What characterizes laryngeal cancer?
Classic symptom triad: hoarseness, sore throat, otalgia (!); 95% are squamous cell carcinomas; risk factors include tobacco and alcohol (if taken both, synergisms will occur); if the vocal cord loses mobility, the cancer will be more progressed – treatment with surgery, chemotherapy, radiation (with metastasis)
What is the treatment for an oropharyngeal papilloma?
Cut it off (no anesthesia necessary)
What causes recurrent respiratory papilloma?
HPV 6 and 11; more common in children (transmitted in birth canal), may require multiple surgical procedures; if they migrate into the trachea, treatment will be complicated
What are causes of hoarseness?
Vocal cord paralysis, vocal cord mass, functional deficits (muscle tension dysphonia/spastic dysphonia)
What characterizes vocal cord nodules (screamers/singers nodules)?
Slight elevations on the vocal cords
What characterizes blastomycosis?
Marked epithelial hyperplasia; Non-specific symptoms; CXR: lobar consolidation, multilobar/ perihilar infiltrates, multiple nodules; Organisms undergo broad-based budding (!)
What distinguishes Mycobacterium tuberculosis from Mycobacterium avium-intracellular complex (MAC)?
TB: pathogen, immunocompromised patients (not severely), florid granulomatous inflammation with caseation, few bacilli noted in tissue and stains;
MAC: opportunistic infection, severely immunocompromised patients, no florid granulomas/ caseation, numerous bacilli noted in tissues and stains
How can a person acquire intestinal TB?
Either by a disseminated form of TB or by drinking contaminated milk
Where in the lungs does TB usually reside?
Distal airways of upper lobes (subpleural)
What are common opportunistic lung infections?
Diffuse: Pneumocystis, CMV; Focal: Gram-negative rods, S. aureus, Aspergillus, Candida
What characterizes coccidioidomycosis?
Ubiquitous in SW US, most asymptomatic; Patient can have lung lesions, fever, cough, pleuritic pain, erythema nodosum/multiforme; Rarely a disseminated form can develop; C. immitis consists of thick-walled, non-budding spherules filled with small endospores within macrophages/giant cells; Can affect lungs, meninges, skin, bones, adrenals, lymph nodes, spleen, liver
What geographical locations are associated with fungal infections responsible for causing chronic pneumonia?
Histoplasma: Ohio, Mississippi, Caribbean; Blastomyces: Central/SE US;
Coccidioides: SW US, Mexico
What characterizes histoplasmosis?
Caused by inhalation of bird/bat droppings; Intracellular parasite of macrophages; Resembles TB (coin lesions, apex, night sweats), but mostly asymptomatic; Patient can have epithelioid granulomas, coagulative necrosis, concentric fibrosis (“tree-barking”)
Which lung cancers are considered central/hilar?
Squamous cell carcinoma, small cell carcinoma – they are more easily diagnosed on sputum
How does squamous cell carcinoma appear on sputum cytology?
Orange-pink cytoplasm with dark, pyknotic nucleus
How does squamous cell carcinoma appear on H&E?
Large, pink cells
How does squamous cell carcinoma appear radiologically?
As cavitary lesions
What genetic changes are seen with squamous cell carcinoma?
p53 overexpression (highest frequency); also Rb loss, p16INK4a mutation, EGFR and Her-2/Neu overexpression
What characterizes the spread of squamous cell carcinomas?
Spread locally, metastasize later; rapid rate of growth; may see associated pre-malignant lesion (i.e. squamous metaplasia, dysplasia)
In what age group does lung cancer occur mostly and what is the 5-year survival rate?
Ages 50-60; 5-year survival: 15%
What is the classification of lung cancers?
Non-small cell carcinomas: squamous cell carcinoma (current smokers), adenocarcinoma (past smokers), large cell carcinoma; Small cell carcinoma (non-surgical disease)
What molecular genetics are associated with lung cancers?
Small/non-small cell carcinoma: p53; Small cell only: c-myc, Rb; Non-small cell: ras, p16INK4a; also, EGFR, Her-2/Neu
What industrial hazards are associated with smoking?
Radon progeny, asbestos
What characterizes neuroendocrine carcinomas (small cell carcinomas)?
Strong association with smoking; Central/hilar location; Aggressive, metastasize early and often (sites of metastases frequently diagnosed before small cell carcinoma in lung);
Associated with paraneoplastic syndromes: ACTH, ADH
What characterizes a small cell carcinoma on H&E?
Small blue cells, round-oval, little cytoplasm, salt&pepper chromatin pattern, high mitotic grade, cells tend to mold against each other; used to be called oat cell carcinomas
What is meant by the Azzopardi effect?
Deposition of DNA around vessels, seen with small cell carcinoma
What is the prognosis for small cell carcinoma?
Untreated: 6-17 weeks; very radio-sensitive; potential cure rates 15-25% for limited disease
What genetic changes are seen in small cell carcinoma?
p53, Rb mutations; Bcl-2 expression
Where do small cell carcinomas originate from?
Argentaffin (Kulchitsky) cells in bronchial epithelium
What characterizes adenocarcinoma?
Incidence is increasing since more common in ex-smokers; They are peripheral in origin and slow growing; k-ras mutations are most common
What characterizes bronchioloalveolar carcinoma?
Subtype of adenocarcinoma; Peripheral location: multiple lesions with diffuse consolidation;
Growth along septae (mucinous spreads aerogenously vs. non-mucinous rarely aerogenously)
How does adenocarcinoma appear on sputum culture?
Contains ample cytoplasm and vacuoles
How does adenocarcinoma appear on H&E?
Glandular, spherical
What secondary pathology is associated with lung cancers?
Partial obstruction (marked focal emphysema), total obstruction (atelectasis), impaired airway drainage, pulmonary abscess, SVC syndrome, pericarditis/pleuritis
What is seen in carcinoid syndrome?
Diarrhea, flushing, cyanosis – due to the secretion of vasoactive amines
How are carcinoids classified?
Typical: <2 mitoses/10hpf; Atypical: 2-10 mitoses/10hpf and/or necrosis
What are tumorlets?
Small, inconsequential hyperplastic neuroendocrine cells
What paraneoplastic syndromes are seen with lung cancer?
ADH (hyponatremia), ACTH (Cushing’s), PTH (hypercalcemia), Calcitonin (hypocalcemia), Gonadotropins (gynecomastia), Serotonin/bradykinin (carcinoid syndrome)
What characterizes the clinical presentation of lung cancers?
Cough > Weigh loss = Chest pain > Dyspnea; may present as metastasis
How does a patient with SVC syndrome often present?
Swollen, edematous neck
Why are small cell carcinomas associated with paraneoplastic syndromes?
They contain secretory granules (seen on EM) that contain neuroendocrine hormones (e.g. ACTH)
What characterizes large cell carcinomas?
Undifferentiated with large, atypical cells
Between what two structures are the palatine tonsils located?
Anterior tonsillar pillar (palatoglossal fold) and posterior tonsillar pillar (palatopharyngeal fold)
How do bacteria cause breakdown of the enamel/cementum?
The bacteria metabolize sugars and in the process liberate acids that lead to decalcification
What characterizes dental caries?
An infectious disease that begins as chalky white decalcification of enamel/cementum, becomes more discolored (yellow, brown, black) eventually leading to brittle enamel/cementum; when the pulp becomes involved, pain and abscess develop
What is the ligament called that goes from the tooth to the alveolar bone in which it sits?
Periodontal ligament
What causes a tooth abscess?
Infection of the pulp of the tooth
What are the layers of the tooth from the outside in?
Enamel, dentin, cementum
How many deciduous teeth and how many permanent teeth do humans normally have?
Deciduous (20 total): 4 incisors, 2 canines, 4 molars (each half --> 2-1-2)
Permanent (32 total): 4 incisors, 2 canines, 4 bicuspids, 6 molars (each half --> 2-1-2-3)
What ducts are associated with the salivary glands, and where do they drain, respectively?
Stenson’s duct: parotid gland, drains out in the buccal mucosa opposite the 2nd maxillary molar;
Wharton’s duct: submandibular gland, drains in the floor of mouth adjacent to lingual frenulum;
The sublingual gland has multiple unnamed/invisible openings in the floor of the mouth
What is the relationship between size of the salivary gland and malignancy of an associated tumor?
The larger the gland (e.g. parotid) the more commonly it will have benign tumors associated with it, whereas minor salivary glands will almost always have malignant tumors associated with it
What is the problem with sialolithiasis and sialadenitis?
Sialolithiasis: stones in the salivary ducts leading to obstruction;
Sialadenitis: infection/inflammation of ducts due to obstruction (pain worse with eating)
What is benign migratory glossitis (geographic tongue)?
Areas of discoloration of the tongue that appear in different locations at different times
What is the treatment associated with aphthous ulcers?
Usually none required; there is no treatment that is beneficial to all patients, but silver nitrate cautery, topical steroids, pasty salves, tetracycline rinse, and other treatments have been used
What characterizes aphthous ulcers?
Small, painful ulcers found on unattached gingiva (alveolar mucosa); disease of unknown cause
What is acute necrotizing ulcerative gingivostomatitis also known as and what characterizes it?
Trench mouth – comes on rapidly and can be very painful and severe; patient gets punched-out-papilla; completely reversible if caught early (use same treatment as for gingivitis)
What is the treatment for gingivitis and periodontitis?
Removal of plaque and calculus; meticulous oral hygiene, surgical debridement of diseased gingiva and bone; antibiotics
What characterizes gingivitis?
An infectious disease of plaque that causes the gingiva to become red, swollen, friable, tender or painful; untreated it will lead to a loss of alveolar bone (i.e. periodontitis), but in its early stages is completely reversible
What are the systemic consequences associated with periodontal disease?
The inflammatory mediators released with periodontal disease can affect multiple organ systems and increase a patient’s risk for conditions such as heart disease, stroke, diabetes
What is the purpose of fluoride in preventing dental caries?
It makes enamel more resistant to decalcification
What are Fordyce spots (granules)?
Yellow-white spots found in the buccal mucosa; they represent ectopic sebaceous glands
What is torus palatinus and torus mandibularis?
Torus palatinus: bony lump (osteoma–benign excrescence of bone) in the palate (usually midline);
Torus mandibularis: osteomas found along the lingual surface of the mandible in the vicinity of the canine and bicuspid teeth (usually symmetric)
What is major aphthous stomatitis also known as and what characterizes it?
Sutton’s disease: large deep ulcers with sharp borders that can last for several months; patients often have several ulcers at a time; etiology is unknown but thought to be autoimmune
In what people is oral candidiasis seen?
Infants and people taking antibiotics or inhaled steroids
What are some triggers associated with secondary HSV infections and what characterizes them?
UV light, menstruation, stress, trauma – most commonly occur as a single lip lesion; lesions are sometimes announced by an initial tingling (at which time treatment should be applied); if lesions occur intraorally, they would most likely be found on keratinized mucosa (as opposed to aphthous ulcers appearing on nonkeratinized mucosa)
What is the treatment associated with a primary HSV infection?
Usually no treatment is needed and symptoms will resolve in 1-2 weeks (however, virus will remain dormant in trigeminal ganglion) – acyclovir, valacyclovir, and famciclovir can help reduce severity of disease (but not eradicate it)
How does a primary HSV infection usually present?
Most common in children/young adults; seen as multiple discrete vesicles on the mucosa of the mouth/lips/face(occasionally) which quickly rupture causing severe pain and surrounding erythema/edema; patients can have a seven day prodrome with slight fever, malaise, and cervical lymphadenopathy
What effect do benzodiazepines have?
Potentiation of the effects of GABA (as opposed to barbiturates, which can act without the presence of GABA)
What are the BZ1-selective (α1) benzodiazepines (associated with sedation)?
Zolpidem, Zaleplon, Zopiclone, Eszopiclone
What is the α2 subunit of the GABAA receptor critical for?
Anxiolytic effects
What is the α1 subunit of the GABAA receptor critical for?
Sedative effects
What effects, besides anxiolytic, are associated with Diazepam?
Sedative, anticonvulsant, cognitive impairing, muscle relaxant, potentiation of alcohol
For which GABAA subunits does Diazepam have a high affinity?
α1, 2, 3, 5 – they share a conserved histidine-101 required for Diazepam binding
What is the difference between BZ1 and BZ2 receptors?
Both are involved in anxiolytic effects; BZ1: associated with sleep, Zaleplon/Zolpidem exhibit selectivity; BZ2: muscle relaxation
What brain areas are associated with anxiety?
Amygdala: activated in fear/anxiety responses, GABA inhibits neuronal activity;
Hippocampus: storage of emotional memories;
Raphe nucleus: serotonergic cell bodies, widespread projections to forebrain
What are the clinical uses of benzodiazepines?
Situational anxiety, generalized anxiety disorder, panic disorder, combined treatment (for early actions)
What are the classes of anxiolytic drugs?
Benzodiazepines, Azapirones, Beta blockers, SSRIs, others
What are contraindications of benzodiazepines?
Acute alcohol intoxication, abrupt discontinuation – Cautions: pregnancy, lactation, children/elderly (increased sensitivity)
What are beta blockers (Propranolol, Nadolol) used for regarding their anxiolytic function?
Situational anxiety: reduced physiological symptoms of anxiety (i.e. sympathetic effects, feedback mechanism to CNS)
What contraindications are associated with Buspirone?
Lacks those of benzodiazepines; Contraindicated with MAOIs, CYP interactions
What are the pharmacological effects of Buspirone?
Decreased motor tension (not a muscle relaxant), decreased autonomic hyperactivity, decreased vigilance and scanning – Lacks may effects of benzodiazepines: not an anticonvulsant, not a muscle relaxant, does not impair psychomotor function, minimal sedation, no physical dependence
What is the mechanism of action of Buspirone?
Partial 5-HT1A agonist; D2 antagonist
What is the only drug in the Azapirones class available for commercial use?
Buspirone: used for generalized anxiety disorder; slow onset of action (3-6 weeks), not used for other anxiety disorders
What drug interactions are seen with benzodiazepines?
CNS depressants: synergistic CNS, cardiovascular, respiratory depression; CYP interactions
What are the BZ2-selective (α2, 3, 5) benzodiazepines (associated with the anti-anxiety/anti-convulsant effects)?
Alprazolam (high potency, short duration), Chlordiazepoxide (low, long),
Clonazepam (high, long), Diazepam (low, long), Lorazepam (high, short), Oxazepam (low, short)
What adverse effects are associated with benzodiazepines used as anxiolytics?
Sedation, impaired cognition and motor function; Cardiovascular and respiratory depression with overdose, Teratogenicity; Paradoxical CNS stimulation; Physiological dependence (especially with short half-life drugs)
What are the pharmacological effects of benzodiazepines?
Sedation, hypnosis, anesthesia, anticonvulsant, anxiolytic, muscle relaxant – effects are rapid
What characterizes the use of SSRIs regarding their anxiolytic function?
Used for most types of anxiety disorders (not for situational anxiety); slow onset of action (weeks) – Fluvoxamine is used for OCD
What characterizes the use of antihistamines regarding their anxiolytic function?
Cause short-term sedation – used in patients who cannot take other anxiolytics
What characterizes Chloral Hydrate?
Occasionally used for situational anxiety, narrow therapeutic window
What characterizes the use of SSRIs regarding their anxiolytic function?
Used for most types of anxiety disorders (not for situational anxiety); slow onset of action (weeks) – Fluvoxamine is used for OCD
What characterizes the use of antihistamines regarding their anxiolytic function?
Cause short-term sedation – used in patients who cannot take other anxiolytics
What characterizes Chloral Hydrate?
Occasionally used for situational anxiety, narrow therapeutic window
What characterizes Group A Streptococci (GAS)?
Spherical cells that grow in chains; they are non-motile and spore-forming; they have fastidious growth (complex nutrient requirements), are facultatively anaerobic, and produce a capsule
What secreted factors are produced by GAS?
Streptolysin O, hyaluronidase, pyrogenic exotoxins, NADase
What are examples of other infections caused by GAS?
Pneumonia, meningitis, sinusitis, otitis
What diseases are caused by GAS exotoxins released into the bloodstream?
Scarlet fever, toxic shock syndrome
What are examples of non-suppurative GAS infections?
Acute rheumatic fever, acute post-streptococcal glomerulonephritis
What are examples of suppurative (pus forming) GAS infections?
Pharyngitis, tonsillitis, impetigo, erysipelas, cellulitis, necrotizing fasciitis, myositis
What diagnostic criteria are associated with GAS?
Gram+ cocci, catalase-negative, β-hemolytic on blood agar; can use Rapid Antigen Detecting Tests (RADT)
What are the classes of Streptococci based on hemolytic patterns?
α (greenish), β (clear), γ (none, non-hemolytic)
What is the classification for Streptococcus pyogenes?
Group A, β-hemolytic
What is the Lancefield classification of Streptococci based on?
Group-specific antigens (A-U)
What is currently used for epidemiological GAS typing?
emm-types: sequencing of 5` end of emm gene (codes for M protein)
What is the purpose of Hyaluronidase?
It hydrolyzes hyaluronic acid thus helping GAS invade deeper tissues
What is the purpose of Streptolysin O?
It is responsible for the characteristic β-hemolysis and it is toxic to a variety of human cells
What is the purpose of Streptolysin S, produced by GAS?
It attacks eukaryotic cell membranes and makes pores
How does GAS cause the breaking up of blood clots?
It produces streptokinase (converts plasminogen to plasmin which degrades the fibrin clot)
What anti-phagocytic proteins are made by GAS?
M-like protein which binds plasma inhibitors and IgGs; C5a peptidase which cleaves the chemoattractant C5a (thus inhibiting the influx of PMNs to the site of infection)
What is the purpose of Fn-binding proteins?
They bind fibronectin and thereby enhance host colonization
What is a purpose of lipoteichoic acid?
It has adhesive properties to mucous membranes
What characterizes M protein of GAS?
Virulence factor; dimeric protein that coils around itself; carboxy-terminal inserts in GAS cell membrane, whereas the amino-terminal contains a hypervariable region (elicits antibody response); M protein has sites that bind factor H, fibrinogen, and IgG, which inhibits opsonization and phagocytosis
What is the most important virulence factor of GAS?
Hyaluronic acid capsule – chemically identical to connective tissue components, so it functions as camouflage; it also has antiphagocytic properties
What is the most common cause of heart disease in children in developing countries?
Acute rheumatic fever
What is the therapy for strep throat?
No licensed vaccine available; single IM injection of penicillin G, or can give penicillin V orally (all strain are susceptible) – erythromycin is used in patients with a penicillin allergy (some strains are resistant)
What is the gold standard for strep throat diagnosis?
Throat culture (presence of GAP colonies confirms diagnosis, but about 10% of culture-negative patients have strep throat); if the condition are severe a rapid antigen-detecting test should be performed (while waiting on blood agar test)
What other agents can cause pharyngitis besides GAS?
Viral, other bacterial infections, mycoplasma, chlamydia
What epidemiology is associated with GAP pharyngitis?
5-15yo children; occurs mostly in late winter/early spring, often in military personnel; it spreads by person-to-person contact and there are many asymptomatic carriers (15-20%)
What causes scarlet fever?
GAP strains that produce erythrogenic exotoxins; associated with pharyngotonsillitis; present with scarlatinal rash (starts on trunk --> extremities), and “strawberry tongue”; in severe forms, septic and toxic scarlet fever can occur (very rare)
What characterizes GAS pharyngitis?
Sudden onset of sore throat, fever, HA, abdominal pain, n/v; Physical findings include pharyngeal erythema/edema, enlarged tonsil with white exudate, uvulitis
What is Streptozyme?
An screening test that looks for GAS antibodies
What can be caused by GAS pyrogenic exotoxins?
They can function as superantigens thus non-specifically activating T cells, inducing a massive inflammatory response – they are the hallmark for toxic shock syndrome
What are the three GAS pyrogenic exotoxins?
SPE A and SPE C: phage encoded; SPE B: cysteine protease that degrades connective tissue
What clinical signs are associated with acute glomerulonephritis?
Glomerular lesions, edema, HTN, hematuria, proteinuria; also immune complexes of IgG and C3 deposited in the glomerular basement membrane – treated with penicillin/erythromycin
What is the treatment of necrotizing fasciitis?
Debridement of fasciotomy, amputation of 1-4 extremities; also penicillin/clindamycin
What characterizes necrotizing fasciitis?
Infection of deeper tissues/fascia with extensive and rapid spreading necrosis and gangrene; mortality can reach 70% within 2-3 days; signs include fever, severe pain, portal of entry not always obvious – diagnosis is made by surgical exploration with Gram-stain and biopsy
What is Erysipelas?
A form of cellulitis which often involves the face (butterfly distribution)
What characterizes streptococcal cellulitis?
Acute, spreading inflammation of the skin, usually as a result from infections of burns/wounds/ surgery; systemic signs include fever, chills, bacteremia – treated with penicillin or vancomycin (when staph cannot be excluded)
What are the signs associated with streptococcus pyoderma?
Crusty lesions (amber crust, honey-yellow crust), GAS culture, anti-strep antibodies
What is streptococcal pyoderma also known as and what is the epidemiology?
Impetigo: found mostly in young children in tropical climates; usually associated with poor hygiene; the GAS skin strains are genetically different from those causing pharyngitis
What are the Jones criteria for acute rheumatic fever (ARF)?
Major: carditis, migratory polyarthritis (most commonly), Sydenham’s chorea (unique to ARF), erythema marginatum (skin rash on trunk, unique to ARF), subcutaneous nodules;
Minor: less specific symptoms – need 2 major or 1 major/2 minor for diagnosis
What are the three requirements for an autoimmune disease to occur?
Significant antigen sharing; Abnormal immune response (humoral/cellular); Genetic predisposition to abnormal reaction
Why does GAS cause autoimmune symptoms in ARF?
Molecular mimicry (antigen sharing)
What is the pathogenesis of C. diphtheria?
Colonizes pharynx after inhalation --> 2-4 days later the patient will have a mild sore throat and a low grade fever --> 12-24 hours later a local lesion in the tonsil area will appear creating white exudate and fibrin network (due to local toxin) known as a pseudomembrane --> patient may develop localized edema (bull neck) --> bacteria do not invade beyond the throat lesion, but the toxin they produce disseminates systemically (heart, muscles, nerves, kidneys, liver, spleen)
What characterizes diphtheria toxin?
One diphtheria toxin can kill an entire cell
How does diphtheria toxin get into the cell and what happens subsequently?
The HB-EGF/CD9 receptor binding domain binds to the DTR and is taken up by endocytosis --> the AB-toxin is cleaved by Furin --> the vessel fuses with a lysosome which acidifies it --> this causes a deformation in the translocation fragment which forms a pore --> the A-fragment is released through this pore --> the A-fragment contains a signal sequence that translocates it to the ER where it is able to transfer ADP-ribose from NAD to EF2 causing protein synthesis to halt
What is the receptor that the B-fragment of diphtheria toxin binds?
Diphtheria toxin receptor (DTR): heparin-binding epidermal growth factor precursor protein/CD9
What is the mechanism of diphtheria toxin?
It transfers an ADP-ribose from NAD to EF2, thus inactivating EF2 --> since EF2 is an elongation factor used in protein synthesis, the cell can no longer make proteins and dies
What are the components of the diphtheria toxin?
AB-toxin --> A-fragment: ADP-ribosyltransferase (EF2); B-fragment: internalization domain (contains many α-helices), HB-EGF/CD9 receptor binding domain
What is the clinical presentation associated with diphtheria?
Sore throat/fever, severe distress, pseudomembrane, lack of immunization, travel to diphtheria endemic region, high WBC count; also, punched-out non-healing ulcers (more rare)
What is the hallmark of diphtheria?
Formation of a pseudomembrane in the mouth
How does diphtheria spread?
By droplets and direct contact
What are the two diseases caused by Corynebacterium diphtheriae?
Localized respiratory tract infection; Localized skin infection – both localized infections produce a systemic toxin
What laboratory tests aid in the diagnosis of C. diphtheria infection?
Gram stain showing Gram+ rods, aerobic, non-motile; Bacteria are club or V-shaped, resembling Chinese letters; Culture from mucosa or skin on selective tellurite medium (exclude normal flora); Non-hemolytic on blood agar; Test for diphtheria toxin via PCR/immunological/cytotoxicity
What A-fragment is used in current immunotoxins and why?
A pseudomonas exotoxin (PE38), since humans are not immunized against it
What is an important function of immunotoxins?
They can selectively localize cytotoxic drugs to tumor cells – can be made by altering the receptor binding domain of AB-toxins
What part of the AB-toxin should be targeted with vaccines?
The receptor binding domain (on the B-fragment)
What is done when a toxoid vaccine made by formaldehyde treatment is ineffective?
Recombinant toxin vaccines are used (mutant, enzymatically inactive forms of toxin)
What therapy is ineffective with diphtheria?
Antibiotics, innate immune response (since treatment has to be antibody-mediated)
What is the treatment for diphtheria?
Treat with antitoxin as soon as possible; Antibiotics (penicillin/erythromycin); Supportive (ventilation); Immunize at discharge (toxoid vaccine); Give booster/antibiotics to patient contacts
How is release of diphtheria toxin regulated by C. diphtheria?
Under normal circumstances, iron is bound to a diphtheria toxin repressor protein which is bound to the promoter sequence associated with diphtheria toxin --> when iron levels drop, this repressor protein will release, allowing the bacterial machinery to produce the diphtheria toxin –
the toxin is encoded on a phage
What is the normal immunization schedule for diphtheria?
5 doses of DTaP (diphtheria, tetanus, acellular pertussis) at 2, 4, 6, 12-15 months, and 4-6 years;
then again at age 11-16 years and subsequently every 10 years
What does the diphtheria vaccine consist of?
Formalin treated purified toxin (made in horses)
What is the most important virulence factor associated with H. flu?
Its capsule: polyribosyl ribitol phosphate (PRP) – there are 7 capsule types (PRP is type B)
How do the non-typeable H. flu strains differ from the capsulated forms?
They are less invasive; they induce inflammatory disease and can be opportunistic; they are associated with otitis media, conjunctivitis, sinusitis, and pneumonia
Besides meningitis, what other complications can be seen with an H. flu infection?
Acute epiglottitis (can result in airway obstruction/suffocation); Childhood septic arthritis, cellulitis
What does the H. flu vaccine consist of?
It is protein conjugated to PRP-polysaccharide (the added protein is important since children <18mo do not have a T-cell independent antigen response to the polysaccharide component only)
What is the treatment for H. flu?
Nafcillin, Cephalosporin (for meningitis), Tetracycline/Sulfa drugs (for non-typeable H. flu), Corticosteroids (for brain inflammation)
What is the diagnosis of H. flu based on?
Presumptive diagnosis is based on H&P; Established diagnosis is based on positive cultures for H. flu (should be serotyped to identify PRP capsule – type B)
What characterizes the transmission of H. flu?
Person-to-person contact (via secretions/aerosol); humans are the only reservoir; peak incidence if unvaccinated is 6-11 months
What is the differential diagnosis associated with acute exacerbation of chronic bronchitis?
Non-typeable Hemophilus influenza; Streptococcus pneumoniae/Moraxella catarrhalis (more likely to see pneumonia), viruses (adeno, influenza, parainfluenza – would not see sputum)
What are the signs/symptoms associated with H. flu?
Initially: runny nose, low-grade fever, HA (1-3 days); invasive organism: enters the circulation, crosses BBB, rapidly progresses to meningitis (stiff neck), convulsions, coma, death (almost 100% fatal without treatment; with treatment 5-10% mortality)
What characterizes Hemophilus influenza?
Blood loving: requires factors V (NAD) and X(hematin); grows on chocolate agar (lysed RBCs); grows better in CO2 enriched atmosphere; small Gram– coccobacillus, non-motile, facultative anaerobe; grows rapidly; nutritionally fastidious; does not grow on blood agar
What complications are associated with Pertussis?
Bronchopneumonia (linked to superinfection); Neurological damage (edema/hemorrhage due to coughing)
How is the diagnosis of Bordetella pertussis made?
Cough lasting > 2weeks, lymphocytosis; definitive diagnosis is made by recovery of organism from nasopharyngeal mucus (fragile organism, must be cultured on Bordet-Gengou agar); PCR
What are the key differences between the AB-toxin of Pertussis and that of Cholera?
The Cholera AB-toxin also have 5 B-fragments, but they are all the same (whereas Pertussis have 4 different fragments) – Cholera toxin binds to the GM1 ganglioside and causes an increase in cAMP by ribosylating Gs (prevents Gs inactivation) whereas Pertussis toxin binds to S2-5 and causes an increase in cAMP by ribosylating Gi, Gt, and Go (causes Gi inactivation)
What kind of toxin in Pertussis toxin?
AB5-toxin: A-fragment contains the ADP-ribosyl transferase that targets trimeric G-proteins, whereas the B fragment consists of 5 segments that bind to different receptors – it is secreted by a type IV secretion mechanism, targets macrophages and lymphocytes and is internalized by receptor-mediated endocytosis
What other virulence factors are associated with Pertussis?
Adenylate cyclase; Tracheal cytotoxin (leads to characteristic cough); Dermonecrotic toxin (causes tracheal tissue death); Pertussis toxin (causes cell death)
What virulence factors are associated with Pertussis?
Fimbriae (pili that bind to β1 integrins); Filamentous hemagglutinin (FHA – target for antibodies); Pertussis toxin (complex) – virulence factors are associated with improving the ability to colonize and destroy ciliated respiratory epithelium
What symptoms are associated with Pertussis?
Local symptoms: acute inflammation, hypoxia, cough; Systemic symptoms: fever, peripheral lymphocytosis
What characterizes H. ducreyi, H. aegypticus, and H. parainfluenza?
H. ducreyi: causes genital ulcers (chancroid), requires only factor X; H. aegypticus: pink eye;
H. parainfluenza: requires only factor V
What characterizes the morphology of Bordetella pertussis?
Tiny, Gram– coccobacillus; strict aerobe, nutritionally fastidious, slow growing
What are the stages associated with Pertussis?
Organism colonizes URT (transmitter through air-droplets, highly contagious) --> 7-10 days later, patient enters catarrhal stage (profuse URT disease that mimics the common cold) --> 7-10 days after that, patient enter the paroxysmal stage (severe, repetitive paroxysmal cough followed by a prolonged inspiratory gasp – whoop) --> 2-4 weeks later patient will recover
What characterizes the Pertussis vaccine?
Inactivated pertussis toxin + FHA; part of DTaP vaccine
What is the treatment for Psittacosis?
Tetracyclines (e.g. Doxycycline)
How is the diagnosis of Psittacosis made?
Clinical history (illness, pet parrot), positive chlamydial Ab titer; for a definitive diagnosis, need serological tests
What is the disease process associated with Psittacosis?
5-14 day incubation period --> fever, chills, HA, malaise, myalgia, respiratory infection (cough, breathing difficulty, chest tightness) – in pregnant women it can cause severe illness, respiratory failure, thrombocytopenia, hepatitis, and fetal death
What disease does Chlamydia psittaci cause?
Psittacosis/Parrot fever: zoonosis (mainly from companion birds via inhalation of the aerosolized organism)
What characterizes Chlamydia psittaci?
Obligate intracellular parasite with a Gram– outer membrane, but no peptidoglycan; it depends on host ATP synthesis (energy parasite); it exists in two forms: elementary body (infectious phage, metabolically inactive), reticulate body (intracellular, non-infectious)
What are the key differences between H. flu and B. pertussis?
H. flu is an invasive organism that causes meningitis/otitis media in children and pneumonia in all age group; B. pertussis is a toxigenic disease that in non-invasive and causes whooping cough
What is the treatment of Pertussis?
Stage 1: erythromycin; Stage 2: provide supportive care; erythromycin can also be used prophylactically
What other diseases, besides pneumonia, can be caused by pneumococcal infections?
Meningitis, sinusitis, otitis media, bacteremia; also, less frequently: endocarditis, septic carditis, peritonitis
What molecules of Strep pneumo aid in its spread?
Autolysin: lyses cells and activates complement; Pneumolysin: inhibits beating of cilia, activity of PMNs, and is pro-inflammatory…
What molecules of Strep pneumo aid in its attachment to host cells?
Teichoic acid, choline-binding proteins, protein A, surface adhesin A, neuraminidase, hyaluronidase
What parts of Strep pneumo do cause activation of the immune system?
Peptidoglycan and teichoic acid --> stimulate cytokine production --> complement activation and infiltration of PMNs
How does Strep pneumo evade the immune system?
Phagocytic cells lack receptors for the capsule; Capsule masks surface antigens, which prevents complement fixation and subsequent ingestion
How is Strep pneumo diagnosed in the lab (4 criteria)?
Catalase negative, α-hemolysis on blood agar (green discoloration – seen with many oral bacteria), susceptible to optochin, soluble in bile salts
What characterizes Strep pneumo?
Gram+ cocci, α-hemolytic, replicate in pairs or chains
In what four categories can pneumonia be divided based on clinical manifestations, what are the major causative agents of each?
Lobar pneumonia (lobe/part): Pneumococci, H. influenza, Legionella;
Bronchopneumonia (patchy): Mycoplasma pneumoniae, respiratory viruses;
Interstitial pneumonia in immunosuppressed: viral etiology (influenza, CMV);
Lung abscess (severe): polymicrobial (normal mouth flora/nosocomial: K. pneumonia, S. aureus)
What is a common cause of the acute form of pneumonia?
Streptococcus pneumoniae
What are the classifications of pneumonia based on the progression of symptoms?
Acute: community-acquired, hospital-acquired (nosocomial); Subacute/chronic: tuberculosis, fungal, lung abscess; Pneumonia in the immunocompromised host
What are the classical symptoms associated with pneumococcal pneumonia?
Sudden onset of chills/sweats; high-grade fever; chest pain; cough with production of mucopurulent sputum; fatigue; grayish, anxious appearance (differentiates it from a mycoplasmal or viral etiology)
What antibiotics have very little/no resistance associated with Strep pneumo infections?
Quinolones; Vancomycin
What specifically has been responsible for Strep pneumo resistance to Erythromycin in the US and Europe, respectively?
US: M-phenotype (changes in cell wall, efflux pump); Europe: ermB-gene polymorphism
What has been responsible for penicillin resistance to Strep pneumo?
Mutations in PBPs – cross-resistance with Cephalosporins
What are predisposing conditions for pneumococcal pneumonia?
Defective antibody formation: agammaglobulinemia (primary reason), lymphoma, HIV infection;
Defective complement; Insufficient/poorly functioning PMNs; Splenectomized patients; Prior respiratory infections; Multifactorial causes (aging, diabetes)
What is the natural niche of Strep pneumo in humans?
Nasopharynx; 5-10% of adults/20-40% of children are carriers – transmission is via direct contact
What lab findings are associated with pneumococcal pneumonia?
Low hemoglobin (25% of cases); Leukocytosis (although normal WBC counts are common too, and even low WBC counts are possible) – can also do Gram stain on sputum (25% positive)
935. What is produced by all clinical strains of Strep pneumo?
Polymerized oligosaccharide capsule – important in human antibody response
What are the radiological findings associated with a pneumococcal pneumonia?
Fluid accumulation seen as a non-lucent area; typically, a single lobe is involved; there is a lack of lung abscess
What is the pathogenesis of Strep pneumo?
Infection of alveoli --> complement activation --> exudate and WBC activation --> severe inflammation --> pneumonia
What characterizes Legionella pneumophila?
Gram– bacillus, fastidious, thermophilic, found in ponds (inside unicellular protozoa), acquired by inhalation following exposure to water systems (hotels/nursing homes)
What is the most common reason for nosocomial pneumonias?
Use of ventilators in the ICU (ventilator-associated pneumonia – VAP)
What are the most common causes of early- and late-
onset nosocomial pneumonia?
Early-onset (<48 hours after admission): Pneumococci, H. influenza;
Late-onset (~6 days after admission): Gram– bacilli, S. aureus
What is the treatment of Legionaire’s disease?
Erythromycin or Quinolones
How does the chest x-ray of a patient with L. pneumophila infection appear?
Initially it appears similar to that of a Mycoplasma pneumoniae infection, but there is a rapid progression to multi-lobe stage with pleural effusion
What lab tests are useful in the diagnosis of L. pneumophila?
DFA, DNA hybridization, ELISA; culture is most specific but takes 3-5 days
What is the pathology of Legionella pneumophila?
Bacteria are inhaled from contaminated water --> invade and survive in alveolar macrophages causing Legionnaire’s disease (severe pneumonia); there is no person-to-person transmission – predisposing factors include old age, smoking, immunosuppression
What characterizes Mycoplasma pneumoniae?
Small, fastidious, slow growing, lack of cell wall, “fried-egg appearance”, milder than acute pneumonia causing sore throat/fever/cough
What is the treatment for a Mycoplasma pneumonia infection?
Erythromycin, Quinolones; (not β-lactams!)
What may be helpful in making the diagnosis of Mycoplasma pneumonia infection?
The presence of cold hemagglutinins: cause clumping of RBCs on ice; unilateral/bilateral patchy distribution in the lower lobes
What is the pathogenesis of nosocomial pneumonia?
Microaspiration (most common): aspiration of secretions of upper airway colonized with Gram– bacteria and S. aureus; Marcoaspiration (less common): infection with esophageal/gastric material (associated with ventilator tubing); Hematogenous spread of bacteria (rare)
What are the signs associated with nosocomial pneumonia?
Fever, leukocytosis, production of purulent sputum, presence of new lobar infiltrate – diagnosed by sputum inspection, radiograph, Gram-stain, blood/sputum cultures
Why does intubation lead to nosocomial pneumonia?
Biofilm development on the tube paves the way for infection
How are the M2 ion pores created by orthomyxovirus?
By alternative splicing of the gene encoding for the M1 matrix protein
What distinguishes between the human and the avian flu?
Human: H1-4; Avian: H5+
What is meant by the term antigenic shift?
Swapping of genes between two different viruses (especially when combining a human virus and a virus from another animal)
What causes the antigenic drift of certain viruses?
Mistakes made by the RNA-dependent RNA polymerase leading to mutations in the virus that enable it to bypass human immune defense mechanisms in a future infection
By what mechanism does assembly and budding of orthomyxoviruses occur?
HA, NA, and M proteins are delivered to the host cell membrane --> the 8 strands of –RNA contact these structural proteins --> budding occurs passively (some host membrane proteins may be acquired in this process) --> neuraminidases cleave off sialic acid from the virion to prevent other viruses from clumping to it
How are the 10 strands of +RNA created from the original 8 segments of viral –RNA?
The –RNA travels to the nucleus along with the RNA-dependent RNA polymerases where it uses small primers from the nucleus as well as host alternative splicing machinery to create the 10 strands of +RNA needed for its replication (it also makes copies of the –RNA for new virions)
How is influenza virus released from the endocytic vesicle in has gained entry to by the receptor mediated endocytosis?
As the pH is pumped down in the endocytic vesicle, HA undergoes a conformational change that reveals a fusion sequences, thus allowing the content to be released into the host cell cytoplasm – the lowered pH also creates an influx in hydrogen ions through the M2 ion pore, thus breaking up the interactions between the various viral proteins
What are the key differences between the influenza types?
Type A: most severe, infects humans/birds/other mammals; Type B: intermediate severity, only affects humans; Type C: common cold, only affects humans
What happens when hydrogen ions flow into the M2 ion pore?
The interactions between hemagglutinin, matrix protein (M1), RNA-dependent RNA polymerase (PB1/PB2), and the RNA are destabilized
What characterizes orthomyxoviruses?
Enveloped, helical, 8 segment, –RNA, contains neuraminidase (cuts through mucus) and hemagglutinin (HA - for host cell entry), has a hydrogen ion pore in membrane (M2 ion pore)
What is the mechanism of action of Ribavirin?
It is a purine analog that gets incorporated into viral RNA and terminates chain elongation – it is not specific for Influenza virus
What is the most contagious human virus known and what characterizes it?
Measles (rubeola): typified by fever, rash, and encephalitis, as well as Koplik’s spots (small, white spots on inside of mouth, multinucleated giant cells/syncytia) – transmission is through respiratory droplets/aerosol; outbreaks are highest in winter months
Which diseases are caused by paramyxoviruses?
Measles, mumps, parainfluenza (croup), respiratory syncytial virus
What are the key differences between paramyxo- and orthomyxoviruses?
Paramyxoviruses do not have a segmented genome, have an excellent RNA-dependent RNA polymerase (no antigenic drift), can cause syncytial formation (host cell fusion – multinucleated cells), and it does not enter by the endocytic route (also, replication occurs in the cytoplasm)
What people should not receive a killed influenza vaccine?
People allergic to eggs; people who have had severe reactions to an influenza vaccine; children less than 6 months old; people with an acute febrile illness; children on aspirin (Reyes syndrome)
What is the killed influenza vaccine made up of?
Two most prevalent Type A serotypes and the most prevalent Type B serotype found in the southern hemisphere during our spring/summer months
What steps in viral replication are blocked by Zanamivir/Oseltamivir?
Viral movement through mucus; Viral processing after release from the host cell (causes viruses to clump together)
Why is the avian flu harder to transmit from human to human?
It resides lower in the lung
Why is the drug Amantadine not used much anymore?
It binds the M2 ion pore and slows the viral life cycle down (hopefully long enough for a vaccine to take effect) – however, it has some cardiovascular side effects that make it less desirable
What are the clinical findings associated with the flu?
24-48 hour incubation period followed by fever, myalgia, HA, cough; resolves in 4-7 days;
in rare cases it can lead to Reyes syndrome and Guillain-Barre syndrome
What proteins are associated with paramyxoviruses?
HN protein (hemagglutinin), F protein (causes fusion of the virus with the host cell),
M protein (matrix proteins that cover the single –RNA strand)
What is the treatment for severe cases of respiratory syncytial virus?
Aerosolized Ribavirin
What is caused by parainfluenza and respiratory syncytial virus?
Pneumonia and bronchitis is infants, common cold in adults; they are mostly transmitted in the spring time; Parainfluenza virus can cause upper and lower respiratory tract infection with croup (hoarse cough)
How is mumps spread?
Coughing, sneezing, talking
What is affected in patients with mumps and what can this lead to?
Virus infects the upper respiratory tract, developing a viremia --> virus moves to glandular tissue (parotids, ovaries, testes, pancreas) – can lead to deafness, meningitis, encephalitis, sterility
What characterizes the MMR vaccine?
It is live, attenuated vaccine, which is highly effective – immunity for measles/mumps is life-long
How does infection with rubella present?
Mild rash and fever
What describes the life cycle of rhinovirus?
Attaches to host cell --> penetrates at the membrane and replicates in the cytoplasm --> makes a single polyprotein from its +RNA which then proteolytically cleaves itself into the individual viral proteins --> emerges from the cell when it dies and ruptures
What is the difference between enterovirus and rhinovirus?
Enterovirus grows optimally at 37 degrees C and pH 6.0 (targets intestinal tract) whereas rhinovirus grows optimally at 33 degrees C and NOT at pH 6.0 (targets respiratory tract)
What virus is responsible for the most common form of the common cold?
Rhinovirus – member of the picornavirus family (ss +RNA, icosahedral capsid, no envelope)
What replication strategy is uniquely associated with the alpha/toga/rubivirus family?
Subgenomic message strategy: replicates from 5` end first (for RNA-dependent RNA polymerase) followed by replication from the 3` end (structural proteins)
What describes the life cycle of rubivirus?
Attaches to host cell --> penetrates via endosome --> replicates in cytoplasm --> assembles at the membrane --> buds and takes membrane as an envelope
What is the biggest problem associated with rubivirus?
If a women contracts rubella during pregnancy (especially in 1st trimester), the virus will slowly kill fetal cells, resulting in a “Down syndrome”-like syndrome (MR, heart damage), cataracts, hearing impairment, and skin manifestations known as “blueberry muffin” lesions
What terms are used to describe German measles?
Rubella, Rubivirus (also togavirus, and it is part of the alphaviruses)
What characterizes rubivirus?
Grows slowly, ss +RNA with icosahedral capsid and envelope; (does not carry its RNA-dependent RNA polymerase with it inside its capsid – rather, it is encoded in its genome)
How is rubivirus transmitted?
Person-to-person via aerosolizes particles
What characterizes hantavirus?
Part of the bunyavirus family: three circular strands of ambisense (mostly –RNA) with a helical capsid (RNA-dependent RNA polymerase included) and an envelope
What is the treatment for adenovirus infection?
No therapy, but there is an inactivated vaccine that is given to military recruits
What form of the common cold is DNA based?
Adenovirus: ds DNA with pentons
How is hantavirus prevention accomplished?
Eliminate rodent food sources; Prevent rodents from entering buildings; Use continuous trapping methods
How is hantavirus transmitted?
Via aerosolized mouse urine (some rodents are chronically infected – deer mouse/cotton rat)
What is the lifecycle of hantavirus?
Attaches to host cell protein --> penetrates by endocytosis --> replicates in cytoplasm --> assembles and buds into vesicles (unique feature) --> vesicles transport viruses out to infect other cells
What disease is caused by hantavirus?
Hemorrhagic fever with renal syndrome; Hantavirus pulmonary syndrome
How is rhinovirus transmitted?
Aerosol or by touching an infected surface (hand to mouth or nose) – virus infects upper respiratory tract
What describes the diagnosis and treatment of rhinovirus?
Diagnosis is presumptive and there is no treatment (although disinfectants and hand washing work well in prevention)