• 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

1/2297

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;

2297 Cards in this Set

  • Front
  • Back
What are the three cancers for which regular screening is recommended?
Cancers of the colon, breast, and cervix.
When should screening for colon cancer begin in the patient with no family history of colon cancer?
Screening for colon cancer should begin at 50 with colonoscopy every 10 years. Annual fecal occult blood testing, sigmoidoscopy every 5 years, and barium enema are also options.
What are the screening mammography recommendations for breast cancer?
Every 1-2 years from age 40 and over. Breast examination by physician for ages 20-30, every 3 yrs. Patients with multiple first-degree relatives with breast cancer should receive prophylactic tamoxifen.
What is the prophylaxis for hepatitis A for travelers?
If a patient is leaving within 2 wks to a nonindustrialized country, vaccine and immune serum globulin are given. A booster shot given 6 months after the initial vaccination confers immunity for 10 years.
What is the most common cause of hepatitis A?
Hepatitis A is the most common vaccine-preventable disease in travelers; from fecal contamination of food or water.
Which travelers should receive hepatitis B vaccine?
Patients who will work with indigenous, engage in sex, receive medical/ dental care, on remain abroad >6 mth in a country where prevalence is intermediate or high.
What is the recommended prophylaxis for malaria for travelers?
Traveling to Mexico, Central America (except Panama), or Caribbean, chloroquine is prophylaxis for malaria. For travelers to areas where chloroquine resistance common, mefloquine recommended.
What is the recommended prophylaxis for malaria for travelers?
Rabies vaccination is recommended for patients traveling to areas where rabies is common (India, Asia, Mexico).
What is the vaccination recommendation for yellow fever for travelers?
Yellow fever vaccine is required for travel to sub-Saharan Africa and South American countries.
What are the vaccination recommendations for meningococcal meningitis?
Persons traveling to Nepal, sub-Saharan Africa, northern India should be immunized with the polysaccharide vaccine. Saudi Arabia requires immunization for pilgrims to Mecca.
At what age is meningococcal vaccine usually given?
Meningococcal is routinely given at age 11.
What are the recommendations for prevention of traveler's diarrhea?
Patients who experience loose stools without fever or blood may take loperamide. Treatment with a fluoroquinolone or azithromycin is reserved for patients with bloody diarrhea.
What are the recommendations for tetanus and diphtheria vaccination in adults who were never vaccinated?
Adults who were never vaccinated should receive three doses, the first two are given 1 to 2 months apart, with the third dose given 6 to 12 months later. A booster vaccination should be given every 10 years for life.
What are the recommendations for influenza vaccination?
Annually for adults >50. Cardiopulmonary disease, diabetes, hemoglobinopathy, or who are in chronic care facilities should receive annual. Pregnant women who will be in 2nd or 3rd trimester during influenza season.
What are the indications for pneumococcal vaccination?
Adults >65; sickle-cell, splenectomy, cardiopulmonary disease, alcoholism, cirrhosis, Alaskan natives, Native Americans; immunocomp (heme malignancies, CRF, nephrotic, HIV; immunosuppressives).
What are the indications for hepatitis B vaccination?
IV drug abuse, male homosexuality, household or sexual contact with hepatitis B carriers, frequent exposure to blood, chronic liver disease. Immunity can be confirmed serologically.
What are the recommendations for hepatitis A vaccine?
Hepatitis A vaccine is recommended for travelers to endemic areas, day-care employees, homosexual men, and chronic liver disease.
What are the indications for varicella vaccine?
Varicella vaccine is recommended for all adults who lack a history of childhood infection; should not be given to immunocompromised or pregnant because it is live attenuated.
What are the recommendations for measles, mumps, rubella vaccine?
Measles, mumps, rubella vaccine is a live attenuated vaccine given in childhood. Healthy adults should receive 1 dose if never vaccinated. Pregnant and immunocompromised not vaccinated.
What are the recommendations for osteoporosis screening?
All women older than age 65 should be given DEXA scans every 1-2 years. Begin at age 60, if there is low body weight or increased risk of fractures.
What are the recommendations for screening for abdominal aortic aneurysm?
Ultrasound should be given once in male smokers older than age 65.
What are the recommendations for cholesterol screening?
Cholesterol screening should begin at age 35 in men and 45 in women who have no risk factors. For patients with risk factors for coronary artery disease, screening should be done routinely after age 20.
What are the recommendations for screening for diabetes?
Obesity, impaired fasting glucose, African-Americans, native Americans and/or positive family history.
What is the criteria for the diagnosis of diabetes?
Diabetes mellitus is diagnosed when two fasting are greater than 126 mg/dL, or when a random glucose is >200 mg/dL.
What clinical test should be used to screen for alcohol abuse?
CAGE: Have you ever felt the need for Cut down? Have you ever felt Annoyed by criticism of your drinking? Have you ever Guilty about your drinking? Have you ever taken Eye opener? Positive is 2 "yes.?
What pathological conditions are associated with hyperprolactinemia?
Excess of prolactin occurs with prolactinomas (most common functioning pituitary adenomas, 60% of pituitary tumors.
What medications cause hyperprolactinemia?
Decreased inhibitory action of dopamine caused by drugs that block dopamine synthesis (phenothiazines, metoclopramide) and dopamine-depleting agents (alpha-methyldopa).
What is the presentation of hyperprolactinemia?
Women have galactorrhea, amenorrhea/oligomenorrhea, osteoporosis, infertility, gynecomastia; men have hypogonadism, erectile dysfunction, decreased libido, gynecomastia and infertility.
How is hyperprolactinemia diagnosed?
Always exclude pregnancy, lactation, hypothyroidism and medications before starting the evaluation of hyperprolactinemia. Prolactin levels >100 ng/mL suggest probable pituitary adenoma.
What is the management of hyperprolactinemia?
For prolactinomas: cabergoline or bromocriptine (dopamine agonist) reduce prolactin levels. Surgery for adenomas not responsive to cabergoline or bromocriptine, or if the tumor causes neurologic effects.
What is acromegaly?
Excessive secretion of growth hormone in an adult. In children, gigantism occurs. In adults acromegaly occurs, which is bony and soft tissue overgrowth. Pituitary adenoma that overproduces growth hormone. Only 1% are malignant.
How is acromegaly diagnosed?
The best initial test is insulin-like growth factor-1 levels. Confirmatory testing involves the measurement of GH after 100 g of glucose is given orally.
What is the management of acromegaly?
Transsphenoidal surgery. Hypopituitarism occurs after surgery in 10-20%. Octreotide is a somatostatin analog that reduces GH in two thirds of patients and causes partial tumor regression in 20-50% of patients.
What are the causes of hypopituitarism?
Tumors, cysts, sarcoidosis, Tb, syphilis, eosinophilic granuloma, Hashimoto, gastric atrophy. Trauma, radiation, surgery, infections, hypoxia. Sheehan postpartum necrosis, hemochromatosis, amyloidosis.
What are the signs of hypopituitarism?
Amenorrhea, genital atrophy, loss pubic hair. Growth fail. Thyrotropin deficiency: hypothyroidism. Adrenocorticotropin def: fatigue, anorexia, wt loss, decreased skin pigment, hypotension, hypoNa.
How is hypopituitarism diagnosed?
Measure GH, TSH, LH, and insulin-like-GF1.
How is ACTH deficiency diagnosed?
Insulin tolerance test: give insulin and measure serum cortisol. Metyrapone tests for decreased ACTH production. Metyrapone blocks cortisol production, which should increase ACTH levels.
How is gonadotropin deficiency diagnosed?
To diagnose gonadotropin deficiency in women, measure LH, FSH, and estrogen. In males, gonadotropin deficiency is detected by measuring LH, FSH, and testosterone.
How is thyroid stimulating hormone deficiency diagnosed?
To diagnose TSH deficiency, measure serum thyroxine (T4), and free triiodothyronine (T3), which are low, with a normal to low TSH.
What is central diabetes insipidus?
CDI is neurohypophyseal deficiency of vasopressin (ADH), which results in excessive, dilute urine and increased thirst with hypernatremia. Nephrogenic DL is caused by renal resistance to vasopressin.
What are the causes of central diabetes insipidus?
Adenomas, craniopharyngiomas; leukemias, sarcoid histocytosis. Pituitary/hypothalamic surgery, radiotherapy, severe head injuries, anoxia, hypertension, and meningitis. Idiopathic; encephalitis, TB, and syphilis.
What are the causes of nephrogenic diabetes insipidus?
Nephrogenic DL: idiopathic or caused by hyperCa, hypoK, sickle cell, amyloidosis, myeloma, pyelonephritis, sarcoidosis, Sjögren syndrome, lithium, demeclocycline, colchicine.
What are the clinical signs of diabetes insipidus?
Polyuria, excessive thirst, polydipsia (16-20 L/d), hypernatremia with high serum osmolarity, decreased urine specific gravity, dehydration, weakness, fever, altered mental status, and death.
How is diabetes insipidus diagnosed?
Water deprivation test: Uosm after dehydration versus Uosm after vasopressin. Normal response to fluid restriction is increased urine osm, decrease urine vol. DL urine vol remains high despite vol depletion.
How is nephrogenic diabetes insipidus differentiated from central diabetes insipidus?
ADH levels will be low in central DL and high in nephrogenic DL.
What is the differential diagnosis of diabetes insipidus?
Psychogenic polydipsia, drug-induced polydipsia from chlorpromazine, anticholinergic drugs, or thioridazine, and hypothalamic diseases.
What is the management of central diabetes insipidus?
Hormone replacement with vasopressin. For nephrogenic DL, HCTZ or amiloride increase the reabsorption of fluid from the proximal tubule.
What are the causes of syndrome of inappropriate secretion of ADH?
Small cell CA, pancreas CA, ectopic ADH; TB, pneumonia, lung abscess; head injury, stroke, encephalitis; chlorpropamide, clofibrate, vincristine, vinblastine, cyclophosphamide, carbamazepine.
What are the clinical signs of syndrome of inappropriate ADH?
Water retention and ECF expansion without edema or hypertension. Hyponatremia. Hyponatremia and concentrated urine (Uosm >300 mOsm), no edema or dehydration.
What are the CNS signs of SIADH?
Cerebral edema from hyponatremia causes irritability, confusion, seizures, coma.
What are the laboratory findings in syndrome of inappropriate secretion of ADH?
Hyponatremia <130 mEq/L, and Posm <270 mOsm/kg. Urine Na >20 mEq/L (inappropriate natriuresis). Low BUN, low creatinine, low urate, low albumin.
What is the management of SIADH?
Treat cause. Fluid restrict 800 mL/d. Demeclocycline. If severe confusion, convulsions, or coma: hypertonic saline (3%) 200-300 mL IV over 3-4 h. The rate of correction 0.5-1 mmol/L/h of serum Na.
What is the most sensitive test for thyroid diseases?
The most sensitive test in thyroid diseases is the TSH. If the TSH is normal, then the patient is euthyroid.
What are the effects of thyroid binding globulin levels on the thyroxine levels?
Increased TBG in pregnancy, OCP will increase total T4 but free T4 level is normal. Decreased TBG levels in nephrotic syndrome and androgens. Decreased TBG will decrease total T4, but free or active T4 normal.
What are the signs of thyrotoxicosis?
Graves disease, diffuse goiter, exophthalmos, dermopathy. More in women in the fourth decade.
What is the cause of Grave disease?
Autoantibodies bind to TSH receptor and stimulate thyroid. Intrinsic thyroid autonomy can result from a hyperfunctioning adenoma (toxic) or toxic multinodular goiter of the elderly.
What are the causes of transient hyperthyroidism?
Subacute thyroiditis (painful) and lymphocytic thyroiditis (painless, postpartum). Thyrotoxicosis factitia and ectopic thyroid tissue (struma ovarii, functioning follicular carcinoma).
What are the cardiovascular signs of Graves disease?
Anxiety, insomnia; atrial fibrillation, tremors, frequent bowel movements, excessive sweating, heat intolerance. Weight loss, increased appetite, weakness. Dyspnea, palpitations, angina, or cardiac failure.
What are the dermatologic signs of Grave disease?
Skin is warm and moist, palmar erythema; fine, silky hair. Staring, infrequent blinking, oligomenorrhea; osteoporosis and hypercalcemia.
What are the laboratory findings in Graves disease?
Low TSH; increased serum free T4 and T. RIAU is increased. Antithyroglobulin and antimicrosomal antibodies are elevated in Graves disease and autoimmune thyroiditis.
What is the treatment of hyperthyroidism?
Popranolol, propylthiouracil (safe in preg) or methimazole. Both can cause agranulocytosis. Ablation with radioactive iodine. Subtotal thyroidectomy only in pregnancy and children.
What is a thyroid storm?
Thyrotoxicosis precipitated by stress, infection, surgery, or trauma. Extreme irritability, delirium, coma, tachycardia, restlessness, vomiting, jaundice, diarrhea, hypotension, dehydration, and fever.
What is the treatment for thyroid storm?
Glucose, hydration, glucocorticoids, oxygen, cooling blanket. Propylthiouracil, beta-blockers, and iodine inhibit hormone release.
What is the affect of antithyroid drugs on uptake of radioactive iodine?
Antithyroid drugs should be stopped several days before radioactive iodine treatment because the antithyroid medications block the uptake of the radioactive iodine.
What are the causes of primary hypothyroidism?
Hypothyroidism is from thyroid in 95%. Can be caused by chronic thyroiditis (Hashimoto), postablative or radioactive iodine, heritable defects, iodine deficiency; lithium, aspirin, amiodarone, interferon, sulfonamides.
What is the most common cause of goitrous hypothyroidism?
Hashimoto disease is most common cause of goitrous hypothyroidism.
What are the clinical signs in congenital hypothyroidism?
Cretinism, jaundice, hoarse, constipation, somnolence, feeding problems; delayed milestones, dwarf, protruding tongue, broad flat nose, wide eyes, sparse hair, dry skin, potbelly, umbilical hernia, impaired mental.
What are the signs and symptoms of hypothyroidism in the adult?
Lethargy, constipation, cold intolerance, cramping, carpal tunnel, menorrhagia. Slow intellectual/motor; appetite decrease, weight increase; dry hair/skin. Deep voice, prolonged DTR. Myxedema: expressionless, sparse hair, periorbital puffiness, Lg tongue.
What is the treatment of hypothyroidism?
Levothyroxine (T4) should be administered with monitoring of TSH/T3 levels (6 weeks to steady state after dosing changes).
What is myxedema coma?
Severe hypothyroidism with hypothermic, stuporous state; frequently fatal; respiratory depression. Precipitated by cold exposure, trauma, infections, and CNS depressants. Treatment is very high doses of T4 and T3.
What are the causes of subacute thyroiditis?
Granulomatous, giant cell, and de Quervain thyroiditis. In the fourth and fifth decades. Viral origin and follows upper respiratory infection; malaise, fever, pain over the thyroid. Thyroid enlarged and firm.
What are the laboratory findings in subacute thyroiditis?
Elevated erythrocyte sedimentation rate (ESR), decreased radioactive iodine uptake, initial elevation in T4 and T3, followed by hypothyroidism.
What is the treatment of subacute thyroiditis?
Aspirin, prednisone, and Popranolol.
What is Hashimoto thyroiditis?
Chronic autoimmune inflammatory process with lymphocytic infiltration. Common disorder most frequently in middle-aged women; most common cause of sporadic goiter in children.
What are the clinical signs of Hashimoto thyroiditis?
Goiter that is painless, rubbery and hypothyroidism.
How is Hashimoto thyroiditis diagnosed?
TSH increases, and T4 and T decrease. Antimicrosomal and antithyroperoxidase antibodies.
What is the management of Hashimoto thyroiditis?
Replacement with L-thyroxine (T4).
What is chronic lymphocytic thyroiditis?
Self-limiting thyrotoxicosis; more common women. Thyroid is painless, nontender, firm, symmetrical, and enlarged. T4 and T3 are elevated, RAIU is low, ESR normal. Lasts for 2-5 months. Treatment with Popranolol.
What is Reidel thyroiditis?
Fibrosis of the thyroid and mediastinal and retroperitoneal fibrosis.
What is papillary carcinoma?
Most common thyroid CA; radiation exposure. 70% of all thyroid CA. Women 3 times more than men. Second decade and later. Treatment is surgery or suppression with levothyroxine.
What is follicular carcinoma of the thyroid?
Follicular carcinoma accounts for 15% of thyroid cancers. Elderly women. More malignant than papillary carcinoma. Spreads hematogenously.
What is the treatment of follicular thyroid cancer?
Treatment is near-total thyroidectomy with postoperative radioiodine ablation.
What is anaplastic thyroid carcinoma?
10% of all thyroid cancer. Elderly women. Highly malignant, painful enlargement. Eighty percent of patients die.
What is medullary thyroid carcinoma?
5% of all thyroid cancers. More malignant than follicular carcinoma. Often produces calcitonin. Medullary carcinoma is a component of two types of multiple endocrine neoplasias.
What is the treatment of medullary thyroid cancer?
Treatment is thyroidectomy. Calcitonin levels can also be increased from cancer of the lung, pancreas, breast, and colon.
What are the clinical signs of thyroid carcinoma?
Growth of thyroid or mass with no tenderness or hoarseness. History of radiation. Men and elderly have a higher incidence of thyroid malignancy. Solitary; may produce calcitonin.
What is the significance of psammoma bodies in the thyroid?
Calcifications such as psammoma bodies suggest papillary carcinoma; increased density is associated with medullary carcinoma.
What is the significance of finding hyperthyroidism in association with a thyroid nodule?
Hyperthyroidism is never associated with cancer.
What is the diagnostic approach to solitary nonfunctioning nodule?
Fine-needle aspiration for cytology is the initial procedure. The first test for a thyroid nodule is the TSH; if TSH is normal, then proceed to an FNA. If TSH is low, the nodule is benign. Ultrasound to distinguish cysts.
What is the function of parathyroid hormone?
Maintains calcium concentration. PTH acts on bone and kidney, indirectly on intestine (effects synthesis 1,25-dihydroxy-cholecalciferol) to increase serum calcium. Regulated by serum-ionized calcium.
What is the effect of parathyroid hormone on the bones?
PTH increases osteoclast, which releases Ca. PTH inhibits PO4 reabsorption kidney which decreases PO4 and favors Ca release from bones. PTH activates vitamin D, increases GI Ca absorption. Calcitonin decreases Ca.
What is the most common cause of hypercalcemia?
Primary hyperparathyroidism. Hypercalcemia of malignancy caused by PTH-like protein. Sarcoidosis, Tb, berylliosis, histoplasmosis, coccidioidomycosis increase vitamin D.
What are the rare causes of hypercalcemia?
Rare causes: vitamin D, thiazides, lithium, Paget, immobilization, hyperthyroidism.
What are the signs of hypercalcemia?
Lethargy, confusion, constipation, anorexia, nausea, vomiting. Pancreatitis because of precipitation of Ca pancreas. Polyuria, polydipsia. Ca precipitates kidney, resulting in nephrolithiasis. HTN in 50%.
What are the ECG signs of hypercalcemia?
ECG shows short QT.
What is the treatment for hypercalcemia?
Saline or half-normal and furosemide, which promote calcium loss. Bisphosphonates such as alendronate, zoledronate, risedronate, pamidronate inhibit osteoclasts and stimulate osteoblasts. Calcitonin.
What are the causes of primary hyperparathyroidism?
Gland adenoma (80%). Parathyroid CA rare (<1%). Hyperparathyroidism can be part of MEN type I: hyperparathyroidism, pituitary tumors, pancreatic tumors. MEN II: hyperparathy, pheo, medullary thyroid CA.
What the effects of hyperparathyroidism on bone?
Osteitis fibrosa cystica occurs because of osteoclastic bone resorption; bone pain, fractures, swelling, deformity, bone cysts, and brown tumors (punched-out lesions). One half asymptomatic.
How is primary hyperparathyroidism diagnosed?
Serum calcium >10.5 mg/dL, with elevated PTH level. In every other cause of hypercalcemia the PTH level will be low. Serum phosphate is low (<2.5 mg/dL).
What is the management of primary hyperparathyroidism?
Removal of parathyroids. Bisphosphonates if surgery is contraindicated or if serum Ca <11.5 mg/dL. Calcium reduced to 400 mg/d. 2-3 L of fluid/d. Phosphor-soda. Cinacalcet simulates effect of Ca parathyroid.
What are the causes of hypocalcemia?
Most common: hypoparathyroidism, renal failure, hyperphosphatemia, hypoMg. Loop diuretics, phenytoin, alendronate, foscarnet. Lack of renal activated 1,25-vitamin D, leads to decreased Ca absorption.
What are the signs of hypocalcemia?
Hypocalcemia results in neural hyperexcitability, seizures, tetany, circumoral numbness, and tingling. Cataracts.
What are the ECG signs of hypocalcemia?
Arrhythmias may develop because of a prolonged QT.
What is the most common cause of hypoparathyroidism?
Thyroidectomy. Low PTH levels are also seen in hereditary hypoparathyroidism and hypoMg. Mg deficiency prevents release of PTH. High PTH levels in renal failure because decreased active vitamin D.
What are the neurologic signs of hypoparathyroidism?
Neuromuscular irritability, tetany, laryngospasm, cramps, seizures, impaired memory. Chvostek sign: percussion of facial nerve contraction face. Trousseau: inflation of cuff elicits flexion metacarpophalangeal.
What are the cardiovascular signs of hypocalcemia?
QT prolongation, CHF, and/or hypotension. Circumoral tingling and tingling of the hands and feet.
How is hypoparathyroidism diagnosed?
Low calcium may be due to a low albumin. Ionized calcium is best test. PTH low. Low Ca with high PO4 can be due to renal failure, tissue destruction, hypoparathyroidism, pseudohypoparathyroidism.
What are the causes of a low calcium with low phosphorous?
Low calcium with low phosphorous is caused by low vitamin D.
What is the management of hypoparathyroidism?
Calcium gluconate IV. Maintenance oral calcium 2-4 g/d, vitamin D, and if there is hyperphosphatemia, diet restriction and phosphate binders (CaCO3).
What is the usual age of onset of type 1 diabetes?
The age of onset is usually <30 years. <10% of first-degree relatives are affected with a 50% occurrence in identical twins. Autoantibodies to islet cells. Lean body build.
What is the usual age of onset of type 2 diabetes?
Usually >40; males > females. >20% of first-degree relatives are affected with 90-100% occurrence in identical twins. Obese.
What is the cause of type 2 diabetes?
90% of diabetes is type 2, caused by abnormal insulin secretion and resistance to insulin.
What are the clinical signs of diabetes?
Hyperglycemia causes polyuria, polydipsia, polyphagia, coma (ketoacidosis for type I diabetes and hyperosmolar coma for type 2 diabetes).
How is diabetes diagnosed?
Polyuria, polydipsia, ketonuria, weight loss, plasma glucose >200 mg/dL in these patients is sufficient for diagnosis. In asymptomatic patients, fasting plasma glucose 126 mg/dL on two occasions is diagnostic of diabetes.
What is glycosylated hemoglobin A1c?
HgA1c is condensation of glucose with free amino groups on Hb. It is used to follow glucose control. HgA1C is high in diabetics with chronic hyperglycemia during the preceding 8-12 weeks.
What is the glycosylated hemoglobin A1c goal?
The goal of therapy is to have an HgA1c <7%.
What is the management of type 2 diabetes?
Metformin in obese. Contraindicated: renal insufficiency. Does not cause hypoglycemia. If diabetes is not controlled with metformin, add a sulfonylurea; then either switch to insulin or add glitazone.
What is the dosage for insulin in type 2 diabetes?
0.5 U/kg, with two-thirds given AM (2/3 NPH, 1/3 reg) and one-third in PM (½ NPH, ½ reg). Glucose measurements before each meal and at bedtime.
What is the dosage for glargine insulin?
Once-a-day injection along with 2-3 times/day ultra short-acting insulin (such as lispro or aspart before meals). Glargine causes less hypoglycemia compared with NPH.
What is the acute complication of uncontrolled type 1 diabetes mellitus?
Diabetic ketoacidosis results from severe insulin insufficiency in type 1 diabetics. Precipitating factors of DKA include interrupted insulin therapy, infection, emotional stress, and excessive alcohol.
What are the clinical signs of diabetic ketoacidosis?
Anorexia, nausea, vomiting, abdominal pain, rapid breathing (Kussmaul), "fruity" breath odor (acetone), dry skin/mucous membranes, poor skin turgor, altered consciousness to coma. Arrhythmias, hyperkalemia fatal.
What is the treatment of diabetic ketoacidosis?
K is low because of the urinary loss of K. As soon as K <4 mEq/L, KCl given. Normal saline should be given in high volume. Bolus with 5-10 units of regular insulin then infuse.
What acid-base disorder is associated with uncontrolled diabetes?
Elevated blood glucose, increased acetoacetate, acetone, hydroxybutyrate result in metabolic acidosis (low serum bicarbonate and low blood pH), and increased anion gap [sodium - (bicarbonate + chloride)].
What is hyperosmolar nonketotic coma?
HONK occurs in type 2 diabetes; severe hyperglycemia without ketosis. Precipitated by noncompliance plus inability to drink. Elderly, infections, strokes, steroids, immunosuppressants, diuretics.
What is the presentation of hyperosmolar nonketotic coma?
Profound dehydration results from hyperglycemic diuresis. Weakness, polyuria, polydipsia, lethargy, confusion, convulsions, and coma.
What are the laboratory findings in hyperosmolar nonketotic coma?
Elevated blood glucose (>1,000 mg/dL) and extremely high serum osmolality.
What is the treatment of hyperosmolar non-ketotic coma?
High-volume fluid and electrolyte replacement and insulin.
What are the chronic ocular complications of diabetes?
Retinopathy is the leading cause of blindness; background or proliferative (microaneurysms, hemorrhages, exudates, retinal edema).
What are the ophthalmic screening recommendations for diabetes?
Diabetics should be screened for retinopathy once a year. Proliferative retinopathy is treated with laser photocoagulation.
What are the renal complications of diabetes?
Nephropathy affects 40% of type 1 and 30% of type 2 diabetics. Proteinuria and end-stage renal disease can develop. Diffuse widening of glomerular basement membrane and mesangial thickening.
What is the treatment of diabetic nephropathy?
Management of nephropathy involves strict control of diabetes, angiotensin-converting enzyme inhibitors, and dialysis or renal transplantation.
What are the screening recommendations for proteinuria in diabetics?
All diabetics should be screened for proteinuria annually. Microalbuminuria is >30/day. Treated with ACE inhibitors. Diabetes is the most common cause of end stage renal disease.
What are the cardiovascular complications of diabetes?
75% of deaths in diabetes are from myocardial infarction, congestive failure, or stroke. Coronary artery bypass should be performed in a diabetic patient even if there is only two-vessel coronary disease.
What is the lipid management of diabetes?
Diabetes is equivalent of coronary disease for management of hyperlipidemia. The target LDL in a diabetes is <100 mg/dL. Diet and exercise should be initiated if LDL >100 mg/dL. Statin for an LDL >100 mg/dL.
What is the management of diabetic retinopathy?
Screening for retinopathy annually. In type 1 diabetes, the first screening should take place 5 years after diagnosis, then annually. Proliferative retinopathy with vitreous hemorrhages or neovascularization is treated with laser.
What is the treatment of diabetic gastroparesis?
Metoclopramide or erythromycin.
What is the treatment of diabetic erectile dysfunction?
Occurs in 50% of patients after 10 years of diabetes. Treatment is sildenafil.
What is the screening for diabetic peripheral neuropathy?
Podiatric examination annually. Neuropathy leads to increased injury. Diabetes is responsible for 50% of all nontraumatic amputations.
What are the characteristics of diabetic peripheral neuropathy?
Peripheral neuropathy is symmetrical, with numbness, paresthesia, pain; absent reflexes and loss of vibratory. Mononeuropathy (mononeuritis multiplex) is foot drop, wrist drop, or paralysis of 3, 4, 6 cranial nerve.
What are the signs of diabetic autonomic neuropathy?
Difficulty swallowing, delayed gastric emptying (gastroparesis), constipation, or diarrhea. Bladder dysfunction or paralysis.
What is the treatment for peripheral neuropathy?
Analgesics, gabapentin, pregabalin, amitriptyline, and carbamazepine
What is the Somogyi effect?
Rebound hyperglycemia in the morning because of counterregulatory hormone release after hypoglycemia at night.
What is the dawn phenomenon?
Early morning rise in plasma glucose, requiring increased amounts of insulin to maintain euglycemia.
What are the symptoms of hypoglycemia?
Sweating, tremor, tachycardia, anxiety, hunger. Dizziness, headache, clouding vision, blunted mental, fine motor loss, confusion, abnormal behavior, convulsions, loss consciousness. Major symptoms <20 mg/dL.
What are the causes of hypoglycemia?
Panhypopituitarism, adrenal insufficiency, enzyme defects, malnutrition, pregnancy, liver disease, alcohol, Popranolol, salicylates. Insulinoma, exogenous insulin, sulfonylureas, quinine, endotoxic shock, insulin receptor Ab.
What are the clinical signs of insulinoma?
Insulinoma (pancreatic B-cell tumor) can cause hypoglycemia. Benign symptoms of hypoglycemia are blurred vision, headache, feelings of detachment, slurred speech, and weakness.
How is insulinoma diagnosed?
Serum insulin level of >8 mg/mL in the presence of blood glucose values <40 mg/dL.
What are the causes of factitious hyperinsulinism?
Administration of insulin or ingestion of Equal or oral sulfonylureas. It is common.
What are the laboratory abnormalities in factitious hyperinsulinism?
Hypoglycemia, high insulin and suppressed plasma C peptide is pathognomonic of exogenous insulin. Ethanol-hypoglycemia with starvation.
What is Cushing syndrome?
Group of abnormalities. Glucocorticoid therapy is most common cause. Others: adrenal hyperplasia, which can be caused by pituitary ACTH in pituitary dysfunction or pituitary ACTH-adenomas
What is Cushing disease?
Pituitary adenoma that secretes ACTH, causing hypercortisolism.
What percentage of Cushing syndrome is caused by ACTH-producing pituitary adenomas?
ACTH-producing pituitary adenomas cause 40% of Cushing cases.
What types of cancers can cause adrenal hyperplasia by producing ACTH?
ACTH or corticotropin-releasing hormone is secreted by bronchogenic, thymus, or pancreatic CA. Adrenal adenoma or carcinoma and adrenal nodular hyperplasia account for 30% Cushing.
What are the clinical signs of Cushing syndrome?
Moon face, buffalo hump, truncal obesity; HTN, weakness, osteoporosis; striae, bruises. Acne, hirsutism, oligomenorrhea. Depression, psychosis. Hyperglycemia, delayed healing, nephrolith; glaucoma, polyuria, infections.
What are the laboratory signs of Cushing syndrome?
Cushing's is associated with hypokalemia and leukocytosis. Hypokalemia occurs because of the mineralocorticoid effect of steroids.
How is Cushing syndrome diagnosed?
1- mg overnight dexamethasone suppression test and the 24-hour urine free cortisol. The tests used to establish the etiology of the cortisol excess are the ACTH level, high-dose Dex suppress test, MRI.
What is the 1-mg overnight dexamethasone suppression test?
Best initial diagnostic test of glucocorticoid excess. Give dexamethasone 11 PM; cortisol at 8 AM should be normal. Single random cortisol levels are not reliable. Abnormal 1-mg test confirmed with 24h urine cortisol.
How is the etiology of the Cushing syndrome established?
ACTH levels are elevated with either a pituitary source of ACTH (adenoma or ectopic). If the ACTH level is low, then the etiology is adenoma, cancer, or adrenal hyperplasia.
When there is a high ACTH level, how is the precise etiology of Cushing syndrome determined?
MRI of the pituitary for an adenoma or a CT scan of the chest looking for an ectopic focus.
What is hyperaldosteronism?
Aldosterone reabsorbs sodium and excretes K and acid (H+). Hyperaldosteronism is caused by: primary aldosteronism from the adrenal; and secondary aldosteronism, in which the stimulus is extraadrenal.
What are the causes of primary hyperaldosteronism?
Unilateral adrenal adenoma (70%). Bilateral hyperplasia accounts for 25-30%. Excessive licorice ingestion (licorice has aldosterone-like qualities).
How is primary hyperaldosteronism diagnosed?
High urine aldosterone and low plasma renin levels confirm a diagnosis of primary hyperaldosteronism. High-salt diet before the test should suppress aldosterone. Confirm the diagnosis with a CT scan of the adrenals.
What is the treatment of bilateral adrenal hyperplasia?
Bilateral adrenal hyperplasia is treated with spironolactone, which blocks aldosterone.
What is Bartter syndrome?
Secondary hyperaldosteronism without edema or hypertension; defect in the loop of Henle; severe hypokalemic alkalosis, defect in renal conservation of sodium or chloride.
What is congenital adrenal hyperplasia?
Most common adrenal disorder of infancy and childhood. CAH is an autosomal recessive mutation, which produce deficiencies of enzymes that are necessary for the synthesis of cortisol.
What are the common enzymatic defects associated with congenital adrenal hyperplasia?
Enzymatic defects include alpha-21 hydroxylase deficiency in 95% of all cases. Alpha-21 hydroxylase deficiency is associated with reduction in aldosterone secretion in one third of patients.
What are the signs of congenital adrenal hyperplasia?
Adrenal virilization with salt-losing tendency caused by aldosterone deficiency (hyponatremia, hyperkalemia, dehydration, hypotension). Ambiguous genitalia, enlarged clitoris, labial fusion, virilization. Males have macrogenitosomia, precocious puberty.
How is congenital adrenal hyperplasia diagnosed?
Failure to thrive, adrenal insufficiency, salt wasting, HTN. Decreased testosterone, androstenedione, dehydroepiandrosterone, 17-hydroxy-progesterone, urinary 17-ketosteroid,, pregnanetriol.
What is the management of congenital adrenal hyperplasia?
Glucocorticoid (hydrocortisone) replacement.
What are the two types of adrenal deficiency disorders?
Adrenal insufficiency is divided into primary adrenocorticoid deficiency (Addison disease) and secondary failure in elaboration of ACTH. Primary adrenocortical insufficiency results from adrenal hypofunction.
What are the causes of Addison disease?
Caused by destruction of gland. Autoimmune destruction accounts for 80%. Destruction by surgical removal, Tb, fungal, CMV, Mycobacterium avium complex, hemorrhagic, trauma, metastatic invasion.
What are the clinical signs of Addison disease?
Weakness, paresthesias, cramping, stress intolerance, irritability. Small heart, wt loss, sparse axillary hair. Hyperpigmentation only with primary insufficiency. Addisonian crisis is fever, hypotension, low Na, high K, acidosis.
How is primary adrenocortical insufficiency diagnosed?
Rapid ACTH, then measurement of cortisol. Moderate neutropenia, lymphocytosis, eosinophilia; elevated K, BUN; low Na; low blood glucose; low plasma cortisol and urinary 17-hydroxycorticosteroid.
What is the cosyntropin or ACTH stimulation test?
A cortisol level is obtained before and after administering ACTH. A normal person should show a rise in cortisol level after ACTH administration.
What is the management of Addison disease?
Glucocorticoid, mineralocorticoid, and sodium chloride replacement.
What is an adrenal crisis?
Fever, vomiting, abdominal pain, altered mental, vascular collapse. Previously undiagnosed adrenal insufficiency after surgery, serious infection, stress; bilateral adrenal infarction/hemorrhage, withdrawal from glucocorticoids.
What is the treatment of adrenal crisis?
Measure cortisol level, then rapidly administer fluids and IV hydrocortisone.
What is pheochromocytoma?
Pheochromocytoma is a rare, usually benign, tumor that arises from the chromaffin cells of the sympathetic nerves. 90% in adrenals; 10% extraadrenal, 10% malignant, 10% in children, 10% bilateral, 10% not HTN.
What substances are secreted by pheochromocytomas?
Epinephrine secretion causes tachycardia, sweating, flushing, and hypertension. Norepinephrine is secreted by all extraadrenal pheochromocytomas.
What are the clinical signs of pheochromocytoma?
Paroxysms lasting a few minutes to several hours, with headache, profuse sweating, palpitations, and apprehension. Pain in the chest or abdomen, nausea and vomiting. Hypertension with tachycardia.
How is pheochromocytoma diagnosed?
24-h urine collection for free catecholamines, urinary metanephrines and vanillylmandelic acid, plasma catecholamine. Plasma metanephrine is initial test. Chromogranin levels correlate tumor size. CT/MRI.
What is the management of pheochromocytomas?
Control BP with alpha-adrenergic blocker (phentolamine, phenoxybenzamine). No other antihypertensive drugs should be used before adequate control of blood pressure. Surgery after BP stabilization.
What are the causes of primary hypogonadism?
Primary hypogonadism (hypergonadotropic: increased LH, FSH) from Klinefelter syndrome (small testes, eunuchoid, XXY), anorchia, castration or radiotherapy, mumps, TB, leprosy, or chemotherapeutic agents.
What are the causes of secondary hypogonadism?
Secondary hypogonadism (low LH, FSH) can result from hypopituitarism caused by idiopathic, tumors, hypothalamic lesions, Kallmann syndrome (hypergonadotrophic hypogonadism, decreased smell).
What are the clinical signs of prepubertal hypogonadism?
External genitalia are underdeveloped, voice is high-pitched, beard does not grow, lacks libido and potency. Youthful appearance, with obesity, long extremities, small Adam's apple, gynecomastia.
What are the laboratory abnormalities in hypogonadism?
Urinary 17-ketosteroid is low to normal, and serum testosterone low. Serum FSH and LH are low in hypothalamic or pituitary origin and elevated in primary testicular failure. Treatment is testosterone.
What are the causes of postpubertal hypogonadism?
Pituitary lesion or testes damage by trauma, radiation, infection, drugs. Loss of libido, potency, hair; sterility. Testosterone low. FSH, LH are low with pituitary lesions and high with testicular failure. Rx testosterone.
What is Klinefelter syndrome?
1 of every 500 males. Caused by a supernumerary X chromosomes. 47,XXY. Gynecomastia with elevated levels of LH and FSH. Sterility and lack of libido. The testes are small. Mental retardation may be present.
What are the laboratory abnormalities in Klinefelter syndrome?
Urinary 17-ketosteroids are low normal; serum testosterone is low to normal, LH and FSH are elevated, and serum estradiol is elevated. 20x increased risk of breast cancer. Treatment is testosterone.
What are the basic synovial fluid tests?
Cell count, crystals, cultures, and Gram stain.
What are the synovial fluid cell count in osteoarthritis?
OA and traumatic arthritis have 200-2,000 white blood cells (WBC)/mm3 in the synovial fluid. Inflammatory diseases (RA, gout) have 5,000-50,000 WBC/mm3. Septic arthritis has >50,000 WBC/mm3.
What are antinuclear antibodies?
Antinuclear antibodies are Ab that bind to structures within nucleus. ANAs are found in SLE, Sjögren syndrome, and systemic sclerosis, and 5% of normal people.
How is the ANA test performed?
The ANA test is performed by exposing the antibodies in the serum to laboratory test cells. It is then determined whether the antibodies react with various parts of the nucleus.
What subsets of ANAs are associated with systemic lupus erythematosus?
Anti ds-DNA and anti-SM antibodies are found in patients with SLE; anti-histone antibodies are found in patients with drug-induced lupus.
What is the significance of a positive ANA test?
95% of SLE have positive ANA. Neg ANA excludes SLE. Pos ANA in asymptomatic patient is a false pos (5% of population); a positive ANA with arthritis, proteinuria, pleural effusion is likely SLE.
What are rheumatoid factors?
Autoantibody against Fc portion of IgG. RF found in 70% of RA. Not specific for RA and are found in 5% of healthy. RF is neither sensitive nor specific for the diagnosis of RA. High titers causes more aggressive disease.
What is the significance of a positive cyclic citrullinated peptide?
Cyclic citrullinated peptide is associated with rheumatoid arthritis.
What are antineutrophil cytoplasmic antibodies?
ANCAs are antibodies directed against cytoplasm of neutrophils in Wegener granulomatosis. Perinuclear ANCA is found in polyarteritis nodosa and Churg-Strauss syndrome.
What is antiphospholipid antibody syndrome?
Hypercoagulable state with Ab against phospholipids or cardiolipins (lupus anticoagulant or anticardiolipin antibodies). Elevated PTT, false-positive VDRL.
What is the presentation of antiphospholipid antibody syndrome?
Presents with spontaneous abortions or thromboembolism. If two first-trimester spontaneous abortions occur, antiphosolipid antibodies should be checked.
A 28-year-old woman with joint swelling and stiffness. Stiffness for 2 h in morning. Fatigue, fever. Wrist, MCPs, and PIPs red and swollen. DIPs are not involved. Fluid in wrist joints. What is the diagnosis?
Rheumatoid arthritis.
What is rheumatoid arthritis?
Chronic, symmetric, inflammatory synovitis. Inflammation destroys cartilage and causes bone erosions, deforming joint. Triggered by infectious agent. Women 3x more than men; 35-50 years.
What is the diagnostic criteria for rheumatoid arthritis?
4 of the following: Morning stiffness. Swelling of wrists, MCPs, PIPs. Swelling of three joints. Symmetric joint swelling. Joint erosions on x-rays. RF positive. Rheumatoid nodules.
Which joints are never involved in rheumatoid arthritis?
Distal interphalangeal joints and joints of the lower back.
What are the systemic symptoms of rheumatoid arthritis?
Two-thirds of the patients present with fatigue, anorexia, weight loss, generalized weakness before the onset of the arthritis.
What are the extraarticular manifestations of rheumatoid arthritis?
Damage to the ligaments and tendons. Radial deviation of the wrist with ulnar deviation of the digits. Boutonniere deformity. Swan-neck deformity. Rheumatoid nodules.
What are the laboratory findings in rheumatoid arthritis?
Rheumatoid factor, anemia, increased ESR, X-rays erosions.
What is the initial treatment for rheumatoid arthritis?
Nonsteroidal antiinflammatory drugs (NSAIDs) have fewer side effects than aspirin.
What is the mechanism of selective COX-2 inhibitors?
Cyclooxygenase 2 inhibitors selectively block the COX-2 enzyme at the site of inflammation. Benefit of COX-2 inhibitors is not inhibiting COX-1, an enzyme that produces protective stomach lining.
What is the mechanism of action of nonselective NSAIDS?
Nonselective of NSAIDs block both COX-2 and COX-1, which can lead to increased risk for gastrointestinal side effects (bleeding).
What is the beneficial effect of selective NSAID COX-2 inhibitors?
Selective COX-2 inhibitors (celecoxib) reduce risk of gastric ulcers, but COX-2 inhibitors double risk of MI and stroke because COX-2 enzyme creates protective fatty acids that prevent thromboses.
What is the treatment of rheumatoid arthritis that does not respond to NSAIDs?
Glucocorticoids. Disease-modifying agents: antimalarials, gold, sulfasalazine, methotrexate, and tumor necrosis factor receptor inhibitors: infliximab (Remicade), adalimumab (Humira), etanercept (Enbrel)
What is atlantoaxial subluxation?
Occurs in rheumatoid arthritis when there is excessive movement between atlas (Cl) and axis (C2). Neck pain, C2 radicular pain (hands), myelopathy, paraplegia, quadriplegia.
What is a ruptured Baker cyst?
Extension of inflamed synovium into the popliteal space; swollen, painful calf.
34-year-old woman with confusion, malar rash, HTN, symmetrical joint swelling of wrists and MCPs. Creatinine of 2.5, proteinuria. What is the diagnosis?
Systemic lupus erythematosus.
What is systemic lupus erythematosus?
Systemic disease in which tissues and multiple organs are damaged by autoantibodies and immune complexes. Ninety percent of cases are women. Ultraviolet light may cause flares.
What is the diagnostic criteria for systemic lupus erythematosus?
4 required: malar/discoid rash, photosensitivity, oral ulcers, arthritis, serositis, renal; neurologic (seizures, psychosis); hematologic (hemolysis, leukopenia); immunologic (ANA).
What are the skin manifestations of systemic lupus erythematosus?
Malar rash and photosensitivity (diffuse, maculopapular) flare with exposure to UV-B light and resolve with no scarring. Discoid lupus is a circular rash with a raised rim over scalp and face; central atrophy and scarring.
What are the joint manifestations of systemic lupus erythematosus?
Arthritis is systemic, non-erosive of wrists, MCPs, PIPs.
What are the psychiatric manifestations of systemic lupus erythematosus?
Change in personality and psychosis with CNS lupus. Libman-Sacks endocarditis is noninfectious endocarditis that is rare in lupus
What are the laboratory findings in lupus?
Positive ANA supports diagnosis but is not specific. Complement levels are decreased in lupus. ds-DNA antibodies with active lupus are very specific: increase likelihood of nephritis, and active SLE.
What is the treatment for systemic lupus?
NSAIDs for arthritis, pleurisy. Corticosteroid for rashes. Hydroxychloroquine and corticosteroids for arthritis. Cytotoxics (azathioprine, cyclophosphamide) for nephritis, carditis, pleuritis. Sunscreen.
What drugs cause drug-induced lupus?
Hydralazine, isoniazid, procainamide, quinidine.
What is the presentation of drug-induced lupus?
Arthritis, fatigue, fever, and rarely pleurisy. There is no skin disease, kidney disease, and milder symptoms than SLE. Causes positive ANA. Symptoms resolve within 2 weeks.
35-year-old woman with skin tightness and painful fingertips with exposure to cold. BP 170/100; shiny, thickened skin. Elevated creatinine. What is the diagnosis?
Scleroderma.
What is systemic sclerosis?
Thickening of the skin caused by accumulation of connective tissue; involvement of GI, lungs, kidneys. All scleroderma patients have Raynaud phenomenon.
What are the gastrointestinal signs of systemic sclerosis?
Esophageal dysmotility and achalasia, hypomotility of the small intestine with bacterial overgrowth and malabsorption, and dilatation of the large intestine with diverticula.
What are the pulmonary manifestations of scleroderma?
Pulmonary fibrosis with restrictive lung disease and cor pulmonale.
What are the renal manifestations of scleroderma?
Renal crisis in which malignant hypertension acute renal failure. Treated with angiotensin-converting enzyme inhibitors.
What is limited scleroderma (CREST syndrome)?
Variant of scleroderma: Calcinosis, Raynaud, Esophageal dysfunction, Sclerodactyly, Telangiectasias. Calcium deposits in fingers, knees, elbows, ulcerate. Sclerodactyly is skin thickening of fingers, toes.
What are the laboratory findings in limited scleroderma?
Limited scleroderma has positive ANA test. Antibodies to Sc-70 are negative in limited scleroderma and positive in diffuse scleroderma.
What is Raynaud phenomenon?
Pallor, cyanosis in cold. Vasoconstriction. Raynaud disease is not associated underlying disease. Secondary Raynaud phenomenon is associated with disease (scleroderma). 5% population Raynaud disease. Young women.
What is the treatment for systemic sclerosis?
No cure. D-penicillamine may be used. Raynaud phenomenon: use calcium-channel blockers; for hypertension use angiotensin-converting enzyme inhibitors.
A 43-year-old woman with dry eyes and sensation of sand in her eyes; painful swallowing. Parotid enlargement. ANA positive. What is the diagnosis?
Sjögren syndrome.
What is Sjögren syndrome?
Autoimmune lymphocytic infiltration of exocrine glands; xerostomia, dry eyes. Sjögren syndrome may be primary or with autoimmune diseases (secondary), RA or SLE. Lungs, kidneys, malignant lymphoma.
What is the presentation of Sjögren syndrome?
Itchy eyes, sandy feeling under their eyes destruction of the corneal epithelium (keratoconjunctivitis sicca), and dysphagia.
What are diagnostic tests for Sjögren syndrome?
Schirmer's test shows decreased tear production. ANA positive; anti-Ro (SSA) and anti-La (SSB) are positive. Treatment is artificial tears.
Which autoimmune diseases have arthritis plus multiple organ involvement?
SLE, systemic sclerosis (scleroderma), and Sjögren syndrome.
25-year-old man with lower back stiffness and pain in morning >2 h, improving with exercise. Murmur over second right intercostal space and decreased range-of-motion of lumbar spine. What is the diagnosis?
Ankylosing spondylitis.
What allele is associated with the spondyloarthropathies?
B-27 allele.
What is ankylosing spondylitis?
Inflammatory disorder that affects axial skeleton and peripheral joints. Second to third decade (rare after 40). Men 3-4 times that of women. Positive for HLA B-27.
What is the presentation of ankylosing spondylitis?
Chronic lower back pain, young man (20s-30s). Morning stiffness 1 h, improves with exercise. Cervical spine not affected. Anterior uveitis, aortic insufficiency, CHF, heart block. Decreased spine mobility, fractures.
What are the x-ray abnormalities in ankylosing spondylitis?
Sacroiliitis and fusing of the sacroiliac joint. Bamboo spine and squaring of the vertebral bodies.
What is the treatment for ankylosing spondylitis?
NSAIDs, physical therapy, exercise. TNF blockers (infliximab, adalimumab, etanercept).
What is reactive arthritis?
Seronegative arthropathy after infection, non-gonococcal urethritis. Keratoderma blennorrhagica (waxy, palms/soles), circinate balanitis, oral/genital ulcers, conjunctivitis, arthritis. Campylobacter, Salmonella.
What is the treatment for reactive arthritis?
Severe form of reactive arthritis in HIV patients improves with antiretroviral medications. NSAIDS, tumor necrosis factor blockers.
What is psoriatic arthritis?
Arthritis involves the DIP joints and associated with psoriatic nail disease (pitting of the nails); sausage-shaped digit.
What is enteropathic arthropathy?
Joint arthropathy with ulcerative colitis or Crohn disease; associated with pyoderma gangrenosum and erythema nodosum.
A 66-year-old man with knee pain and pain in distal interphalangeal joints. Knee crepitations. What is the diagnosis?
Osteoarthritis
What is osteoarthritis?
Most common joint disease. Articular cartilage destruction with hypertrophy of bone. Osteoarthritis, unlike RA, is not an inflammatory disease. Knee osteoarthritis is leading cause of chronic disability in elderly.
What are the major risk factors for osteoarthritis?
Age, female sex, genetic factors, major joint trauma, repetitive stress, and obesity. The most common joint to be affected is the knee; base of the thumb.
What is the presentation of osteoarthritis?
Weight-bearing joints (hip, knee), small finger joints (PIPs, DIPs). Oligoarticular-asymmetric. Progressive, irreversible. Pain with exercise, relieved by rest. Morning stiffness <30 min. Crepitations, no systemic.
What X-ray findings are associated with osteoarthritis?
Osteophytes and unequal joint space PIP osteophytes are called Bouchard's nodes, in the DIP are called Heberden's nodes.
What is the treatment of osteoarthritis?
Acetaminophen, analgesic ibuprofen is 1,200 mg/d, antiinflammatory dose is 2,400 mg/d. Caution in elderly with NSAIDs. COX-2 inhibitors may be used in patients at high risk for ulcers (celecoxib).
What are the crystal-induced arthropathies?
Monosodium urate, calcium pyrophosphate, calcium oxalate, calcium hydroxyapatite caused by deposition in joints; they have identical presentations and can only be distinguished by synovial fluid analysis.
What is gout?
Gout affects middle-aged men and presents with acute monoarthritis. Deposition of urate crystals in connective tissue (tophi) and kidneys.
What is the presentation of acute gout?
First toe is commonly affected (podagra); knee, ankle, PIPs, or DIPs may be initially involved. First episode occurs at night with severe joint pain. Warm, red, tender. Pain resolves within 3-14 days.
What are the factors that can precipitate gout?
Excessive alcohol, trauma, surgery, infection, steroid withdrawal, hydrochlorothiazide, furosemide; pyrazinamide and ethambutol, and serious medical illnesses.
How is gout diagnosed?
Uric acid maybe normal. Many have elevated uric acid levels and never develop gout. Serum uric acid level is of no value in diagnosis of gout. Synovial fluid: negatively birefringent needles. WBCs 5000-50000.
What is the treatment of acute gouty arthritis?
Indomethacin 50 mg three times a day. Colchicine, 0.6 mg every hour until symptoms resolve. Steroids, intraarticular or oral in elderly patients who cannot tolerate NSAIDs, colchicine or in renal impairment.
What is the treatment of recurrent gouty attacks?
Hypouricemic therapy in recurrent gouty. Probenecid used in undersecretors (>80%). Allopurinol in overproducers, undersecretors, with renal failure/stones. Allopurinol should not be initiated during attack.
What is pseudogout?
Calcium pyrophosphate crystal deposition is more common in the elderly population and preexisting joint damage. Hyperparathyroidism, hemochromatosis, hypophosphatemia, hypomagnesemia.
What are the clinical signs of pseudogout?
The knee is the most commonly affected joint; other joints are the wrist, shoulder, and ankle.
How is pseudogout diagnosed?
Rhomboid, positive birefringent crystals on synovial fluid evaluation. Linear radiodense deposits in joint menisci or articular cartilage (chondrocalcinosis).
What is the treatment for calcium pyrophosphate crystal deposition disease?
NSAIDS, colchicine, corticosteroid. Prevent frequent recurrences with low doses of colchicine.
A 65-year-old woman with history of rheumatoid arthritis with left shoulder pain for 2 days; low-grade fever. decreased range of motion, erythema. What is the next step?
Perform arthrocentesis
What is the most common cause of infectious arthritis in patients younger than 40 years old?
Gonorrhea, which accounts for 70% of episodes in patients younger than 40 years old.
What is the most common cause of infectious arthritis in older patients?
Staphylococcus aureus is most common cause of infectious arthritis and occurs in preexisting joint destruction from other rheumatic diseases. RA is highest risk.
What are the synovial fluid findings in gonococcal infectious arthritis?
Infected joints with gonococci will not have positive cultures, and the Gram stain will be negative.
What treatment should be prescribed for a young woman with acute monoarticular arthritis with >50,000 WBCs in synovial fluid without crystals?
A young woman with acute monoarticular arthritis who is found to have >50,000 WBCs in the synovial fluid without crystals should be treated with ceftriaxone.
What treatment should be ordered for an elderly man with RA and acute monoarticular arthritis and >50,000 WBCS in synovial fluid?
An elderly man with infectious arthritis should be treated with IV nafcillin or vancomycin.
What is Wegener granulomatosis?
Small vessel vasculitis of any organ, but mainly the sinuses, nose, trachea, lungs and kidneys. Rhinitis, sinusitis, and nasal ulcers. Cough, hemoptysis, and dyspnea. Kidney involvement in 80%. Arthritis in 60%.
How is Wegener granulomatosis diagnosed?
Associated with antineutrophil cytoplasmic antibodies (C-ANCA). Confirm diagnosis by biopsy of an involved organ (nasal septum), vasculitis and granulomas.
What is the treatment of Wegener granulomatosis?
Glucocorticoid and immunosuppressive agent (cyclophosphamide).
What is polyarteritis nodosa?
Multisystem disease with fever, malaise, weight loss, anorexia, and abdominal pain. Affects any site in the body, except the lungs. Predisposition for skin, kidney, nerves, GI.
What is the presentation of polyarteritis nodosa?
Peripheral neuropathies. abdominal pain and gastrointestinal bleeding. Biopsy of skin, symptomatic nerves, or muscles medium-size arteritis.
What is the treatment of polyarteritis nodosa?
Corticosteroids, immunosuppressive drugs (cyclophosphamide).
What is Churg-Strauss syndrome?
Vasculitis can involve any organ; asthma, eosinophilia, lung involvement.
What is the presentation of Churg-Strauss syndrome?
Middle aged individual with new-onset asthma, mononeuropathy, transient pulmonary infiltrates, paranasal sinus abnormalities, nasal polyps, allergic rhinitis.
What are the biopsy findings in Churg-Strauss syndrome?
Biopsy: medium arteritis. Treatment is prednisone and cytotoxic agent.
What is temporal arteritis?
Temporal arteritis is a vasculitis of large arteries that supply the head, eyes, and optic nerves. New-onset headache in any patient older than 50 years; may cause permanent vision loss.
What is the presentation of temporal arteritis?
Headache, pain in temples. Scalp tenderness, jaw claudication, decreased vision, tongue numbness, sudden loss of vision. 25% also have polymyalgia rheumatica.
How is temporal arteritis diagnosed?
ESR is first test (100% sensitive); always confirm by biopsy of temporal artery for giant cells. If temporal arteritis is suspected and the ESR elevated, prednisone should be started immediately (before biopsy).
A 73-year-old woman with new right-sided headache, fever, decreased vision. What is the first step?
Check the ESR; if elevated, start prednisone 60 mg daily.
43-year-old woman with proximal weakness for 2 months. Diffuse lilac rash over sun-exposed areas. Weakness in upper and lower proximal muscles. CPK and aldolase positive. What is the diagnosis?
Dermatomyositis
What are the inflammatory myopathies?
Inflammatory muscle diseases with progressive muscle weakness. Polymyositis, dermatomyositis, and inclusion body myositis.
What are the clinical signs of inflammatory myopathies?
Proximal muscle weakness: lifting objects, combing hair, rising from chair. Ocular muscles never involved, which differentiates inflammatory myopathies from myasthenia.
What is the presentation of dermatomyositis?
Dermatomyositis will have weakness with heliotrope rash, which is a purple-lilac discoloration of the face, eyelids, and sun-exposed areas of body.
What are the laboratory findings in inflammatory myopathies?
Inflammatory destruction of muscles causes an elevation of the muscle enzymes creatine kinase and aldolase. EMG shows of myopathic potentials; confirmed by biopsy.
What is the treatment for polymyositis and dermatomyositis?
Steroids are useful in polymyositis and dermatomyositis.
What is achalasia?
Idiopathic loss of nerves of lower esophageal sphincter; inhibitory neurons lost, inability to relax LES. Unknown etiology. Very small number can be from Chagas disease, gastric carcinoma, lymphoma.
What is the presentation of achalasia?
Achalasia presents with progressive dysphagia to both solids and liquids; regurgitation.
How is achalasia diagnosed?
Barium esophagography shows dilation of esophagus, which narrows into "bird's beak" at end. Esophageal manometry shows increased lower esophageal sphincter resting pressure. Treatment is dilation.
A 65-year-old man with difficulty swallowing solids, weight loss. reflux esophagitis, smoking, enlarged left supraclavicular lymph node. What is the diagnosis?
Esophageal cancer.
What are the causes of esophageal cancer?
Alcohol, tobacco for squamous cell cancer in proximal two-thirds of esophagus. Adenocarcinoma in the distal third, associated with gastroesophageal reflux disease and Barretts esophagus.
What is the presentation of esophageal cancer?
Progressive dysphagia first for solid food, then liquids, weight loss. Halitosis, regurgitation, hoarseness, and hypercalcemia.
How is esophageal cancer diagnosed?
Barium swallow first; endoscopy is mandatory for biopsy. CT scan assesses local spread; bronchoscopy detects spread into the bronchi. Endoscopic ultrasound for staging.
What is the presentation of esophageal spasm?
Intermittent chest pain, dysphagia, with no relationship to exertion or eating. The pain is precipitated by drinking cold liquids.
How is esophageal spasm diagnosed?
Barium studies show a "corkscrew" pattern. The most accurate test is manometric studies.
What is the treatment of esophageal spasm?
Nifedipine and nitrates.
What is Schatzki's ring?
Schatzki's rings are thin, epithelial membranes; intermittent dysphagia; painless. More distal and located at the squamocolumnar junction proximal to the lower esophageal sphincter.
What is Plummer-Vinson syndrome?
Plummer-Vinson syndrome causes a more proximal ring in the hypopharynx. Associated with iron-deficiency anemia and progression to squamous cell cancer. Middle-aged women.
How is Plummer-Vinson syndrome diagnosed?
Diagnosed with a barium swallow.
What is the treatment of Plummer-Vinson syndrome?
May respond to treatment of iron deficiency. Dilation procedures are used.
What is the most common infectious cause of esophagitis?
Candida albicans. HIV positive with a CD4 <200/mm3. Diabetes mellitus is the second most common risk. Herpes simplex, cytomegalovirus, aphthous ulcers.
What is the treatment of esophagitis?
If the patient is HIV positive, empiric treatment is with fluconazole.
What medications are associated with esophagitis?
Pills that cause esophagitis are alendronate, quinine, risedronate, vitamin C, potassium chloride, doxycycline, NSAIDs, and iron sulfate.
What is a Zenker diverticulum?
Outpocketing of posterior pharynx. Older patients have bad breath and difficulty initiating swallowing. Repeated clearing of throat; regurgitation of food eaten several days ago.
How is Zenker diverticulum diagnosed?
The diagnosis is made by barium studies. Treated with surgical resection.
What is Mallory-Weiss syndrome?
Tear of the lower esophagus caused by retching and vomiting. Painless upper gastrointestinal bleeding. Melena or hematemesis. Diagnosed by upper endoscopy.
What is the treatment for Mallory-Weiss tears?
Resolve spontaneously. Injection of the tear with epinephrine or cauterization.
What are the causes of epigastric pain?
Ulcer disease, pancreatitis, gastroesophageal reflux disease, gastritis, and gastric cancer.
What organism causes gastric ulcers, duodenal ulcers, and gastritis?
Helicobacter pylori is most strongly associated with the development of duodenal ulcers, gastric ulcers, and gastritis.
What is the most common cause of epigastric tenderness?
Pancreatitis is the most common cause for epigastric tenderness and pain. Ulcer disease is associated with epigastric tenderness in <20% of patients.
What is the most common cause of epigastric pain?
The most common etiology of epigastric pain is nonulcer dyspepsia, a functional disorder of persistent pain with normal tests.
How is Helicobacter pylori disease diagnosed?
H. pylori can be diagnosed with serology, urea breath testing, stool antigen detection. Endoscopy not needed, but biopsy and is the most accurate. When testing for eradication, use breath test or stool antigen.
When is endoscopy indicated for patients with ulcer disease?
All patients with epigastric pain and >45 years or alarm symptoms, such as weight loss, dysphagia, odynophagia, or heme-positive stool, should undergo endoscopy to exclude gastric cancer.
What is the treatment of Helicobacter pylori disease?
Ulcers, reflux disease, and gastritis treated empirically. Young, healthy patients can be treated empirically with H2 blockers, antacids, or proton-pump inhibitors, and then endoscopy if there is no improvement.
What is the cause of gastroesophageal reflux disease?
Abnormal flow of the acid gastric contents backward from the stomach up into the esophagus through the lower esophageal sphincter.
What factors decrease tone of the lower esophageal sphincter?
Nicotine, alcohol, caffeine, peppermint, chocolate, anticholinergics, calcium-channel blocking agents, and nitrates.
What is the clinical presentation of GERD?
Epigastric pain, sore throat; a bad, metal-like taste in the mouth; hoarseness; and cough and wheezing. Pain in the substernal area. 24-hour pH monitoring is not necessary.
What is the treatment for GERD?
PP-inhibitors: omeprazole, esomeprazole, lansoprazole, pantoprazole, rabeprazole increase pH. Avoid nicotine, alcohol, caffeine, chocolate, late meals. Elevate bed. 5% require Nissen fundoplication.
What is Barrett esophagus?
Complication of long-standing reflux disease where epithelium of lower esophagus changes from normal squamous epithelium to columnar epithelium. Treatment is proton-pump inhibitors.
What are the causes of peptic ulcer disease?
Ulcers are caused by NSAIDs, H pylori, stomach CA, Zollinger-Ellison, Crohn, burns, head trauma, mechanical ventilation. Ulcers not caused by smoking, alcohol, steroids.
What is the pathophysiology of peptic ulcer disease?
Prostaglandins stimulate mucous production that forms a protective barrier. Prostaglandins are inhibited by NSAIDs. Parietal cells produce acid.
What hormones stimulate acid production?
Stimulants of acid production are gastrin, acetylcholine, histamine. Histamine is released by stomach cells and stimulates acid release.
What is the mechanism of action of H2 blockers?
H2 blockers, such as cimetidine, famotidine, and ranitidine, inhibit acid release by blocking histamine effect on parietal cells.
What is Zollinger-Ellison syndrome?
Zollinger-Ellison syndrome is the excessive production and release of gastrin from G cells.
What is the most common cause of ulcer disease?
The most common cause of ulcer disease is Helicobacter pylori followed by the use of NSAIDs. Eighty to 90% of duodenal ulcers and 70 to 80% of gastric ulcers are associated with H. pylori.
How is ulcer disease diagnosed?
Ulcer is best diagnosed with endoscopy. In patients <45 with epigastric pain, endoscopy performed only if symptoms persist. If >45, wt loss, anemia, or heme-positive, endoscopy should be performed.
How is H. pylori disease diagnosed?
Diagnosis of H. pylori is based on serology, urea breath test, stool antigen, or biopsy. Serology is the least expensive and has high sensitivity. Serology does not distinguish between old disease and new disease.
What is the advantage of breath testing for H pylori over serology?
Advantage of breath testing and stool antigen detection is same sensitivity as serology and are able to distinguish new versus old disease. After treatment, breath/stool tests become neg and used as a test for cure.
What is the treatment of H pylori ulcer disease?
PPI, clarithromycin and amoxicillin. Omeprazole, lansoprazole, pantoprazole, rabeprazole, and esomeprazole are all equal. PPI/clarithromycin/amoxicillin is >90% effective. Therapy 10-14 d.
What is the approach to patients who fail therapy for H pylori?
In those who fail therapy, a urea breath test should be performed. If the organism was not eradicated, then re-treat with PPI, tetracycline, metronidazole and bismuth subsalicylate.
What is gastritis?
Inflammation, erosion of the gastric lining. Unlike ulcer disease, gastritis can be caused by alcohol; also NSAIDs, Helicobacter, head trauma, burns, mechanical ventilation. Associated with increased gastric acid.
What is the presentation of gastritis?
Most patients with gastritis present with asymptomatic bleeding. Other patients have epigastric pain, nausea and vomiting, hematemesis or melena.
How is gastritis diagnosed?
The diagnosis of H. pylori is based on either serology, urea breath testing, stool antigen testing, or biopsy with histology.
When should endoscopy be performed in patients with gastritis?
If <45 years of age with epigastric pain, endoscopy can be performed if the symptoms persist. In those older than 45 years or with weight loss, anemia, heme-positive stools, dysphagia, endoscopy should be performed.
How is gastritis treated?
PPI combined with clarithromycin and amoxicillin. Omeprazole, lansoprazole, pantoprazole, rabeprazole, esomeprazole are equal. Duration of therapy is 10 to 14 days.
How is pernicious anemia diagnosed?
B12 deficiency and pernicious anemia are diagnosed with low B12 level and increased methylmalonic acid level. Pernicious anemia is confirmed by antiparietal cell Ab and anti-intrinsic factor Ab.
What is Zollinger-Ellison syndrome?
Hypergastrinemia caused by cancer of gastrin-producing cells. Half of gastrinomas are in duodenum, and quarter are in pancreas. <20% are associated with MEN 1 or parathyroid, pituitary, pancreatic tumors.
What is the presentation of Zollinger-Ellison syndrome?
Present with ulcer. <1% of ulcer disease has a Zollinger-Ellison syndrome or gastrinoma. Ulcers are recurrent after therapy, multiple in number, and occur in the distal duodenum, or resistant to routine therapy. Diarrhea.
How is Zollinger-Ellison syndrome diagnosed?
Gastrin level must be elevated while the patient is off antisecretory therapy for several days. Nuclear somatostatin-receptor scintigraphy is sensitive metastatic disease. Most sensitive test endoscopic ultrasound.
What is gastroparesis?
Weak stomach. Cause is diabetes. Presents with early satiety, postprandial nausea, and sense of increased abdominal fullness. Abdominal pain and bloating and diabetes, with retinopathy, neuropathy, nephropathy.
What is the treatment for gastroparesis?
Increase motility of the stomach with erythromycin or metoclopramide.
What are the symptoms of inflammatory bowel disease?
Fever, diarrhea, weight loss, and abdominal pain and bleeding. Episcleritis, scleritis and iritis, sclerosing cholangitis, joint pains, and pyoderma gangrenosum or erythema nodosum.
What are skip lesions in inflammatory bowel disease?
"Skip lesions" are areas of normal tissue in between the areas of disease; associated with Crohn disease.
What are the characteristics of ulcerative colitis?
UC that is a mucosal disease which is limited to the large bowel. UC has no skip lesions, no fistula formation, and no oral or perianal involvement. UC is more likely to cause bloody diarrhea.
Which type of inflammatory bowel disease can lead to colon cancer?
Both forms of IBD can lead to colon cancer after 8 to 10 years of involvement of the colon.
What are the complications of Crohn disease?
Complications of Crohn disease are calcium oxalate kidney stones, diarrhea, and cholesterol gallstones.
How is inflammatory bowel disease diagnosed?
Colonoscopy. Antineutrophil cytoplasmic antibody is associated with UC.
What laboratory abnormalities are associated with Crohn disease?
B12, calcium, vitamin K, iron deficiencies because malabsorption. Anti-saccharomyces cerevisiae antibodies. PT increased because vitamin K malabsorption. Kidney stones because increased absorption oxalate.
What is the treatment of inflammatory bowel disease?
Pentasa is a form of mesalamine released in upper and lower bowel; hence, it is used in CD. Asacol is a form of mesalamine released in the large bowel, and it is most useful for UC. Rowasa is used for rectal disease.
What is the treatment of exacerbations of inflammatory bowel disease?
Acute exacerbations of IBD are treated with high-dose steroids. Budesonide has a strong local effect when used orally, but is largely cleared by the liver in a first-pass effect.
What is the treatment of Crohn perianal disease?
Ciprofloxacin and metronidazole are used for CD with perianal disease. Infliximab is used for CD fistulae or refractory disease.
What is the role of surgery in inflammatory bowel disease?
Surgery is curative in UC; almost 60% will require surgery within 5 years after the diagnosis of UC due to refractory symptoms or severe disease. Surgery is less effective in CD and the disease reoccurs after surgery.
What is the laboratory evaluation of inflammatory bowel disease?
Stool should be evaluated for fecal leukocytes, culture, and ova and parasite examination. Clostridium difficile toxin and stool Giardia-antigen testing.
What are the most common causes of infectious diarrhea?
Campylobacter and Salmonella.
What is the presentation of Bacillus cereus diarrhea?
Ingestion of refried Chinese food. Short incubation period (1-6 hours). Vomiting is prominent. Blood is never present.
What type of diarrhea is associated with reactive arthritis and Guillain-Barre syndrome?
Campylobacter
What type of diarrhea occurs in HIV-positive patients with <100/mm3 CD4 cells?
Cryptosporidium and Isospora.
What type of food is associated with E. coli 0157:H7?
Hamburger meat. Shiga toxin may cause hemolytic uremic syndrome when organism dies; therefore, antibiotics are contraindicated. Platelet transfusions exacerbate the disease.
What type of ingestion is associated with Giardia diarrhea?
Unfiltered water on a camping trip. Giardia never causes blood in stool. There is abdominal fullness, bloating, and gas. Giardia can also simulate celiac disease by causing a fat and vitamin malabsorption.
What type if diarrhea is associated with ingestion of chicken and eggs, dairy products?
Salmonella
What type of ingestion is associated with scombroid poisoning?
Contaminated fish, resulting in vomiting, diarrhea, flushing, wheezing within minutes. Bacteria invade, producing and then releasing histamine into the flesh of fish, such as tuna, mahi mahi, mackerel and amberjack.
What is the presentation of Yersinia diarrhea?
Yersinia can mimic appendicitis. Also common in people with iron overload, e.g., hemochromatosis.
What type of ingestion is associated with Vibrio parahaemolyticus?
Ingestion of raw shellfish, such as mussels, oysters, and clams.
What type of ingestion is associated with Vibrio vulnificus?
Raw shellfish; high incidence in people with underlying liver disease or disorders of iron metabolism. Associated with the development of skin bullae.
What population is predisposed to viral diarrhea?
Children in day-care centers; absence of blood and white cells
Ingestion of what type of foods is associated with Staphylococcus aureus gastroenteritis?
Ingestion of dairy products, eggs, salads. Short incubation period (1-6 hours); nausea/vomiting predominate; rarely diarrhea.
Ingestion of what type of food is associated with ciguatera-toxin?
2-6 hours after ingestion of grouper fish, red snapper, and barracuda. Neurological symptoms include paresthesia, weakness, and reversal of heat and cold.
How is acute diarrhea diagnosed?
Fecal leukocytes is most useful test. Found when there has been invasion of the intestinal mucosa, as in dysentery, which is a bacterial infection of the bowel, producing diarrhea and bloody stool.
What are the invasive organisms that cause infectious diarrhea?
Salmonella, Shigella, Campylobacter, V parahaemolyticus, Yersinia, E coli, Vibrio vulnificus. Invasive organisms need 24 h to produce effect. The most definitive test for these bacteria is stool culture.
How is cryptosporidiosis diagnosed?
Cryptosporidiosis is diagnosed with a modified acid-fast test.
How is giardia diagnosed?
Giardia is diagnosed with an ELISA stool antigen test. 90% sensitivity.
What is the treatment of acute infectious diarrhea?
Antibiotics are used if there is abdominal pain, blood in the stool, and fever. Empiric therapy for infectious diarrhea is ciprofloxacin.
What is the treatment for scombroid poisoning?
Scombroid poisoning is treated with diphenhydramine.
What is the treatment of Giardia?
Giardia is treated with metronidazole. A newer agent for Giardia is tinidazole, which is effective in a single dose.
What is the treatment of Cryptosporidiosis?
Cryptosporidiosis is treated with paromomycin and raise the CD4 count to >100/mm3 with antiretrovirals.
What is antibiotic-associated diarrhea?
Most common cause is Clostridium difficile. Any antibiotic may cause diarrhea, although clindamycin may have one of the highest frequencies. Stool test for C. difficile toxin.
What is the treatment for antibiotic-associated diarrhea?
Metronidazole. Oral vancomycin only if not resolve with metronidazole.
What is the presentation of lactose intolerance?
Diarrhea with gas and bloating; never has blood or leukocytes. Weight loss does not occur. High prevalence. Increased stool osmolality and increased osmolar gap.
How is lactase deficiency diagnosed?
Remove milk, cheese, ice cream, and all other dairy products (except yogurt) from the diet; resolution of symptoms should occur within 24 to 36 hours. Dietary changes are the best therapy; lactase supplements.
What is irritable bowel syndrome?
Pain syndrome in which there is increased frequency of peristaltic contractions.20% have constipation. Some have diarrhea alone or diarrhea alternating with constipation. No nocturnal symptoms. History of abuse.
How is irritable bowel syndrome diagnosed?
No specific diagnostic test. Exclude lactose intolerance, inflammatory bowel disease, celiac disease, carcinoid, Giardia infection, and anatomic defects of the bowel.
What is the Rome criteria for irritable bowel syndrome?
Pain relieved by a bowel movement or by a change in bowel habit. Fewer symptoms at night. Diarrhea alternating with constipation. No fever, weight loss, anorexia, or anemia.
What is the treatment for irritable bowel syndrome?
High-fiber, loperamide, diphenoxylate. Antispasmodics: hyoscyamine, dicyclomine, belladonna. Tricyclics. Tegaserod in constipation-predominant; may cause diarrhea. Alosetron reduces diarrhea.
What is carcinoid syndrome?
Tumors that cause neuroendocrine syndrome. Located in appendix and ileum. Symptoms appear after liver has been overwhelmed by metastatic disease. Bronchial carcinoids are rare but are highly symptomatic.
What is the presentation of carcinoid syndrome?
Diarrhea, flushing, tachycardia, hypotension. Tricuspid insufficiency and pulmonic stenosis.
How is carcinoid syndrome diagnosed?
Urinary 5-hydroxyindolacetic acid level (5-HIAA).
What is the treatment of carcinoid syndrome?
Control diarrhea with octreotide (somatostatin analog). Very few carcinoids are localized enough for surgical resection.
What are causes of fat malabsorption?
Celiac disease and chronic pancreatitis; tropical sprue and Whipple disease are extremely rare causes.
What is the presentation of fat malabsorption?
Greasy diarrhea, oily, floating, foul smelling. Weight loss. Malabsorption of the fat-soluble vitamins A, D, E, K. Hypocalcemia, easy bruising; prolongation PT. Iron malabsorption, macrocytic anemia.
What type of malabsorption causes Vitamin B12 deficiency?
Vitamin B12 malabsorption occurs from damage or loss of the mucosal surface of the terminal ileum from celiac disease.
What is the most common presentation of celiac disease?
Iron deficiency anemia. Dermatitis herpetiformis is a vesicular skin rash on the extensor surfaces of the body in 10%.
What is the most common cause of fat malabsorption?
Celiac disease is the most likely cause of fat malabsorption because it is the most common.
What is the presentation of malabsorption caused by chronic pancreatitis?
Patients with chronic pancreatitis will give a history steatorrhea with repeated episodes of pancreatitis from alcohol or gallstones.
What type of malabsorption is associated with travel to a tropical country?
Tropical sprue is suspected when there is a history of being in a tropical country.
What is the presentation of Whipple disease?
Whipple disease is the rarest cause of fat malabsorption; dementia (10%), arthralgias (80%), and ophthalmoplegia. Caused by the bacterium Tropheryma whipplei.
How is celiac disease diagnosed?
Antigliadin, antiendomysial, and antitransglutaminase antibodies. Small bowel biopsy shows flattening of villi. Just removing gluten (wheat, oats) from diet is not effective for diagnosis because antibodies.
What is the presentation of chronic pancreatitis?
Chronic pancreatitis is diagnosed from the history of repeated episodes of pancreatitis and is confirmed by finding calcification of the pancreas on x-ray and CT scan.
How is tropical sprue diagnosed?
Tropical sprue and Whipple disease are diagnosed by finding organisms on a bowel-wall biopsy. A positive Tropheryma Whippelii biopsy shows foamy macrophages that are PAS positive.
What is the treatment of malabsorption?
Chronic pancreatitis can be managed by orally replacing, amylase, lipase, trypsin. Tropical sprue is treated with sulfamethoxazole/trimethoprim or doxycycline.
What is the treatment of Whipple disease?
Whipple disease is treated with sulfamethoxazole/trimethoprim or doxycycline or ceftriaxone for 1 year.
What is diverticulosis?
Diverticulosis is caused by a lack of fiber in the diet and an increase in intracolonic pressure, leading to outpocketing of the colon. 50% of persons are over the age of 50.
What is the presentation of diverticulosis?
Asymptomatic. Left lower quadrant abdominal pain that can be colicky in nature.
How is diverticulosis diagnosed?
Colonoscopy. Diverticula are more on left in sigmoid, but bleeding occurs more often from diverticula on the right because of thinner mucosa and more fragile blood vessels. Bleeding is painless.
What is the treatment for diverticulosis?
Dietary fiber in bran, or bulking agents, such as psyllium husks.
What is the presentation of diverticulitis?
Fever, tenderness, pain, and elevation of white blood cell count.
How is diverticulitis diagnosed?
CT scanning. Barium studies and endoscopy are relatively contraindicated in diverticulitis.
What is the treatment for diverticulitis?
Cipro and metronidazole. Other choices are ampicillin/sulbactam, piperacillin/ tazobactam, or the combination of cefotetan or cefoxitin with gentamicin. Mild disease treated with amoxicillin/ clavulanate.
What are the causes of constipation?
Lack of fiber; calcium-channel blockers, oral ferrous sulfate, hypothyroidism, opiate analgesics, anticholinergics, and tricyclic antidepressants.
What is the lifetime risk of colon cancer?
>6%. Most cases occur sporadically. Diets that are high in red meat, fat and smoking increase the risk.
What is the presentation of colon cancer?
Heme-positive, brown stool; chronic anemia when the cancer in right side of colon. Left-sided lesions cause obstruction and decreased stool caliber.
What type of cancer is associated with endocarditis caused by Streptococcus bovis?
Colon cancer is associated with endocarditis by Streptococcus bovis and Clostridium septicum.
What is the treatment for colon cancer?
Localized disease easily be cured. Once the disease has spread, then surgical resection will not be effective. Widespread disease is treated with resection and 5-FU.
What is the screening for colon cancer?
Annual fecal occult blood testing, colonoscopy every 10 years, and sigmoidoscopy every 5 years. Screening should occur after the age of 50 years.
What are the causes of a false-positive stool guaiac test?
False-positive stool guaiac tests can be caused by aspirin, NSAIDs, red meat, and poultry. False-negative tests can be caused by vitamin C.
What is hereditary polyposis colorectal cancer syndrome?
Genetic defect causing colon cancer. 3 family members in at least 2 generations with colon CA. 30% develop endometrial/ovarian cancer. Screening should start at the age of 25 with colonoscopy every 1 to 2 years.
What is familial adenomatous polyposis?
100% adenomas by 35 yrs and of colon cancer by 50. Polyps by age 25. Screening flexible sigmoidoscopy should be done every 1 to 2 years beginning at age 12. If polyps are found, a colectomy should be performed.
What is the colon cancer risk in juvenile polyposis syndrome?
Juvenile polyposis syndrome has 10% risk of colon cancer. Hamartomas have very little risk of developing into cancer.
What is Gardner syndrome?
Gardner syndrome is colon cancer with multiple, osteomas, lipomas, cysts, and fibrosarcomas. The osteomas have predilection for the mandible.
What is Peutz-Jeghers syndrome?
Hamartomatous polyps in the large and small intestine with hyperpigmented spots. Melanotic spots on lips, buccal, skin. Risk of cancer increased. Intussusception, obstruction.
What is the usual outcome of gastrointestinal bleeding?
80% of cases of gastrointestinal bleeding will resolve spontaneously with supportive management, irrespective of etiology.
What is the initial management of gastrointestinal bleeding?
Fluid resuscitation with NS. CBC, PT, T&C. If PT is elevated, give FFP. Platelets if count is <50,000. NG tube is only useful to determine site of bleeding.
What medications should be given to patients with gastrointestinal bleeding?
Patients with gastritis or ulcer disease should be treated with PPIs empirically. H2 blockers have no efficacy in acute gastrointestinal bleeding.
What medication reduces portal hypertension in patients with cirrhosis of the liver and gastrointestinal bleeding?
If a history of cirrhosis of the liver, or if long-term alcoholic, octreotide should be added to this initial management plan to decrease portal hypertension.
What are the indications for transfusion in acute bleeding?
Hematocrit should be maintained >30% in older patients or CAD. Younger patients do not need to be transfused, unless their hematocrit is <20%.
What is the treatment of bleeding esophageal varices?
Octreotide to lower portal pressure. If ineffective, endoscopy to place bands. If banding is not effective, then transjugular intrahepatic portosystemic shunting.
What is transjugular intrahepatic portosystemic shunting?
Creates shunt between the systemic circulation of hepatic vein and portal circulation of portal vein.
What is the primary complication of TIPS?
Complication of transjugular intrahepatic portosystemic shunting is worsening of encephalopathy.
What are the indications for a Blakemore tube?
A Blakemore tube tamponades the site of bleeding in the stomach or esophagus; rarely used and is only a temporary bridge to surgery.
What medication reduces risk of bleeding in portal hypertension?
Propranolol is a nonselective beta-blocker that used in the long-term management of portal hypertension to decrease the frequency of bleeding.
What are the most common causes of upper gastrointestinal bleeding?
Ulcers, gastritis, Mallory-Weiss, esophagitis, gastric cancer. Variceal bleeding is common in portal hypertension from cirrhosis.
What is the anatomic definition of upper gastrointestinal bleeding?
Upper gastrointestinal bleeding is defined as bleeding proximal to the ligament of Treitz.
What are the causes of lower gastrointestinal bleeding?
Diverticulosis, angiodysplasia, hemorrhoids, cancer, and inflammatory bowel disease.
What is the clinical presentation of lower gastrointestinal bleeding?
Red blood in stools; upper gastrointestinal bleeding presents with black stool or melena.
What degree of blood loss is indicated by the finding of orthostasis?
Orthostasis is a >10 rise in pulse or >20 drop in systolic BP when the patient goes from supine to standing or sitting. Orthostasis indicates a 15-20% blood loss.
If the resting pulse is >100 bpm, what amount of blood loss has occurred?
Pulse above 100/min or a systolic blood pressure under 100 indicates >30% blood loss.
How is lower gastrointestinal bleeding diagnosed?
Endoscopy is the most accurate test for upper and lower GI bleeding. If endoscopy is unrevealing, nuclear bleeding scan can detect low volume bleeds 0.1-0.5 mL/min. Small bowel bleeding detected capsule endoscopy.
What are the causes of acute pancreatitis?
Majority from alcoholism/gallstones. Pentamidine, didanosine, azathioprine, sulfas (sulfamethoxazole/trimethoprim thiazides). Hypercalcemia, hypertriglyceridemia. ERCP, trauma, mumps.
What is the pathophysiology of pancreatitis?
Premature activation of trypsinogen into trypsin while in the pancreas. Autodigestion of the pancreas.
What is the clinical presentation of acute pancreatitis?
Midepigastric pain with tenderness, nausea, and vomiting; radiates to the back. Septic shock; fever, hypotension, respiratory distress from ARDS, elevation of the white cell count; rigid abdomen.
How is acute pancreatitis diagnosed?
Lipase is more specific than amylase. Elevated WBC, hypoxia, glucose, LDH, AST. Calcium decreases because malabsorption of fat allows fat to bind with Ca. BUN increases because volume depletion.
What test determines the severity of pancreatitis?
The most accurate test to determine the severity of pancreatitis is the CT scan, which will demonstrate inflammation, necrosis, pseudocysts, abscesses, and ductal stones. Findings on the CT scan are a more
What are the signs of severe necrotizing pancreatitis?
Cullen sign: blue around umbilicus caused by hemoperitoneum. Turner's: Purple flanks caused by tissue Hb. Best test for biliary/ pancreas is ERCP. Urinary trypsinogen activation peptide predicts severity.
What are the complications of pancreatitis?
Ascites (high in amylase); pleural effusion (transudate, amylase); splenic vein thrombosis.
What is the treatment of pancreatitis?
Intravenous fluids, bowel rest, and pain medication. ERCP removes stone in the pancreatic duct or and to dilate a stricture.
What as the management of pancreatic necrosis?
Necrosis on CT is an indication imipenem or cefuroxime. Fever is an indication to perform a percutaneous needle biopsy. Infection of the pancreas and necrosis requires debridement.
What is the management of pancreatic pseudocysts?
Pseudocysts develop 2 weeks after pancreatitis. Pseudocysts should be drained if pain, fistula, rupture, or if expanding. Small, asymptomatic pseudocysts <5 cm do not need drainage.
What is the pathophysiology of cirrhosis of the liver?
Cirrhosis develops when there is chronic inflammation of the liver. When at least 70 to 80% of liver function has been lost, the synthetic capacity of the liver is diminished.
What are the causes of cirrhosis of the liver?
The most common cause of cirrhosis is alcohol. Other causes of cirrhosis are primary biliary cirrhosis, sclerosing cholangitis, alpha-1 antitrypsin deficiency, hemochromatosis, and Wilson disease.
What are the signs of hepatic cirrhosis?
Esophageal varices, ascites, edema; spider angiomata, palmar erythema, asterixis, encephalopathy, jaundice, low albumin. PT prolonged because of inability to make clotting factors.
What is spontaneous bacterial peritonitis?
Idiopathic infection of ascites. Gram stain is rarely positive. Culture of the fluid is the most specific test. White cell count of >500 or the presence of >250 neutrophils are criteria for infection.
What is the treatment of spontaneous bacterial peritonitis?
Cefotaxime and ceftriaxone are the drugs of choice for SBP and albumin infusion decreases risk of hepatorenal syndrome.
What is the serum-ascites albumin gradient?
Ascitic albumin is normally < serum. The difference is the serum-ascites albumin gradient. When SAAG is >1.1, portal hypertension is cause. When SAAG is <1.1, cancer or infection is cause.
What is the treatment of cirrhotic edema?
Edema and fluid overload in third spaces, such as ascites, are managed with spironolactone.
What is the treatment of cirrhotic portal hypertension?
Portal hypertension and varices are managed with propranolol to prevent bleeding. Encephalopathy is managed with neomycin or lactulose. Vitamin K is not effective.
What is primary biliary cirrhosis?
Idiopathic autoimmune disorder in middle-aged women. Bilirubin levels do not elevate until 5 years. Association with Sjögren syndrome, rheumatoid arthritis, and scleroderma.
What is the presentation of primary biliary cirrhosis?
Fatigue, pruritus. Asymptomatic with an elevated alkaline phos. Osteoporosis. Transaminases normal. GGTP, IgM elevated. Antimitochondrial Ab specific. Biopsy diagnostic.
What is the treatment for primary biliary cirrhosis?
Bile acid-binding medication, such as ursodeoxycholic acid and cholestyramine. Ultraviolet light. Liver transplant.
What is the presentation of primary sclerosing cholangitis?
Fatigue, pruritus; increased alkaline phos. Idiopathic disorder of biliary system associated more often ulcerative colitis, also with Crohn. Cancer of biliary system in 15%.
How is primary sclerosing cholangitis diagnosed?
Elevated alkaline phosphatase; the antimitochondrial antibody test is negative. The most specific test for primary sclerosing cholangitis is an ERCP or transhepatic cholangiogram.
What is the treatment for primary sclerosing cholangitis?
Bile acid-binding resins. Liver transplant.
What is hemochromatosis?
Common genetic disease of overabsorption of iron in duodenum. Hepatic inflammation, fibrosis, cirrhosis. Hepatocellular CA in 20%. Restrictive cardiomyopathy 15%. Arthralgias, skin hyperpigmentation, diabetes.
What are the laboratory findings in hemochromatosis?
Decreased iron-binding capacity. Ferritin is elevated. Liver biopsy.
What is the treatment of hemochromatosis?
Phlebotomy to remove iron.
What is Wilson disease?
Autosomal recessive; diminished ability to excrete copper. Copper in liver, brain, cornea. Basal ganglia movement disorder. Kayser-Fleischer rings in eye on slit-lamp. Fanconi syndrome and type II RTA.
How is Wilson disease diagnosed?
Low ceruloplasmin level. High urinary copper level. Liver biopsy is most specific.
What is the treatment for Wilson disease?
Penicillamine is a copper chelator. Liver transplantation is curative.
What is alpha-1 antitrypsin deficiency?
Autosomal recessive disorder leading to deficient alpha-1 antitrypsin levels. Cirrhosis, emphysema developing at a young age in a nonsmoker.
How is alpha-1 antitrypsin deficiency diagnosed?
Low level of the enzyme in a person with COPD confirms diagnosis.
What is the treatment of alpha-1 antitrypsin deficiency?
Replacement of the enzyme and stop smoking.
How is chronic hepatitis B and C diagnosed?
Persistence of hepatitis B surface antigen for more than 6 mth confirms chronic hepatitis B. Ab to hepatitis C and elevated viral load by PCR confirms chronic hepatitis C. Liver biopsy most accurate.
What is the treatment for chronic hepatitis B?
Chronic hepatitis B is treated with interferon, lamivudine, or adefovir.
What is the treatment for chronic hepatitis C?
Chronic hepatitis C is treated with the combination of pegylated interferon and ribavirin.
What are the important aspects of the chest pain history?
Duration, quality, location, radiation, frequency, alleviating or precipitating factors (exercise), associated symptoms. For acute coronary syndromes the quality of chest pain is described as "tightness," or "pressure."
What is the clinical presentation of inferoposterior myocardial ischemia?
Nausea and vomiting are sometimes the main symptoms in inferoposterior wall ischemia. In inferoposterior wall ischemia, vagal reflexes may cause bradycardia and hypotension.
What are the characteristics of pain that is less likely to be caused by ischemia?
"Sharp" chest pain that the patient can pinpoint is not likely caused by ischemia, especially if pain is reproduced by position or palpation. Diaphoresis, tachypnea, anxiety indicate a life-threatening process.
What is the usual amount of time that myocardial infarction pain is present?
Myocardial infarction lasts more than 20-30 minutes.
What etiology is suggested if pain is relieved by nitroglycerine?
Relief of chest pain with nitroglycerin indicates ischemia or esophageal spasm. Chest pain that worsens with nitroglycerin occurs with gastroesophageal reflux disease.
What is the presentation of acute coronary syndrome in women?
Acute coronary syndromes in women present with atypical symptoms: dyspnea, shortness of breath, fatigue.
What are the physical examination findings that suggest aortic dissection?
Blood pressure a difference between arms of over 20 mm Hg systolic suggests aortic dissection.
What etiologies of chest pain are suggested by hypotension?
Massive pulmonary embolism or cardiac shock. Fever may suggest pneumonia or esophageal rupture. Corneal lipid rings, narrowed retinal arteries, murmurs; pigment and hair-loss on legs indicate coronary syndrome.
What etiology is suggested by wide splitting of the second heart sound?
Wide physiologic splitting of the second heart sound (splitting wider with inspiration) can be found in right ventricular infarction.
What is the significance of fourth heart sound in patients with chest pain?
A new fourth heart sound can occur with angina or infarction. An S3 is may be caused by underlying heart failure.
What etiology is suggested by the finding of a murmur of aortic regurgitation in patients with ischemic chest pain?
Significant aortic regurgitation occurs in over half of patients with aortic dissection, while mitral regurgitation can occur in patients with angina or infarction and is due to papillary muscle dysfunction.
What are the 12-lead ECG signs of an acute myocardial infarction?
50% of persons with acute MI will have diagnostic findings (ST elevation or Q waves >0.04 sec), while 35% will have findings consistent with ischemia (ST depression and/or T wave inversion).
What is the role of creatine kinase MB isoenzyme in patients with an acute coronary syndrome?
CK-MB isoenzyme is cardiac specific and is useful for early diagnosis of MI.
At what time after an acute myocardial infarction does the CK-MB become elevated?
CK-MB detectable 4-6 hrs after ischemia onset, peaks 12-24 hrs, normalizes 2-3 d; can be used to detect early reinfarction. CK-MB levels are obtained at admission to ED and repeated 6-12 hrs.
What are cardiac troponins?
Troponins (T, I, C) are in striated and cardiac muscle. T and I are preferred markers for diagnosis of myocardial injury. Troponin T and I have similar sensitivity and specificity for MI.
What is the difference between troponin T and I?
Unlike troponin I levels, troponin T levels may be elevated in patients with renal disease, polymyositis, or dermatomyositis.
What is the significance of an elevated troponin level and a normal CK-MB in a patient with chest pain?
Patients with a normal CK-MB level but elevated troponin levels have sustained minor myocardial damage, or microinfarction. Elevations of both CK-MB and troponins is acute MI.
How much time is required before cardiac troponins return to normal after an acute myocardial infarction?
The cardiac troponins may remain elevated up to two weeks after symptom onset, which makes them useful as late markers of recent acute myocardial infarction.
What is the role of the chest x-ray in patients with chest pain?
Pneumothorax, pneumomediastinum, effusion, infiltrates. Aortic dissection can cause widening of mediastinum. Loss of lung volume or unilateral loss of vascular markings suggest PE.
What are the symptoms of aortic dissection?
Pain is sharp, tearing, severe; radiates to back; loss of pulses; aortic insufficiency murmur; mediastinum is widened; confirmed by MRI, CT scan, transesophageal echocardiogram, or aortography.
What are the symptoms of pulmonary embolism?
Dyspnea, tachycardia, hypoxemia; pleuritic pain.
How is pulmonary embolism diagnosed?
ECG may show an S wave in lead I, Q wave in lead III, or inverted T wave in lead III. Diagnosis is confirmed by spiral chest CT, lung scan, or pulmonary angiogram.
What are the symptoms of pericarditis?
Pain is sharp, positional, pleuritic, and relieved by leaning forward; preceded by viral illness; pericardial rub.
What are the ECG signs of acute pericarditis?
Diffuse ST elevation occurs without evolution of Q waves; CK level usually normal.
What are the symptoms of myocarditis?
Pain is vague and mild if present preceded by viral illness; MB fraction of CK are often elevated; tachycardia, conduction abnormalities and sometimes Q waves occur.
What are the musculoskeletal causes of chest pain?
Musculoskeletal causes are the most common cause of chest pain. Costochondritis, cervical osteoarthritis, radiculitis; pain is stabbing, localized; pleuritic; reproduced by motion or palpation; ECG changes absent.
What are the gastrointestinal causes of chest pain?
Esophageal reflux is worse with recumbency or meals; regurgitation; relieved by antacids, nitroglycerin. Ulcers, cholecystitis. Pancreatitis causes abdominal tenderness, with radiation to back, hyperamylasemia.
What are the symptoms of pneumothorax?
Onset abrupt with sharp pleuritic chest pain and dyspnea; breath sounds are absent, chest x-ray confirms.
What are the symptoms of pleuritis?
Pain is sharp and increases on inspiration; pleural friction rub; underlying pulmonary infection.
What are the major modifiable risk factors for ischemic heart disease?
Hypercholesterolemia, smoking hypertension, physical inactivity, obesity, diabetes.
What subgroup of hypercholesterolemia is the most important risk factor for ischemic heart disease?
LDL cholesterol is single most important subgroup that carries risk for ischemic heart disease. Low HDL cholesterol, hypertriglyceridemia, increased total-to-HDL-cholesterol ratio, and increased lipoprotein A increase risk.
What is the risk of myocardial infarction in smokers?
Cigarette smoker's risk of heart attack is more than twice that of a nonsmoker. The risk for myocardial infarction in patients who quit smoking is reduced to that of nonsmokers within two years of cessation.
What degree of hypertension is a risk factor for cardiovascular events?
Risk for cardiovascular events increases at BP above 110/75 mm Hg. HTN is a risk factor for myocardial ischemia, stroke, kidney failure, and heart failure.
What percentage of patients with diabetes die of cardiovascular disease?
Almost 75% of patients with diabetes die of some form of cardiovascular disease. Elevated blood glucose levels and insulin resistance are associated with ischemic heart disease and overall cardiovascular events.
What are the clinical signs of stable angina?
Substernal pressure lasting 5-15 min; not affected by respiration/position. Stable angina occurs when myocardium becomes ischemic during exercise, hypotension, anemia.
What is the most common ECG sign in patients with angina?
Most commonly ST segment depression is seen. ST segment elevation typically occurs in variant angina (Prinzmetal angina) where coronary artery spasm is responsible.
How is ischemic heart disease diagnosed?
The exercise treadmill test is the most useful test in the evaluation of the cause of chronic chest pain when ISCHEMIC HEART DISEASE (stable angina) is a consideration.
What is the criteria for a positive exercise stress test for myocardial ischemia?
>2 mm ST-segment depressions or hypotension (a drop of more than 10 mm Hg in systolic pressure) occur either alone or in combination.
What type of noninvasive test for ischemic heart disease is indicated for patients who are unable to exercise?
Patients who are unable to exercise or walk should be considered for chemical stress testing, such as dipyridamole (Persantine) or dobutamine stress tests.
What type of noninvasive test for ischemic heart disease is indicated for patients with bundle branch block?
Nuclear stress imaging. These tests may also be used in patients who are taking digoxin or beta-blockers. Beta-blockers may blunt the heart rate during exercise.
What is a nuclear stress test?
Radioactive substance is injected and perfusion of heart tissue is visualized at rest and after exercise. Abnormal amount of thallium in areas of heart that have decreased blood.
What are the dobutamine and adenosine stress tests?
This test is used in people who are unable to exercise. A drug is given to induce tachycardia.
What is a stress echocardiogram?
The stress echocardiogram combines a treadmill stress test and an echocardiogram (ECHO). Abnormal movement of the walls of the left ventricle are induced by exercise.
What are the indications for cardiac catheterization?
Patients with stable angina for (1) diagnosis and (2) prognosis/risk stratification. Angiography is also used after stress test are positive to identify patients that will benefit form stent or bypass.
What is the treatment of ischemic heart disease?
Nitroglycerin SL alleviates pain. Long-acting nitrates, beta blockers. Aspirin and statins. All patients with stable angina need cardiac angiography or stress testing.
What are the target goals for treatment of hyperlipidemic patients with coronary artery disease?
LDL less than 100 mg/dL. HDL 40 mg/dL or greater. Triglycerides less than 150 mg/dL.
What is the optimal LDL-cholesterol goal for patients at very high risk of coronary artery disease?
<70 mg/dL for very high risk with cardiovascular disease plus diabetes, smoking, HTN; metabolic syndrome (triglycerides >200, HDL <40); and patients with ACS. Statin therapy regardless of levels.
What are the indications for coronary bypass graft in ischemic heart disease?
Left main coronary disease or three vessel disease and left ventricular (LV) dysfunction. More efficacious in diabetes. More beneficial in low ejection fraction.
What are the types of acute coronary syndromes?
Acute coronary syndromes (ACS) include a range of thrombotic coronary diseases, including unstable angina (UA), non-ST elevation myocardial infarction, and STEMI.
What is a non ST-segment elevation myocardial infarction?
Ischemia causes damage detectable by troponin I, troponin T, or CK-MB. If there are no detectable serum markers after 24 h, the patient has UA. UA and NSTEMI may be indistinguishable.
What are the indications for thrombotic therapy in patients with acute coronary syndrome?
Thrombolytic therapy is beneficial for ST elevation MI (STEMI), but is not effective in UA or NSTEMI because the infarct-related artery is not occluded.
What is unstable angina?
Angina of increasing severity, frequency, duration; angina with increasing resistance to nitrates; or angina occurring at rest or new-onset angina. Untreated unstable angina progresses to MI in 50% of cases.
What are the ECG findings in non-segment elevation MI?
Dynamic ST-segment depression (>5 mm), or new T-wave inversion ( 2 mm), but the ECG may be normal or show minor changes.
What are the high-risk features for patients with UA/NSTEMI?
Repetitive or prolonged chest pain (>10 min), elevated markers, persistent ST depression >0.5 mm or new T-wave inversion, SBP <90, VT; syncope, ejection fraction <40%, prior PTCA or CABG, diabetes, kidney disease.
What is the general management of NSTEMI?
Aspirin is recommended in all patients unless contraindicated. High-risk patients should be treated with aggressive medical management and arrangements should be made for coronary angiography.
What are the drugs that are used in NSTEMI?
Aspirin, clopidogrel, heparin, tirofiban, metoprolol, nitroglycerine.
What antiplatelet therapy should be given to patients with NSTEMI?
Aspirin and clopidogrel. Clopidogrel should be avoided if likely to require bypass surgery (severe ST-segment depression, SBP <90). Clopidogrel should be given before percutaneous coronary intervention.
What are the indications for anticoagulants in NSTEMI?
Unfractionated heparin or subcutaneous enoxaparin should be given until angiography or for 48-72 hours. Enoxaparin dose must be reduced in patients with impaired renal function.
What is the mechanism of the glycoprotein (GP) Hb/IIIa inhibitors?
Inhibit platelet function by blocking a receptor involved in platelet aggregation, providing a more comprehensive platelet blockade than the combination of aspirin and heparin.
What is the indication for tirofiban or eptifibatide in NSTEMI?
High-risk patients in whom an invasive strategy is planned. Tirofiban is particularly beneficial and recommended in patients with diabetes. Complications are bleeding and thrombocytopenia.
What are the indications for a beta-blocker in patients with NSTEMI?
A beta-blocker should be given unless contraindicated. Intravenous nitroglycerin is given for refractory pain.
Which patients with NSTEMI should receive invasive management?
Early angiography (48 h) and revascularization is recommended in NSTEMI with high-risk features. Refractory ischemia, and high-risk features on early exercise testing need early invasive.
What are the clinical signs of ST elevation myocardial infarction?
Substernal, diffuse, pressure pain. Radiation neck, jaw, shoulders, arms. Lightheadedness, nausea, vomiting, diaphoresis, dyspnea. Pain lasts longer than 20 min, does not respond completely to nitroglycerin.
What are the signs of STEMI in elderly and diabetic patients?
Elderly and diabetic patients are prone to atypical symptoms, such as nausea or dyspnea. 20% of MI are "silent".
What are the physical signs of ST elevation myocardial infarction?
Bradycardia in inferior MI. Tachycardia with large MI. BP elevated. Left ventricular failure, valve dysfunction. S4 of stiffened ventricle. Mitral regurgitation if papillary muscle malfunction. S2 paradoxical split as LV contraction time increases due to
What are the ECG criteria for STEMI?
Persistent ST-segment elevation of >1 mm in two contiguous limb leads. ST-segment elevation of >2 mm in two contiguous chest leads. New LBBB. Initially, markers are not elevated in STEMI (eventually positive).
What is the initial nonspecific management of acute coronary syndrome?
Initial nonspecific management for all patients with possible MI (chest pain) is oxygen therapy, aspirin (unless contraindicated), nitroglycerin and morphine.
What is the management of STEMI in patients who present within 12 hours of the onset of ischemic symptoms?
Completely occluded coronary artery with thrombus at the site of a ruptured plaque. Reperfusion strategy with fibrinolytic therapy or percutaneous coronary intervention.
Which ECG leads have ST elevation in an inferior myocardial infarction?
II, III, aVF
Which coronary artery is occluded in an inferior myocardial infarction?
Right coronary.
Which ECG leads have ST elevation in an anteroseptal myocardial infarction?
V1-V3
Which coronary artery is occluded in an anteroseptal myocardial infarction?
Left anterior descending
Which ECG leads have ST elevation in an anterior myocardial infarction?
V2-V4
Which coronary artery is occluded in an anterior myocardial infarction?
Left anterior descending artery.
Which ECG leads have ST elevation in a lateral myocardial infarction?
I, aVL, V4, V5, and V6
Which coronary artery is occluded in an anterior myocardial infarction?
Left anterior descending or circumflex artery.
Which ECG leads have ST elevation in a posterior MI?
V1-V2: Tall broad initial R wave, ST depression, tall upright T wave; usually occurs in association with inferior or lateral MI.
Which coronary artery is occluded in a posterior myocardial infarction?
Posterior descending
What is the time of onset of hyperacute T waves (tall, peaked T waves) after a myocardial infarction?
Immediately. Disappear in 6-24 hours.
What is the time of onset of ST-segment elevations after a myocardial infarction?
Immediately
At what time do ST-segment elevations disappear after a myocardial infarction?
1-6 weeks
What is the time of onset of Q waves longer than 0.04 seconds after a myocardial infarction?
One to several days. 0.04 seconds is one small ECG box.
At what time do Q waves longer than 0.04 seconds disappear after a myocardial infarction?
Years to never
What is the time of onset of T wave inversions after a myocardial infarction?
6-24 hours
At what time do T wave inversions disappear after a myocardial infarction?
Disappear months to years later.
What is the best emergent reperfusion therapy?
PCI is the best available treatment because of improved short-term and long-term reduction of deaths and MI in patients with STEMI presenting within 12 hours when compared with thrombolytic therapy.
What are the indications for thrombolytics?
tPA is the most common agent used in the US. Thrombolytic agent is an alternative to primary PCI in suitable candidates with: ST-elevation MI (>1 mm ST elevation in two contiguous leads). New LBBB
What are the contraindications to thrombolytic therapy?
Active bleeding or bleeding diathesis, head or facial trauma within 3 months, suspected aortic dissection, prior intracranial hemorrhage, ischemic stroke within 3 months.
What are the relative contraindications to thrombolytic therapy?
Recent major surgery (<3 weeks); traumatic or prolonged cardiopulmonary resuscitation; recent internal bleeding; active peptic ulcer. Severe, poorly controlled HTN. Ischemic stroke.
What are the indications for antiplatelet agents in STEMI?
Aspirin (300 mg) should be given to all STEMI; 75 mg/d long term. Clopidogrel (300-600 mg loading) in addition to aspirin if undergoing PCI. If fibrinolytic therapy, clopidogrel given in addition to ASA. If CABG planned, clopidogrel withheld.
What are the indications for antithrombin in STEMI?
With PCI: Antithrombin therapy should be used in conjunction with PCI. Enoxaparin in acute STEMI in conjunction with PCI is effective.
What are the indications for enoxaparin with fibrinolysis?
Enoxaparin may be used in conjunction with fibrin-specific fibrinolytic agents in patients under the age of 75 years if there is no significant renal dysfunction.
What are the indications for glycoprotein IIb/IIIa inhibitors in STEMI?
Use abciximab with primary PCI in patients with STEMI.
What are the recommended discharge medications after ACS?
Aspirin, clopidogrel, beta-blocker, statin, nitrates.
What discharge antiplatelet therapy should be given to patients after ACS?
Aspirin: All patients should take 75-325 mg daily unless contraindicated. Clopidogrel should be prescribed for up to 9-12 months after ACS, particularly after stent placement.
What are the indications for a beta-blocker after ACS?
Beta-blocker should be prescribed for all patients after an ACS unless contraindicated, and continued indefinitely. Metoprolol and carvedilol should be used in patients after ACS who have heart failure.
What are the indications for ACE inhibitors after ACS?
ACE inhibitors should be given in patients with ACS in congestive heart failure, left ventricular dysfunction (ejection fraction 40%).
What are the indications for statin therapy after ACS?
Statin therapy should be initiated in the hospital in all patients with ACS.
What are the indications for nitrates after ACS?
All patients should be prescribed short-acting nitrates. Long-acting nitrates (isosorbide) are given if there is continuous and frequent chest pain.
What are the indications for warfarin after ACS?
Warfarin is recommended after ACS only for those at high risk of systemic thromboembolism because of atrial fibrillation, mural thrombus, or congestive heart failure.
What are the recommendations for exercise ECG testing after ACS?
Submaximal testing 4-7 days after infarction. A maximal test can be performed at 3-6 weeks postinfarction. Myocardial perfusion imaging can be performed before hospital discharge to assess residual ischemia.
What are the complications of ACS?
Pump dysfunction, ischemia, pericarditis (Dressler syndrome), thromboembolism, sudden cardiac death due to arrhythmia (most commonly ventricular fibrillation). Ventricular tachycardia.
What is right ventricular infarction?
Accompanies 30% of inferior MIs. It is diagnosed with right side leads and treated with fluids.
What is Prinzmetal angina?
Uncommon condition: episodes of angina are triggered when a coronary artery goes into spasm with ST-segment elevation. May cause acute MI, ventricular arrhythmias, sudden death.
What is the treatment of Prinzmetal angina?
Calcium channel blockers or nitrates.
81-year-old with HTN, dyslipidemia dyspnea, edema. Jugular venous distention 8 cm, S3 gallop, apical impulse displaced. Cardiomegaly. Dilated left ventricle with an EJECTION FRACTION 30%. Diagnosis?
Congestive heart failure
What is systolic heart failure?
Systolic dysfunction is caused by a loss of contractile strength of the myocardium with ventricular dilatation. Ischemic cardiomyopathy and dilated cardiomyopathy.
What is diastolic heart failure?
Filling of one or both ventricles is impaired while the emptying capacity is normal (the ejection fraction is normal). Infiltrative cardiomyopathies (amyloidosis).
What are the causes of decompensated heart failure?
Exacerbation of HF can be caused by nonadherence to medication, increase in dietary salt, acute ischemia, tachycardia or pulmonary infection, arrhythmias, hypertension, thyrotoxicosis, and anemia.
What are the causes of congestive heart failure?
Ischemic heart disease causes 70%. Other:: hypertensive heart disease, cardiomyopathies (idiopathic, alcohol), valvular, congenital. With exacerbation of HF, always check troponin to exclude myocardial infarction.
What are the symptoms of left ventricular heart failure?
Dyspnea, orthopnea, paroxysmal nocturnal dyspnea, fatigue, and weakness.
What is the New York Heart Association Functional Classification of heart failure?
Class I: no limitation. Class II: slight, mild limitation of activity. Class III: marked limitation. Class IV: confined to bed or chair.
What is the best test to confirm the diagnosis of heart failure?
Echocardiography is the test-of-choice for diagnosis and to classify the type (systolic vs. diastolic). Echocardiography measures ejection fraction.
What are the chest x-ray findings in congestive heart failure?
The chest x-ray may show cardiomegaly, vascular redistribution, Kerley B-lines, and interstitial edema.
What are the ECG findings in congestive heart failure?
Electrocardiogram identifies ventricular hypertrophy, ischemic heart disease, arrhythmias or conduction delays which may cause HF.
What is B-type natriuretic peptide?
BNP is a polypeptide secreted by the heart in response to excessive stretching of the myocytes. The BNP is elevated (97% sensitivity) in decompensated HF.
What are the beneficial effects of ACE inhibitors in congestive heart failure?
ACE reduce preload and afterload through vasodilation, reducing right atrial, pulmonary arterial, pulmonary capillary wedge pressures. Angiotensin receptor blockers if unable to tolerate ACEI (cough).
What are the side effects of captopril?
Rash, nonproductive cough, proteinuria, renal failure, taste disturbance, agranulocytosis, hypotension
What are the side effects of enalapril?
Hypotension, angioedema, renal failure, laryngeal edema, cough.
What are the side effects of lisinopril?
Hypotension, proteinuria, renal insufficiency, rash, cough.
What are the side effects of nitroprusside?
Thiocyanate toxicity, methemoglobinemia.
What are the side effects of nitroglycerin?
Headache, postural hypotension, methemoglobinemia.
What are the side effects of isosorbide dinitrate?
Headache, postural hypotension
What are the side effects of hydralazine?
Headache, positive ANA, SLE-like syndrome (10-20%) drug fever, rash.
What is the treatment of congestive symptoms in heart failure?
Loop diuretics. Furosemide is most commonly used. Spironolactone and eplerenone (aldosterone antagonists) are used as add-on therapy to ACE inhibitors in severe heart failure.
What is the mechanism of action of thiazides?
Inhibits NaCl cotransport; used mostly for treatment of hypertension; hydrochlorothiazide, chlorothiazide
What are the side effects of thiazides?
HypoNa, hypoK, hyperlipidemia, hyperCa, metabolic alkalosis, hyperuricemia, reduced GFR, allergy, thrombocytopenia, agranulocytosis, leukopenia, anemia, pancreatitis, sexual dys, glucose intolerance.
What are the side effects of indapamide?
Hypokalemia and lipid abnormalities are less common than with other thiazides. Hyponatremia, hypercalcemia, metabolic alkalosis, hyperuricemia, reduced glomerular filtration rate, allergy.
What are the side effects of phthalimidine derivatives?
Similar to thiazides, but hypokalemia may be profound. Chlorthalidone, metolazone.
What is the mechanism of action of loop diuretics?
Inhibitors of Na/K/Cl cotransport; most commonly used diuretics in heart failure; furosemide, ethacrynic acid, bumetanide.
What are the side effects of loop diuretics?
Hyponatremia, hypokalemia, hypocalcemia, metabolic alkalosis, hyperuricemia, interstitial nephritis, ototoxicity, thrombocytopenia, agranulocytosis, leukopenia, anemia.
What are the side effects of potassium-sparing diuretics?
Hyperkalemia, mental confusion, nausea, and gynecomastia (spironolactone only).
What is the initial therapy for congestive heart failure?
ACE inhibitor and furosemide is considered first line for all HF. Once stable, add on carvedilol or metoprolol. Chronic adrenergic activation is associated with HF.
What treatment is recommended for black patients with severe heart failure who have already been treated with the standard therapy?
Reduction in death and a decrease in hospitalization occurs with the combination of hydralazine and isosorbide.
What is the treatment of severe HF if there is no improvement of the EJECTION FRACTION and/or symptoms on standard therapy (diuretic, ACE inhibitor and beta blocker)?
Addition of spironolactone significantly reduces death and hospitalizations.
What is the treatment of severe refractory heart failure?
Inotropic agents for severe heart failure improves symptoms, quality of life and reduces hospitalizations, but does not improve survival.
What is the mechanism of digitalis?
Inhibits Na+/K+ - ATPase pump which results in increased intracellular Na+ and decreased intracellular Ca. Increase in intracellular Ca results in improved cardiac contractility.
What is the effect of potassium levels on the effect of digitalis?
Hyperkalemia will decrease digitalis activity, whereas hypokalemia results in toxicity.
What conditions predispose to digitalis toxicity?
Renal insufficiency, hypokalemia, hypercalcemia, hypomagnesemia, advanced age, sinoatrial and atrioventricular block; hypothyroidism, hypoxic states, pulmonary disease; multiple drug therapy.
What drug interaction occurs with digoxin and quinidine?
Decreases renal clearance of digoxin.
What drug interaction occurs with digoxin and verapamil/diltiazem?
Decreases renal clearance of digoxin.
What drug interaction occurs with digoxin and cholestyramine/colestipol?
Binds digoxin in gastrointestinal tract; interferes with enterohepatic circulation.
What drug interaction occurs with digoxin and spironolactone?
Inhibits tubular secretion of digoxin.
What drug interaction occurs with digoxin and thiazides/furosemide?
Diuretic-induced hypokalemia and/or bumetanide. Hypomagnesemia potentiates digitalis action.
What are the side effects of digoxin?
Nausea, vomiting, gynecomastia, blurred vision; yellow halo around objects; paroxysmal atrial tachycardia with block, PVCs, and bradycardia.
What is the treatment for digoxin toxicity?
Stop the drug. K+ administration if needed. Lidocaine and phenytoin. Digibind only for acute overdose.
What is the treatment of severe heart failure that does not respond to standard therapy?
Sympathomimetic amines (dopamine, dobutamine) and phosphodiesterase inhibitors (amrinone, milrinone) IV infusion. Other options: pacing, implantable defibrillator, heart transplantation.
What is the treatment of diastolic heart failure?
Diastolic HF may worsen when diuretics and vasodilators. Reducing the heart rate with beta blockers and calcium channel blockers (verapamil, diltiazem) to allow adequate diastolic filling.
What are the signs of pulmonary edema?
Increased respiratory rate, nocturnal dyspnea, cough with expectoration of pink frothy sputum, cyanosis, crackles, rhonchi, wheezing.
What are the chest x-ray findings in pulmonary edema?
Prominent pulmonary vasculature, enlarged cardiac silhouette, Kerley B lines, effusions.
What is the treatment of pulmonary edema?
Morphine sulfate. Furosemide reduces preload. Nitroglycerin reduces preload. Digoxin if atrial fibrillation. Intravenous ACE inhibitors.
What is mitral stenosis?
Thickened valve leaflets impede left ventricular filling; increased left atrial pressure causes pulmonary congestion; cardiac output reduced, secondary pulmonary vasoconstriction and RV failure occurs
What is the most common lesion caused by rheumatic fever?
Mitral stenosis is the most common lesion caused by rheumatic fever. Rarely congenital.
What are the early clinical signs of mitral stenosis?
Dyspnea, orthopnea, paroxysmal nocturnal dyspnea, fatigue, wasting, hemoptysis (rupture of pulmonary vessels); systemic embolism from stasis of blood in an enlarged left atrium
What are the late clinical signs of severe mitral stenosis?
Hoarseness due to impingement of an enlarged left atrium on recurrent laryngeal nerve; right-sided heart failure; hepatomegaly, ascites; peripheral edema.
What are the physical signs of mitral stenosis?
Atrial fibrillation (irregular rhythm); pulmonary rales; decreased pulse pressure; loud S1, opening snap following S2, diastolic rumble, sternal lift (right ventricular enlargement).
What are the ECG findings in mitral stenosis?
Right ventricular hypertrophy; left and right atrial enlargement; atrial fibrillation may occur.
What are the chest x-ray findings in mitral stenosis?
Large left atrium (double-density right heart border, straightening of the left heart border). Signs of pulmonary hypertension, including Kerley B lines and increased vascular markings. Large pulmonary artery.
What is the treatment of mitral stenosis?
Diuretics and salt-restricted diet; digitalis, anticoagulants if atrial fibrillation; when symptomatic: mitral commissurotomy or valve replacement.
What are the hemodynamic findings in mitral regurgitation?
LV stroke volume is pumped backward into LA, resulting in decreased CO. Volume overload, increased preload. LV failure causes dyspnea, orthopnea, paroxysmal nocturnal dyspnea. Pulmonary HTN.
What are the acute causes of mitral regurgitation?
Rupture chordae tendineae; papillary muscle rupture; endocarditis, trauma.
What are the chronic causes of mitral regurgitation?
Rheumatic heart disease, papillary muscle dysfunction, mitral prolapse/calcification, endocarditis; HCM; endocardial cushion defect, corrected transposition; endocardial fibroelastosis; LV dilatation.
What are the physical signs of mitral regurgitation?
Hyperdynamic and downward left ventricular impulse. Holosystolic apical murmur radiating to the axilla with a thrill (palpable turbulence). S3 with a soft S1 and widely split S2. Distended neck veins.
What are the ECG signs of mitral regurgitation?
Left ventricular hypertrophy and left atrial enlargement. Chest x-ray: cardiac enlargement vascular congestion.
What is the treatment of mitral regurgitation?
Digitalis, diuretics, arteriolar vasodilators (ACE inhibitors), warfarin; mitral valve replacement when symptoms.
What is mitral valve prolapse?
Most common congenital valvular abnormality; also in young women with connective tissue disease (e.g., Marfan or idiopathic).
What is the presentation of mitral valve prolapse?
Asymptomatic. Lightheadedness, palpitations, syncope, chest pain. Arrhythmias. Mid-to-late systolic click, late systolic murmur at apex which worsens with Valsalva; improves with squatting.
What are the complications of mitral valve prolapse?
Serious arrhythmias, sudden death, congestive heart failure, bacterial endocarditis, calcifications, transient cerebral ischemic attacks, mitral regurgitation.
What is the treatment of mitral valve prolapse?
Endocarditis prophylaxis only in severe cases. Beta-blocker for chest pain. Mitral valve replacement is rarely necessary.
What is aortic stenosis?
Calcification and degeneration; more common in elderly. Calcification and fibrosis of a congenitally bicuspid aortic valve. Rheumatic valvular disease.
What is the pathophysiology of aortic stenosis?
Elevation LV systolic pressure and LV hypertrophy. Noncompliant left ventricle causes a prominent S4 gallop. LV diastolic pressure increases; pulmonary congestion.
What are the clinical signs of aortic stenosis?
Angina, syncope, dyspnea. Pulsus tardus et parvus (carotid pulse weak, late), carotid thrill. Systolic ejection murmur aortic area; harsh, S4 gallop; A2 decreased, S2 single or paradoxically split; aortic ejection click.
What are the ECG signs of aortic stenosis?
Left ventricular hypertrophy. Chest x-ray: calcification, cardiomegaly, pulmonary congestion.
What is the treatment of aortic stenosis?
Endocarditis prophylaxis is not recommended. Valve replacement when the valve area is reduced below 0.8 cm or symptoms. Balloon valvuloplasty if too ill to tolerate surgery.
What is aortic valve sclerosis of the elderly?
Systolic murmur may mimic aortic stenosis. Systolic murmur ends early and does not peak late. Carotids do not have delayed upstrokes. No left ventricular hypertrophy by ECG. There is no stenosis on echo.
What are the clinical signs of hypertrophic obstructive cardiomyopathy?
Brisk bifid carotid. Systolic crescendo-decrescendo-murmur at left sternal border, which does not radiate to neck. Murmur increases with Valsalva (all other murmurs decrease). Pseudoinfarct (large septal Q).
What are the clinical sings of mitral regurgitation?
Murmur is holosystolic and radiates to axilla and not carotids. Carotid upstroke is normal; dilated left ventricle.
What are the clinical signs of pulmonic stenosis?
Murmur does not radiate into neck; loudest along the left sternal border; increases with inspiration.
What are the causes of aortic regurgitation?
Hypertension is the most common cause; infectious endocarditis, syphilis, ankylosing spondylitis; Marfan syndrome, rheumatic fever, aortic dissection, aortic trauma.
What is the pathophysiology of aortic regurgitation?
Aortic regurgitation results in a volume overload of the left ventricle.
What are the clinical signs of aortic regurgitation?
Dyspnea is most common. Diastolic decrescendo murmur. Systolic flow murmur. S3 in left ventricular failure, Austin-Flint murmur: low pitched, late diastolic apex murmur
How is aortic regurgitation diagnosed?
ECG: LV hypertrophy; chest x-ray: LV and aortic dilation; echocardiogram
What is the treatment of aortic regurgitation?
Endocarditis prophylaxis is not recommended. Salt restriction, diuretics, ACE inhibitors. Aortic valve replacement when symptoms worsen.
What is hypertrophic obstructive cardiomyopathy?
Marked hypertrophy of left ventricle; usually disproportionate hypertrophy of septum
What is restrictive cardiomyopathy?
Reduced ventricular compliance; usually caused by infiltration of myocardium (by amyloid, hemosiderin, or glycogen deposits).
What is obliterative cardiomyopathy?
Thickened endocardium or mural thrombi, or both, act as space-occupying lesions and reduce left ventricular filling.
What is dilated cardiomyopathy?
Characterized by diminished myocardial contractility, usually involving both ventricles; most common cause for heart transplants.
What are the causes of dilated cardiomyopathy?
Idiopathic, alcoholic, peripartum; viruses, parasites, mycobacteria, Rickettsiae; toxins (cobalt, lead, arsenic); doxorubicin cyclophosphamide, vincristine. Hypophosphatemia, hypoK, hypoCa, uremia.
What are the clinical signs of dilated cardiomyopathy?
Symptoms of left and right ventricular failure: dyspnea, orthopnea; peripheral edema.
What are the x-ray signs of dilated cardiomyopathy?
Cardiomegaly with pulmonary congestion. ECG: sinus tachycardia, arrhythmias, conduction disturbances.
What is the cause of hypertrophic obstructive cardiomyopathy?
Hypertrophic cardiomyopathy can develop sporadically; it is hereditary in 60%; autosomal dominant.
What is the pathophysiology of hypertrophic obstructive cardiomyopathy?
Thickening of interventricular septum. LV compliance reduced. Heart is hypercontractile. EJECTION FRACTION 80% (normal is 60%). Obstruction worsened by increased contractility.
What is the effect of digitalis glycosides on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of increased contractility.
What is the effect of beta-adrenergic stimulation (eg, isoproterenol, epinephrine) on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of increased contractility.
What is the effect of premature beats on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of increased contractility.
What is the effect of Valsalva maneuver on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of decreased preload.
What is the effect of decrease in intravascular volume (eg, hemorrhage, diuresis, gastrointestinal losses) on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of decreased preload.
What is the effect of standing on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of decreased preload.
What is the effect of nitroglycerin on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of decreased preload.
What is the effect of vasodilator drugs on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of decreased preload.
What is the effect of tachycardia on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of decreased preload and increased contractility.
What is the effect of hypovolemia on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of decreased afterload.
What is the effect of nitroglycerin and related drugs on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of decreased afterload.
What is the effect of vasodilator drugs on hypertrophic obstructive cardiomyopathy?
Increase obstruction because of decreased afterload.
What is the effect of beta-adrenergic blockade (eg, propranolol) on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of decreased contractility.
What is the effect of heavy sedation and general anesthesia on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of decreased contractility.
What is the effect of calcium channel blockers, disopyramide, and other drugs that depress myocardial function on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of decreased contractility.
What is the effect of intravascular volume expansion on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of increased preload.
What is the effect of squatting on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of increased preload.
What is the effect of bradycardia on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of increased preload.
What is the effect of beta-adrenergic blockade on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of increased preload.
What is the effect of intravascular volume expansion on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of increased afterload.
What is the effect of squatting on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of increased afterload.
What is the effect of alpha-adrenergic stimulation (e.g., phenylephrine, mephentermine) on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of increased afterload.
What is the effect of handgrip exercise on hypertrophic obstructive cardiomyopathy?
Decreased obstruction because of increased afterload.
What are the symptoms of hypertrophic obstructive cardiomyopathy?
Dyspnea, angina, presyncope, syncope, and palpitations.
What are the physical signs of hypertrophic obstructive cardiomyopathy?
Large jugular A wave, bifid carotid pulse, palpable S4 gallop, systolic murmur and thrill, mitral regurgitation murmur; sudden death.
What are the ECG signs of hypertrophic obstructive cardiomyopathy?
Left ventricular hypertrophy, pseudo Q waves (V1-V3), ventricular arrhythmias.
What is the treatment of hypertrophic obstructive cardiomyopathy?
Beta-blockers, calcium channel blockers, surgery.
What is restrictive cardiomyopathy?
Least common cause of cardiomyopathy; rigid noncompliant ventricular walls.
What are the causes of restrictive cardiomyopathy?
Sarcoidosis; amyloidosis, hemochromatosis, neoplasia, scleroderma, radiation
What is the pathophysiology of restrictive cardiomyopathy?
Myocardium is rigid and noncompliant, impeding ventricular filling and raising cardiac filling pressures. Systolic performance reduced.
What are the signs of restrictive cardiomyopathy?
Dyspnea, exercise intolerance, weakness. Elevated jugular venous pressure, edema, hepatomegaly, ascites, S4 and S3 gallop, Kussmaul sign.
What are the x-ray signs of restrictive cardiomyopathy?
Mild cardiomegaly, pulmonary congestion. Echocardiogram is diagnostic.
What is the treatment of restrictive cardiomyopathy?
Heart transplantation; death from congestive heart failure or arrhythmias.
What are the causes of acute pericarditis and myopericarditis?
Idiopathic, infections (viral), vasculitis (connective tissue disease), disorders of metabolism, neoplasms, trauma, drug reactions.
What are the clinical symptoms of acute pericarditis?
Chest pain at left of sternum, worsened by lying down, coughing, and deep inspiration, relieved by sitting up and leaning forward.
What are the physical signs of acute pericarditis?
Scratchy, high-pitched rub that has 1-3 components. The rub is best heard with the diaphragm of the stethoscope as the patient sits forward at forced-end expiration.
What are the ECG signs of acute pericarditis?
Diffuse ST-segment elevation with upright T waves. The diffuseness of the ST-segment elevation, and absence of Q waves distinguish pericarditis from acute MI.
What is the treatment of acute pericarditis?
NSAIDs, aspirin, corticosteroids.
What are the types of pericardial effusions?
Transudates, serosanguineous, blood.
What are the causes of transudative pericardial effusions?
Transudates occur with congestive heart failure, overhydration, or hypoproteinemia. Exudates occur in the presence of pericardial injury.
What are the causes of serosanguineous pericardial effusions?
Serosanguineous pericardial fluid is sign in tuberculosis and neoplastic diseases.
What are the causes of bloody pericardial effusions?
Blood in the pericardial space may occur in aortic aneurysm or aortic dissection, trauma, rupture of the heart in acute MI, and bleeding caused by coagulation defects.
How is pericardial effusion diagnosed?
Echocardiography. Chest x-ray may show a "water-bottle" cardiac silhouette.
What is the treatment of pericardial effusion?
Pericardial fluid aspiration.
What is cardiac tamponade?
A life-threatening condition in which a pericardial effusion compresses the heart.
What are the causes of cardiac tamponade?
Neoplasia, idiopathic (viral) pericarditis, tuberculosis, suppurative, intrapericardial hemorrhage, central line extravasation, surgery, postpericardiotomy syndrome, uremia, radiation therapy, vasculitis.
What are the clinical manifestations of pericardial tamponade?
Dyspnea, fatigue, orthopnea. Pulsus paradoxus (decrease in systolic blood more than 10 with inspiration or disappearance pulse with inspiration).
What is the triad of pericardial tamponade?
Triad of neck vein distension with clear lung, hypotension, and muffled heart sounds.
What is the treatment of cardiac tamponade?
Pericardiocentesis; subxiphoid surgical drainage.
What is constrictive pericarditis?
Diffuse thickening of the pericardium caused by inflammation; reduced distensibility of the cardiac chambers. Cardiac output is limited and filling pressures are increased; abnormal diastolic filling.
What are the causes of constrictive pericarditis?
Idiopathic; following open-heart surgery; thoracic radiation; postviral infection.
What are the clinical manifestations of constrictive pericarditis?
Dyspnea on exertion, orthopnea; ascites, edema, jaundice, hepatic tenderness, and hepatomegaly (right-side failure). Jugular venous distension that increases with inspiration (Kussmaul sign).
How is constrictive pericarditis diagnosed?
CT: thickened pericardium; pericardial calcifications in tuberculous constriction. Left ventricular ejection fraction is decreased in restrictive cardiomyopathy, but not in constrictive pericarditis.
What is the treatment of constrictive pericarditis?
Sodium restriction, diuretics, pericardiectomy.
What is sinus bradycardia?
Ventricular complexes are normal width, evenly spaced, rate is <60/min.
What are the causes of sinus bradycardia?
Acute MI, carotid sinus pressure, vomiting, Valsalva maneuver, phenothiazines, digitalis glycosides, beta-adrenergic blocking agents, calcium blockers, marathoners and swimming, hypothyroidism, normal variant.
What is the treatment of sinus bradycardia?
Atropine if symptoms. Pacemaker.
What are the types of atrioventricular block?
The three classic clinical types are first-, second-, and third-degree (or complete) atrioventricular block.
What is first-degree atrioventricular block?
PR interval greater than 0.20 s at a heart rate of 70 beats/min.
What are the causes of first-degree heart block?
Digitalis; exaggerated vagal tone after tracheal suctioning; ischemia, inflammation (myocarditis, acute rheumatic fever), cardiomyopathies.
What are the types of second-degree atrioventricular block?
Type I (Mobitz I, Wenckebach) and type II (Mobitz II).
What is type I atrioventricular block (Mobitz I, Wenckebach)?
There is progressive prolongation of the PR interval until a P wave is blocked and a ventricular beat is dropped. The PR interval of the next conducted beat is shorter than the preceding PR interval.
What is type II (Mobitz II) atrioventricular block?
An intermittent blocked beat occurs suddenly and is not preceded by a change in the duration of the PR interval.
What is the site of the block in type I second-degree atrioventricular block?
Usually AV nodal (supra-Hisian)
What is the site of the block in type II second degree atrioventricular block?
Infranodal (infra- or infra-Hisian).
What are the causes of type I second degree atrioventricular block?
Degenerative changes in AV node; diaphragmatic myocardial infarct; digitalis toxicity; myocarditis; rheumatic fever; increased vagal tone.
What are the causes of type II second degree AV block?
Extensive anterior myocardial infarct; degenerative changes in His-Purkinje system; calcification of mitral or aortic valve annulus.
What are the ECG findings in type I second degree AV block?
PR interval lengthens progressively until ventricular beat dropped. PR shortens after dropped beat. RR narrows progressively up to the dropped beat. RR after blocked beat is < twice the RR interval of conducted beats.
What are the ECG findings in type II second degree AV block?
PR interval is normal duration and constant length; blocked beats occur suddenly without progressive lengthening of PR. RR interval of conducted beats is constant or a multiple of a basic RR interval.
What is third-degree atrioventricular block?
In 3rd degree block, all atrial beats are blocked, and ventricles are driven by an escape focus.
What are the most common causes of 3rd degree atrioventricular block?
Most common is fibrous degeneration of conduction system from aging; inferior/ posterior infarction; infection, inflammation, digitalis, ankylosing spondylitis, Adams-Stokes, heart failure.
What are Adams-Stoke attacks?
Sudden asystole or transient ventricular tachycardia or ventricular fibrillation.
What is the treatment of third degree AV block?
Pacing.
What is sinus tachycardia?
Rhythm with a rate >100. Ventricular complexes are normal width, evenly spaced, normal P precedes a QRS. Caused by fever, hypotension, volume depletion, anxiety, pain. Thyrotoxicosis, anemia, beta-agonists.
What is paroxysmal supraventricular tachycardia?
Group of ectopic tachyarrhythmias that are characterized by sudden onset and abrupt termination. Includes paroxysmal atrial tachycardia. 80% are caused by re-entry, mainly in the AV node.
What are the ECG signs of paroxysmal supraventricular tachycardia?
Paroxysmal supraventricular tachycardia has an absolutely regular rhythm at a rate between 130 and 220 beats/min. P-wave is abnormal.
What is the treatment of paroxysmal supraventricular tachycardia?
Right side carotid sinus massage to increase vagal tone; adenosine is effective in more than 90%. IV propranolol, esmolol, digitalis; synchronized external cardioversion if patient is unstable.
What are the ECG signs of multifocal atrial tachycardia?
Irregular supraventricular rhythm rate 100-200. At least 3 different P wave morphologies; PR interval varies. Each QRS complex is preceded by a P wave. Occurs in elderly patients or chronic lung disease.
What are the causes of atrial flutter?
Chronic obstructive lung disease, pulmonary embolism, thyrotoxicosis, mitral valve disease, alcohol.
What are the ECG signs of atrial flutter?
Atrial flutter has an absolutely regular rhythm with a ventricular rate of 125-150 beats/min and an atrial rate of 250-300 beats/min (ie, 2:1 block).
What is the initial treatment of atrial flutter?
Cardioversion if hemodynamically unstable (e.g., hypotension); digitalis, verapamil, diltiazem, beta-blockers.
What is the most common sustained cardiac rhythm disturbance?
Atrial fibrillation, which is often associated with heart disease but occurs commonly in patients with no disease. AF may cause thromboembolic events and hemodynamic impairment.
What are the ECG signs of atrial fibrillation?
No p waves. Fibrillatory waves with an irregular, rapid ventricular response (irregularly, irregular).
What is the sign of Wolf-Parkinson White syndrome with atrial fibrillation?
Extremely rapid rates (over 200 bpm) suggest the presence of an accessory pathway (W-P-W syndrome), which may manifest as AF. There is a delta wave.
What is the rate of ischemic stroke in atrial fibrillation?
The rate of ischemic stroke among patients with nonrheumatic atrial fibrillation averages 5% per year.
What are the signs of atrial fibrillation?
Elderly patients with shortness of breath, dizziness, or palpitations. Acute fatigue or exacerbation of congestive heart failure. Irregularly irregular pulse.
What is "lone atrial fibrillation"?
Atrial fibrillation with no evidence of structural heart disease.
What are the causes of atrial fibrillation?
Rheumatic mitral disease, CAD, CHF, HTN (atrial dilation). Hyperthyroidism, hypoxemia, alcohol. In younger patients, 30-45% of paroxysmal cases and 20-25% of persistent cases of AF occur as lone AF.
What tests are use in the evaluation of atrial fibrillation?
ECG: AF, left ventricular hypertrophy, pre-excitation, prior myocardial infarction. Echocardiogram: identifies LVH, valvular disease, atrial size, and left atrial thrombus. Thyroid function tests.
What drugs are effective for pharmacologic cardioversion of atrial fibrillation?
Amiodarone, dofetilide, flecainide, ibutilide, propafenone, and quinidine.
Which drugs are used to maintain sinus rhythm in patients with atrial fibrillation?
Drugs used to maintain sinus rhythm in patients with atrial fibrillation include amiodarone, disopyramide, dofetilide, flecainide, propafenone, and sotalol.
What is the first step in managing atrial fibrillation?
Control rate to <100 with beta-blockers, calcium-blockers, or digoxin. These agents do not convert AF to sinus rhythm. Digoxin is the drug of choice in HF. Diltiazem, atenolol, metoprolol, verapamil.
What is the recommended treatment strategy for atrial fibrillation?
Rate control with chronic anticoagulation for the majority of patients with chronic AR. Rhythm control has not been shown to be superior to rate control in reducing morbidity and mortality.
A 40-year-old man with shortness of breath. Blood pressure is 70/50 mm Hg. Jugular vein distention and bilateral crackles on exam. ECG atrial fibrillation waves. What is the next appropriate step in management?
Electrical cardioversion.
What is Wolff-Parkinson-White syndrome?
Ventricle is activated by atrial impulses early by accessory pathway (Kent bundle). Short PR interval followed by a wide QRS complex with a slurred initial deflection (delta), which represents early ventricular activation.
What is the treatment of Wolff-Parkinson-White syndrome?
If unstable, then synchronized cardioversion. If stable, then procainamide. Ablation.
What drugs should be avoided in Wolff-Parkinson-White syndrome?
Avoid digoxin, beta-blockers, calcium-blockers because inhibit conduction in the normal conduction pathway and cause ventricular or supraventricular tachycardia via in the aberrant conduction pathway.
What is ventricular tachycardia?
VT is three or more consecutive beats of ventricular origin at a rate greater than 120 beats/min. QRS complexes are wide and bizarre.
What are the causes of ventricular tachycardia?
Ischemic heart disease after an acute MI; cardiomyopathies, hypokalemia, hypercalcemia, hypomagnesemia, hypoxia, digitalis toxicity and thioridazine drugs.
What are the clinical manifestations of ventricular tachycardia?
Hypotension, congestive heart failure, syncope, or cardiac arrest.
What are the ECG signs of torsade de pointes ventricular tachycardia?
Undulating rotations of QRS complexes around baseline. Initiated by a PVC in setting of abnormal ventricular repolarization, characterized by prolongation of QT interval. Episodes of dizziness or syncope.
What are the causes of torsade de pointes ventricular tachycardia?
Antiarrhythmics that prolong ventricular repolarization (prolong QT); quinidine, procainamide, disopyramide, phenothiazines, thioridazine, tricyclics, lithium, hypoK hypoMg, intracerebral hemorrhage.
What is the treatment of torsade de pointes ventricular tachycardia?
Treat underlying electrolyte disorders. Magnesium and potassium are first-line therapies. Isoproterenol and short-acting beta-blockers. Overdrive pacing. If unstable, perform electrical cardioversion.
What are the indications for amiodarone?
Amiodarone is a very effective antiarrhythmic drug, used in ventricular tachycardia, AF, and atrial flutter. Amiodarone has a very long half-life (over 50 days).
What are the side effects of amiodarone?
Cough, fever; painful breathing can be fatal. 20% neurotoxicity with imbalance, tremor, numbness, weakness. Hypothyroidism/ hyperthyroidism; blue-gray skin; halos, blurred vision; corneal deposits.
What are the indications for disopyramide?
Ventricular ectopic activity and atrial ectopic activity.
What are the adverse effects of disopyramide?
Anticholinergic effects; hypotension, heart failure, heart block, tachyarrhythmia.
What are the indications for lidocaine?
Ventricular ectopic activity
What are the adverse effects of lidocaine?
Drowsiness, agitation, seizures; rarely congestive heart failure or heart block.
What are the cardiac indications for phenytoin?
Ventricular ectopic activity.
What are the adverse effects of phenytoin?
Ataxia, nystagmus, drowsiness; hypotension and heart block with rapid IV injection
What are the indications for procainamide?
Ventricular ectopic activity, atrial ectopic activity
What are the adverse effects of procainamide?
Lupus-like syndrome; nausea/vomiting; insomnia; rash; hypotension; aggravation of arrhythmia; blood, dyscrasias
What are the indications for quinidine?
Ventricular ectopic activity, atrial ectopic activity
What are the adverse effects of quinidine?
Aggravation of arrhythmias; thrombocytopenia; fever, rash; cinchonism; nausea; digoxin-quinidine interaction (elevation of digoxin levels).
What are the antiarrhythmic indications for beta-adrenergic blocking agents?
Atrial ectopic activity, ventricular ectopic activity.
What are the adverse effects of beta-blockers?
Heart block, hypotension, heart failure, asthma, hypoglycemia, lethargy, impotence.
What are the antiarrhythmic indications for verapamil?
Atrial ectopic activity.
What are the adverse effects of verapamil?
Congestive heart failure, asystole, constipation
What are the indications for adenosine?
Ventricular tachycardia
What are the adverse effects of adenosine?
Transient dyspnea, noncardiac chest pain, rarely hypotension.
What are the indications for mexiletine?
Ventricular ectopic activity
What is mexiletine?
Lidocaine-like drug. Local anesthetic; half-life, 8-14 h.
What are the indications for tocainide?
Ventricular ectopic activity
What is tocainide?
Lidocaine-like drug. Half-life 14-16 h in patients with high-grade ventricular ectopic activity.
What are the indications for amiodarone?
Atrial ectopic activity (especially WPW), ventricular ectopic activity.
What are the adverse effects of amiodarone?
Very long half-life (20-40 d). May increase digoxin level. May worsen existing cardiac conduction disturbances. May prolong Coumadin effect.
What are the indications for encainide?
Ventricular tachycardia, Wolff-Parkinson-White syndrome.
What are the adverse effects of encainide?
Negative inotropism; QRS and PR prolongation. Hepatic metabolism to active metabolites.
What are the indications for flecainide?
Ventricular tachycardia, supraventricular tachycardia, Wolff-Parkinson-White syndrome.
What are the adverse effects of flecainide?
Negative inotropism, QRS, PR prolongation
What are the indications for propafenone?
Ventricular tachycardia, atrial fibrillation
What the adverse effect of propafenone?
Negative inotropism
What are the physiologic effects of low doses of nitrates?
In low doses, nitrates increase venous dilation and reduce preload.
What are the physiologic effects of medium doses of nitrates?
In medium doses, nitrates increase arteriolar dilatation and decrease afterload and preload.
What are the physiologic effects of high doses of nitrates?
In high doses, nitrates increase coronary artery dilatation and increase oxygen supply.
What are the side effects of nitrates?
Vasodilation can lead to orthostatic hypotension, reflex tachycardia, throbbing headache, and blushing.
What is the contraindication to nitrates?
Contraindicated if systolic blood pressure is <90 mm Hg.
What is the dosing pattern for nitrates?
Requires a window-free period of more than 8 hours with nitrate therapy to reduce tachyphylaxis.
What are the beneficial effects of beta-blockers?
Beta-blockers have been shown to improve survival after an acute myocardial infarction and in CHF.
What are the adverse effects of beta-blockers?
Fatigue, mental depression, insomnia, adverse effects on lipid panel; hallucinations, Raynaud phenomenon, bronchoconstriction, mask signs and symptoms of insulin-induced hypoglycemia, sexual dysfunction.
What are the adverse effects of nonselective beta-blockers?
Nonselective beta-blockers may mask hypoglycemic symptoms in insulin-dependent diabetics.
What is the contraindication to beta-blockers?
Contraindicated in severe asthma.
What are the physiologic effects of calcium-channel blockers?
Produces decreases in preload and afterload. May be harmful in postinfarction period, especially if left ventricular failure.
What are the adverse effects of calcium channel blockers?
Congestive heart failure, reflex tachycardia, hypotension, lightheadedness; flushing, headache, weakness, nausea, constipation, nasal congestion, wheezing, peripheral edema.
What is anemia?
A hematocrit less than 41% in men or less than 36% in women, or a hemoglobin less than 13.5 gm/dL in men or less than 12 gm/dL in women.
What is microcytic anemia?
Microcytic anemia is a low mean corpuscular volume (MCV) less than 80; caused by iron deficiency, thalassemia, sideroblastosis, and lead poisoning. Anemia of chronic disease can be either microcytic or normocytic.
What is macrocytic anemia?
Elevated MCV >100 caused by vitamin B12 or folic acid deficiency, alcohol, liver disease, methotrexate, zidovudine, phenytoin. Normal MCV in early micro/macrocytic anemia and hemolysis.
What is the presentation of anemia?
Fatigue, tiredness, poor exercise tolerance. Dyspnea on exertion and light-headedness, confusion. Anemia may cause death from myocardial ischemia. Older patient will develop dyspnea or angina at a Hct <30%.
What is the first step in diagnosing anemia?
First step is to determine the MCV. Iron studies, reticulocyte count, peripheral smear, red cell distribution of width (RDW), Coombs test, B12, folate levels, and occasionally a bone marrow biopsy.
What is the treatment for anemia?
Packed red blood cells are used to maintain a hematocrit >25-30%. A healthy young patient can have transfusion withheld until the Hct is in the low 20%s. Older patient needs Hct >30%.
How does one unit of packed red blood cells affect the hematocrit?
The hematocrit should rise three points for every unit of packed red blood cells given.
What is iron deficiency anemia?
Diminished RBC production and an MCV <80, hypochromic cells, low iron. Iron deficiency anemia is caused by blood loss, either GI or menstrual. Hematuria, malabsorption, hemolysis, poor intake.
What is the presentation of iron deficiency anemia?
Hct 30% causes fatigue, poor exercise tolerance. Hct 25% cause tachycardia, palpitations, dyspnea, pallor. Older and CAD dyspneic at 30%. Lightheadedness, confusion, syncope, chest pain, death.
What are the physical signs of iron deficiency?
Brittle nails, spoon shaped nails, glossitis, and pica.
How is iron deficiency anemia diagnosed?
Low serum ferritin <10 ng/mL. MCV is low. The serum iron is low and the total iron binding capacity is high. The RDW is elevated. The reticulocyte count is low.
What is anemia of chronic disease?
A defect in the ability to use iron; either microcytic or normocytic; caused by any chronic inflammatory, infectious, or neoplastic condition.
How is anemia of chronic disease diagnosed?
Serum ferritin level is normal or elevated. The serum iron level and total iron binding capacity (TIBC) are low. The reticulocyte count is low.
What is the treatment for anemia of chronic disease?
Correct underlying disease. Iron and erythropoietin are not effective, except in renal disease and anemia caused by chemotherapy or radiation therapy for malignancies.
What is sideroblastic anemia?
A microcytic anemia caused by a disorder in the synthesis of hemoglobin. Hereditary and acquired forms from chloramphenicol, isoniazid, or alcohol. Lead poisoning.
How is sideroblastic anemia diagnosed?
Ferritin is elevated. Transferrin saturation very high, and TIBC very low. Serum iron level is high. Prussian Blue stain of red blood cells in the marrow reveals ringed sideroblasts.
What is thalassemia?
Hereditary underproduction of either the alpha or beta globin chains of the hemoglobin molecule, resulting in a hypochromic, microcytic anemia.
What populations more commonly have thalassemia?
Alpha thalassemia is more common in Asian populations. Beta thalassemia is more common in Mediterranean populations.
What is the presentation of thalassemia?
Individuals with two genes deleted have a mild anemia with hematocrits ranging from 30-40% with a low MCV. In beta thalassemia there is a mild anemia with microcytosis (low MCV).
How is thalassemia diagnosed?
Thalassemia trait: mild anemia with microcytosis. Beta thalassemia major has severe symptoms, large spleen, bone abnormalities.
How is thalassemia diagnosed?
Microcytic anemia with normal iron studies, and hemoglobin electrophoresis. Beta thalassemia shows an increased level of hemoglobin F and hemoglobin A2.
What is the treatment for thalassemia?
Thalassemia traits do not require treatment. Beta thalassemia major patients require blood transfusions once or twice a month.
A 73-year-old alcoholic man memory loss and tingling in his feet. Hematocrit of 32% with an MCV of 110. What is the diagnosis?
Folic acid deficiency.
What are the causes of vitamin B12 deficiency?
Most common cause: pernicious anemia (hereditary, autoimmune, decreased intrinsic factor). Gastrectomy, atrophic gastritis, sprue, enteritis, blind loop. Pancreatic insufficiency, tapeworm. Decreased intake.
What is the presentation of vitamin B12 deficiency?
Peripheral neuropathy, decreased position and vibratory sense; psychiatric, autonomic, motor, cranial nerve, bowel, bladder, and sexual dysfunction. Glossitis, diarrhea, and abdominal pain.
How is vitamin B12 deficiency diagnosed?
Anemia with macrocytosis (increased MCV). Hypersegmented neutrophils. Indistinguishable from folate deficiency. Reticulocyte count is reduced. Pancytopenia. Most specific test is low B12 level.
How is pernicious anemia diagnosed?
Antibodies to intrinsic factor and parietal cells confirm the etiology as pernicious anemia. The Schilling test is not necessary if there is a low B12 level combined with the presence of antibodies to intrinsic factor.
What is the treatment for vitamin B12 deficiency?
Replace the vitamin B12 by the intramuscular route.
What is folic acid deficiency?
Deficiency in folic acid levels leading to anemia because of decreased dietary intake. Alcoholics have decreased folate intake.
How is folic acid deficiency diagnosed?
The hematologic presentation of folic acid deficiency is identical to B12 deficiency. Low red-blood-cell folic-acid level. Treatment is oral replacement of folic acid.
What are some chronic causes of hemolytic anemia?
Sickle cell disease, paroxysmal nocturnal hemoglobinuria, and hereditary spherocytosis
What are some acute causes of hemolytic anemia?
Drug-induced hemolysis, auto-immune hemolysis, or glucose 6-phosphate dehydrogenase deficiency.
What is the presentation of hemolytic anemia?
Fatigue and weakness. Dyspnea and confusion. Jaundice and dark urine. Fever, chills, chest pain, tachycardia, and backache may occur.
How is hemolytic anemia diagnosed?
Hemolytic anemias have a normal MCV. The reticulocyte count is elevated. LDH and indirect bilirubin are elevated. Peripheral smear shows fragmented cells. Hemoglobinuria.
What is the treatment of hemolytic anemia?
Transfusion when the hematocrit becomes low. Hydration prevents toxicity to the kidney tubule from free hemoglobin.
What is sickle cell disease?
A hereditary chronic hemolysis ranging from asymptomatic to severe crisis; irreversibly sickled cells and recurrent painful crises. Autosomal recessive.
What is the pathophysiology of sickle cell disease?
Hemoglobin S is due to a substitution of a valine for glutamic acid as the sixth amino acid of the beta globin chain.
What is the prevalence of sickle cell disease?
The heterozygous trait is present in 8% of the African-American population, and the homozygous form (disease) is present in 1 in 400 African Americans.
What conditions may precipitate a painful crisis?
Painful crisis can be precipitated by hypoxia, dehydration, acidosis, infection, and fever. Sudden drops in hematocrit may also be caused by Parvovirus B19 infection or folate deficiency.
What is the presentation of sickle cell disease?
Renal concentrating defects, ulcerations of legs, bilirubin gallstones, aseptic necrosis of femoral head, osteomyelitis, retinopathy, recurrent infections from Pneumococcus, Haemophilus, growth retardation, splenomegaly, autosplenectomy.
What is an acute painful crisis of sickle cell disease?
Back, rib, chest, leg pain. Acute chest syndrome is chest pain, fever, leukocytosis, hypoxia, and infiltrates. Stroke and TIA. Priapism, blindness, MI, cardiomyopathy. Spontaneous abortion, low birth weight.
How is sickle cell disease diagnosed?
Anemia with a normal MCV. Reticulocyte count 10-20%. LDH, bilirubin elevated. Hb electrophoresis most specific. Peripheral smear shows sickled cells. UA blood. WBC elevated.
What is the treatment of acute sickle cell pain crisis?
Fluids, analgesics, and oxygen. Antibiotics with infection, fever; leukocytosis. Ceftriaxone and cefotaxime are preferred because cover Pneumococcus and Haemophilus influenza.
What is the treatment of sickle cell acute chest syndrome, CNS manifestations, priapism, and acute cardiac ischemia?
PRBC if Hct low. Folic acid and vaccinations against Pneumococcus and influenza. Hydroxyurea to decrease frequency of pain crises. Bone marrow transplantation can be curative.
What are autoimmune, cold agglutinin, and drug-induced hemolytic anemias?
Various forms of acquired hemolytic anemias resulting from production of IgG, IgM, or activation of complement C3 against the red cell membrane. Often sudden and idiopathic.
What are the causes of autoimmune hemolytic anemias?
Antibodies in leukemia, viral infections, lymphoma, collagen vascular diseases or penicillins, cephalosporins, sulfa drugs, quinidine, alpha methyldopa, procainamide, rifampin, and thiazides.
What percentage of autoimmune hemolytic anemias are idiopathic?
50% do not have an underlying disorder.
What is cold agglutinin disease?
IgM against the red cell in association with lymphoma, Waldenstrom macroglobulinemia, Mycoplasma or mononucleosis.
Which inflammatory bowel disease is associated with autoimmune hemolytic anemia?
Ulcerative colitis.
What is the presentation of autoimmune hemolysis?
Sudden fever, syncope, congestive failure, hemoglobinuria. Splenomegaly; cold agglutinin disease results in cyanosis of the ears, nose, fingers, and toes. Weakness, pallor, jaundice, and dark urine.
How is autoimmune hemolysis diagnosed?
Normocytic anemia, reticulocytosis, increased LDH, increased indirect bilirubin. Coombs test is specific for autoimmune, cold agglutinin, drug-induced hemolysis. Spherocytes on smear.
What is the treatment for autoimmune hemolysis?
Steroids. Splenectomy for those unresponsive to steroids. Cold agglutinin disease is managed by avoiding cold.
What is hereditary spherocytosis?
Hemolysis with spherocytes, jaundice, splenomegaly. Autosomal dominant; loss of spectrin in RBC membrane results in spheres, which are not able to pass spleen. Anemia; splenomegaly jaundice; bilirubin stones.
How is hereditary spherocytosis diagnosed?
Decreased MCV anemia, elevated LDH, indirect bilirubin, reticulocytes. Hemolysis; negative Coombs. RBC sensitivity to lysis in osmotic fragility test. The mean corpuscular hemoglobin concentration is elevated.
What is the treatment for hereditary spherocytosis?
Folate replacement. Splenectomy.
What is paroxysmal nocturnal hemoglobinuria?
A red cell membrane defect leading to intermittent dark urine and venous thrombosis and a chronic hemolysis. A red cell membrane defect allows binding of complement to the red cell.
What is the presentation of paroxysmal nocturnal hemoglobinuria?
Dark urine from intravascular hemolysis. Thrombosis of the hepatic vein (Budd-Chiari syndrome). Hemoglobinuria in the first morning urine.
How is paroxysmal nocturnal hemoglobinuria diagnosed?
Increased LDH, bilirubin, and reticulocyte count, hemoglobinuria. Sugar-water test and the acidified-hemolysis (Ham) test. Decay accelerating factor is diminished in PNH.
What is the treatment for paroxysmal nocturnal hemoglobinuria?
Iron replacement. Corticosteroids.
What is glucose-6-phosphate dehydrogenase deficiency?
Hereditary deficiency of an enzyme for neutralizing oxidant stress to the red cell, resulting in acute hemolysis. Oxidant stress from infections, sulfa drugs, primaquine, dapsone, quinidine, and nitrofurantoin.
What is the presentation of glucose-6-phosphate dehydrogenase deficiency?
Sudden, severe, intravascular hemolysis; jaundice, dark urine, weakness, and tachycardia.
How is glucose-6-phosphate dehydrogenase deficiency diagnosed?
High LDH, bilirubin, and reticulocyte count with a normal MCV and hemoglobinuria. Heinz bodies are inclusions in red cells. Definitive test is the G6PD level.
What is the treatment for glucose-6-phosphate dehydrogenase deficiency?
Hydration and transfusion. Avoid oxidant stress.
What is aplastic anemia?
Failure of bone marrow, resulting in anemia, leukopenia, and thrombocytopenia (pancytopenia). The marrow is empty.
What are the causes of aplastic anemia?
Radiation, benzene, NSAIDs, chloramphenicol, alcohol, chemotherapeutics. Tb, lymphoma. Hepatitis, HIV, CMV, EBV, Parvovirus B19. The most common etiology is idiopathic.
What is the presentation of aplastic anemia?
Bleeding from thrombocytopenia. Fatigue from anemia and infections from neutropenia. Pancytopenia on CBC. Marrow biopsy confirms.
What is the treatment for aplastic anemia?
Bone marrow transplantation or immunosuppressive agents.
What is acute leukemia?
Derangement of pluripotent stem cell, resulting in the destruction of marrow. Leukemic blasts crowd out marrow cells, resulting in pancytopenia. Blood cells lose the ability to mature and function.
What are the causes of acute leukemia?
Most cases are idiopathic; radiation exposure, benzene, melphalan and etoposide, retroviruses.
What is the presentation of acute leukemia?
Fatigue from anemia. Bleeding from thrombocytopenia. Infection from the underproduction of white blood cells.
Which type of anemia is more common in children?
Acute lymphocytic leukemia (ALL) is more common in children. Acute myelogenous leukemia (AML) is more common in adults. Enlargement of the liver, spleen, and lymph nodes and bone pain are common at presentation.
How is acute leukemia diagnosed?
WBC can be low, normal, or elevated. Thrombocytopenia, anemia; leukemic blasts in peripheral blood. Marrow biopsy >20% blasts confirms.
What are the markers of acute myelogenous leukemia?
AML is characterized by Auer rods, myeloperoxidase, and esterase.
What are the markers of acute lymphocytic leukemia?
ALL is characterized by common ALL antigen and terminal deoxynucleotidyl transferase (TdT).
What is chronic myelogenous leukemia (CML)?
Massive overproduction of myeloid cells. Philadelphia chromosome is characteristic, producing tyrosine kinase.
What is the presentation of chronic myelogenous leukemia?
Markedly elevated white blood cell count fatigue, night sweats, and low-grade fever. Abdominal pain from massive enlargement of the spleen. Bone pain.
How is chronic myelogenous leukemia diagnosed?
Elevated WBC consisting of neutrophils. Leukocyte alkaline phosphatase score is diminished. Basophilia characteristic of CML and all myeloproliferative disorders such as polycythemia vera. Platelets elevated.
What is the treatment for chronic myelogenous leukemia?
Imatinib which is direct inhibitor of the tyrosine kinase produced by the Philadelphia chromosome. 90% hematologic response.
What is chronic lymphocytic leukemia?
Massive overproduction of mature; lymphocytes from production of B lymphocytes.
What is the presentation of chronic lymphocytic leukemia?
CLL can often present as an asymptomatic elevation of white cells. 90% being over the age of 50. Fatigue, lethargy, and enlargement of lymph nodes. Infiltration of spleen, liver, and bone marrow.
How is chronic lymphocytic leukemia diagnosed?
Older patient with a marked elevation in the white cell count with a marked lymphocytic predominance. The marrow is infiltrated with the leukemic lymphocytes with CD19 antigen.
What is multiple myeloma?
Plasma cell overproduction, replacing the bone marrow; large quantities of functionless immunoglobulins.
What is the presentation of multiple myeloma?
Bone pain. Pain in back, ribs; fractures. Radiculopathy. Encapsulated organisms (Pneumococcus, Haemophilus). Renal failure, anemia. Hypercalcemia: polyuria, polydipsia, altered mental. Weakness, pallor.
How is multiple myeloma diagnosed?
Normochromic, normocytic anemia. Protein electrophoresis monoclonal immunoglobulin spike. Skull punched out lesions. beta2 microglobulin. Hypercalcemia from bone destruction. Elevated BUN, creatinine from kidney damage. Biopsy: 10% plasma cells.
What is Bence-Jones protein?
Urinary Bence-Jones protein is nonfunctional gamma globulin found in multiple myeloma. It will not react with urine dipstick test.
What is the treatment for multiple myeloma?
Bone marrow transplantation. Older patients should receive melphalan and prednisone.
What is monoclonal gammopathy of uncertain significance?
Overproduction of immunoglobulin by plasma cells without manifestations MGUS is present in 1% > 50 and in 3% of those > age 70. Monoclonal immunoglobulin spike on electrophoresis. Treatment is not necessary.
What is Hodgkin disease?
A neoplastic transformation of lymphocytes in the lymph node with Reed-Sternberg cells; spread to contagious areas of lymph nodes. Bimodal age peaks in the 20s and 60s.
What is the presentation of Hodgkin disease?
Enlarged, rubbery, nonerythematous, nontender lymph nodes. "B" symptoms are night sweats, weight loss, fevers. Pruritus; cervical, supraclavicular, axillary lymphadenopathy most common. Spleen, skin, gastric, lung, CNS.
What is the treatment for Hodgkin disease?
Localized disease is managed with radiation. All patients with "B" symptoms and stage III or stage IV disease are managed with chemotherapy.
What is non-Hodgkin lymphoma?
Neoplastic transformation of B and T lymphatic cells. Accumulation of neoplastic cells in both the lymph nodes and diffusely in extra-lymphatic organs and the bloodstream. Reed-Sternberg cell is absent.
What conditions predispose to non-Hodgkin lymphoma?
Infections such as HIV, hepatitis C, Epstein-Barr, HTLV-I, and Helicobacter pylori predispose to the development of NHL.
What is the presentation of non-Hodgkin lymphoma?
Enlarged, rubbery, nonerythematous, nontender lymph nodes. "B" symptoms are night sweats, weight loss, fevers. Pruritus may occur.
What sites are affected by non-Hodgkin lymphoma?
NHL is the same as HD except that HD is localized to cervical and supraclavicular nodes. NHL involves extra-lymphatic sites and blood. CNS is more common NHL. HIV patients often have CNS NHL.
What is the treatment for non-Hodgkin lymphoma?
Local disease is treated with radiation, and those with "B" symptoms or Stages III and IV receive combination chemotherapy.
A 21-year-old woman with epistaxis and heavy periods. She has a PT of 12 seconds, a PTT of 40 seconds (prolonged), and 200,000/mm3 platelets. What is the diagnosis?
Idiopathic thrombocytopenic purpura. Also known as primary immune thrombocytopenic purpura.
What is idiopathic thrombocytopenic purpura?
Thrombocytopenia caused by idiopathic production of an antibody to platelets; often associated with lymphoma, chronic lymphocytic leukemia, HIV, and connective tissue diseases.
What is the presentation of idiopathic thrombocytopenic purpura?
Bleeding skin, nasal, oral, GI, urine, vagina. Young, female; epistaxis, bruising, hematuria, uterine and GI bleeding. Petechiae, purpura, ecchymoses. Splenomegaly absent.
How is idiopathic thrombocytopenic purpura diagnosed?
Thrombocytopenia. Antiplatelet Ab have a high sensitivity, poor specificity. Marrow filled with megakaryocytes, indicating platelet destruction. Smear and creatinine are normal, excluding HUS, TTP, DIC.
What is the treatment for idiopathic thrombocytopenic purpura?
Prednisone. Splenectomy if very low platelet counts.
What is Von Willebrand disease?
Platelet-type bleeding, autosomal dominant, decreased von Willebrand factor; VWD is the most common congenital disorder of hemostasis. Decreased ability of platelets to adhere to endothelial lining of blood vessels.
What is the presentation of Von Willebrand disease?
Epistaxis, petechiae, bruising, and menstrual abnormalities; gastrointestinal and urinary tract bleeding.
How is Von Willebrand disease diagnosed?
Platelet count is normal. The bleeding time is increased. Von Willebrand factor is low. Ristocetin platelet aggregation test is abnormal. PTT may be elevated.
What is the treatment for von Willebrand disease?
Desmopressin acetate (DDAVP) for mild bleeding. Factor VIII replacement is used if desmopressin is not effective and the bleeding continues. Aspirin contraindicated. FFP is not useful.
Which coagulopathies cause a prolonged prothrombin time?
Factor VII deficiency, vitamin K deficiency, liver disease, warfarin use, factor VII inhibitor.
Which coagulopathies cause a prolonged PTT?
vWF and factors VIII, IX, XI, or XII deficiencies; heparin, antiphospholipid antibody
Which coagulopathies cause a prolonged PT and PTT?
Prothrombin, fibrinogen, factor V, factor X or combined factor deficiencies, liver disease, DIC, supratherapeutic heparin or warfarin; combined heparin and warfarin.
What is hemophilia?
Deficiency of clotting factor 8 in hemophilia A and deficiency of factor 9 in hemophilia B resulting in increased bleeding. X-linked recessive, resulting in disease in males. Females are carriers. Hemophilia A is far more common than B.
Why does hemophilia not express in females?
Females do not express the disease because homozygosity results in intrauterine death of the fetus.
What is the presentation of hemophilia?
Mild deficiencies result in absence of symptoms or with symptoms only during surgery or trauma. Severe deficiency results in spontaneous bleeding. Factor-type bleeding is deeper than bleeding caused by platelets.
What type of bleeding is associated with factor deficiency?
Factor deficiencies cause hemarthrosis, hematoma, gastrointestinal bleeding, urinary bleeding, bruising, CNS bleeding. Severe hemophilia presents by age two or circumcision.
How is hemophilia diagnosed?
A prolonged PTT with a normal PT. Specific factor VIII or IX levels allow for a precise diagnosis of hemophilia A or B.
What is the treatment of hemophilia?
Mild hemophilia can be treated with desmopressin (DDAVP). Severe deficiencies are treated with replacement of the specific factor. Desmopressin is ineffective for hemophilia B.
What factor deficiency results from vitamin K deficiency?
The deficiency of vitamin K results in decreased production of factors 2, 7, 9, and 10. Caused by dietary deficiency, malabsorption, and antibiotics that kill the colon bacteria that produce vitamin K.
What is the presentation of vitamin K deficiency?
Bleeding may occur at any site. Oozing at venipuncture sites.
How is vitamin K deficiency diagnosed?
PT and PTT are elevated. A correction of PT and PTT in response to giving vitamin K is the most common method of confirming the diagnosis.
What is the treatment for vitamin K deficiency?
Severe bleeding is treated with fresh frozen plasma. Vitamin K is given at the same time.
Why do patients with liver disease have increased PT and PTT?
Coagulopathy is caused by decreased production of clotting factors by the liver. Any severe liver disease or cirrhosis can cause decreased production of clotting factors.
What are the effects of coagulopathy of liver disease?
Bleeding at any site, gastrointestinal tract is the most common site. Vitamin K does not result in improvement. Low platelet counts are often caused by hypersplenism that accompanies the liver disease.
What is the treatment of coagulopathy of liver disease?
Fresh frozen plasma severe bleeding.
What is disseminated intravascular coagulation?
Consumptive coagulopathy from major illness, resulting in consumption of platelets and clotting factors; bleeding with a microangiopathic hemolysis and fibrin products; occasionally thrombosis.
What are the causes of disseminated intravascular coagulation?
Sepsis is most common. Any cell destruction and release of tissue factor. Rhabdomyolysis, adenocarcinomas, heatstroke, transfusion reactions, burns, head trauma, abruption, amniotic embolism, trauma, pancreatitis, snakebites, leukemia.
What is the presentation of disseminated intravascular coagulation?
Bleeding from any site. Thrombosis is less common. Hemolysis often may lead to acute renal failure, jaundice, confusion.
How is disseminated intravascular coagulation diagnosed?
Serious underlying disorder with bleeding, elevation PT, PTT; decrease platelets. Fibrinogen low. D-dimers and fibrin- split products are increased. Schistocytes with intravascular hemolysis.
What is the treatment for disseminated intravascular coagulation?
Fresh frozen plasma and platelet transfusions. Heparin is rarely used except in thrombosis.
What are the names of the semisynthetic penicillinase-resistant penicillins?
Oxacillin, cloxacillin, dicloxacillin, methicillin, nafcillin
Which bacteria are susceptible to the semisynthetic penicillins?
Staphylococci and streptococci. Oxacillin, nafcillin, dicloxacillin, cloxacillin. Exclusively effective against Gram-pos cocci, particular staph. Methicillin not used because interstitial nephritis.
What are the indications for the semisynthetic penicillins?
Used when Staphylococcus is sensitive to the semisynthetic penicillins and if concurrent Gram-negative infection is not suspected. More efficacious than vancomycin when the organism is sensitive.
What antibiotic is used to treat resistant-resistant Staph aureus?
MRSA is treated primarily with vancomycin. Oral linezolid or IV quinupristin-dalfopristin are alternatives for MRSA.
What is the coverage of linezolid?
Drug-resistant enterococcus, staphylococcus, and pneumococcus.
Which bacteria are sensitive to penicillin G, penicillin VK, ampicillin, and amoxicillin?
Effective against streptococci, such as S. pyogenes, viridans group streptococci, and S. pneumonia, but not against staphylococci.
When can ampicillin and amoxicillin be used against staphylococcal infections?
Ampicillin and amoxicillin are only effective against staph when ampicillin is combined with the beta-lactamase inhibitor sulbactam or when amoxicillin is combined with clavulanate.
What is the coverage of ampicillin against E coli?
Ampicillin also has some activity against E. coli. Ampicillin and amoxicillin are effective against enterococci and Listeria. Useful against Gram-negative bacteria, such as Neisseria.
What are the first-generation cephalosporins?
Cefazolin, cefadroxil, cephalexin.
What are the second-generation cephalosporins?
Cefoxitin, cefotetan, cefuroxime, cefprozil, loracarbef.
What is the coverage of the first-generation cephalosporins?
Same organisms that the semisynthetic penicillins will cover. In addition to staphylococci and streptococci, 1st and 2nd gen cephalosporins will also cover some Gram-neg. 1st gen agents will only reliably cover Gram-neg Moraxella and E. coli.
What is the coverage of second-generation cephalosporins?
Cover everything a first-generation cephalosporin covers, as well as a few more Gram-negative bacilli such as Providencia, Haemophilus, Klebsiella, Citrobacter, Morganella, and Proteus.
What is the coverage of the third-generation cephalosporins?
3rd gen, particularly ceftazidime, are not reliable against staphylococcus. 4th gen cephalosporin, cefepime, will cover staph and strep, but should not be used for exclusively Gram-pos infections.
What is the allergic cross-reactivity of penicillins with cephalosporins?
For persons allergic to penicillin, there is only 5% risk of cross-reaction with cephalosporins. When the allergic reaction was a rash, a cephalosporin can be used. When the reaction was anaphylaxis, a cephalosporin should not be used.
What antibiotic should be used in patients with a history of an anaphylactic reaction to a penicillin?
A macrolide (clarithromycin or azithromycin) or levofloxacin, gatifloxacin, or moxifloxacin can be used. For serious infections, use vancomycin, linezolid, or daptomycin.
What are the indications for macrolides (erythromycin, clarithromycin, azithromycin), extended fluoroquinolones (levofloxacin, gatifloxacin, moxifloxacin), and clindamycin?
Alternatives to penicillin and cephalosporins for Gram-pos infection. Macrolides should not be used for serious staph. Extended quinolones very good for strep, particularly S pneumoniae in absence of penicillin resistance. Good against staph; not MRSA.
Why should ciprofloxacin not be used to treat pneumonia?
Ciprofloxacin does not cover Strep pneumoniae.
What are the indications for vancomycin, linezolid, quinupristin, dalfopristin?
Alternatives for Gram-pos infections when penicillin allergy or MRSA. Linezolid is the only oral MRSA.
What is the coverage of linezolid, quinupristin, dalfopristin?
Linezolid, quinupristin, dalfopristin are effective against MRSA and vancomycin-resistant enterococci.
What is the activity of antipseudomonal penicillins (piperacillin, ticarcillin, mezlocillin) against Gram-negative bacilli?
Fully active against range of Gram-negative bacilli, such as the Enterobacteriaceae including Pseudomonas. Enterobacteriaceae include E. coli, Proteus, Enterobacter, Citrobacter, Morganella, Serratia, Klebsiella.
What is the activity of ticarcillin and mezlocillin against MSSA?
Antipseudomonal penicillins are only active against staph when combined with a beta-lactamase inhibitor, such as piperacillin/tazobactam or ticarcillin/clavulanate. Not MRSA.
What is the coverage of ampicillin/sulbactam and amoxicillin/clavulanate?
Cover strep and staph and Gram-neg bacilli, but not Pseudomonas. All penicillins will cover sensitive streptococci.
What are the third-generation cephalosporins?
Ceftazidime, cefotaxime, ceftriaxone, cefotaxime.
What is the fourth generation cephalosporin?
Cefepime
What is the coverage of third- and fourth-generation cephalosporins?
Fully active against full range of Gram-neg bacilli, such as the Enterobacteriaceae. Only ceftazidime and cefepime will cover Pseudomonas. Cefepime also covers staph.
What is the Gram-negative coverage of second-generation agents?
Cover some of the Enterobacteriaceae, but not Pseudomonas.
What is the coverage of the third generation cephalosporins?
Predominantly for use against Gram-negative organisms. Ceftriaxone and cefotaxime cover best for penicillin-insensitive pneumococci which cause meningitis or pneumonia.
What is the coverage of quinolones (ciprofloxacin, levofloxacin, gatifloxacin, moxifloxacin, ofloxacin)?
Cover most Enterobacteriaceae, such as E. coli, Proteus, Enterobacter, Haemophilus, Moraxella, Citrobacter, Morganella, Serratia, and Klebsiella. Only ciprofloxacin will cover Pseudomonas.
What is the coverage of extended-spectrum quinolones?
Moxifloxacin, levofloxacin, gatifloxacin are also active against Gram-positive cocci, in particular Strep pneumoniae. Good for pneumonia because also cover Mycoplasma, Chlamydia, Legionella.
What is the coverage of aminoglycosides (gentamicin, tobramycin, amikacin)?
Gram-negative coverage. Aminoglycosides are synergistic with ampicillin in treatment of staph.
What is the coverage of monobactams?
Aztreonam is exclusively a Gram-negative agent, with no strep or staph coverage.
What is the coverage of carbapenems (imipenem, meropenem, ertapenem)?
Enterobacteriaceae and Pseudomonas with Gram-neg coverage. Excellent staph and anaerobic coverage. Used in Gram-negative infections, but also effective polymicrobial. Ertapenem not cover Pseudomonas.
What agent is most active against intraabdominal anaerobes?
Metronidazole. Clindamycin is less active against intraabdominal anaerobes. Metronidazole has good anaerobic Gram- neg in bowel, such as B fragilis. Metronidazole is first-line against C difficile.
What is the coverage of clindamycin?
Clindamycin may have some advantages over metronidazole against the anaerobic streptococci found in the mouth.
What agents have intraabdominal anaerobic coverage?
Metronidazole, carbapenems and beta-lactam/beta-lactamase combinations (piperacillin/tazobactam, ticarcillin/clavulanate, ampicillin/sulbactam, or amoxicillin/clavulanate). 2nd gen cephalosporins cefoxitin and cefotetan, have fair activity against.
A 15-year-old male with fever, headache, nausea, and vomiting. Neck stiffness and photophobia. What is the next step?
Lumbar puncture.
What is the most common cause of meningitis after the neonatal period?
Streptococcus pneumoniae is the most common cause of meningitis for all patients beyond the neonatal period. Neisseria meningitidis is the most common cause of meningitis in adolescents.
What factors predispose to listeria monocytogenes meningitis?
Listeria monocytogenes is more common with immune system defects, HIV, steroid use, leukemia, lymphoma, and various chemotherapeutic agents. Neonates and the elderly.
What is the most common cause of meningitis in the elderly and infants?
Streptococcus pneumoniae is the most common etiology. Listeria is more common in the very old and infants.
Which patients are at risk for staphylococcal meningitis?
Staphylococcus aureus is more common in those who have had any form of neurosurgery.
Which patients are at risk for cryptococcus meningitis?
Cryptococcus is more common in those who are HIV positive and who have profound decreases in their T-cell counts to levels less than 100 cells/mm3.
Which persons are at risk for Rocky Mountain spotted fever meningitis?
Rocky Mountain spotted fever is common in those who have been exposed to ticks in the mid-Atlantic areas, such as the Carolinas, Kentucky, Tennessee, Delaware, Maryland, Virginia, Georgia, Florida, Oklahoma, etc.
What area is associated with Lyme meningitis?
Lyme disease can also cause meningitis and is more common in the Northeast. Tuberculosis and syphilis are also associated with meningitis.
What are the CSF characteristics in aseptic meningitis?
CSF has lymphocytic pleocytosis and bacterial cultures are negative.
What are the causes of aseptic meningitis?
Enteroviruses, arboviruses (St. Louis encephalitis virus, West Nile virus), HIV, herpes simplex, and lymphocytic choriomeningitis virus.
What is the most common cause of neonatal meningitis?
Group B Streptococcus (Streptococcus agalactiae) is the most common cause of meningitis in the neonatal period.
What is the clinical presentation of meningitis?
Fever, photophobia, headache, nuchal rigidity (Kernig, Brudzinski), nausea, vomiting, seizures. The most common deficit from meningitis is damage to 8th cranial nerve (hearing).
What type of meningitis is associated with a petechial rash?
Neisseria. A rash on the wrists and ankles with centripetal spread toward the body suggests Rocky Mountain spotted fever.
What type of meningitis is associated with facial palsy?
Facial nerve palsy is suggestive of Lyme disease. Pulmonary symptoms or an abnormal chest x-ray suggest tuberculosis.
When should a CT scan be done before lumbar puncture in patients with suspected meningitis?
CT should be done before lumbar puncture if papilledema, focal motor deficits, new onset seizures, mental status abnormalities, or immunocompromise (HIV, immunosuppressive).
What should be done if the lumbar puncture is delayed more than 20-30 minutes?
An empiric dose of antibiotics should be given.
What are the CSF findings in bacterial meningitis?
Protein elevated with bacterial meningitis. Opening pressure elevated. Gram stain has a limited sensitivity; positive 50%. The most useful test is the cell count. Only bacterial meningitis causes thousands of cells, all neutrophils.
What types of meningitis are associated with an elevation in lymphocytes?
Viral infection, Rickettsia, Lyme disease, tuberculosis, syphilis, or fungal (cryptococcal) etiology. Normal CSF cell count is <5 cells, lymphocytes.
What is the empiric treatment of bacterial meningitis in adults?
Vancomycin (pneumococci are resistant penicillin) plus a 3rd gen cephalosporin, such as ceftriaxone. Ampicillin is added to cover Listeria if immune defects, >50, or <1 month. Listeria is resistant cephalosporins.
What is the treatment of Lyme meningitis?
Lyme disease is treated with ceftriaxone.
What is the treatment of Cryptococcal meningitis?
Cryptococcus is treated with amphotericin. Followed by fluconazole in HIV-positive patients for life or until the patient is on HAART and is asymptomatic with a CD4 count >100.
What is the treatment of neurosyphilis?
Neurosyphilis is treated with high-dose intravenous penicillin.
What is the treatment of tuberculosis meningitis?
TB meningitis is treated the same as pulmonary tuberculosis (though a longer duration of 9-12 months).
What are the indications for corticosteroid in meningitis?
Steroid use in adult meningitis is appropriate for TB meningitis and bacterial meningitis.
What are the recommendations for adjunctive dexamethasone for bacterial meningitis?
Reduces inflammatory response to bacterial lysis; given with antibiotics for 4 days if bacterial meningitis is confirmed by positive Gram stain or >1000 WBCs in CSF.
What is encephalitis?
Infection of brain parenchyma usually caused by any viruses. The most common cause is HSV type 1. Varicella-zoster virus, CMV, enteroviruses, Eastern and Western equine encephalitis, St. Louis, and West Nile.
What is the presentation of encephalitis?
Altered mental status with fever and headache. Confusion to lethargy or coma. Focal deficits. Neck stiffness. Seizures.
How is HSV encephalitis diagnosed?
Lumbar puncture PCR has eliminated the need for biopsy. PCR for HSV has a 98% sensitivity and >95% specificity. HSV has a predilection for temporal lobes.
What is the treatment for HSV encephalitis?
Intravenous acyclovir.
What is the treatment of cytomegalovirus encephalitis?
Ganciclovir or foscarnet are active against CMV.
An HIV-negative man with a seizure. Aphasia and weakness of the right hand and leg. A CT scan of the head with contrast shows a ring enhancing lesion. What is the diagnosis?
Brain abscess.
What are the causes of brain abscesses?
Bacteria can spread into the brain from otitis media, sinusitis, mastoiditis, or dental infections. Organisms may also spread through the bloodstream from endocarditis or pneumonia.
Which organisms cause brain abscesses?
Brain abscesses most commonly Streptococcus 60-70%, Bacteroides 20-40%, Enterobacteriaceae 25-35% and Staph 10%, often polymicrobial. Toxoplasmosis can reactivate in HIV when CD4 <100.
What is the presentation of brain abscess?
Headache. Fever, focal neurologic deficits, seizures.
How is brain abscess diagnosed?
Brain abscess and malignancy enhance with contrast. MRI more accurate than CT. Stereotactic aspiration for Gram stain, culture. In HIV, 90% of brain lesions will be toxoplasmosis or lymphoma.
What is the treatment for brain abscess?
Surgical drainage. Penicillin, metronidazole, and a third-generation cephalosporin, such as ceftazidime. HIV patients are treated empirically with pyrimethamine and sulfadiazine.
What are the causes of otitis media?
The most common organisms are Strep pneumoniae (35-40%), H. influenzae (nontypeable; 25-30%), and Moraxella catarrhalis (15-20%). Viruses.
What is the presentation of otitis media?
Ear pain, fever, and decreased hearing. Immobility of the membrane on insufflation.
What is the treatment for otitis media?
High-dose amoxicillin. Amoxicillin-clavulanate is used if recent amoxicillin or if no response to amoxicillin. Alternatives: Cefuroxime, loracarbef, cefprozil, or cefdinir or cefixime.
What is the treatment for otitis media in severe allergy to penicillin?
Macrolides, such as azithromycin or clarithromycin. Levofloxacin, moxifloxacin or gatifloxacin are acceptable. TMP/SMZ is poorly active against Streptococcus pneumoniae.
What is the most common site of sinusitis?
The most common site of infection is the maxillary sinus.
What are the cause of sinusitis?
Viruses are responsible for most cases. Bacterial organisms are strep pneumonia, H influenza (non-typable), Moraxella catarrhalis.
What is the presentation of sinusitis?
Facial pain, headache, postnasal drainage, and purulent nasal drainage. Headache, fever, tooth pain.
How is sinusitis diagnosed?
If imaging is required because of concern of complications, a CT scan is the best test.
What is the treatment for sinusitis?
High-dose amoxicillin. Amoxicillin-clavulanate if recent amoxicillin use or if no response amoxicillin. Alternatives: cefuroxime, loracarbef, cefprozil, cefdinir, cefixime.
What is the treatment of sinusitis in patients with severe penicillin allergies?
Patients with severe penicillin allergies should receive azithromycin or clarithromycin. Levofloxacin, moxifloxacin, or gatifloxacin are acceptable.
What are the causes of pharyngitis?
Usually viruses; group A beta-hemolytic streptococci (S. pyogenes) may cause rheumatic fever or glomerulonephritis. S. pyogenes causes 15-20% of cases of pharyngitis.
What is the presentation of pharyngitis?
Sore throat with cervical adenopathy and inflammation of the pharynx with an exudate is highly suggestive of S. pyogenes. Viruses do not cause an exudate.
How is pharyngitis diagnosed?
Rapid Streptococcal antigen test is 60-100% sensitive and >95% specific. A positive test can be considered the equivalent of a positive culture, whereas a negative test should be confirmed with a culture.
What is the treatment for pharyngitis?
Penicillin or amoxicillin. Azithromycin, clarithromycin, and oral, second-generation cephalosporins are alternatives in penicillin-allergic.
What is influenza?
A systemic viral illness from influenza A or B, usually occurring in an epidemic pattern, leading to sinusitis, otitis media, bronchitis, and pneumonia.
What is the presentation of influenza?
Fever, myalgias, headache, and fatigue. Upper respiratory symptoms predominate. Coryza, nonproductive cough, sore throat, and conjunctival injection.
How is influenza diagnosed?
Rapid antigen detection methods of swabs or washings of nasopharyngeal secretions. Viral culture.
What is the treatment for influenza?
Specific antiviral medications for both influenza A and B are the neuraminidase inhibitors: oseltamivir and zanamivir. Amantadine and rimantadine are only effective against influenza A.
What are the indications for influenza vaccinations?
>50 chronic lung and cardiac disease, pregnant in the second and third trimesters; chronic facilities, health-care workers, immunosuppressed, diabetes, renal dysfunction. Contraindicated in allergy to eggs.
A 63-year-old man with a cough sputum smoked 30 years. Temperature of 100 degrees F. Chest x-ray is normal. What is the diagnosis?
Bronchitis
What are the causes of bronchitis?
Viruses. Small percentage caused by M. pneumoniae, C. pneumoniae, B. pertussis. Most common organisms responsible for chronic bronchitis are S pneumoniae, H influenzae, and Moraxella. CXR normal.
What is the treatment for acute exacerbations of chronic bronchitis?
Amoxicillin, doxy, or TMP/SMZ. Patients not responding to amoxicillin should be treated with amoxicillin/clavulanate, clarithromycin, azithromycin, 2nd/3rd-gen cephalosporins, or gatifloxacin, levofloxacin, or moxifloxacin.
A 65-year-old alcoholic with cough, sputum, fever. Lost 15 pounds. Febrile. Poor dentition. Putrid, foul breath. CXR: cavitary lesion. What is the diagnosis?
Lung abscess.
What is a lung abscess?
Necrosis of the pulmonary parenchyma caused by microbial infection.
What is the bacteriology of lung abscesses?
90% anaerobes. Peptostreptococcus, Prevotella, and Fusobacterium species (oral anaerobes). Aerobic bacteria, most frequently are S. aureus, E. coli, Klebsiella, and Pseudomonas.
What conditions predispose to lung abscess?
Association with periodontal disease or aspiration (e.g., altered sensorium, seizures).
What is the presentation of lung abscess?
Fever, cough, chest pain; putrid, foul-smelling sputum in 60-70%; chronic course with weight loss and fatigue.
How is lung abscess diagnosed?
Sputum Gram stain and culture. Cavitary lesion. Lower lobes in upright patients, and the posterior right upper lobe in supine patients. Aspiration of abscess fluid is necessary for diagnosis.
What is the treatment of lung abscess?
Clindamycin has good empiric coverage for the "above the diaphragm" anaerobes. Penicillin is also acceptable.
What conditions predispose to pneumonia?
Cigarette smoking, diabetes, alcoholism, malnutrition, obstruction from tumors, and immunosuppression. Neutropenia and steroid use predispose to Aspergillus infection.
What is the most common cause of community-acquired pneumonia in all groups?
S. pneumoniae (viruses <5 years). Hospital-acquired or ventilator-associated pneumonia: Gram-neg bacilli such as E. coli, the other Enterobacteriaceae, or Pseudomonas, and MRSA.
What conditions predispose to Haemophilus influenzae pneumonia?
Smokers, COPD.
What factors are associated with Mycoplasma pneumonia?
Young, otherwise healthy patients.
What conditions predispose to Legionella?
Epidemic infection in older smokers, particularly when located near infected water sources, such as air-conditioning systems.
What conditions predispose to Pneumocystis jiroveci pneumonia?
HIV-positive persons with <200 CD4 cells/mm3 not on PJP prophylaxis.
What conditions predispose to Coxiella burnetii (Q-fever) pneumonia?
Exposure to animals, particularly at the time the animals are giving birth.
What condition predisposes to Klebsiella pneumonia?
Alcoholics
What condition predisposes to Staphylococcus aureus pneumonia?
Occurs after viral syndromes or viral bronchitis, especially influenza
What condition predisposes to Coccidioidomycosis pneumonia?
Exposure to the deserts of the American Southwest, particularly Arizona
What conditions predispose to Chlamydia psittaci pneumonia?
Bird exposure.
What conditions predispose to Histoplasma capsulatum pneumonia?
Exposure to bat or bird droppings, spelunking (recreational cave exploration)
What are the clinical signs of Bordetella pertussis pneumonia?
Cough with whoop and post-tussive vomiting.
What conditions predispose to Francisella tularensis pneumonia?
Hunters or exposure to rabbits, bioterrorism.
What activity predisposes to severe acute respiratory syndrome (SARS, Avian influenza pneumonia)?
Travel to southeast Asia.
Which bacteria may be utilized in bioterrorism?
Bacillus anthracis, Yersinia pestis, and Francisella tularensis.
What is the clinical presentation of pneumonia?
Cough, fever, sputum, dyspnea. Bacterial infections (pneumoniae, Haemophilus, Klebsiella) have purulent sputum. S. pneumoniae causes rusty hemoptysis.
What are the characteristics of the sputum in Klebsiella pneumonia?
Klebsiella pneumoniae sputum is like currant jelly.
What are the characteristics of pneumonia caused by Pneumocystis jiroveci, viruses, Mycoplasma, and Legionella?
Interstitial infections are caused by Pneumocystis jiroveci pneumonia, viruses, Mycoplasma, and Legionella; often cause nonproductive or "dry" cough.
What type of pain is associated with pneumococcal pneumonia?
Pleuritic pain is associated with lobar pneumonia, caused by Pneumococcus, because of inflammation of the pleura. S. pneumoniae usually causes a lobar pneumonia with pleural effusion.
What are the physical signs of pneumonia?
Rales, rhonchi, lung consolidation, dullness to percussion, bronchial breath sounds, increased vocal fremitus, egophony. Severe pneumonia leads to hypoxia.
What is the presentation of Mycoplasma pneumonia?
Dry cough and chest soreness. Bullous myringitis; anemia from hemolysis from cold agglutinins. Rarely need to be admitted to the hospital.
What is the presentation of Legionella pneumonia?
Confusion, headache, and lethargy. Diarrhea and abdominal pain.
What is the presentation of Pneumocystis jiroveci pneumonia?
Marked dyspnea, particularly on exertion, with chest soreness with cough in an HIV-positive person with a CD4 count of <200/mm3.
Which types of pneumonia are associated with interstitial infiltrates?
Interstitial infiltrates are associated with PJP, viral, Mycoplasma, Chlamydia, Coxiella, and Legionella pneumoniae.
What is the most specific test for lobar pneumonia?
Sputum culture and Gram stain. S pneumoniae, Staph, Klebsiella, Haemophilus. Viral, Mycoplasma, Chlamydia, Coxiella ("atypicals") will not appear on a Gram stain or bacterial culture.
What is the best diagnostic method for Mycoplasma pneumonia?
Specific serologic antibody titers. Cold agglutinins have limited specificity and sensitivity.
What are the diagnostic methods for Legionella pneumonia?
Charcoal yeast extract media, urine antigen tests, direct fluorescent antibodies, and antibody titers.
What is the best diagnostic method for Pneumocystis jiroveci pneumonia?
Bronchoalveolar lavage; increased LDH.
What is the best diagnostic method for Chlamydia pneumoniae, Coxiella, Coccidioidomycosis, and Chlamydia psittaci?
Specific antibody titers.
What are the signs of severe pneumonia?
pO2 <60 mm Hg, O2 sat <94%, resp >30, disorientation, uremia, systolic BP <90. High fever, hypothermia, leukopenia (<4,000), tachycardia (>125), hyponatremia, dehydration, elevated BUN.
Which pneumonia patients should be hospitalized?
Patients with severe pneumonia or serious underlying diseases such as cancer, liver disease, renal disease, or chronic lung disease should be hospitalized for intravenous medications.
What is the empiric therapy for outpatient pneumonia?
Macrolide, azithromycin or clarithromycin. Levofloxacin, moxifloxacin, gatifloxacin are alternatives. Cephalosporins and amoxicillin/clavulanate do not cover atypical pathogens.
What is the treatment for hospitalized patients with community-acquired pneumonia?
Levofloxacin, moxifloxacin, or gatifloxacin or cefotaxime or ceftriaxone combined with a macrolide such as azithromycin or clarithromycin (or doxycycline).
What organisms cause hospital-acquired pneumonia?
Drug-resistant, Gram-negative bacilli (Pseudomonas, Klebsiella, E. coli), or gram-positives, including methicillin-resistant Staphylococcus aureus.
What is the treatment of hospital-acquired pneumonia?
3rd gen cephalosporin with antipseudomonal activity (ceftazidime, cefotaxime) or carbapenems (imipenem) or beta-lactam/beta-lactamase combinations (piperacillin/tazobactam), and vancomycin or linezolid. Gentamicin added for Gram-neg synergy.
What is the specific coverage for Haemophilus influenzae?
Second- or third-generation cephalosporin.
What is the specific coverage for Mycoplasma pneumoniae?
Macrolide, doxycycline, or a quinolone.
What is the specific coverage for Legionella?
Macrolide, doxycycline, or a quinolone
What is the specific coverage for pneumocystis jiroveci pneumonia?
Trimethoprim/sulfamethoxazole. Steroids should be used if arterial pO2 <70 mm Hg. If the patient is allergic to TMP/SMZ, intravenous pentamidine or atovaquone should be used.
What is the specific coverage for Coxiella burnetii (Q-fever)?
Doxycycline (or erythromycin)
What is the specific coverage for klebsiella?
Third-generation cephalosporin.
What is the specific coverage for staphylococcus aureus?
Semisynthetic penicillin (nafcillin) if methicillin sensitive. In the nosocomial setting, isolates are methicillin-resistant, and vancomycin or linezolid is used.
What is the specific coverage for coccidioidomycosis?
Treatment only for disseminated disease or pulmonary disease with immunosuppression. Life-threatening disease is treated with amphotericin. Mild disease is treated with fluconazole or itraconazole.
Which patients should receive pneumococcal vaccine?
>65. Lung, cardiac, liver, renal disease. Immunocompromised. Re-dosing in 5 y is only necessary with severe immunocompromise or if vaccinated <65. >65, a single dose confers lifelong immunity.
What is bacillus Calmette-Guerin?
Bacillus Calmette-Guerin vaccination is used outside the United States to prevent tuberculosis. Only 50% effective and is never used in the US. BCG vaccination does not alter PPD test interpretation.
Which persons are at risk for tuberculosis infection?
Immigrants, alcoholics, healthcare workers, prisoners, homeless shelters, nursing homes, chronically debilitated, impairment of T-cell immunity. Steroid use, organ transplantation, leukemia, lymphoma, and HIV.
What is the clinical presentation of tuberculosis?
Cough, sputum, fever, and an abnormal lung examination. Weight loss. Tuberculosis usually takes up to five years to become fatal. Night sweats. TB occurs outside of the lungs in 15-20%.
What sites may be affected by extrapulmonary tuberculosis?
Any part of the body, although the lymph nodes, meninges, GI and GU are the most frequent extrapulmonary TB. Adenitis is the most common extrapulmonary site.
How is tuberculosis diagnosed?
CXR: apical infiltrates, cavitation. Adenopathy, effusion, calcified nodules (Ghon). 3 sputum smears 90% sensitive. Culture is most specific; 4 wks. PPD not used to diagnose acute TB unless other tests neg.
What is the treatment of tuberculosis?
Initial 4-drug: INH, rifampin, pyrazinamide, ETB. 4 drugs continued for 2 mo or until sensitivity known. PZA/ETB are discontinued, and continue INH, rifampin for 4 mo. ETB given if sensitivity not known.
What is the length of treatment of tuberculosis meningitis?
12 mths. TB in pregnancy is treated for 9 mths. HIV- positive treated for 6-9 months.
What drug should be taken with isoniazid?
INH use should generally be combined with vitamin B6 (pyridoxine) to prevent peripheral neuropathy.
Which antituberculosis drugs are contraindicated in pregnancy?
Pregnant patients should not receive PZA or streptomycin.
When are steroids used in tuberculosis?
Steroids are used for TB meningitis and TB pericarditis.
What are the side effects of antituberculosis medications?
All TB medications can cause liver toxicity, except streptomycin. INH causes peripheral neuropathy because of pyridoxine deficiency. Rifampin is associated with orange/red bodily fluids .
What are the adverse effects of ethambutol?
Ethambutol is associated with optic neuritis, color blindness and other visual disturbances. PZA can cause a benign hyperuricemia, which does not require treatment.
What is the PPD test?
PPD test is used to screen asymptomatic populations at risk of TB to determine if they have been exposed and are at increased risk of re-activating. PPD should not be used to diagnose TB in an acutely ill patient.
What is the physical indication of a positive PPD?
A PPD test is positive if there is induration of the skin 48-72 h after intradermal injection. A positive PPD test indicates a 10% risk of TB in HIV-negative. BCG vaccination does not alter recommendations.
What is two-stage PPD testing?
Those in whom there has not been a recent PPD test and now show some reactivity that is <10 mm should have a second test within two weeks. This ensures that the first test was not a false negative.
What is the criteria for a positive PPD in close contacts of active TB cases, HIV-positive persons, persons with an abnormal chest x-ray consistent with old TB, and steroid use or organ transplantation recipients?
>5 mm
What is the criteria for a positive PPD in high-risk groups, such as healthcare workers, prisoners, nursing home residents; immigrants; homeless; leukemia, lymphoma, diabetics, dialysis, IVDU who are HIV-neg; and children exposed to adults at high risk?
>10 mm
What is the criteria for a positive PPD in low risk populations?
>15 mm
What is the management of a positive PPD?
Chest x-ray to determine if they have early signs of TB. Those with abnormal chest x-rays should have sputum AFB stains to see if they have active disease. Positive AFB smears need the start of four TB drugs.
What is the treatment for patients with positive PPD and no evidence of active disease (negative chest x-ray)?
9 months of INH and vitamin B6. A normal chest x-ray or an abnormal x-ray and three negative AFB stains of sputum are sufficient to exclude active disease.
What is the cause of most infectious diarrhea?
Contaminated food and water. Bacillus cereus and Staphylococcus aureus, food poisoning present predominantly with vomiting. Food poisoning is most commonly Campylobacter.
What type of diarrhea is associated with poultry and eggs?
Salmonella is most commonly associated with contaminated poultry and eggs.
What is the most common cause of travelers' diarrhea?
E. coli is the most common cause of travelers' diarrhea. E. coli 0157:H7 is associated with eating undercooked hamburger meat.
What organism is associated with fried rice?
Bacillus cereus is associated with fried rice.
What types of diarrhea are associated with water from streams?
Giardia lamblia and cryptosporidiosis are acquired from contaminated water from streams.
What type of diarrhea is associated with AIDS?
Cryptosporidiosis is associated with HIV, particularly when the CD4 count has dropped below 50 cells. Vibrio cholera is very rare in the United States.
What type of diarrhea is associated with shellfish?
Vibrio parahaemolyticus is associated with contaminated shellfish.
What is the most common cause of diarrhea in children?
Viral infections such as with rotavirus or Norwalk agents are associated with outbreaks in children.
What type of diarrhea is associated with previous antibiotic use?
Clostridia difficile is associated with previous antibiotic use.
What type of food is associated with clostridia botulinum?
Clostridia botulinum is associated with ingestion of infected canned foods, and Clostridia perfringens is associated with meats that have been contaminated with spores.
What organisms are associated with blood in the stool?
Blood in the stool is most commonly associated with invasive enteric pathogens, such as Salmonella, Shigella, Yersinia, invasive E. coli, and Campylobacter.
What type of diarrhea is associated with Guillain-Barre syndrome?
Campylobacter is rarely associated with Guillain-Barre syndrome.
What are the symptoms of ciguatera toxin poisoning?
Ingestion of barracuda, red snapper, grouper within 2 h; paresthesias, numbness, nausea, vomiting, cramps; weakness, reversal of hot-cold, hypotension.
What is the treatment for ciguatera toxin poisoning?
No therapy.
Which infectious agents are associated with hemolytic uremic syndrome?
E. coli 0157:H7 and Shigella are associated with hemolytic uremic syndrome.
What is the presentation of staphylococcal food poisoning?
Bacillus cereus and Staphylococcus predominantly present with vomiting within 1-6 hours of their ingestion because they contain a preformed toxin. Diarrhea occurs later.
What is the characteristic of viral diarrhea?
Viruses can cause voluminous watery diarrhea but do not result in bloody diarrhea.
What type of diarrhea occurs after ingestion of tuna, mackerel, or mahi mahi?
Scombroid poisoning from histamine in flesh. Minutes: rash, diarrhea, vomiting, wheezing, burning in mouth, dizziness, paresthesias.
How is infectious diarrhea diagnosed?
Test the stool for white blood cells with methylene blue testing. WBCs indicate an invasive pathogen. Culture is necessary to determine specific type.
Which test is used to detect Giardia and Cryptosporidium?
Giardia and Cryptosporidium are detected by direct examination for ova and parasites, as well as for their eggs. A special modified AFB stain is necessary to detect Cryptosporidium.
What is the treatment of infectious diarrhea?
More severe infections with high fever, abdominal pain, tachycardia and hypotension, require intravenous fluids and oral ciprofloxacin.
What are the causes of viral hepatitis?
Viral hepatitis is an infection of the liver caused by hepatitis A, B, C, D, or E.
What are the symptoms of hepatitis A?
Fever, malaise, headache, anorexia, vomiting, dark urine, jaundice.
What are the symptoms of hepatitis B?
Fever, malaise, headache, anorexia, vomiting, dark urine, jaundice, 10-20% with serum sickness-like (joint pain, rash).
What are the symptoms of hepatitis C?
Only 20% are acutely symptomatic with fever, malaise, headache, jaundice.
What are the symptoms of hepatitis D?
Fever, malaise, headache, anorexia, vomiting, dark urine, jaundice
What are the symptoms of hepatitis E?
Fever, malaise, headache, anorexia, vomiting, dark urine, jaundice
What types of hepatitis have a carrier state?
Hepatitis B, C, D. There is no carrier state for hepatitis A or hepatitis E.
What is the serology of hepatitis A?
Anti-HAV, IgM fraction, IgG fraction.
What is the serology of hepatitis B?
HBsAg, HBsAb, HBeAg, anti-HBs, anti-HBc, anti-HBe
What is the serology of hepatitis C?
Antibody to hepatitis C, PCR-RNA
What is the serology of hepatitis D?
Anti-delta, IgM fraction, IgG fraction
What is the serology of hepatitis E?
Anti-Hep E IgM, IgG
What is the mode of transmission of hepatitis A and E?
Transmitted by contaminated food and water. Incubation period 2-6 weeks. Hepatitis A and E cause symptomatic disease for several days to weeks, no chronic form, cause cirrhosis or hepatocellular CA.
What is the mode of transmission of hepatitis B, C and D?
Transmitted parenterally, perinatally, or through sexual contact; blood transfusion, needlestick, and needle sharing. Hepatitis B and C cause a chronic form, cirrhosis and hepatocellular carcinoma.
What is the most common disease leading to the need for liver transplantation?
Hepatitis C. All forms of hepatitis can occasionally present with fulminant hepatic necrosis and acute liver failure.
What is the presentation of acute hepatitis?
Jaundice, dark urine, light stool, fatigue, malaise, weight loss, tender enlarged liver. B and C can also cause joint pain, rash, vasculitis, glomerulonephritis. B with polyarteritis nodosa.
How is acute hepatitis diagnosed?
Viral and drug hepatitis cause elevated bilirubin. Viral hepatitis causes elevation of ALT and AST, but the ALT is greater than AST. AST is more elevated in alcohol hepatitis.
What is the significance of a positive IgM antibody in the diagnosis of hepatitis?
Hepatitis A, C, D, and E are diagnosed by acute IgM antibody. IgG antibody to hepatitis A, C, D, and E indicates old, resolved. Hepatitis C can be followed with a PCR-RNA viral load.
What is the first hepatitis B marker to elevate?
Hepatitis B is diagnosed as acute with the presence of the hepatitis B surface antigen, which is the first viral marker to elevate. The hepatitis B e antigen and the IgM core antibody also indicate an acute infection.
What is the significance of the hepatitis B e antigen?
The e antigen indicates high levels of viral replication and increased infectivity.
What laboratory findings indicate resolution of hepatitis B?
Resolution of the infection is definitively indicated by the loss of surface antigen activity and the development of hepatitis B surface antibody.
What is the significance of the hepatitis B core IgG antibody?
B core IgG and e antibody indicate that the acute infection is about to resolve and may be the only markers present in window period 2-6 weeks between loss of surface antigen and development of surface Ab.
What is the treatment for chronic hepatitis B?
Interferon, entecavir, adefovir, or lamivudine.
What is the treatment of chronic hepatitis C?
Chronic hepatitis C is treated with interferon combined with ribavirin. The only therapy for cirrhosis is liver transplantation.
What is the postexposure prophylaxis after hepatitis B needlestick?
If surface-antigen-positive patient, the person stuck should receive hepatitis B immunoglobulin (HBIg) and B vaccine. If the person stuck already has protective levels of surface antibody, then no therapy.
What is the postexposure prophylaxis for hepatitis C?
There is no effective postexposure prophylaxis to hepatitis C, and there is no vaccine?
What are the indications for hepatitis B vaccine?
All healthcare workers, IV drug users, and others at risk should be vaccinated. Newborn children are vaccinated.
What are the causes of urethritis?
Neisseria gonorrhoeae or nongonococcal urethritis is caused by either Chlamydia trachomatis (50%), Ureaplasma urealyticum (20%), Mycoplasma hominis (5%), Trichomonas (1%), or herpes simplex.
What are the clinical signs of urethritis?
Purulent urethral discharge; dysuria, urgency, and frequency in urination
How is urethritis diagnosed?
Urethral smear Gram-neg intracellular, coffee bean-shaped diplococci. Fluorescent antibodies for chlamydia by urethral swab, or by ligase chain reaction test of urine. Culture is the most specific test for gonorrhea.
What is the treatment of urethritis?
Combination of ceftriaxone IM and single-dose azithromycin PO. An alternative regimen is ceftriaxone and PO doxycycline for 7 d. Gonorrhea can also be treated with single-dose Cipro or cefixime.
What is pelvic inflammatory disease?
Infection of the fallopian tubes, uterus, ovaries, or ligaments of the uterus caused by N. gonorrhoeae, Chlamydia, Mycoplasma, anaerobic bacteria or Gram-negative bacteria. Intrauterine devices predispose to PID.
What are the clinical signs of pelvic inflammatory disease?
Pelvic pain on palpation of the cervix, uterus, or adnexa; fever, and leukocytosis. Cervical motion tenderness. Discharge from the cervix.
How is pelvic inflammatory disease diagnosed?
Cervical culture on Thayer-Martin for gonococcus. Laparoscopy. A pregnancy test should be done.
What is the therapy for pelvic inflammatory disease?
Doxycycline and cefoxitin (or cefotetan) IV. Outpatient therapy is single-dose ceftriaxone intramuscularly and doxycycline orally for 2 weeks. Outpatient therapy can also be 2 weeks ofloxacin and metronidazole.
What are the sequelae of pelvic inflammatory disease?
Infertility and ectopic pregnancy.
A 22-year-old man with painless, ulcerated genital lesion, adenopathy. What is the next step?
Dark-field microscopy for syphilis, VDRL.
What are the clinical signs of primary syphilis?
Chancre within 3 weeks; disappears 10-90 days; regional lymphadenopathy is painless, nontender. Chancres on penis, anus, rectum, vulva, cervix, perineum. Caused by the spirochete, Treponema pallidum.
What are the signs of secondary syphilis?
6-12 wks after infection, symmetrical rash more on flexor and volar (pinkish in whites; copper blacks). Lymphadenopathy; papules at mucocutaneous junctions and moist areas are condylomata lata (extremely infectious), alopecia.
What are the signs of latent syphilis?
Asymptomatic; persist for life, and one-third develop late or tertiary syphilis.
What are the signs of late or tertiary syphilis?
Most neurologic. Symptomatic but not contagious. Tertiary develops 3-20 y after initial infection, and the typical lesion is the gumma (chronic granulomatous reaction); any tissue or organ. Cardiovascular syphilis.
What is the Argyll Robertson pupil?
Small irregular pupil that accommodates but does not react to light.
What are the signs of tabes dorsalis?
Tabes dorsalis (locomotor ataxia) is pain, ataxia, sensory changes, and loss of tendon reflexes.
How is syphilis diagnosed?
Screening tests are VDRL and RPR; specific tests are FTA-ABS, MHA-TP, and Darkfield exam of chancre.
What are the causes of a false positive VDRL?
False positives VDRL occurs with EBV, collagen vascular disease, TB, subacute bacterial endocarditis.
What is the Jarisch-Herxheimer reaction?
Malaise, fever, headache, sweating rigors, and temporary exacerbations of the syphilitic lesions 6-12 hours after initial treatment.
What is the treatment of primary, secondary, and latent syphilis?
Im benzathine penicillin once a week. Primary and secondary syphilis require one week of therapy. Late latent syphilis is treated for three weeks and diagnosed when VDRL or RPR >1:8 without symptoms.
What is the treatment for tertiary syphilis?
Penicillin 10-20 million units/day IV for 10. Penicillin-allergics receive doxycycline for primary and secondary syphilis, but must be desensitized in tertiary syphilis. Pregnant must also undergo desensitization.
What is chancroid?
Acute, localized, contagious disease of painful genital ulcers and suppuration of the inguinal lymph nodes caused by Haemophilus ducreyi (Gram negative bacillus)
What are the clinical signs of chancroid?
Small, soft, painful papules that become shallow ulcers with ragged edges. Inguinal lymph nodes become very tender and enlarged.
How is chancroid diagnosed?
Gram stain shows Gram-negative bacillus, culture; PCR testing.
What is the treatment of chancroid?
Azithromycin single dose or ceftriaxone intramuscularly (single dose). Erythromycin for 7 days or Cipro for 3 days are alternatives.
What is lymphogranuloma venereum?
Contagious, sexually transmitted disease with primary lesion followed by suppurative lymphangitis caused by chlamydia trachomatis.
What are the clinical signs of lymphogranuloma venereum?
Small, transient, nonindurated, ulcerates, heals; tender, unilateral enlargement inguinal nodes; multiple draining sinuses (buboes); scar, sinuses; fever, malaise, joint pains, headaches.
How is lymphogranuloma venereum diagnosed?
Complement fixing antibodies; isolate chlamydia from pus in buboes
What is the treatment of lymphogranuloma venereum?
Doxycycline or erythromycin.
What is granuloma inguinale?
Chronic granulomatous condition, spread by sexual contact; caused by Donovania granulomatis, Calymmatobacterium granulomatis
What are the clinical signs of granuloma inguinale?
A painless, red nodule that develops into an elevated granulomatous mass. In males, on penis, scrotum, groin, thighs; in females on vulva, vagina, perineum. In homosexual males, the anus and buttocks. Scar formation.
How is granuloma inguinale diagnosed?
Giemsa or Wright stain (Donovan bodies) or smear of lesion; punch biopsy. Treatment with doxycycline ceftriaxone, TMP/SMZ, or erythromycin.
What virus causes genital herpes?
Herpes virus, type II; type I can be seen in genital herpes.
What are the clinical signs of genital herpes?
Vesicles skin or mucous membranes; become painful, circular ulcers, red areola. Soreness precedes lesions. Inguinal lymphadenopathy. Lesions on penis or labia, clitoris, perineum, vagina, cervix.
How is genital herpes diagnosed?
Tzanck test and viral culture.
What is the treatment of genital herpes?
Acyclovir, famciclovir, or valacyclovir. Herpes may recur; sexual contacts should be examined. Acyclovir-resistant herpes is treated with foscarnet.
What are genital warts?
Also known as condylomata acuminata; papilloma virus. Soft, moist, minute, pink, or red; grow rapidly and become pedunculated. Cauliflower appearance. Differentiate from flat warts and condylomata lata.
What is the treatment of genital warts?
Destruction (curettage, trichloroacetic acid), cryotherapy, podophyllin, imiquimod for 14 weeks (an immune stimulant), laser.
What conditions predispose to cystitis?
Predisposed by tumors, stones, strictures, prostatic hypertrophy, neurogenic bladder. Sexual intercourse in women. Catheters.
What is the etiology of cystitis?
E. coli in >80%; second are other coliforms (Gram-negative bacilli) such as Proteus, Klebsiella, Enterobacter; enterococci occasionally; Staph saprophyticus in young women.
What are the symptoms of cystitis?
Dysuria, frequency, urgency, and suprapubic pain. Hematuria, low-grade fever; foul-smelling and cloudy urine. Suprapubic tenderness without flank tenderness.
How is cystitis diagnosed?
Urinalysis for WBCs, RBCs, protein, bacteria. Nitrites indicate Gram-negative. Urine culture with >100,000 colonies/mL of urine is confirmatory but not always necessary.
What is the treatment of cystitis?
Uncomplicated cystitis: 3 days of trimethoprim/sulfamethoxazole, or any quinolone. Seven days of therapy for cystitis in diabetes. Quinolones should not be used in pregnancy.
What are the predisposing factors for pyelonephritis?
Strictures, tumors, calculi, prostatic hypertrophy, or neurogenic bladder, vesicoureteral reflux; more common in women, childhood, pregnancy, after urethral catheterization or instrumentation
What are the clinical signs of pyelonephritis?
Chills, fever, flank pain, nausea, vomiting, costovertebral angle tenderness, increased frequency in urination, and dysuria.
How is pyelonephritis diagnosed?
Clean-catch urine for urinalysis, culture; >100,000 bacteria/mL of urine. If the patient does not improve in 48-72 hours, ultrasound or CT scan can be done to detect obstruction renal or perinephric abscess.
What is the treatment of pyelonephritis?
Antibiotics for 10-14 days with fluoroquinolone, or ampicillin and gentamicin, or a third-generation cephalosporin. Most treated as outpatients. Resistance to TMP/SMZ 20%.
What is the bacteriology of pyelonephritis?
E. coli is the most common pathogen, others include: Klebsiella, Proteus, and Enterococcus. Immunosuppressed and indwelling catheters are more prone to Candida.
What is perinephric abscess?
Infected material surrounding kidney and within the Gerota fascia; from pyelonephritis. Stones in 20-60%. Other structural abnormalities, recent surgery, trauma, diabetes.
What is the microbiology of perinephritic abscess?
E. coli most common, then Klebsiella, Proteus; Staph aureus sometimes accounts for hematogenous cases.
How is perinephritic abscess diagnosed?
Urinalysis normal 30% and culture normal 40%. Fever, pyuria with a negative culture or a polymicrobial culture. Ultrasound; CT or MRI scan are better tests. Aspiration of the abscess.
What is the treatment of perinephric abscess?
Third-generation cephalosporins, antipseudomonal penicillin, or ticarcillin/clavulanate with an aminoglycoside percutaneous drainage
What is impetigo?
Superficial, pustular skin infection in children (ecthyma is ulcerative form of impetigo), oozing, crusting, draining; S. aureus (bullous impetigo) and group A beta-hemolytic Streptococcus
What are the clinical signs of impetigo?
Arms, legs, and face after skin trauma. Maculopapules, rapidly progresses to vesicular pustular lesions or bullae. Progress to lymphangitis, furunculosis, or cellulitis, and acute glomerulonephritis.
What is the treatment of impetigo?
Oral first-generation cephalosporin or semisynthetic penicillin (oxacillin, cloxacillin, dicloxacillin). Topical mupirocin or bacitracin. Penicillin-allergic: clarithromycin or azithromycin.
What is erysipelas?
Superficial cellulitis caused by group A beta-hemolytic Streptococcus.
What are the clinical signs of erysipelas?
Bilateral, shiny, red, indurated, edematous, tender lesions on the face, arms, and legs. Lesions are sharply demarcated.
What is the treatment of erysipelas?
Semisynthetic penicillin or first-generation cephalosporin; penicillin.
What are dermatophyte infections?
Superficial infection caused by a dermatophyte fungus that invades outer layer of skin nails, hair, stratum corneum; caused by microsporum, Trichophyton, and Epidermophyton
How are dermatophyte infections diagnosed?
Potassium hydroxide (10%) prep; culture.
What are the signs of tinea corporis?
Papulosquamous annular lesions of the body with a raised border, which expands peripherally and clear centrally.
What are the signs of tinea pedis?
Macerated and scaling borders (third and fourth interdigital space) of the foot.
What are the signs of tinea unguium?
Thickened and lusterless nails.
What are the signs of tinea capitis?
Small, scaly, semi-bald, grayish patches with broken lusterless hair on head
What are the signs of tinea cruris?
Ringed lesion that extends from the crural folds over the adjacent inner thigh
What are the signs of tinea barbae?
Inflammatory, deep plaques and noninflammatory patches on the bearded area of the face.
What is the treatment for dermatophyte infections?
Terbinafine or itraconazole orally for capitis, corporis, and unguium. Miconazole, clotrimazole and ketoconazole in cream or lotion for mild cruris, pedis, and corporis.
What is candidiasis?
Infection of skin, mucous membranes by Candida albicans. Immunodeficiency antibacterial therapy, obesity, diabetes, corticosteroid, antimetabolite, pregnancy, debilitate, blood dyscrasias, HIV.
What are the clinical signs of candidiasis?
Intertriginous infection: Well-demarcated, erythematous, itchy, exudative patches, rimmed with small red-based pustules in the groin, gluteal folds (diaper rash), axilla, umbilicus, and inframammary areas
What are the signs of candida vulvovaginitis?
White discharge with inflammation of the vaginal and vulva. Pregnancy and diabetes mellitus.
What are the signs of oral candidiasis (thrush)?
White patches of exudates on tongue or buccal mucosa.
What is candidal paronychia?
Painful red swelling around the nail.
How is candidiasis diagnosed?
Potassium hydroxide exam. Culture is definitive.
What is the treatment of candidiasis?
Topical nystatin, clotrimazole, miconazole, ciclopirox, econazole, terconazole. Amphotericin for invasive infection. Fluconazole for less serious infections and paronychia.
What is tinea versicolor?
Multiple macules (usually asymptomatic), varying in color from white to brown caused by Pityrosporum orbiculare (Malassezia furfur)
What are the clinical signs of tinea versicolor?
Tan, brown, white, scaling macular lesions that tend to coalesce; found on chest, neck, abdomen, or face. Lesions do not tan.
How is tinea versicolor diagnosed?
Skin scrapings with 10% KOH. "spaghetti and meatballs," appearance of hyphae and spores.
What is the treatment of tinea versicolor??
Topical selenium sulfide, clotrimazole, ketoconazole, or oral itraconazole.
What are the signs of scabies?
Skin infection with superficial burrows, intense pruritus, and secondary infections, caused by Sarcoptes scabiei; skin-to-skin transmission.
What are the clinical signs of scabies?
Pruritus, burrows, papules on flexor surfaces of wrists, finger webs, elbows, axilla, areola of breast and genitals. Immunocompromised may develop severe, crusted Norwegian scabies.
How is scabies diagnosed?
Parasite in scrapings in mineral oil.
What is the treatment of scabies?
Permethrin, lindane (Kwell). Ivermectin oral therapy if the disease is extensive. Lindane in contraindicated in pregnancy.
What is pediculosis?
Skin infestation caused by lice. Itching, excoriations, erythematous macules and papules; secondary bacterial infection
How is pediculosis diagnosed?
Examination of the pubic area, axillae, scalp, and other hair-bearing surfaces for the louse or nits.
What is the treatment for pediculosis?
Permethrin or lindane (Kwell).
What is molluscum contagiosum?
Skin-colored, waxy, papules caused by poxvirus. Small papules anywhere on the skin (genital and pubic area). Transmitted by skin-to-skin contact or sexually. Seen in children; increased with HIV.
What is the treatment of molluscum contagiosum?
Freezing, curettage, electrocautery, or cantharidin.
61-year-old man with painful leg for 2 weeks. Ulcer over the proximal portion of his tibia just below the knee. Peripheral vascular disease and diabetes. Sinus tract in the ulcer draining purulent material. What is the diagnosis?
Osteomyelitis
What is acute hematogenous osteomyelitis?
S aureus. Long bones of lower extremities children; tibia, femur, metaphyseal. In adults, hematogenous osteomyelitis accounts for 20%, lumbar vertebrae. Fever, back tenderness. Injection drugs.
What is the most common organism in osteomyelitis that is caused by spread of a contiguous infection?
A contiguous infection after recent trauma or placement of a prosthetic joint may cause osteomyelitis. S. aureus is the most common organism; sometimes polymicrobial.
What is vascular insufficiency osteomyelitis?
Majority over 50, with diabetes or peripheral vascular disease. Bones of the lower extremities. Polymicrobial, but most common organism is S. aureus.
What is the presentation of osteomyelitis?
Pain, erythema, swelling, and tenderness. Nearby ulceration or wound. Draining sinus tract.
How is osteomyelitis diagnosed?
Technetium bone scan and MRI. Plain x-ray: Periosteal elevation. Bone biopsy and culture is the best test.
What is the treatment of osteomyelitis?
Children treated with antibiotics; osteomyelitis in adults requires surgery and nafcillin or vancomycin (MRSA) plus an aminoglycoside or a 3rd gen cephalosporin for 12 weeks.
70-year-old woman with a swollen right knee; effusion and decreased mobility. Redness and tenderness. What is the next step?
Arthrocentesis.
What are the causes of septic arthritis?
Neisseria gonorrhoeae, staphylococci or streptococci; Rickettsia, viruses, spirochetes. Bacterial arthritis is divided into gonococcal and nongonococcal types?
What is the pathogenesis of gonococcal septic arthritis?
Sexual activity is risk factor for gonococcal septic arthritis. 1-5% of people with gonorrhea will develop disseminated disease.
What is the pathogenesis of nongonococcal bacterial arthritis?
Hematogenous spread. Bites, surgery, trauma, spread from surrounding infection. Normal or damaged joints can be infected after rheumatoid arthritis, osteoarthritis, surgery, prosthesis, gout; sickle, IVDU, diabetes, HIV.
What is the microbiology of nongonococcal arthritis?
Gram-positive (>85); (S. aureus [60%], Streptococcus [ 15%], Pneumococcus [5%]), Gram-negative (10-15%), polymicrobial (5%).
What is the presentation of nongonococcal septic arthritis?
Monoarticular in >85%, with a swollen, tender, erythematous joint with decreased range of motion. Knee most common. Petechiae or purpura?
How is nongonococcal septic arthritis diagnosed?
Nongonococcal. Culture of joint aspirate fluid and Gram stain. The cell count of the synovial fluid is high (>50,000) and is predominantly PMNs with a low glucose. Blood culture is positive in 50%.
How is gonococcal septic arthritis diagnosed?
Only 50% of aspirates have positive culture. Less than 10% of blood cultures are positive. Cervix, pharynx, rectum, and urethra may be positive.
What is the treatment for nongonococcal septic arthritis?
Good empiric coverage is nafcillin or oxacillin (or vancomycin if MRSA is suspected) combined with an aminoglycoside or a third-generation cephalosporin.
What is the treatment for gonococcal septic arthritis?
Ceftriaxone is the drug of choice.
What is gas gangrene (clostridial myonecrosis)?
Necrotizing infection of muscle by gas-producing organisms with sepsis. 80% caused by the spread of infection from C perfringens after traumatic wound (50%). Postoperative (30%), nontraumatic (20%).
What are the signs and symptoms of clostridial myonecrosis?
<1-4 days of incubation after the wound, then pain, swelling, and edema. Hypotension, tachycardia, and fever. Crepitation, renal failure, death.
How is clostridial myonecrosis diagnosed?
Gram-positive rods, but no white cells. Culture positive for C. perfringens.
What is the treatment for clostridial myonecrosis?
High-dose penicillin or clindamycin; surgical debridement or amputation. Hyperbaric oxygen 38-year-old man with fever, intravenous drug use, systolic murmur at the lower left sternal border?
What is the next step?
Blood cultures for infective endocarditis.
Which heart valves are most frequently affected by endocarditis?
Left-sided lesions of the aortic and mitral valves are the most common.
What procedures are associated with infective endocarditis?
Invasive and dental procedures. Oral and upper respiratory tract surgery. GI procedures. GU surgery. Alimentation catheters. Pressure-monitoring catheters. IV drug use.
What is the most common cause of acute infective endocarditis?
S. aureus is the most common cause. Seed previously normal valves. IV drug use is the major risk factor.
What are the signs of acute infective endocarditis?
Rapid onset of fever and sepsis. Splenomegaly of invasion of myocardium and rapid valve destruction. Embolic complications.
What is the most common cause of subacute infective endocarditis?
4-5% have negative blood cultures. Viridans group streptococci is the most common organism; low virulence. Seed abnormal valves.
Which cardiac conditions are risk factors for infective endocarditis?
1) Ventricular septal defect, 2) stenosis, 3) prosthetic valves, 4) indwelling catheters, 5) bicuspid aortic valve, 6) mitral valve prolapse, and 7) Marfan syndrome
What are the clinical signs of subacute infective endocarditis?
Slow onset with vague symptoms of malaise, low-grade fever, weight loss; and survival 80-90% with treatment.
What are the complications of infective endocarditis?
CHF (most common cause of death). Septic infarctions and metastatic infections to brain; spleen; kidneys; coronary arteries. Glomerulonephritis with nephrotic syndrome or renal failure (immune complex).
How is infective endocarditis diagnosed?
Positive blood cultures; transesophageal echo is >90% sensitive.
What is the treatment of infective endocarditis?
Empiric therapy with antistaphylococcal drug (nafcillin), and streptococcal drug (ampicillin and gentamicin). Vancomycin and gentamicin are empiric treatment if MRSA suspected.
What is the prophylactic treatment of bacterial endocarditis?
Dental procedures: Amoxicillin. For penicillin-allergic patients: clindamycin or (azithromycin, or clarithromycin or cephalexin). Urinary or gastrointestinal procedures: no prophylaxis is required.
What cardiac conditions require prophylactic antibiotics?
Prosthetic cardiac valves. Previous bacterial endocarditis, even in the absence of heart disease. Congenital cardiac malformations.
What cardiac conditions do not require prophylactic therapy?
Rheumatic and other acquired valvular disease. Hypertrophic cardiomyopathy; mitral valve prolapse with valvular regurgitation; surgically repaired shunts or intracardiac defects.
What are the causes of pericarditis?
Viruses/bacteria/tuberculosis/fungal/spirochetes; viral is most common (Coxsackie, ECHO). Acute HIV, SLE; RA, scleroderma. Uremia; neoplasia adjacent heart; hypothyroidism; irradiation; rheumatic fever, trauma, Dressler.
What is the clinical presentation of pericarditis?
Sharp, pleuritic, chest pain that improves when sitting up and leaning forward. Pericardial rub. Best at the apex with the patient sitting up. Low fever.
What is pulsus paradoxus?
Tamponade causes 10-mm drop in BP with inspiration. Blood is drawn into the right heart. Heart is compressed by fluid around it, and the septum bulges into left. Distended neck veins, tachycardia, hypotension also.
What are the ECG signs of pericarditis?
ST segment elevation in almost every lead, except aVR. The ST elevation progresses to T-wave inversion. PR depression may occur. Electrical alternans. Echocardiogram demonstrates effusion.
What is the treatment for viral pericarditis?
Indomethacin, ibuprofen, or Naprosyn. Prednisone if there is no response, or for tuberculosis with antitubercular medications. Pericardiocentesis and pericardial window for large effusion tamponade.
What is myocarditis?
Infection or inflammation of heart muscle by viruses (Coxsackie B). Other causes: bacteria, Rickettsia, fungi, parasites (Chagas, toxoplasmosis), Lyme. Radiation, drugs, collagen-vascular, hyperthyroidism.
What are the signs of myocarditis?
Myocardial dysfunction, dyspnea and fatigue. Asymptomatic, subclinical infection or a rapid progression to chest pain, arrhythmia, and death. S-3 gallop and murmurs.
How is myocarditis diagnosed?
Nonspecific ST-T changes are most common. Heart block. CK-MB, LDH, troponin elevated. LV systolic dysfunction. Viruses from stool, saliva, nasopharynx. Ab to viruses. Biopsy rarely done.
What is the vector for Lyme disease?
Lyme disease is spread by the bite of the Ixodes scapularis tick.
What is the clinical presentation of lyme disease?
Rash 3-30 d after tick bite. 80% develop erythema migrans rash at bite (red patch, central clearing, "bull's-eye"). Rash resolves in several weeks. Flulike illness with fever, chills, myalgias.
What are the neurologic symptoms of lyme disease?
Neurologic symptoms in 10-20% of patients, most commonly paralysis of the seventh cranial nerve (facial paralysis), which may be bilateral. Meningitis, encephalitis, headache, and memory disturbance may develop.
What are the cardiovascular signs of lyme disease?
AV heart block, myocarditis, pericarditis, and various arrhythmias. Joint involvement may develop months to years later in up to 60%, most commonly a migratory polyarthritis.
What is the diagnostic criteria of Lyme disease?
Erythema migrans with 1 late manifestation and lab confirmation. Treat based on rash. Serology by ELISA and western blot does not distinguish current and previous infection. Early test is often negative.
What is the treatment of Lyme disease?
The rash, the facial palsy, and joint pain can be treated with oral doxycycline or amoxicillin. Heart block, meningitis, myocarditis, or encephalitis are treated with intravenous ceftriaxone.
Which cell is the primary target of HIV?
The primary mechanism of HIV is to infect a subset of T lymphocytes called CD4 cells, often just referred to as T cells.
What are the symptoms of pneumocystis jiroveci pneumonia?
CD4 count <200. Pneumonia; dyspnea on exertion, dry cough, fever, chest pain; usually subacute onset and progression.
What is the diagnostic test for PJP?
Bronchoscopy with bronchoalveolar lavage for direct identification of the organism. Chest x-ray reveals bilateral, interstitial infiltrates. PJP may occur with a normal chest x-ray. Serum LDH is usually elevated.
What is the treatment of pneumocystis jiroveci pneumonia?
Trimethoprim-sulfamethoxazole (TMP-SMZ) IV is the first-line therapy for mild-severe disease and may cause a rash.
What is the alternative treatments for PJP if TMP-SMZ is not tolerated?
For mild-moderate disease: dapsone and trimethoprim, or primaquine and clindamycin; or atovaquone, or trimetrexate (with leucovorin).
What are the adverse effects of pentamidine?
Pancreatitis, hyperglycemia, hypoglycemia.
What are the indications for steroids in PJP?
Steroids are used as adjunctive therapy for any patient with severe pneumonia. Severe is defined with a PaO2 of <70 mm Hg or an A-a gradient of >35 mm Hg.
How is cytomegalovirus diagnosed?
Funduscopy; colonoscopy with biopsy for diarrhea; or upper gastrointestinal endoscopy with biopsy of ulcers.
What is the treatment of cytomegalovirus retinitis and esophagitis?
Valganciclovir, an oral prodrug of ganciclovir; achieves levels comparable to IV ganciclovir. IV ganciclovir is reserved for serious CNS infections; foscarnet and cidofovir for ganciclovir resistance.
What are the side effects of ganciclovir?
Ganciclovir may cause neutropenia.
What is the prophylactic therapy for cytomegalovirus retinitis?
Valganciclovir can be used for maintenance therapy.
What is Mycobacterium avium complex?
Occurs in HIV infected with CD4 <50/mm. Atypical mycobacteria found in the environment; mode of infection is inhalation or ingestion. Fevers, night sweats, bacteremia, wasting, anemia, diarrhea.
What is the testing for Mycobacterium avium complex?
Blood culture, culture of bone marrow, liver, or other body tissue or fluid.
What is the treatment of Mycobacterium avium complex?
Clarithromycin, ethambutol, rifabutin. Prophylaxis is azithromycin once a week or clarithromycin bid; rifabutin is alternative. Prophylaxis discontinued if antiretrovirals raise CD4 >100 for several months.
At what level of CD4 cell count does toxoplasmosis occur?
Occurs in HIV infected patients with CD4 <100/mcL.
What are the signs of cerebral toxoplasmosis?
Brain mass lesion: Headache, confusion, seizures, and focal neurologic deficits
What tests are used to detect toxoplasmosis?
CT of head showing a "ring" (contrast enhancing) with edema and mass effect. Trial of anti-toxoplasmosis therapy is given for 2 weeks, and scan repeated. Shrinkage of lesion is diagnostic. Toxoplasmosis serology, CSF PCR.
What is the treatment of toxoplasmosis?
Pyrimethamine and sulfadiazine. Clindamycin can be substituted for sulfadiazine. Leucovorin prevents marrow suppression. Prophylaxis with TMP/SMZ or dapsone/pyrimethamine.
What is cryptococcosis?
Occurs in HIV infected persons with CD4 <100/mcL. Meningitis; fever, headache, and malaise
How is cryptococcosis diagnosed?
Lumbar puncture with India ink and cryptococcal antigen testing. Serum cryptococcal antigen testing.
What is the treatment of cryptococcosis?
Amphotericin intravenously for 10-14 days (with flucytosine), followed by fluconazole orally for suppressive therapy.
What is the normal CD4 cell count?
700-1,500/mcL.
What CD4 cell count is associated with oral thrush, Kaposi sarcoma, tuberculosis, zoster, lymphoma?
200-500/mcL
What CD4 cell count is associated with pneumocystis jiroveci pneumonia, dementia, progressive multifocal leukoencephalopathy, disseminated histoplasmosis and coccidiomycosis?
100-200/mcL
What opportunistic infections occur when the CD4 count is <100/mcL?
Toxoplasmosis, Cryptococcus, cryptosporidiosis, disseminated herpes simplex
What opportunistic infections occur when the CD4 count is <50/mcL?
Cytomegalovirus, Mycobacterium avium complex. Progressive, multifocal leukoencephalopathy, CNS lymphoma.
What test indicates an adequate response to antiretroviral therapy in HIV-infected persons?
The goal is complete suppression of viremia with <50 to 70 copies of HIV-RNA/mcL.
What are the indications for viral sensitivity testing?
Patient failing a combination of medications and a change in therapy is necessary. Any pregnant woman who has not been fully suppressed on the initial combination of medications.
What are the side effects of zidovudine?
Leukopenia, anemia, gastrointestinal
What are the side effects of didanosine (DDI)?
Pancreatitis, peripheral neuropathy.
What are the side effects of stavudine (D4T)?
Peripheral neuropathy
What are the side effects of lamivudine (3TC)?
No side effects have been reported in additional to placebo.
What are the side effects of emtricitabine?
Structurally related to lamivudine; few side effects.
What is the structure of tenofovir?
Tenofovir is a nucleotide analog as compared to the others that are nucleoside analogs.
What are the side effects of abacavir?
Hypersensitivity reaction in the first 6 weeks of therapy. Rash, fever, nausea/vomiting, muscle and joint aches, dyspnea. The drug should be stopped and never restarted.
What are the side effects of zalcitabine (DDC)?
Pancreatitis, peripheral neuropathy, lactic acidosis
What are the nucleoside reverse transcriptase inhibitors?
Zidovudine (ZDV or AZT), didanosine (DDI), stavudine (D4T), lamivudine (3TC), emtricitabine, abacavir, zalcitabine (DDC).
What are the side effects of protease inhibitors?
Hyperlipidemia, hyperglycemia, elevated liver enzymes; fat loss (lipoatrophy) from the face and extremities with redistribution of fat to the back of the neck and abdominal viscera.
What are the side effects of nelfinavir?
Nausea, diarrhea.
What are the side effects of indinavir?
Nephrolithiasis (4%), hyperbilirubinemia (10%).
What are the side effects of ritonavir?
Severe nausea, diarrhea.
What are the side effects of saquinavir?
Gastrointestinal effects.
What are the side effects of amprenavir?
Rash, headache, diarrhea, and nausea.
What are the side effects of the lopinavir/ritonavir combination?
Diarrhea
What are the side effects of atazanavir?
Diarrhea, asymptomatic hyperbilirubinemia.
What are the names of the protease inhibitors?
Nelfinavir, indinavir, ritonavir, saquinavir, amprenavir, atazanavir.
What are the names of non-nucleoside reverse transcriptase inhibitors?
Efavirenz, nevirapine, delavirdine.
What are the side effects of efavirenz?
Neurologic; somnolence, confusion, or psychiatric disturbance; non-nucleoside reverse transcriptase inhibitor.
What are the side effects of nevirapine?
Rash, hepatotoxicity.
What are the side effects of delavirdine?
Rash.
When should antiretroviral therapy be started in HIV infected patients?
Antiretroviral medication should be started when the CD4 count has dropped or the viral load has risen.>55,000 copies/mL of RNA.
What antiretrovirals should be started in HIV infected patients?
Use two nucleosides combined with a protease inhibitor or, Use two nucleosides combined with efavirenz or, Use two nucleosides combined with two protease inhibitors.
What is highly active antiretroviral therapy?
Uses medications with activity at different sites; 2 NRTIs (tenofovir/emtricitabine or ZDV/lamivudine) combined with NNRTI (efavirenz) or 2 PI (atazanavir/ritonavir, fosamprenavir/ritonavir, lopinavir/ritonavir).
What is "boosted protease inhibitor therapy"?
Practice of giving protease inhibitors in combination with a low dose of ritonavir (a PI). Ritonavir when given in a low dose with other PIs, decreases metabolism of the second PI and increases drug level.
What is considered adequate antiretroviral therapy?
Any regimen that increases the CD4 count and reduces the viral load to undetectable amounts or close to undetectable amounts. Drop of at least 50% of viral load in the first month indicates adequate therapy.
What percentage of children born to HIV positive mothers will be infected with HIV?
Without treatment, 30% of children born to HIV-positive mothers will be infected. All children at birth will carry the maternal antibody by ELISA testing.
What treatment should be given to pregnant HIV infected women?
Women with low CD4 or high viral load should receive triple antiretrovirals. ZDV, a second nucleoside and a protease inhibitor should begin in pregnant women at 14 weeks if not already on therapy.
What are the indications for cesarean section in pregnant HIV-infected women?
C-section is used routinely in those whose CD4 count is low or whose viral load >1000 copies.
Which antiretroviral in a known teratogen?
The only known teratogen is efavirenz in animal studies.
What treatment is recommended for HIV infected pregnant women?
If already on antiretrovirals, continue. If low CD4 or opportunistic infection, start immediately. If high CD4, combination therapy can begin at 14 weeks. Can deliver vaginally if the viral load <1000.
What is the effect of breastfeeding on transmission of HIV to the infant?
Breast feeding is associated with transmission of virus to the infant.
What postexposure prophylaxis for HIV after needlestick injury should be given?
Persons with serious exposure to blood of HIV-pos should receive ZDV, lamivudine, nelfinavir (or any other fully suppressive 3-drug combo) for 4 wks. ZDV alone will decrease the risk of transmission by 80%.
What is Q-Fever?
Coxiella burnetii infection, which is transmitted by inhalation; found in cattle, sheep, and goats.
What are the clinical signs of Q-fever?
Febrile illness, atypical pneumonia, hepatitis, and hepatomegaly, endocarditis. Treatment is doxycycline. Diagnosed by specific serology.
What is Rocky Mountain spotted fever?
R. rickettsi is transmitted by the wood tick from the mid-Atlantic coast, upper South, and Midwest. In spring and summer.
What is the triad of Rocky Mountain spotted fever?
Abrupt fever, headache and erythematous maculopapules. Rash starts wrist, ankles and spreads centripetally, palms and soles. Confusion, lethargy, dizziness, irritability, stiff neck, GI symptoms.
How is Rocky Mountain spotted fever diagnosed?
Specific serology; biopsy of skin lesion.
What is the treatment of Rocky Mountain fever?
Doxycycline
What is toxoplasmosis?
Intracellular parasite. 50% of the US population is antibody positive. Prevalence increases with age. Undercooked meat, especially pork and lamb. Also from domestic cat feces; transplacental infection.
What is the presentation of toxoplasmosis?
80-90% asymptomatic in the immunocompetent. In AIDS: chorioretinitis, CNS mass lesion, encephalitis. Headache, fever, nausea. The retinal lesion can cause visual complaints.
How is toxoplasmosis diagnosed?
Serology is the most common method used. The best test is to visualize the parasite in biopsy tissue.
What is the treatment of toxoplasmosis?
Pyrimethamine and sulfadiazine.
What is tetanus?
Infectious complication of wounds caused by the toxin of Clostridium tetani; takes 1-7 days to develop; spore forming, Gram-positive rod.
What are the clinical signs tetanus infection?
Tonic spasms of voluntary muscles, respiratory arrest, difficulty in swallowing (dysphagia), restlessness, irritability, stiff neck, arms, and legs; headache, lockjaw, high mortality.
What is the prophylaxis of tetanus?
Tetanus toxoid (boosters every 10 years). Débride wound. Antitoxin is tetanus immunoglobulin; penicillin 10-14 days.
What is aspergillosis?
Fungus widespread in environment; pulmonary disease in immunocompromised. 90 species, with A. fumigatus the most common in decaying organic matter, ceiling tile, ventilation systems.
What are the signs of aspergillosis?
Asthma with cough, fever, wheezing. Mycetoma, a "fungal ball": in a pre-existing cavity, with hemoptysis. Invasive pulmonary. Disseminates to any organ from lung.
What are the risk factors for aspergillosis?
Risks: 1) neutropenia <500, 2) steroids 3) cytotoxic drugs (e.g., azathioprine, cyclophosphamide).
How is aspergillosis diagnosed?
Abnormal chest x-ray and Aspergillus in sputum.
What are voriconazole and caspofungin?
Drugs used to treat aspergillosis and other fungal infections.
What is the treatment of aspergillosis?
Invasive disease is treated with voriconazole, a broad-spectrum azole. Itraconazole for very mild disease. Caspofungin is superior to amphotericin. Caspofungin is an echinocandin.
What is blastomycosis?
A fungus that is strongly associated with rotting organic material and soil. Occurs in southeast and central US. It has a 10:1 male:female ratio. Begins with inhalation of decaying wood and vegetation.
What are the signs of blastomycosis?
Fever, cough, chest pain, and weight loss. After inhalation, disseminates anywhere in body, but skin is by far the most common.
What is the treatment of blastomycosis?
Prolonged amphotericin for severe disease (8-12 weeks); itraconazole or ketoconazole for mild disease.
What is the cause of toxic shock syndrome?
Staphylococcus aureus toxin TSST-1. Infected tampons, sponges, and surgical wound infections.
What are the clinical signs of toxic shock syndrome?
Hypotension, fever, mucosal changes, desquamative rash on hands and feet. Gastrointestinal, renal, hepatic, and muscular symptoms.
What is the treatment of toxic shock syndrome?
Beta-lactamase stable penicillin (nafcillin or oxacillin). Restoration of hypovolemic shock, remove infected tissue, prosthesis, tampons, diaphragm.
What is acute renal failure?
Rapid rise in blood urea nitrogen or creatinine over a period of several hours to days. Decrease in glomerular filtration rate.
What is renal insufficiency?
Renal insufficiency is renal failure, but not to the point of needing dialysis. The term azotemia can be used interchangeably with the term renal insufficiency.
What is uremia?
Very severe renal failure requiring dialysis. Severe acidosis, mental status changes, hyper, fluid overload, anemia, hypocalcemia, pericarditis. Bleeding. Uremia is same as "end stage renal disease.?
What is blood urea nitrogen?
BUN becomes abnormally elevated in all forms of renal failure. BUN rises after a protein meal or from gastrointestinal bleeding. BUN is derived from protein waste products.
What is creatinine?
Creatinine is the closest approximation of glomerular filtration rate. Creatinine is a metabolic product of skeletal muscle. Creatinine can be falsely low just because of a decrease in muscle mass.
What is prerenal azotemia?
Renal insufficiency caused by diminished perfusion, hypovolemia (dehydration, burns, diuretic, vomiting, diarrhea, hemorrhage), hypotension (septic, cardiogenic, anaphylactic), third spacing in peritonitis.
What is the laboratory criteria for prerenal azotemia?
BUN:creatinine ratio of 20:1. There is also a low urine sodium and low fractional excretion of sodium (FeNa <1%). High urine osmolality with a high specific gravity (>1.010) and a high urine osmolality (>500).
What are the signs of renal artery stenosis?
High BUN and creatinine with a high BUN:creatinine ratio. HTN, prerenal azotemia. ACE inhibitors will markedly diminish renal perfusion in renal artery stenosis because of the high aldosterone.
What is hepatorenal syndrome?
Renal failure caused by hepatic failure secondary to intense vasoconstriction of afferent arteriole, resulting in decreased renal perfusion.
What are the laboratory abnormalities in hepatorenal syndrome?
Prerenal azotemia with a high BUN:creatinine ratio >20:1. Urine Na <10 and a fractional excretion of sodium <1%. No improvement in renal failure after 1.5 L saline is diagnostic of hepatorenal syndrome.
What is the effect of ACE inhibitors on the kidney?
Renal failure can be caused by vasodilation of the efferent arteriole. In elderly, diabetic, hypertensive, or renal disease such as from myeloma, an ACEI can result in a decreased renal function. Rise in BUN and creatinine.
What is the hepatopulmonary syndrome?
Pulmonary failure is caused by hepatic disease. The oxygen saturation drops when the patient sits up and there is an increase in the A-a gradient.
What is postrenal azotemia?
Bilateral blockage in outflow of urine. Small stone or clot in bladder. Bladder CA, prostate hypertrophy or CA, retroperitoneal fibrosis, neurogenic bladder. Creatinine will only begin to rise when 80% of renal function lost.
What are the laboratory abnormalities in postrenal azotemia?
BUN and creatinine will elevate in a ratio of 20:1 as with prerenal azotemia. Low fractional excretion of sodium and low urine sodium.
What are the consequences of prolonged postrenal obstruction?
Permanent damage occurs, and the kidney tubule cells die, then the BUN:creatinine ratio will lower to 10:1, such as that seen in acute tubular necrosis. Recovery is possible until 10 days of obstruction.
What are the clinical findings in postrenal azotemia?
Distended bladder, bilateral hydronephrosis on renal sonogram or CT, or large volumes of urine in bladder after passing Foley catheter. After voiding, there should be no more than 50 mL of residual.
What is acute tubular necrosis?
85% of acute renal failure is caused by intrinsic renal disease, such as ATN caused by either hypoperfusion of kidney or toxic injury (aminoglycoside). ATN is most common cause of ARF in hospitalized.
What are the urine microscopic findings in acute tubular necrosis?
Severe ischemia causes the tubular cells to necrose and slough off into the urine and become visible as granular, muddy brown, or pigmented casts. At the point of necrosis, the renal insufficiency can be permanent.
How is acute tubular necrosis diagnosed?
BUN:creatinine ratio 10:1. High urine sodium (>40), high fractional excretion of sodium (>1%), and low urine osm (<350). If tubular cells die of ischemia, then kidney can neither concentrate nor dilute the urine.
What is the treatment of acute tubular necrosis?
Underlying cause must be corrected. Hydration is given to exclude possibility of prerenal component. Furosemide does not reverse ATN. Dopamine at low doses does not increase renal perfusion. Dialysis.
What is allergic interstitial nephritis?
Allergic interstitial nephritis causes 10-15% of intrinsic renal; failure fever and rash, eosinophils in the urine. AIN is usually caused by an adverse effect of medications. Infections can also cause AIN.
What medications can cause allergic interstitial nephritis?
Penicillin, cephalosporins, sulfas, allopurinol, rifampin, quinolones. Calcium blockers. Sulfa drugs include thiazides, furosemide, acetazolamide.
What infections can cause allergic interstitial nephritis?
AIN is also caused by infections by viruses, bacteria, and fungi. Leptospirosis, legionella, CMV, rickettsia, and streptococci.
What autoimmune disorders can cause allergic interstitial nephritis?
Systemic lupus erythematosus (SLE), Sjögren syndrome, sarcoidosis, and cryoglobulins.
What laboratory abnormalities are associated with allergic interstitial nephritis?
Eosinophilia, eosinophiluria, hematuria, proteinuria, increased IgE. Best initial test is a urinalysis for white cells. Urine eosinophils on Wright stain. Kidney biopsy not necessary.
What is the treatment of allergic intestinal nephritis?
AIN resolves spontaneously after stopping the offending agent. Short course of steroids may be administered.
A 25-year-old man was exercising vigorously became very weak with painful muscles and dark urine. What is the most important test to do first?
Electrocardiogram
What are the causes of rhabdomyolysis?
Rhabdomyolysis is caused by sudden, severe crush injury, seizures, severe exertion, hypokalemia, hypophosphatemia, and medications such as statins.
What are the laboratory signs of rhabdomyolysis?
The most important test after a severe crush injury or seizure and rhabdomyolysis is an ECG or potassium level. UA is positive for blood but with no red cells because myoglobin reacts with the dipstick.
What action should be taken if peaked T-waves are present on an ECG?
Acidosis and hyperkalemia can cause arrhythmia. If there are peaked T-waves, an amp of calcium gluconate should be given.
How is rhabdomyolysis confirmed in the laboratory?
CPK skeletal muscle necrosis. Metabolic acidosis with a decreased serum bicarbonate, hyperphosphatemia, hypocalcemia. Severe hyperuricemia. Rapidly rising creatinine.
What is the treatment for rhabdomyolysis?
If there are ECG abnormalities from the hyperkalemia, therapy is calcium chloride or gluconate. Hydration, mannitol. Alkalinize urine with bicarbonate to prevent precipitation of pigment in tubule.
What is the most common cause of hyperoxaluria resulting in acute renal failure?
Ethylene glycol overdose from antifreeze. An intoxicated person with a metabolic acidosis with an elevated anion gap and renal insufficiency. Oxalate crystals on UA. Oxalate crystals are shaped like envelopes.
What is the treatment of acute ethylene glycol overdose?
Fomepizole infusion to prevent the formation of the toxic metabolite of ethylene glycol (oxalic acid). Dialysis to then remove the ethylene glycol. Sodium bicarbonate to correct acidosis.
What renal complication occurs in patients being given chemotherapy?
Acute renal failure from uric acid toxicity occurs in the setting of tumor lysis syndrome. Vigorous hydration and allopurinol should be given before chemotherapy.
What is the effect of hypercalcemia on the kidneys?
Calcium precipitates in tubule, forming stones. Hypercalcemia can lead to distal RTA and nephrogenic diabetes insipidus. Most common cause of hypercalcemia is primary hyperparathyroidism.
Which drugs are toxic to the kidneys?
NSAIDs, aminoglycosides, cephalosporins, contrast agents, amphotericin, cisplatin, radiation effect, lead, mercury, or gold, and cyclosporine.
What method of administration of aminoglycosides reduces the risk of toxicity?
Ability of antibiotics to kill is associated with peak level, but toxicity is associated with the trough level. Once-a-day dosing allows high bactericidal levels and very low trough levels, which reduce toxicity.
What is the affect of amphotericin B on the kidneys?
Renal insufficiency and distal renal tubular acidosis. After several weeks of amphotericin, the patient will develop high creatinine and decreased magnesium.
How does atheroembolic disease affect the kidneys?
Vascular procedures (angioplasty) cause renal failure several days later. Atheroemboli are associated with eosinophilia, low complement, bluish discoloration of fingers, toes, livedo reticularis.
What are the common nephrotoxic medications?
Radiocontrast for CT can result in renal failure 12 hours after. Pentamidine is associated with renal failure and pancreatitis. Vancomycin, aminoglycosides, cyclosporine, and lithium.
Which antiretroviral is associated with renal failure?
Indinavir results in renal failure from the drug precipitating in the kidney tubules. Indinavir stones require contrast to be visualized on a spiral CT scan.
Which persons are at risk for analgesic nephropathy?
NSAIDs are a frequent cause of renal failure. A rise in creatine occurs in persons with significant impairment such as the elderly or those with hypertension or diabetes.
What are the causes of acute papillary necrosis?
Acute papillary necrosis occurs in sickle cell disease, diabetes, urinary obstruction, chronic pyelonephritis, and NSAIDs.
What is the presentation of acute papillary necrosis?
Sudden flank pain, hematuria, pyuria, fever. Similar to pyelonephritis. Necrotic material in urine. Papillary necrosis will have a negative culture. The best test for papillary necrosis is CT.
What is glomerulonephritis?
Glomerulonephritis is inflammation of the glomerulus caused by autoimmunity, antibodies, or vasculitis.
What is the presentation of glomerulonephritis?
Edema, hematuria, red cell casts, and hypertension. The urine red cells are dysmorphic.
What are the physical signs of glomerulonephritis?
Edema of GN initially appears in areas of low tissue tension, such as periorbital or scrotum. Edema can be diffuse. Salt and water retention leads to edema and hypertension.
What amount of proteinuria is associated with glomerulonephritis?
Modest amounts of protein appear in the urine with the daily total being <2 grams per 24 hours. When the proteinuria rises to >3.5 grams per 24 hours the patient has nephrotic syndrome.
What are the laboratory signs of glomerulonephritis?
Low urine Na with a fractional excretion of Na <1%.Many forms of GN are associated with a positive ANCA, basement membrane antibodies, ANA, or antistreptolysin. The most important test is renal biopsy.
What is Wegener granulomatosis?
Vasculitis of kidney, lung, upper respiratory, sinuses, middle ear. Chronic upper/lower respiratory illness, not responding to antibiotics, with renal disease suggests WG. Involves skin, joints, eyes, GI; neuropathy.
What are the laboratory abnormalities associated with Wegener granulomatosis?
Elevated ESR, anemia, leukocytosis. Rheumatoid factor in 50%. The best initial test that is specific for WG is the cytoplasmic antineutrophil cytoplasmic antibody. Complement levels are normal.
What is the most accurate test for Wegener granulomatosis?
The most accurate test for WG is a biopsy of the kidney or lung looking for granulomas. Biopsy of the nasal septum is less sensitive and has more false negative results.
What is the treatment of Wegener's granulomatosis?
Cyclophosphamide and steroids.
What is Churg-Strauss syndrome?
Characterized by lung involvement, neuropathy, skin lesions, GI, cardiac, and renal involvement.
What is the presentation of Churg-Strauss syndrome?
Fever, weight loss, malaise. Asthma, eosinophilia, atopic diseases. Elevated eosinophil, positive P-ANCA or antimyeloperoxidase. Most accurate test is lung biopsy: granulomas, eosinophils.
What is the treatment of Churg-Strauss syndrome?
Glucocorticoids and cyclophosphamide.
What is Goodpasture syndrome?
Idiopathic disorder of renal and lung disease characterized by a unique anti-basement membrane antibody. GP does not affect multiple organs or sites in the body other than the lung and the kidney.
What are the clinical signs of Goodpasture syndrome?
Hematuria, proteinuria, hemoptysis, cough, dyspnea. Best test is anti-basement membrane Ab to type IV collagen. Lung or kidney biopsy shows hemosiderin macrophages and linear deposits.
What is the treatment of Goodpasture syndrome?
Plasmapheresis and steroids. Cyclophosphamide.
What is polyarteritis nodosa?
Systemic vasculitis of small and medium arteries of every organ exception lung. Renal involvement manifests as HTN, renal insufficiency, and hemorrhage due to microaneurysms. Biopsy.
What is the treatment of polyarteritis nodosa?
Cyclophosphamide and steroids.
What are the symptoms of polyarteritis nodosa?
Fever, weight loss, malaise. Involvement of skin, eyes, muscles, GI, heart, kidneys, and neurologic system. Abdominal pain and joint pain. Anemia and an elevated sedimentation rate.
What is the treatment of polyarteritis nodosa?
Steroids and cyclophosphamide.
What is Henoch-Schönlein purpura?
Systemic deposition of IgA, resulting in renal insufficiency, skin lesions, GI symptoms.
What is the tetrad of Henoch-Schönlein purpura?
Tetrad of palpable purpura, arthralgias, abdominal pain, and renal disease. IgA deposited in walls of blood vessels. Biopsy is usually not performed. HSP most often resolves spontaneously.
What is the treatment of Henoch-Schönlein purpura?
Treatment is supportive. Steroids can be used.
What is IgA nephropathy (Berger disease)?
IgA nephropathy with mild hematuria that resolves spontaneously in 30%. About 40-50% of patients progress to end stage renal disease. Deposition of IgA only in kidney.
What are the signs of Berger disease?
HTN. Usually an Asian patient under 35 years of age who has had a recent viral illness or pharyngitis, then develops hematuria 1-2 days later.
How is Berger disease diagnosed?
The diagnosis is based on finding IgA deposited in the kidney on biopsy.
What is the treatment of Berger disease?
Proteinuria is treated with ACE inhibitors or angiotensin receptor blockers (ARB).
What are the causes of postinfectious glomerulonephritis?
Group A beta-hemolytic streptococci (S pyogenes) and other infections can cause postinfectious glomerulonephritis. Any infectious agent, including hepatitis B and C, CMV, staph endocarditis.
What type of infection can cause post-streptococcal glomerulonephritis?
Poststreptococcal glomerulonephritis can occur with throat or skin infection with S pyogenes. Rheumatic fever only occurs after pharyngitis in 10-15% of patients with GABS pharyngitis.
What are the signs of postinfectious glomerulonephritis?
Smoky, cola urine from hematuria, red cell casts, proteinuria. Periorbital edema and HTN. The best initial test is antistreptolysin (ASO) and the antihyaluronic acid. Complement low. Renal biopsy rarely.
What is the treatment of postinfectious glomerulonephritis?
Treatment is management of fluid overload and HTN with diuretics. Resolves spontaneously. Antibiotics should be given to eradicate the organism from the pharynx.
What are thrombotic thrombocytopenic purpura and hemolytic uremic syndrome?
TTP and HUS are two different varieties of the same disease of hemolytic anemia, uremia, and thrombocytopenia.
What are the causes of thrombotic thrombocytopenic purpura and hemolytic uremic syndrome?
TTP is associated with HIV, but it is most often idiopathic. HUS is more common in children and with E. coli 0157:H7 food poisoning.
What is the triad of hemolytic uremic syndrome?
HUS is a triad of hemolytic anemia, uremia, and thrombocytopenia.
What are the signs of thrombotic thrombocytopenic purpura?
TTP has anemia, uremia, and thrombocytopenia, and is also associated with fever and headache, confusion, seizures, focal deficit.
What are the laboratory findings in hemolytic uremic syndrome and thrombotic thrombocytopenic purpura?
Anemia is intravascular, and the blood smear shows schistocytes, helmet cells, fragmented red cells. LDH and reticulocyte count will be elevated and the haptoglobin decreased.
What is the treatment of thrombotic thrombocytopenic purpura and hemolytic uremic syndrome?
Plasmapheresis. Dipyridamole may preventing platelet aggregation in TTP. Antibiotics contraindicated in HUS; organism releases toxins. Administering platelets will worsen CNS and renal.
What is Alport syndrome?
Glomerular disease with anterior lenti conus (lens protrusion), and sensorineural hearing loss.
What is idiopathic rapidly progressive glomerulonephritis?
RPGN may occur with any glomerular disease. Idiopathic form is associated with crescent formation in the kidney, which is ANCA negative. Diagnosis is with renal biopsy.
What us the treatment of idiopathic rapidly progressive glomerulonephritis?
Treatment is with steroids and cyclophosphamide.
What is amyloid?
Amyloid is a proteinaceous material associated with chronic infectious or inflammatory conditions, such as rheumatoid arthritis, inflammatory bowel disease, or myeloma.
How does amyloidosis affect the organs?
Amyloid builds up in kidney, causing glomerulonephritis, accumulates in GI, nerves, muscles. Restrictive cardiomyopathy, rhythm disorders, heart block. Macroglossia, carpal tunnel, malabsorption.
How is amyloidosis diagnosed?
Biopsy of the involved organ, such as the kidney. Congo red testing shows green birefringence. Amyloidosis treatment consists of controlling the underlying disease. Melphalan and prednisone.
What is nephrotic syndrome?
Renal disease sufficient to cause proteinuria >3.5 gm/24 h, hyperlipidemia, edema, and a low serum albumin.
What are the causes of nephrotic syndrome?
1/3 nephrotic syndrome associated diabetes, HTN, myeloma. Glomerulonephritis becomes nephrotic syndrome if massive proteinuria, low albumin. Nephrotic syndrome describes severity, not etiology.
What are the signs of nephritic syndrome?
Hyperlipiduria with droplets in shape of Maltese crosses in urinalysis. Hypercoagulability from urinary loss of anticoagulant proteins. Arterial or venous thrombosis.
How is nephrotic syndrome diagnosed?
High protein in urine, low protein in blood, edema, hyperlipidemia. 24-hour urine protein >3.5 gm. A protein:creatinine ratio of >3.5 on a single urine. The most accurate test is a renal biopsy.
What is the treatment for nephrotic syndrome?
Control the underlying disease. Steroids, cyclophosphamide, mycophenolate, azathioprine. ACE inhibitors or ARBs are used for all patients with proteinuria.
What are the causes of proteinuria?
Microalbuminuria is 30-300 mg/24 h. Mild amounts of proteinuria <1 gm/d can be seen in 10%. Proteinuria can also occur from fever, CHF, exercise, standing (orthostatic). Dipstick only detects albumin.
What are the causes of hematuria?
Nephrolithiasis, cancer, bleeding disorders, trauma, cyclophosphamide (hemorrhagic cystitis). Cystitis or prostatitis. The red cells become dysmorphic in glomerular disease.
What is the cause of nitrites on urinalysis?
Gram-negative bacteria reduce nitrate to nitrite.
What is the significance of asymptomatic bacteriuria?
The isolated finding of bacteria in the urine in pregnant women should be treated. About 30% of pregnant women with bacteriuria progress to pyelonephritis.
What is the significance of hyaline casts in the urinalysis?
Dehydration. These casts develop as an accumulation of the normal amount of tubular protein. Hyaline casts may occur in normal persons.
What is the significance of red cell casts on the urinalysis?
Glomerulonephritis
What is the significance of broad, waxy casts in the urinalysis?
Chronic renal failure
What is the significance of granular casts in the urinalysis?
Also called "dirty" or "muddy." They are associated with acute tubular necrosis and represent accumulated epithelial cells.
What is the significance of white cell casts in the urinalysis?
Pyelonephritis, interstitial nephritis.
What are the most common causes of end stage renal disease that require dialysis?
Diabetes and HTN. Glomerulonephritis is the etiology of about 15%, with cystic disease and interstitial nephritis, causing 4-5% each.
What are the indications for dialysis?
Life-threatening abnormalities that cannot be corrected, such as fluid overload refractory to diuretics. Acidosis, pericarditis, encephalopathy, neuropathies, hyperkalemia, persistent nausea, vomiting, bleeding.
What type of dialysis is used most frequently?
Hemodialysis is used in 85% of patients and peritoneal dialysis in 15%. The most common complication of peritoneal dialysis is peritonitis.
What is the cause of anemia in end-stage renal disease?
Decreased erythropoietin from the kidney. Treated with replacement of erythropoietin. The anemia of ESRD is normochromic and normocytic.
What is the cause of hypocalcemia/hyperphosphatemia in end-stage renal disease?
Loss of 1,25-dihydroxyvitamin D. Hyperphosphatemia is from inability of kidneys to excrete PO4. High PO4 contributes to low Ca by precipitating out in tissues in combination with the calcium.
What is the treatment of hyperphosphatemia in renal disease?
High phosphate levels are treated with phosphate binders, such as calcium carbonate or calcium acetate.
What is the treatment of hyperphosphatemia with hypercalcemia?
Sevelamer and lanthanum are phosphate binders. Used when calcium is high because of vitamin D. Aluminum-containing binders should not be used because associated with dementia and bone abnormalities.
What is osteodystrophy of end-stage renal disease?
Osteitis fibrosa cystica occurs because kidneys fail to produce 1,25 vitamin D. Low Ca leads to hyperparathyroidism, which removes Ca from bones. Controlled by increasing calcium levels.
What is the treatment of hypermagnesemia in renal insufficiency?
Magnesium accumulates because of decreased renal excretion. Treatment is restriction of magnesium intake.
What is the effect of end-stage renal disease on the cardiovascular system?
HTN, accelerated atherosclerosis, coronary artery disease. This is the most common cause of death for patients on dialysis. Goal BP is <130/80 for those with renal impairment.
What is the effect of end-stage renal disease on the immune system?
Increased infection because WBCs do not function in uremia. Infection is the second most common cause of death in dialysis patients. Most common organism is Staphylococcus.
What is the effect of end-stage renal disease on the coagulation system?
Bleeding occurs because uremic platelet dysfunction. Bleeding treated with desmopressin, which releases subendothelial von Willebrand factor and factor VIII, which increases platelet adherence.
What is the dietary treatment for severe renal disease?
Diet restricted in potassium, sodium, protein, magnesium, and phosphate.
What is hyponatremia?
Hyponatremia is defined as a low serum sodium <135 mEq. This generally occurs from either increased free water retention or urinary sodium loss. About 85-90% of sodium is extracellular.
What is the formula for serum osmolality?
Serum osmolality is largely a function of the serum sodium level. Serum osmolality = (2 x sodium) + BUN/2.8 + glucose/18. When the glucose and BUN are normal, this equation becomes 2 x sodium + 10.
What is the presentation of hyponatremia?
Neurologic symptoms range from mild confusion and memory loss to disorientation and obtundation to seizure or coma, depending on the severity.
What factor is associated with hyponatremic seizures?
Symptoms depend on rate of decline. Acute 15-20 point drop can result in a seizure or coma. If the level drops gradually, the patient can sustain an extremely low sodium. There should be no symptoms unless below 125.
What is the treatment of mild hyponatremia?
Mild asymptomatic hyponatremia should resolve with fluid restriction.
What is the treatment of moderate hyponatremia?
Moderate hyponatremia can be managed with normal saline administration combined with a loop diuretic such as furosemide. The saline provides sodium, and the loop diuretic causes a net free water loss.
What is the treatment of severe hyponatremia?
3% hypertonic saline. Severe symptoms do not occur unless Na <120. Central pontine myelinolysis occurs if Na is corrected rapidly. Rate should not exceed 0.5-1 mEq/h. No more than a 12-point rise in a 12-h.
What is the maximum rate of correction of severe hyponatremia?
Hyponatremia can be corrected as rapidly as 2 mEq per hour if the patient is having seizures. Fludrocortisone is given for cerebral salt wasting disease.
What is pseudohyponatremia?
Na level is artificially low because of hyperglycemia. Na is decreased by 1.6 mEq/L for every 100 increase in glucose. Glucose load causes a transcellular shift of water. Hyperlipidemia causes low Na.
What hypervolemic states (increased ECF) are associated with hyponatremia?
There is a decrease in intravascular volume resulting in an increase in ADH secretion from the posterior pituitary in congestive heart failure; nephrotic syndrome and low albumin states; and cirrhosis
What is the cause of hyponatremia in renal insufficiency?
When renal failure becomes advanced, the impaired free water excretion will reduce the sodium level and result in hypervolemia.
What is the cause of hyponatremia in hypovolemic states?
Loss of sodium through body fluids and replacement with free water. Sweating and replacement only with free water causes the sodium level to drop.
What are the gastrointestinal causes of hyponatremia?
Vomiting, diarrhea, gastric suction; hypovolemia.
What are the causes of sodium loss through the skin?
Skin loss: burns, sweating with replacement with free water, cystic fibrosis.
How does renal failure cause renal sodium loss?
The kidney can lose the ability to reabsorb sodium in the proximal convoluted tubule as the kidney is damaged. Damaged tubules cannot reabsorb sodium and hypovolemia results.
How does adrenal insufficiency (Addison disease) cause hyponatremia?
Aldosterone reabsorbs sodium from the kidney. Without aldosterone, sodium is lost.
What are the causes of hypovolemic hyponatremia with a urine sodium <10?
Dehydration, vomiting, diarrhea, sweating
What are the causes of hypovolemic hyponatremia with a urine sodium >10?
Diuretics, ACE inhibitors, renal salt wasting, Addison disease, cerebral sodium wasting
What are the causes of euvolemic hyponatremia?
Psychogenic polydipsia with >15-20 liters/d. Hypothyroidism. Diuretics: can be both hypovolemic and euvolemic. ACE inhibitors. Endurance exercise. Syndrome of inappropriate secretion of ADH.
What are the causes of syndrome of inappropriate secretion of ADH (SIADH)?
CNS infections, stroke, tumor, trauma, vasculitis, pain, pneumonia, TB, PE, asthma. Lung CA, pancreas CA. SSRIs, tricyclics, haloperidol, cyclophosphamide, vincristine, carbamazepine.
How is syndrome of inappropriate secretin of antidiuretic hormone diagnosed?
Neurologic symptoms; elevated urine osmolality and elevated urine sodium. If the urine osmolality is >100 Osm/L in the presence of hyponatremia, the person has SIADH. Elevated ADH level.
What is the treatment of SIADH?
Restrict fluids. Give hypertonic saline for severe disease. Normal saline with a loop diuretic. For chronic disease, demeclocycline or lithium to inhibit the effect of ADH on the kidney tubule.
What are the insensible causes of hypernatremia?
Insensible losses: extrarenal loss of water without intake of hypotonic fluids; increased skin loss of water (sweating, burns, fever, exercise) or respiratory infections
What are the gastrointestinal causes of hypernatremia?
Osmotic diarrhea (e.g., lactulose, malabsorption), infectious diarrhea. Transcellular shift caused by Rhabdomyolysis or seizures can cause muscles to take up water and Na.
What are the renal causes of hypernatremia?
Nephrogenic diabetes insipidus (NDI), causing by renal disease, increased calcium, decreased potassium, lithium, demeclocycline, sickle cell disease, and others
What are the causes of central diabetes insipidus?
Idiopathic, trauma, infectious, tumor, granulomatous, hypoxic brain damage or neurosurgical brain injury. Idiopathic most common.
What types of osmotic diuresis can cause hypernatremia?
Osmotic diuresis: diabetic ketoacidosis (DKA), nonketotic hyperosmolar coma, mannitol, diuretics.
What are the signs of hypernatremia?
Lethargy, weakness, irritability, seizures, and coma are present with severe hypernatremia of any cause. Diabetes insipidus causes a dilute diuresis of 3-20 L per day.
How is central diabetes insipidus distinguished from nephrogenic diabetes insipidus?
Decrease in urine volume after administering ADH distinguishes central diabetes insipidus from NDI.
What is the treatment of acute hypernatremia?
IV isotonic saline. Correction should not be >1 mEq every 2 h or 12 mEq/d. 1 mEq/h is acceptable if the patient is having seizures.
What are the complications of overly rapid correction of hypernatremia?
Complications of overly rapid correction include cerebral edema, permanent neurologic damage, or seizures.
What is the treatment of central diabetes insipidus?
Correct the underlying cause. Give vasopressin (ADH) subcutaneously, intravenously, intramuscularly, or by nasal spray.
What is the treatment of nephrogenic diabetes insipidus?
Correct underlying cause if possible. Diuretic or NSAIDs. NSAIDs function by inhibiting prostaglandins, which impair concentrating ability. NSAIDs will increase the action of ADH at the kidney.
What are the gastrointestinal causes of hypokalemia?
Gastrointestinal losses can be from vomiting, diarrhea, or tube drainage.
What types of transcellular shift can cause hypokalemia?
Increased entry into cells can be from alkalosis, increased levels of insulin, beta adrenergic, and the replacement of B12. Trauma causes increased beta adrenergic activity.
What are the causes of urinary potassium losses?
Diuretics. Increased aldosterone states, such as Conn, licorice, Bartter, or Cushing. Aldosterone is the most important regulator of potassium. Renal artery stenosis. Hypomagnesemia will cause potassium loss.
What is the presentation of hypokalemia?
Affects muscles and the heart. Weakness, paralysis, fatal arrhythmias, rhabdomyolysis. Potassium is necessary for ADH effect on the kidney, and hypokalemia causes nephrogenic diabetes insipidus.
What are the ECG signs of hypokalemia?
T-wave flattening and U-waves. A U-wave is an extra wave after the T-wave caused by Purkinje fiber repolarization.
What is the treatment of hypokalemia?
Correct the underlying cause. IV potassium maximum 10-20 mEq/h; do not use dextrose containing fluids, because dextrose causes increase insulin release and lower potassium.
What are the complications of excessively rapid repletion of potassium?
Fatal arrhythmia. Idioventricular rhythm, asystole.
How many mEq of potassium is needed to raise the potassium level by one point?
Very large amounts of potassium may be necessary to raise the body potassium level by 1 or 2 points. The total body requirement is to give 4-5 mEq per kg per point.
What are the causes of hyperkalemia related to movement of K+ from cells to extracellular fluid?
Pseudohyperkalemia: venipuncture, platelets >1,000,000, WBC >100,000. Acidosis. Insulin deficiency. Rhabdomyolysis, tumor lysis, seizures, exercise. Periodic paralysis: weakness, hyperK, family history.
What is the relationship between pH and potassium level?
H+ moves into cells, K+ out. For every 0.1-point decrease in the pH, the potassium level will increase by 0.7 points because of the transcellular shift.
What are the renal causes of hyperkalemia?
Renal failure. Hypoaldosteronism: ACE inhibitors, type IV RTA, adrenal enzyme deficiency; heparin. Primary adrenal insufficiency (Addison) or adrenalectomy. K-sparing diuretics. NSAIDs.
What is the presentation of hyperkalemia?
Muscular weakness can begin usually with K+ levels >6.5.Abnormal cardiac conduction is the most common cause of death, hypoventilation.
How is hyperkalemia diagnosed?
ECG findings: peaked T waves, widened QRS, short QT, or prolonged PR interval.
What is the acute treatment of hyperkalemia?
Calcium chloride one ampule for membrane stabilization. Effect is immediate and short lived. Bicarbonate one ampule. Do not give in same IV line as calcium. Glucose one ampule and insulin, takes 30 min.
What is the non-acute treatment of hyperkalemia?
Furosemide, beta agonists. Cation exchange resin (Kayexalate) absorbs 1 mEq K+/gm. Given with sorbitol. Kayexalate can also be given as a retention enema. Dialysis.
What are the causes of distal renal tubular acidosis (type I)?
Sporadic. Also caused by Sjögren, SLE. Amphotericin, lithium, analgesics, cyclophosphamide. Nephrocalcinosis, sickle cell, chronic infection. Familial, chronic hepatitis.
What is the presentation of distal renal tubular acidosis (type I)?
Inability to concentrate H+ in urine. Urine pH is >5.4. Secondary hyperaldosteronism and hypokalemia. Nephrocalcinosis and nephrolithiasis.
How is distal renal tubular acidosis (type I) diagnosed?
Acid load test; give ammonium, which should lower urine pH. With type I RTA, urine pH remains elevated. Bicarbonate <10. Hypokalemia because of inability to secrete hydrogen ions. Kidney excretes K+.
What is the treatment of distal renal tubular acidosis (type I)?
Oral bicarbonate is the treatment because bicarbonate reabsorption in the proximal tubule remains intact; potassium replacement.
What are the causes proximal (type II) renal tubular acidosis?
Fanconi, Wilson, amyloidosis, myeloma, acetazolamide, vitamin D deficiency, secondary hyperparathyroidism, hypocalcemia, heavy metals, hepatitis, SLE and Sjögren syndrome.
What is the presentation of proximal (type II) renal tubular acidosis?
Inability to absorb bicarbonate. Urine pH is basic. Hypokalemia and bicarbonate of 18-20, and malabsorption of glucose, phosphate, urate, amino acids. Bone lesions (osteomalacia and rickets).
How is proximal (type II) renal tubular acidosis diagnosed?
Patients are have a basic urine in the presence of acidemia.
What is the treatment of proximal (type II) renal tubular acidosis?
Give potassium; mild volume depletion will enhance proximal bicarbonate reabsorption. Thiazide diuretics and very large amounts of bicarbonates.
What are the causes of hyporeninemic/hypoaldosteronism (type IV renal tubular acidosis)?
An aldosterone deficiency of any cause or adrenal insensitivity to angiotensin II, diabetes (50%), Addison disease, sickle cell disease, renal insufficiency.
What is the presentation of hyporeninemic/hypoaldosteronism (type IV renal tubular acidosis)?
Usually asymptomatic hyperkalemia. Mild to moderate renal insufficiency. Hyperchloremic metabolic acidosis (nonanion gap).
How is hyporeninemic/hypoaldosteronism (type IV) diagnosed?
Presence of high urine sodium with oral salt restriction establishes the diagnosis.
What is the treatment of hyporeninemic/hypoaldosteronism (type IV)?
Administration of fludrocortisone. Fludrocortisone has a high degree of mineralocorticoid effect and is similar to administering aldosterone.
What is the relationship of serum bicarbonate and pCO2?
For every 1-point increase in the level of serum bicarbonate, there is a 0.7-point increase in pCO2. Dehydration results in increased aldosterone, which leads to metabolic alkalosis.
What are the causes of alkalosis from H+ ion loss?
Exogenous steroids. GI loss (vomiting, nasogastric suction). Renal loss (Conn syndrome, Cushing, ACTH overproduction, licorice, Bartter syndrome). Decreased chloride intake. Diuretics.
What are the causes of alkalosis from HCO3 retention?
Bicarbonate administration. Contraction alkalosis. Milk-alkali syndrome?
What is the cause of alkalosis from H+ movement into cells?
Hypokalemia.
What are the respiratory causes of alkalosis?
Hyperventilation, pulmonary embolus, sarcoid, anxiety, pain. Progesterone, catecholamines, hypoxia, cirrhosis, pregnancy, and salicylates.
What are the causes of acidosis (low pH) with a low anion gap?
Myeloma, low albumin level, lithium.
What is the formula for anion gap?
Anion gap = (Na + K) - (HCO3 + Cl) Normal: 8-14. Na+ and cations = HCO3- and Cl- and anions
What are the causes of a normal anion gap acidosis?
Diarrhea, renal tubular acidosis, ureterosigmoidostomy.
What are the causes of an increased anion gap acidosis?
Lactate (sepsis, ischemia). Aspirin. Methanol. Uremia. Diabetic ketoacidosis. Paraldehyde, Propylene glycol. Isopropyl alcohol, INH. Ethylene glycol (antifreeze), low calcium.
What are the causes of respiratory acidosis?
Hypoventilation, chronic obstructive pulmonary disease (COPD), Pickwickian, obesity, suffocation, opiates, sleep apnea, kyphoscoliosis, myopathies, neuropathy, effusion, aspiration.
What are the causes of nephrolithiasis?
1-5% of the population. Calcium oxalate stones in 70%. Calcium phosphate in 10%. Mg/Aluminum/phosphate (Struvite) 5%. Uric acid 5%. Cysteine 1%. Indinavir.
What are the causes of hypercalciuria?
Vitamin D intoxication with sarcoid and granulomatous disease. Familial. Idiopathic renal hypercalciuria. Hyperparathyroidism (10%). Multiple myeloma, metastatic disease to bone, hypercalcemia of malignancy.
What are the causes of hyperoxaluria?
Primary familial, enteric hyperoxaluria (chronic diarrhea causes excessive absorption of oxalate).
What complication may result from hypocitraturia?
Citrate binds calcium and prevents calcium absorption. Low citrate leads to increase in calcium absorption and kidney stones. Causes of hypocitraturia: renal tubular acidosis; chronic diarrhea, thiazides.
What are the causes of uric acid stones?
Uric acid stones are associated with diseases like gout, hematologic malignancies, and Crohn disease. Radiolucent on x-rays.
Infection with which organisms can cause struvite kidney stones?
Urinary infection with urease-producing organisms like Proteus, Staphylococcus, Pseudomonas, and Klebsiella causes a highly alkaline urine that produces struvite stones.
What is the presentation of nephrolithiasis?
Constant flank pain (not colicky), hematuria, and pain radiating to groin. Stones <5 mm should pass spontaneously
How is nephrolithiasis diagnosed?
Ultrasound. Strain the urine. Serum and urine calcium. Helical (spiral) CT scan is the best test. No contrast needed for stones. Plain x-ray (80% yield) is rarely used. Intravenous pyelogram is not done.
What is the treatment of nephrolithiasis?
Analgesia, hydration, and bed rest. Shockwave lithotripsy for stones <2 cm. The fragments may cause obstruction. Ureteroscopy. Percutaneous removal.
What is the presentation of adult polycystic kidney disease?
Flank pain, hematuria (micro and gross), infections, and calculi. Genetic. Hepatic cysts (40-60%), colonic diverticula, HTN (50%), intracranial aneurysm (10-20%), mitral valve prolapse (25%).
How is adult polycystic kidney disease diagnosed?
Ultrasound and CT scan.
What is the treatment of adult polycystic kidney disease?
Management of complications (UTI, calculi, and hypertension).
What are simple renal cysts?
Very common; if smooth-walled with no debris in cyst, no treatment necessary. Cysts with irregular walls or debris inside should be aspirated to exclude malignancy. Dialysis causes cysts.
What is essential hypertension?
A systolic blood pressure of 140 mm Hg or a diastolic >90 on multiple readings in the absence of a specific, identifiable underlying cause.
What is the criteria for hypertension in patients with diabetes or renal disease?
Any blood pressure above 130/80 mm Hg is defined as hypertension.
What is the treatment of stage 2 hypertension?
Patients with stage 2 hypertension (blood pressure >160/100 mm Hg), should receive initial therapy with two medications.
What is the criteria for normal BP?
120/80 mm Hg.
What is the criteria for pre-hypertension?
Systolic 120-139. Diastolic 80-89 mm Hg.
What is the criteria for stage 1 hypertension?
Systolic 140-159. Diastolic 90-99 mm Hg?
What is the criteria for stage 2 hypertension?
Systolic >160. Diastolic >100 mm Hg.
What is a hypertensive emergency?
HTN with stroke, subarachnoid hemorrhage, encephalopathy, myocardial ischemia, retinopathy. Requires substantial reduction of blood pressure within one hour.
What are the symptoms of a hypertensive emergency?
Headache, dizziness, chest pain, dyspnea, blurred vision, and palpitations. Malignant HTN is defined as encephalopathy or nephropathy.
What are the long-term cardiac complications of hypertension?
Myocardial ischemia or infarction, congestive heart failure, left ventricular hypertrophy, aortic aneurysm, and dissection. S4 gallop, accentuated A2 heart sound, and prominent left ventricular impulse.
What are the long-term cerebrovascular complications of hypertension?
Transient ischemic attack (TIA) and stroke.
What are the long-term renal complications of hypertension?
Proteinuria, microscopic hematuria, and elevation of BUN/creatinine.
What are the long-term ocular complications of hypertension?
Retinopathy: Hemorrhages, exudates, arteriolar narrowing, and papilledema. Blurred vision, scotomata, and blindness.
What are the signs of secondary hypertension?
<5% secondary. Renovascular: bruit; Cushing: weight gain, moon-face, striae, ecchymoses; pheochromocytoma: episodic HTN with headache, palpitations, sweating; aldosteronism: polyuria/ polydipsia, hypoK.
How is hypertension diagnosed?
Allow the patient to sit 5 min before BP measured. Should not diagnose HTN after only a single reading. Repeat the reading 3-6 times over several months before confirming diagnosis and initiating therapy.
What basic studies are obtained in the evaluation of hypertension?
Urinalysis for protein, glucose, and red blood cells. Hematocrit. Potassium to exclude hyperaldosteronism. Serum creatinine and BUN. ECG to evaluate for left ventricular hypertrophy. Glucose and lipid analysis.
What is the lifestyle modification for essential hypertension?
Weight reduction, sodium restriction, aerobic exercise, avoid alcohol. Low-fat, increased fiber. Severe HTN (diastolic >100 mm Hg) should be start drug therapy with 2 medications.
What is the drug treatment of essential hypertension?
If DBP >90 despite a 3-months nonpharmacologic, start antihypertensive. Diuretics are initial treatment. For stage 2 HTN, BP >160/100, use a diuretic with an ACE/ARB/calcium-blocker or beta-blocker.
What medication should be added if a diuretic dose not control the patient's blood pressure?
If diuretics do not control BP, then add a beta-blocker, calcium-blocker, ACE inhibitor, or angiotensin-receptor blocker. beta-Blockers avoided in asthma, COPD, heart block, or depression.
What is the initial antihypertensive for diabetics?
Diabetics should be treated with ACEI or ARBs, which prevent nephropathy. BP goal in a diabetic is lower, at <130/80; also lower with renal insufficiency, CHF, retinopathy, stroke. Microalbuminuria receive ACEI.
What is the initial antihypertensive for patients who have ischemic heart disease?
After a myocardial infarction (ischemic heart disease), beta-blockers should be started.
What is the initial antihypertensive for patients with decreased left-ventricular systolic function?
Decreased left-ventricular systolic function (congestive heart failure or postmyocardial infarction) should receive ACE inhibitors and beta blockers.
Which antihypertensive should be avoided in African American patients?
African American patients are least effectively treated with ACE inhibitors.
What are the initial antihypertensive choices for pregnant patients?
Alpha-methyldopa, labetalol, hydralazine, or calcium blockers. ACE inhibitors and angiotensin-receptor blockers are contraindicated in pregnancy. Diuretics are relatively contraindicated.
What is a hypertensive emergency?
The acute onset of severe hypertension with severe and rapidly worsening symptoms of end-organ damage. This usually happens with a diastolic pressure >120-130 mm Hg.
What are the neurologic manifestations of hypertensive emergencies?
Encephalopathy, headache, confusion, seizures, and subarachnoid or intracerebral hemorrhage.
What are the cardiac manifestations of hypertensive emergencies?
Chest pain, myocardial infarction, palpitations, dyspnea, pulmonary edema, jugular venous distension, and gallops.
What are the renal manifestations of hypertensive emergencies?
Nephropathy: Acutely progressive hematuria, proteinuria, and renal dysfunction.
What are the retinal manifestations of hypertensive emergencies?
Retinopathy: Papilledema, hemorrhages, and blurred vision.
What is the laboratory evaluation for hypertensive emergencies?
The laboratory evaluation is the same as with essential hypertension. CT scan of the head to exclude hemorrhage. ECG is as an initial test to exclude myocardial infarction.
What is the treatment of hypertensive emergencies?
Intravenous nitroprusside and labetalol are the two best agents. Nitroglycerin if myocardial ischemia. Enalaprilat is IV ACEI. Initial goal is to reduce BP by no more than 25% within the first 1 to 2 hours.
What is secondary hypertension?
HTN with underlying cause. 5% of cases of HTN are caused by an identifiable underlying cause. Renal artery stenosis is the most common of these causes.
Which patients should be screened for secondary hypertension?
Hypertension in the very young or very old (<25 or >55)
What is the cause of renal artery stenosis?
Caused by arthrosclerotic disease in elderly persons and fibromuscular dysplasia in young women. Upper abdominal bruit is present in 50-70% of patients.
How is renal artery stenosis diagnosed?
Screening ultrasound. Captopril renogram measures the uptake of radioisotope before and after captopril. Positive test is decreased uptake (decreased GFR) after captopril. Arteriogram is the best method of diagnosis.
What is the treatment of renal artery stenosis?
Percutaneous transluminal angioplasty. If stenosis recurs, then the procedure should be repeated. If angioplasty fails, surgical resection. ACE inhibitors if angioplasty or surgery is not possible.
What is the cause of primary hyperaldosteronism (Conn Syndrome)?
Conn Syndrome is most commonly due to a unilateral adrenal adenoma. Bilateral adrenal hyperplasia is less common. Cancer is rare as a cause of hyperaldosteronism.
What are the symptoms of primary hyperadosteronism?
Hypertension in association with hypokalemia. Symptoms of hypokalemia such as muscular weakness and polyuria and/or polydipsia from a nephrogenic diabetes insipidus.
How is primary hyperaldosteronism diagnosed?
Elevated aldosterone levels in urine and blood.
What is the treatment of Conn syndrome?
Surgical resection of adenoma. Potassium sparing diuretics such as spironolactone in adrenal hyperplasia.
What is pheochromocytoma?
Pheochromocytoma is most often due to a benign tumor of the adrenal gland; 10% are bilateral, 10% are malignant, and 10% are extra-adrenal.
What is the presentation of pheochromocytoma?
Episodic hypertension with headaches, sweating, palpitations, and tachycardia. Pallor or flushing may also occur.
How is pheochromocytoma diagnosed?
Initial test is urinary vanillylmandelic acid (VMA), metanephrines, and free urinary catecholamines. Plasma catecholamine evaluation. CT and MRI scanning is used to localize tumor.
What is the treatment of pheochromocytoma?
Alpha-adrenergic blockade with phentolamine or phenoxybenzamine, followed by surgical removal.
What is the most common cause of Cushing disease?
Cushing Disease is commonly caused by ACTH hypersecretion by a pituitary adenoma. HTN in association with cushingoid truncal obesity, buffalo hump, menstrual abnormalities, striae.
How is Cushing disease diagnosed?
Dexamethasone suppression testing and 24-hour urine cortisol are the best initial tests.
What is the treatment of Cushing disease?
Surgical resection of the pituitary adenoma.
What is the clinical sign of coarctation of the aorta?
Hypertension greater in the upper extremities compared with the lower extremities.
What are the endocrinologic and renal causes of secondary hypertension?
Oral contraceptives, acromegaly, congenital adrenal enzyme deficiencies, glomerulonephritis, polycystic disease, diabetic nephropathy, pyelonephritis.
What are the commonly used thiazide diuretics?
Hydrochlorothiazide, chlorthalidone, metolazone, indapamide.
What are the names of the potassium sparing diuretics?
Spironolactone, amiloride, triamterene.
What are the indications for diuretics in hypertension?
Congestive heart failure, edematous states, African-American patients; least expensive.
What are the major side effects of diuretics?
Decreases in potassium and magnesium; increases in calcium, uric acid, glucose, LDL-cholesterol; gynecomastia.
What are the relative contraindications to diuretics?
Diabetes, gout, hyperlipidemia.
What are the two most commonly used beta-blockers for hypertension?
Metoprolol and atenolol are the most commonly used.
What are the specific indications for beta-blockers in hypertension?
Myocardial infarction or ischemic heart disease, supraventricular arrhythmias, migraine headaches, glaucoma, anxiety (resting tachycardia), congestive failure from diastolic dysfunction.
What are the major side effects of beta-blockers?
Bronchospasm, heart block, bradycardia, Raynaud phenomenon, depression, impotence, fatigue, decreased HDL, increased triglycerides, hyperglycemia.
What are the relative contraindications to beta blockers?
Asthma, COPD, atrioventricular conduction defects, congestive heart failure from systolic dysfunction; diabetes because of masking signs of hypoglycemia.
What are the names of the angiotensin converting enzyme (ACE) inhibitors?
Benazepril, captopril, enalapril, fosinopril, lisinopril, quinapril, ramipril, moexipril, enalaprilat (only IV form).
What are the specific indications for ACE inhibitors?
Diabetics with HTN to prevent nephropathy; BP goal in a diabetic is <130/80. Congestive heart failure as afterload reduction. Postmyocardial infarction with left ventricular impairment.
What are the major side effects of ACE inhibitors?
Cough, angioneurotic edema, neutropenia, hyperkalemia, taste disturbances, anaphylactoid reactions. Less effective in African-American patients.
What are the absolute contraindications to ACE inhibitors?
Bilateral renal artery stenosis, pregnancy.
What are the names of the calcium channel blockers?
Amlodipine, diltiazem, felodipine, isradipine, nicardipine, nifedipine, verapamil.
What are the specific indications for calcium channel blockers?
Angina pectoris, supraventricular arrhythmia, migraine, Raynaud phenomenon, esophageal spasm.
What are the side effects of calcium-channel blockers?
Peripheral edema, constipation, heart block, reflex tachycardia.
What are the relative contraindications to calcium channel blockers?
Atrioventricular conduction defects, congestive heart failure from systolic dysfunction.
What are the angiotensin receptor antagonists?
Losartan, valsartan, irbesartan, candesartan, telmisartan
What are the indications for angiotensin receptor antagonists?
Patients who are intolerant to ACE inhibitors (because of cough).
What are the names of the central-acting sympatholytics?
Clonidine, guanabenz, guanfacine, methyldopa.
What are the side effects of central-acting sympatholytics?
Depression, fatigue, dry mouth, impotence, bradycardia, heart block, memory loss. Methyldopa causes hepatitis and Coombs-positive hemolytic anemia.
What is the criteria for an abnormal pulmonary flow rate?
<80% of predicted in any lung volume or flow rate is abnormal; lung volume >120% of predicted is consistent with air trapping.
What is vital capacity?
The volume of gas exhaled with maximal forced expiration.
What is total lung capacity?
Volume of gas in the lungs after maximal inspiration.
What is residual volume?
Volume of gas remaining in the lungs after forced maximal expiration (unused space).
What is the formula for total lung capacity?
TLC = RV + VC or VC = TLC - RV
What is the purpose of measuring forced expiratory volumes (FEVs)?
FEVs determine degree of obstruction by comparing forced volume expired at 1 sec to the forced vital capacity. 80% of predicted is normal. It is decreased in patients with COPD or asthma. FEVs normal in restrictive.
What is the purpose of measuring carbon monoxide diffusing capacity (DLco)?
Lung diffusion testing determines how well oxygen passes from the alveolar space into blood. Diffusing capacity is reduced by emphysema, edema, consolidation, or fibrosis.
If pulmonary function tests show an obstructive pattern and decreased DLco, what is the diagnosis?
Emphysema.
If pulmonary function tests show a restrictive pattern and decreased DLco, what is the diagnosis?
Interstitial lung disease (intrapulmonary restriction) or mild left heart failure. Increased DLco may be seen in pulmonary hemorrhage caused by Goodpasture syndrome.
What is the methacholine challenge test?
Bronchoprovocation with methacholine is done to evaluate patients with cough or wheezing and who have a normal PFT; tests for asthma (bronchial reactivity).
What is the formula for oxygen delivery?
The most important factor in gas exchange is oxygen delivery (Do2) to the vital organs. Do2 = Cardiac Output x (1.34 x Hb x HbSat) + 0.0031 x PaO2
What are the two most important factors in maintaining the delivery of oxygen to the vital organs?
The two most important factors in the delivery of oxygen to the vital organs are the cardiac output and hemoglobin. In a critically ill patient, the hemoglobin and cardiac output should be kept near normal.
What is the formula for alveolar-arterial O2 gradient?
PAO2-PaO2 gradient is an assessment of oxygenation. The A-a gradient is valid on room air and increases with age. PAO2-PaO2 gradient = (150 -1.25) x pCO2 -PaO2orA - a = [150 - (1.25 x PaCO2) - PaO2]
What is the normal alveolar-arterial O2 gradient?
The normal gradient is between 5 and 15 mm Hg. It increases with all causes of hypoxemia except hypoventilation and high altitude.
What percentage of solitary pulmonary nodules are malignant?
One third of all solitary nodules are malignant. Calcification indicates benign. Popcorn calcifications are caused by hamartomas. Bull's-eye calcifications are caused by granulomas.
What is the first step in the evaluation of a solitary pulmonary nodule?
The first step in the evaluation of a pulmonary nodule is to look for a prior x-ray. Finding the same pulmonary nodule on an x-ray done years ago eliminates the need for further evaluation.
What is the approach to evaluation of a solitary pulmonary nodule if no prior x-ray is available and the patient is low-risk?
Low risk patients are <35 and nonsmokers with calcified nodules, which may be followed with chest x-rays or chest CTs every 3 months for 2 years. Follow-up may be discontinued after 2 years if no growth.
What is the approach to the evaluation of a solitary pulmonary nodule if no prior x-ray is available and the patient is high risk?
High-risk patients: >50 smokers with a nodule, are likely to have bronchogenic cancer. Diagnostic procedure is open-lung biopsy and removal.
What are the causes of exudative effusions?
Pneumonia, cancer, and tuberculosis. An exudative effusion will cause unilateral effusions. Exudative effusions need further investigation.
What are the causes of transudative pleural effusions?
Heart failure, nephrotic syndrome, liver disease, pulmonary embolism, atelectasis.
What are the causes of exudative pleural effusions?
Parapneumonic effusions (pneumonia), malignancy (lung, breast, lymphoma), tuberculosis, pulmonary embolism, collagen vascular disease (RA, SLE), drug-induced, pancreatitis.
What is the diagnostic approach to pleural effusions?
Thoracentesis should be performed for new and unexplained pleural effusions when sufficient fluid is present. Observe pleural effusions when there is CHF, viral pleurisy, or recent thoracic or abdominal surgery.
What is the laboratory testing for thoracentesis fluid?
Lactate dehydrogenase (LDH) and protein. Measure serum LDH and protein. Calculate the ratios of effusion to serum for these measurements.
What is the Light criteria for exudative pleural effusion?
LDH of effusion >200 IU/mL, LDH effusion/serum ratio >0.6, and protein effusion/serum ratio >0.5. If one criteria is met, it is an exudative effusion, and further evaluation is required.
What is the Light criteria for transudative pleural effusion?
All three are required: LDH of effusion <200 IU/mL, LDH effusion/serum ratio <0.6, and protein effusion/serum ratio <0.5.
What kind of pleural effusion is associated with pulmonary embolism?
Pulmonary embolism can cause a transudate or exudate. A patient with a transudative effusion but no apparent cause may have a PE.
What is the management of parapneumonic effusions?
Thoracentesis to rule out an empyema (pus in the pleural space), which needs chest-tube drainage. Uncomplicated parapneumonic effusions respond to antibiotics alone.
What are the most common causes of malignant pleural effusion?
Lung cancer, breast cancer, and lymphoma. Cytologic examination is diagnostic.
What are the causes of hemorrhagic pleural effusions?
Hemorrhagic pleural effusion may be seen in mesothelioma (a pleural-based cancer associated with asbestos), metastatic lung or breast cancer, pulmonary thromboembolism (with infarction), and trauma.
What are the causes of lymphocytic predominant, exudative pleural effusions?
Caused by tuberculosis. Adenosine deaminase is elevated, and the PCR for tuberculous DNA is positive. Acid-fast stain and culture for Tb are positive in less than 30%. Pleural biopsy confirms diagnosis.
What amount of fluid is required for thoracentesis to be done safely?
If decubitus chest x-ray detects 1 cm or more of free-flowing fluid, the thoracentesis can be performed. If non-free fluid (loculated) an ultrasound-guided thoracentesis should be done.
What are the causes of respiratory compromise?
Airway obstruction (asthma, COPD, foreign object), parenchymal lung disease (bacterial or viral pneumonia, lung injury), heart failure, pulmonary embolism, opiates, myasthenia gravis.
What are the steps in treating respiratory compromise?
The first task is to ensure that the patient's airway is patent and that breathing is adequate. Administer supplemental oxygen.
What are the clinical manifestations of respiratory compromise?
Acute cough, fever, sputum suggest infection. Sudden dyspnea without fever suggests airway obstruction, cardiac disease, or thromboembolism. Chronic dyspnea is usually CHF, interstitial disease, COPD.
What are the physical signs of respiratory compromise?
Resp >30 is severe respiratory compromise. Wheezing indicates asthma, COPD. Localized wheezing: foreign object, mass. Rales indicate pneumonia, interstitial disease, CHF. Consolidation: pneumonia, atelectasis.
What disorders are associated with respiratory compromise with a normal lung exam?
Normal lung examination may be seen in thromboembolic disease, Pneumocystis jiroveci, and disorders of central respiratory drive.
What are the most important laboratory tests in the evaluation of acute respiratory compromise?
An arterial blood-gas measurement is the most important initial laboratory test in determining the presence and severity of respiratory compromise.
What are the laboratory signs of acute respiratory failure?
Rise in pCO2 accompanied by a drop in pH. The bicarbonate level will decrease over 24 hours with renal compensation. Metabolic acidosis (lactic acidosis) in the presence of hypercapnia is an indication for ventilation.
What is the target O2 saturation in COPD exacerbations?
In the setting of acute-on-chronic respiratory failure, the administration of supplemental oxygen is often accompanied by a rise in PaCO2. The target range should be oxygen saturation 88-92%.
What is the significance of an elevated B-type natriuretic peptide?
An elevated BNP is seen in patients with left heart failure. Cor pulmonale and acute right ventricular failure (thromboembolism) may also cause a rise in the BNP.
What are the causes of respiratory failure with a clear chest x-ray?
Chest x-ray without parenchymal infiltrates accompanies respiratory failure due to thromboembolism, central respiratory depression, neuromuscular disease, and upper airway obstruction.
What are the chest x-ray findings in asthma and COPD?
Hyperinflation (hyperlucency). Focal infiltrates suggest bacterial, viral, or fungal pneumonia, aspiration, or pulmonary hemorrhage. Heart failure and ARDS present with diffuse edema.
What blood gas results indicate the need for mechanical ventilation in patients with an asthma exacerbation?
Respiratory acidosis and hypercapnia with asthma exacerbation requires ventilation. Indications for intubation include upper-airway injury, airway compromise, neurologic depression.
What are the signs of pulmonary embolism in ICU patients?
Dyspnea, tachypnea, hypoxemia.
What complication may result from gastric aspiration?
Aspiration, which may precipitate respiratory failure directly or through the development of pneumonia or acute respiratory distress syndrome.
What are the risk factors for aspiration?
The risk factors for aspiration include impaired consciousness and upper airway instrumentation (nasogastric tubes). Respiratory depression from opiates.
What is acute respiratory distress syndrome?
Diffuse inflammatory response of lungs within 24 hours of the onset of severe illness or injury. Tachypnea and hypoxemia. Diffuse pulmonary infiltrates of noncardiogenic pulmonary edema.
22-year-old with asthma, wheezing after URI. Resp 28, pulse 120; afebrile. Oxygen by nasal cannula is administered. What should be the next treatment?
Beta-Agonist (albuterol) by nebulizer.
What is asthma?
Inflammatory hyperreactivity of the respiratory tree, resulting in reversible airway obstruction. Mucosal inflammation, bronchial musculature constriction, excessive mucus; episodic pattern.
What factors precipitate attacks in intrinsic asthma?
Occurs in 50% of asthmatics who are nonallergic. Attacks are precipitated by infections, irritants, cold air, exercise, emotional upset. Prognosis is poor.
What is the pathophysiology of extrinsic asthma?
Extrinsic (allergic, atopic) asthma results from sensitization. IgE elevated. Family history. Precipitated by allergens and accounts for 20% of asthma. Allergic rhinitis, urticaria, eczema. Prognosis good.
What are the most common stimuli that cause asthma exacerbations?
Respiratory infections with respiratory syncytial virus in young children, rhinoviruses in adults. Aspirin, coloring agents such as tartrazine, and beta-adrenergic antagonists.
What are the signs of asthma?
Tachypnea, tachycardia, prolonged expirations, diffuse wheezing. Use of accessory muscles, diminished breath sounds, hyperresonance, intercostal retraction.
What are the signs of poor prognosis in asthma?
Poor prognostic factors include fatigue, diaphoresis, pulsus paradoxus, inaudible breath sounds, decreased wheezing, cyanosis, bradycardia.
What are the arterial blood gas findings in acute asthma?
In the acute phase, PaCO2, increase in pH, and normal or low PaCO2. In severe asthma or status asthmaticus there will be a decreased PaCO2, increased PaCO2, and decreased pH.
What is the significance of a normal PaCO2 in acute asthma?
A normal PaCO2 may indicate respiratory muscle fatigue in acute asthma.
What are the chest x-ray findings in asthma?
Chest x-ray shows hyperinflation. Acute infection may be visible as the cause of an acute attack.
What are the pulmonary function test abnormalities in asthma?
PFTs show obstructive pattern that reverses with bronchodilation. PFTs may be normal because asthma is episodic; in this case a provocative challenge may be performed with methacholine or cold air.
What is the treatment of acute exacerbations of asthma?
beta-Adrenergic agonist inhalers (albuterol, terbutaline). Inhaled beta-adrenergic agonists are preferred. Side effect is tremor. beta-Adrenergic agonists terminate 70% of asthmatic attacks.
What are the indications for salmeterol?
Salmeterol is a long-lasting (12 h) type of albuterol that is effective in nocturnal cough variant and exercise-induced asthma. Salmeterol has no benefit in acute episodes.
What diseases may be adversely affected by beta-agonists?
beta-Adrenergic agonists must be used with caution in cardiovascular disorders, hypothyroidism, diabetes mellitus, hypertension, and coronary insufficiency.
What is the role of theophylline in the treatment of asthma?
Aminophylline and theophylline are only modest bronchodilators. Sometimes of benefit for nocturnal cough. Improve contractility of diaphragm. Not routinely used in asthma because no added benefit.
What are the indications for ipratropium and tiotropium in the treatment of asthma?
Anticholinergics (ipratropium, tiotropium) are especially beneficial with heart disease when beta-adrenergic agonists may be dangerous. Slow to achieve bronchodilation (>90 min), medium potency.
What is the role of supplemental oxygen in the treatment of acute asthma exacerbations?
Supplemental oxygen, by nasal cannula or mask, should be given immediately for acute asthma exacerbation. Always maintain an oxygen saturation above 90%.
When should antibiotics be given to patients with an exacerbation of asthma?
Antibiotic treatment should be considered in patients with purulent sputum and chest x-ray infiltrates consistent with bacterial pneumonia.
What medication is used for long-term control of asthma?
Corticosteroids reduce airway inflammation. Used in acute exacerbations in short bursts for 10-14 days, along with bronchodilators.
What are the side effects of systemic corticosteroids?
Oral candidiasis, weight gain, HTN, glaucoma, cataracts, diabetes, muscle weakness, and osteoporosis.
What is the role of mast cell stabilizers in the treatment of asthma?
Cromolyn and nedocromil. Used for prophylaxis, and exercise and allergic asthma. Adults will benefit less than children. Cromolyn is first-line chronic treatment in children.
What is the mechanism of the leukotriene modifiers?
Inhibit 5-lipoxygenase, the enzyme of leukotriene production, or competitively antagonize LTD4. Zileuton is the only leukotriene inhibitor. The LTD4 receptor antagonists are zafirlukast and montelukast.
What is the role of leukotriene modifiers in the treatment of asthma?
This group of drugs is approved for severe asthma resistant to maximal doses of inhaled steroids. Leukotriene modifiers are not first-line drugs.
What is the treatment of mild asthma?
Mild asthma: Inhaled, short-acting bronchodilators as needed (albuterol inhaler).
What is the treatment of moderate asthma?
Inhaled steroids. Children with mild symptoms benefit from cromolyn, along with inhaled beta-agonists as needed. Nocturnal symptoms, despite short-acting beta agonists, will benefit from salmeterol.
What is the treatment of severe asthma?
Inhaled steroids daily, inhaled long-acting beta agonists (salmeterol), inhaled short-acting agonists for breakthrough symptoms, along with antileukotriene drugs (montelukast) and oral steroids.
What is the most common cause of chronic obstructive pulmonary disease?
Cigarette smoking; 10-15% of smokers develop COPD. 90% of COPD patients are cigarette smokers. COPD symptoms usually begin after at least 20 pack-years of tobacco exposure.
What is alpha-1 antitrypsin deficiency?
Rare hereditary autosomal recessive disease which can cause emphysema and liver abnormalities.
What are the physical signs of emphysema?
Distant breath sounds will be heard on auscultation.
What are the physical signs of chronic bronchitis?
Rhonchi and wheezes. Right heart failure (cor pulmonale) and clubbing.
What are the chest x-ray findings in chronic bronchitis?
Increased pulmonary markings can be seen?
What are the chest x-ray findings in emphysema?
Hyperinflation with diaphragm flattening, small heart size, and increase in retrosternal space. Cor pulmonale in COPD is caused by chronic pulmonary HTN.
How is COPD diagnosed?
Reduction in FEV1/FVC ratio and FEF 25. RV and TLC are increased. Emphysema will has decreased DLco; chronic bronchitis has normal DLco. After a bronchodilator the FEV1/FVC increase.
What are the complications of chronic obstructive pulmonary disease?
Hypoxemia with nocturnal desaturation. Erythrocytosis. Pulmonary HTN, cor pulmonale and right HF. Ventilatory failure and CO2 retention in early chronic bronchitis and end-stage emphysema.
What is the treatment of chronic obstructive pulmonary disease?
Anticholinergic agents (ipratropium bromide and tiotropium) are the first-line drugs in COPD via MDI. Anticholinergic agents can be used synergistically with beta2-adrenergic agonists in COPD.
What medication is given if chronic obstructive pulmonary disease remains symptomatic after treatment with ipratropium?
beta2 agonists (albuterol) are used after anticholinergic. Beta-agonists are not first-line agents in COPD because many have heart disease. Inhaled corticosteroids are not used routinely in COPD.
What is the role of theophylline in the treatment of COPD?
Theophylline is used if beta2 adrenergics and anticholinergics are not effective.
What medications interact with theophylline?
Increased theophylline clearance is seen in smokers, rifampin, phenytoin, phenobarbital. Decreased theophylline clearance is seen with erythromycin, ciprofloxacin, or H2-blockers (cimetidine, ranitidine).
What are the indications for home oxygen in patients with COPD?
Home O2 is given to patients with hypoxemia (PaO2 <55 mm Hg or saturation <88%); goal is O2 saturation >90%. Cor pulmonale will benefit from home oxygen when PaO2 <59 mm Hg.
What immunizations should be given to patients with chronic bronchitis or emphysema?
All patients with COPD should receive the pneumococcal vaccine (Pneumovax) every 5 years and the influenza vaccine yearly. H. influenzae vaccine if they were not previously immunized.
What are the most common causes of COPD exacerbations?
Viral lung infections. Other: bacterial infections, heart failure, myocardial ischemia, pulmonary embolism, lung cancer, esophageal reflux disease, and beta-blockers.
What is the initial management of COPD exacerbation?
Measure O2 saturation. Arterial blood gas determination of hypercapnia and hypoxia. Chest x-ray to identify pulmonary infiltrates pneumonia. Pulmonary edema indicates heart failure.
What is the laboratory evaluation of COPD exacerbations?
Check theophylline levels. CBC for elevated WBCs and polycythemia; ECG may reveal atrial fibrillation, that may precipitate heart failure and exacerbate COPD.
What are the indications for mechanical ventilation in COPD?
Intubation and mechanical ventilation for decreased consciousness, cyanosis, or hemodynamic instability, persistent hypoxemia.
What is the role of antibiotics in patients with COPD exacerbations?
Beneficial in exacerbations. Second-gen macrolides (clarithromycin, azithromycin), extended-spectrum fluoroquinolones (levofloxacin, moxifloxacin), cephalosporins (2nd/3rd gen), amoxicillin-clavulanate.
What medications are used to treat exacerbations of COPD?
O2 supplementation titrated to >90% saturation. Inhaled bronchodilators are the most effective. In acute COPD exacerbations, use albuterol and anticholinergics (ipratropium) simultaneously.
What medications should be avoided in patients with COPD?
There is no benefit to intravenous aminophylline. Avoid opiates and sedatives that may suppress respiratory system.
What is the role of corticosteroids in the treatment of COPD?
Corticosteroids may be given intravenously or orally because the efficacy is similar. 60 mg prednisone is continued for 2 weeks. Severe exacerbation is treated with IV methylprednisolone.
What is bronchiectasis?
Permanent dilation of small- and medium-sized bronchi from destruction of bronchial elastic and muscle. Can be caused by tuberculosis, fungal infections, abscess, cystic fibrosis, immotile cilia syndrome.
What are the clinical signs of bronchiectasis?
Chronic cough, hemoptysis, copious, foul-smelling sputum, sinusitis, immune deficiencies. Wheezes, crackles. Recurrent pneumonias with gram-negative bacteria, especially Pseudomonas. Polycythemia.
How is bronchiectasis diagnosed?
Chest x-ray: advanced cases may show 1- to 2-cm cysts and crowding of the bronchi (tram-tracking). High- resolution CT scan of the chest detects bronchiectasis.
What is the treatment for bronchiectasis?
Bronchodilators, chest physical therapy, postural drainage. TMP-SMZ, amoxicillin, amoxicillin/clavulanate when sputum production increases. A different antibiotic each time.
What bacterial coverage is required in the treatment of bronchiectasis?
Symptoms should be treated IV for gram-negative bacteria (quinolones, ceftazidime, aminoglycosides. Surgical therapy with localized bronchiectasis or massive hemoptysis.
What vaccines are indicated for patients with bronchiectasis?
Yearly vaccination for influenza and vaccination for pneumococcal infection with a single booster at 5 years.
What are the complications of bronchiectasis?
Complications hemoptysis, amyloidosis, cor pulmonale, and visceral abscesses.
What is interstitial lung disease?
Group of heterogeneous diseases of chronic inflammation and fibrosis of interstitium and lung parenchyma. The worst prognosis is with idiopathic pulmonary fibrosis and interstitial pneumonitis.
What is the pathophysiology of interstitial lung disease?
Inflammation and scarring of the interstitium that disrupts normal gas exchange the area in and around the small blood vessels and alveoli.
What are the clinical signs of interstitial lung disease?
Exertional dyspnea and nonproductive cough. Coarse crackles, pulmonary HTN (increased pulmonic sound, right heart failure), and clubbing.
What are the chest x-ray findings in interstitial lung disease?
Reticular (branching) or reticulonodular pattern ("ground-glass" appearance). PFTs show intrapulmonary restrictive pattern.
What are the causes of interstitial lung disease?
Idiopathic pulmonary fibrosis, sarcoidosis, pneumoconiosis, occupational; autoimmune; hypersensitivity pneumonitis; eosinophilic, Wegener, hemosiderosis, bronchiolitis obliterans, lymphangioleiomyomatosis.
What is the diagnostic evaluation of interstitial lung disease?
High-resolution CT scan and, eventually, biopsy via bronchoscopy or open lung biopsy.
55-year-old man with exercise intolerance. Dyspnea on exertion; nonsmoker. Resp 25, JVD, crackles, clubbing, pedal edema. Diffuse reticular disease. What is the diagnosis?
Idiopathic pulmonary fibrosis.
What is idiopathic pulmonary fibrosis?
Inflammatory lung disease of unknown origin that causes lung fibrosis and restrictive lung disease. Only the lung and has no extrapulmonary manifestations except clubbing.
What is the age of onset of idiopathic pulmonary fibrosis?
IPF occurs in patients in the fifth decade of life, with an equal distribution between men and women.
What are the clinical signs of idiopathic pulmonary fibrosis?
Progressive exercise intolerance and dyspnea. Coarse dry crackles.
What are the radiographic findings in idiopathic pulmonary fibrosis?
Reticular or reticulonodular disease. CT shows ground-glass appearance. A restrictive intrapulmonary process on PFTs. Bronchoalveolar lavage will show increased macrophages.
What is the treatment for idiopathic pulmonary fibrosis?
Treatment with steroids with or without azathioprine will benefit 20% of patients; 80% progress to fatal lung fibrosis. There is a 20 to 40% five-year survival.
A 29-year-old woman, painful erythematous papules. Joint swelling, pain. Fever, symmetric swelling of knees, PIP and MCP joints, and 3 cm papules over the anterior legs. What is the diagnosis?
Sarcoidosis
What is sarcoidosis?
Systemic disease, characterized by noncaseating granulomas in the lung and other organs. Increased incidence of sarcoidosis among blacks and patients 20-40 years.
What are the clinical signs of sarcoidosis?
Sarcoidosis can involve any organ system. Hilar adenopathy in an asymptomatic patient. Erythema nodosum, arthritis. Fever, parotid enlargement, uveitis, facial palsy. Lung involvement in 90%.
What are the chest x-ray findings in sarcoidosis?
Hilar adenopathy, reticulonodular parenchyma, or honeycombing with fibrosis.
What are the laboratory findings in sarcoidosis?
Hypercalcemia or hypercalciuria caused by vitamin D produced by macrophages. Elevation in ACE in 60%. Increased LFTs in 30%. Skin anergy; PFT restrictive pattern. Uveitis and conjunctivitis in >25%.
How is sarcoidosis diagnosed?
Lung biopsy shows noncaseating granulomas.
What is the prognosis in sarcoidosis?
Eighty percent of patients with lung involvement from sarcoidosis remain stable, or the disease resolves. Twenty percent of patients develop progressive disease with end-organ compromise.
What is the treatment for sarcoidosis?
Trial of steroids. Mandatory for uveitis, sarcoidosis involving the CNS, and hypercalcemia.
What are the pneumoconioses?
Occupational lung diseases in which inhalation of fibers initiate an inflammatory process that leads to fibrosis. Appears 20-30 years after exposure to mining of gold, silver, lead, copper.
What is the pathology of pneumoconioses?
Macrophages engulf offending agents, causing inflammation and fibrosis of the lung. Respiratory insufficiency.
What are the signs of pneumoconioses?
Dyspnea, shortness of breath, cough, sputum production, cor pulmonale, and clubbing. PFTs show a restrictive pattern with a decreased DLco. Hypoxemia with an increased PAO2-PaO2 gradient.
What are the chest x-ray finding in pneumoconiosis?
Small irregular opacities, interstitial densities, ground glass appearance, and honeycombing.
What is asbestosis?
Lung disease caused by inhalation of asbestos fiber in mining, milling, foundry work, shipyards, or the application of asbestos to pipes, brake linings, insulation, and boilers.
What are the signs of asbestosis?
Exertional dyspnea and reduced exercise tolerance, cough and wheezing, chest wall pain, and respiratory failure.
What are the chest x-ray findings in asbestosis?
Pleural thickening, pleural plaques, and calcifications of the diaphragm. Pleural effusions and the interstitial lung process associated with asbestosis usually involves the lower lung fields.
What cancer is most commonly associated with asbestosis?
The most common cancer associated with asbestosis is bronchogenic carcinoma (adenocarcinoma or squamous cell). Mesotheliomas are also associated with asbestos.
How is asbestosis diagnosed?
Lung biopsy shows barbell-shaped asbestos fiber.
What is silicosis?
Lung disease caused by inhalation of silica dust during mining, quarrying, tunneling, glass and pottery making, and sandblasting.
What are the signs of silicosis?
Exertional dyspnea, cough, wheezing.
What are the chest x-ray findings in silicosis?
Nodules (1-10 mm) throughout the lungs, most prominent in the upper lobes. Eggshell calcifications.
What percentage of miners contract coal miner's lung/coal worker's pneumoconiosis?
12% of all miners.
What is the presentation of coal worker's pneumoconiosis?
Coal worker's pneumoconiosis presents with exertional dyspnea, cough, wheezing.
What are the chest x-ray findings in coal worker's pneumoconiosis?
Small round densities in parenchyma, upper half of the lungs.
What are the laboratory abnormalities associated with coal worker's pneumoconiosis?
Increased IgA, IgG, C3, antinuclear antibodies, and rheumatoid factor are seen in coal worker's pneumoconiosis.
38-year-old woman with sudden onset of shortness of breath and pleuritic chest pain; oral contraceptives. Respirations 28, pulse 110. Mild hypoxemia (7.52/70/25/90%). Chest x-ray is normal. What is the diagnosis?
Pulmonary embolism
What are the high-risk factors for embolism and DVTs?
Surgery, cancer, CHF, travel. Lupus anticoagulant, nephrotic syndrome (loss of antithrombin III), OCPs (smoker). Factor V Leiden mutation; protein C, S, antithrombin III deficiency. Pregnancy.
What are the symptoms and signs of pulmonary embolism and deep vein thrombosis?
Sudden onset of dyspnea (shortness of breath) and tachypnea. Thigh or calf swelling. Pleuritic chest pain, hemoptysis. Increased respiratory rate with tachycardia; increased pulmonic sound (P2).
What patients are considered to be high-risk for pulmonary embolism?
Patients with one risk factor and consistent symptoms and no alternative diagnosis. Low-risk patients have no risk factors, atypical symptoms, and an alternative diagnosis (eg, pneumonia).
What are the arterial blood gas findings in pulmonary embolism?
Hypoxemia with an elevated A-a gradient. The A-a gradient may be normal.
What are chest x-ray findings in pulmonary embolism?
Usually normal. Other nonspecific findings include atelectasis and pleural effusion. Westermark sign is the loss of vascular markings. Hampton hump is a wedge-shaped infiltrate caused by pulmonary infarction.
What diagnosis should be considered in all patients with dyspnea and normal chest x-ray?
Pulmonary embolus should be considered in all patients with dyspnea and normal chest radiography.
What are the ECG signs of pulmonary embolism?
Right heart strain: large S wave in lead I and deep Q in lead III with T-wave inversion in the same lead. The most common finding is sinus tachycardia. The ECG excludes acute pericarditis and myocardial ischemia.
What diagnostic tests are used to diagnose pulmonary embolism?
Spiral CT is initial test for PE. Ventilation-perfusion scan is a nuclear test. Pulmonary embolus will cause perfusion defects. Angiogram is most accurate test, but risk of pulmonary artery rupture is 1%.
What diagnostic tests are used to diagnose deep vein thrombosis?
Compression or duplex ultrasound. Venogram is rarely done. MRI.
What is the role of D-dimer testing in evaluating thromboembolic disease?
D-dimer is the most sensitive test for PE. Indicates fibrin degradation from thromboembolism, surgery, infection, trauma, pregnancy, DIC. Normal test means no thrombus; only be used to rule out PE.
What is the diagnostic approach to suspected pulmonary embolism?
CXR, then spiral CT. Normal CT and normal D-dimer in low-risk excludes PE. Normal CT and normal Doppler ultrasound in low-risk excludes PE. If tests are negative, but high risk, angiogram is done.
What treatment should begin to patients suspected of having a pulmonary embolism or deep vein thrombosis?
All patients should be on anticoagulation while completing diagnostic evaluations. Heparin should always be started before sending that patient for CT or V/Q scan.
What is the treatment of pulmonary embolism?
Give oxygen and start heparin immediately while the diagnostic evaluation is being completed. LMWH or unfractionated heparin for 7-10 d. Warfarin should be started with heparin and continued for 6 mo.
What is the mechanism of action of low molecular weight heparin?
LMWH inactivates factor Xa but has no effect on thrombin (no need PTT). Dosing is based on weight QD or bid. LMWH is equal to unfractionated heparin in DVT and PE.
What are the advantages of low molecular weight heparin over unfractionated heparin?
Less hemorrhage or heparin-induced thrombocytopenia.
What is heparin-induced thrombocytopenia?
Heparin-induced thrombocytopenia (HIT) is a common complication of heparin 5 days after starting in 5%. Associated more with thrombotic events than bleeding. Stop heparin when platelets decrease.
What is the cause of recurrent pulmonary embolism despite treatment with heparin?
Consider heparin-induced thrombocytopenia in a patient with recurrent pulmonary embolism or DVT despite heparin.
What is the treatment of heparin-induced thrombocytopenia?
HIT is treated with the new anticoagulants (argatroban, lepirudin).
What are argatroban and lepirudin?
Anticoagulants that are used in patients who can not use heparin.
What is the mechanism of warfarin?
Warfarin inhibits the vitamin K-dependent factors (II, VII, IX, and X). Factor VII has the shortest half-life of all the affected factors.
What test is used to assess the anticoagulant effect of warfarin?
PT is monitored to assess the warfarin anticoagulant effect. INR is used to control for variability in PT. Warfarin should be titrated to an INR of 2-3.
What complication occurs when warfarin is given to patients with protein C deficiency?
Rare procoagulant effect that occurs in preexisting protein C deficiency. Warfarin causes transient hypercoagulable state and diffuse thrombosis. Risk reduced by starting heparin and warfarin simultaneously.
What are the contraindications to anticoagulation?
Anticoagulation is contraindicated in patients with recent neurosurgery or eye surgery.
What is the anticoagulant treatment of patients who have had recent neurosurgery or eye surgery?
An inferior vena cava filter (Greenfield filter) should be used to prevent further embolism in these patients.
Want is the anticoagulation method for pregnant patients with deep vein thrombosis?
Warfarin is contraindicated in pregnant patients. LMWH for 6 months is the best alternative with injections once or twice a day.
What is the indication for thrombolytics in patients with pulmonary embolism?
Thrombolytics (tPA) for hemodynamically unstable (hypotension, right heart failure) or for massive DVT to prevent the postphlebitic syndrome.
What is postthrombotic syndrome?
Most common complication of DVT, occurring in up to two-thirds. Destruction of the valves. Pain, edema, hyperpigmentation, and skin ulceration. Compression stockings prevent the postthrombotic syndrome.
What is the duration of anticoagulation for noncomplicated proximal deep vein thrombosis?
6 months. In patients with thrombophilias (hypercoagulable states), lifelong anticoagulation is with warfarin if there have been two or more episodes of thrombosis.
Why should protein C or protein S levels not be checked during acute thrombosis?
Both warfarin and acute clot formation cause lower protein C and S.
What disorders should be suspected in patients who develop recurrent thrombosis while on anticoagulants?
Heparin-induced thrombocytopenia or cancer-related thrombosis. Place an inferior vena cava filter or use the hirudin derivative anticoagulants. IVC filters are associated with clot formation around the filter site.
What is the management of distal deep vein thromboses?
Below-the-knee DVT is not a cause of pulmonary embolism unless extend to proximal veins. Monitor for extension to the proximal veins by serial ultrasound or anticoagulation for 3 mo.
What is fat embolism syndrome?
Fat embolism is a rare type of embolism that occurs 3 days after long bone fracture (femur fracture). It may rarely occur after CPR. Acute dyspnea, petechiae (neck and axilla), and confusion.
What is the treatment of fat embolism syndrome?
The treatment is supportive (no anticoagulation).
A 50-year-old man with gram-negative sepsis severe dyspnea. Crackles; ABG shows hypoxemia and hypercarbia. Diffuse alveolar densities. What is the diagnosis?
Acute respiratory distress syndrome
What is acute respiratory distress syndrome?
Increased permeability of alveolar-capillary membrane, edema, hypoxemia caused by sepsis, trauma, DIC, drug overdose, inhalation of toxins, Goodpasture, SLE, drowning, bypass surgery.
What are the signs and symptoms of acute respiratory distress syndrome?
Dyspnea, increased respiratory rate, diffuse crackles, and rhonchi.
What are the chest x-ray findings in acute respiratory distress syndrome?
Diffuse interstitial or alveolar infiltrates; whiteout of both lung fields.
What are the blood gas findings in acute respiratory distress syndrome?
Decreased PaO2 and increased or normal PaCO2. Swan-Ganz catheter findings will reveal normal cardiac output and normal capillary wedge pressure, but increased pulmonary artery pressure.
What is the treatment for acute respiratory distress syndrome?
Mechanical support with increased positive end-expiratory pressure and permissive hypercapnia. Mortality rate 70%.
What is sleep apnea?
Cessation of airflow (>10 seconds) that occurs at least 10-15 times per hour during sleep. Oxygen saturation decreases. Daytime somnolence. Systemic HTN. Pulmonary HTN and cor pulmonary.
What is obstructive sleep apnea?
Airway closure despite adequate ventilatory effort. Usually obese and have abnormal airways. Treatment is weight loss and nasal continuous positive airway pressure.
What is the central sleep apnea?
Central sleep apnea occurs in <5% with sleep apnea; caused by inadequate ventilatory drive. Treatment is acetazolamide, progesterone, and O2.
How is sleep apnea diagnosed?
Polysomnography (sleep studies).
64-year-old man with headache and blurry vision. Neck vein distension and darker coloration over his face and neck. Confusion. Right upper lobe lung mass. Hypercalcemia. What is the diagnosis?
Bronchogenic carcinoma
What is the leading cause of cancer death in men and women?
Bronchogenic carcinoma is the leading cause of cancer death in men and women. 5-year survival rate for small cell cancer is 5% and non-small cell cancer is 8%.
What is the most common cause of bronchogenic carcinoma?
90% of bronchogenic carcinoma are directly related to smoking. 2 most common bronchogenic lung cancers are adenocarcinoma (40%) and squamous cell carcinoma.
What are the characteristics of squamous cell carcinoma?
Squamous cell carcinoma is centrally located. Cavitary lesions. Usually metastasizes by direct extension to hilar node and mediastinum. Hypercalcemia from the secretion of a PTH-like substance.
What are the characteristics of small cell carcinoma?
Central, rapidly growing, early metastasis to liver, adrenals, brain, bone. Associated with Eaton-Lambert syndrome (weakness), and paraneoplastic syndromes. Most common cause of venocaval obstruction syndrome.
What are the characteristics of large-cell carcinoma?
Peripherally located. Metastasize late in the course of disease.
What are the characteristics of adenocarcinoma?
Peripheral lesion metastasizes to liver, adrenals, brain, bone. Bronchioalveolar carcinoma is a subtype of adenocarcinoma; low-grade, may be caused by asbestos.
What are the symptoms of lung cancer?
Cough (74%), weight loss (68%), dyspnea (58%). Hemoptysis, chest wall pain, and post-obstructive pneumonia. Hoarseness indicates a nonresectable bronchogenic carcinoma.
How is lung cancer diagnosed?
Sputum cytology. Bronchoscopy for centrally located lesions. If there is a high degree of suspicion for carcinoma and the bronchoscopy results are nonspecific, a biopsy must be done.
What is the clinical evaluation of benign solitary pulmonary nodules?
75% of solitary nodules are benign. Benign nodules <35 years, nonsmokers, <2 cm, smooth distinct margins, calcification, nodule that has not changed size.
What is the management of solitary pulmonary nodules in low-risk persons?
If a patient is at low-risk for carcinoma, follow-up CT scans are indicated. In a high-risk patient the lesion should be removed.
What is the significance of bull's-eye lesions and popcorn bull lesions on chest x-ray?
Bull's-eye lesions are seen in granulomas, popcorn ball lesions are seen in hamartomas.
What are the symptoms that suggest an unresectable lesion?
Wt loss >10%, bone pain, extrathoracic metastases, CNS, superior vena cava syndrome, hoarseness, mediastinal adenopathy, TV <800, tumor class M1, tracheal, esophagus, pericardium, chest wall.
What is the treatment of small cell carcinoma?
Resectable lesions are treated with VP16 (etoposide, platinum). Non-small cell lesions are treated with chemotherapy and radiation therapy or cyclophosphamide, Adriamycin, and platinum. Effusions can be sclerosed.
67-year-old man dyspneic after cholecystectomy. Resp 24, pulse 100. Fever and decreased breath sounds in left lower lobe. Leukocytosis of 27,000. What is the diagnosis?
Atelectasis
What is atelectasis?
Collapse of part or the entire lung. Most commonly postoperative; caused by poor inspiration or lack of coughing, mucous plug, tumor, or foreign body.
What are signs of atelectasis?
Tachycardia, dyspnea, fever, hypoxemia. Tracheal deviation caused by volume loss. Diaphragm elevation . Mediastinal shift. Lobe will appear to be densely consolidated and smaller.
What is the treatment for atelectasis?
Induce deep breathing and stimulate coughing. Incentive spirometry and pulmonary toilet. Bronchoscopy with subsequent removal of mucous plugs.
After determining that the unresponsive patient is truly unresponsive, what is the next step?
Call for help. After calling for help, position the patient on a firm, flat surface, and roll the patient so that he is face up. Open the airway by performing the chin lift or jaw-thrust maneuver.
After opening the airway of an unresponsive patient, what is the next step?
After opening the airway, assess the patient's breathing by looking, listening, and feeling for air movement. If there is no air movement, then perform two rescue breaths with about two seconds per breath.
After delivering two rescue breaths to a non-breathing patient, what is the next step?
Check to see if there is a pulse by feeling for at least 5-10 seconds at the carotid artery.
If a patient has no pulse, what is the next action that should be taken?
If no pulse, perform chest compressions at a rate of 100 per minute, "push hard and push fast:" Provide 30 compressions and then two ventilations, regardless of whether one or two rescuers is present in adults.
What is the compression to ventilation ratio for CPR on a child?
In children, perform 30 compressions and 2 ventilations if one rescuer is present, and give 15 compressions and 2 ventilations if two rescuers are present.
What are the causes of asystole?
Ischemia and severe underlying cardiac disease. Metabolic derangements, drug overdose, trauma.
How is asystole diagnosed?
Asystole should always be confirmed by observing the rhythm in more than one lead on the ECG.
What is the treatment for asystole?
1 mg epinephrine IV q3-5min. Atropine 1 mg IV q 3-5min (max 0.04 mg/kg). Bicarbonate if cause of asystole is acidosis, or if the cause is tricyclic or aspirin overdose, hyperkalemia, or DKA. Pacing for very slow bradycardia.
What are the "H's and T's" of the causes of arrhythmias?
Hypoxia, Hyper/Hypokalemia, Hypothermia, Hypoglycemia, Hypovolemia, Trauma, Toxins (overdose), Tamponade, Tension pneumothorax, Thrombosis (coronary and pulmonary)
What are the ECG signs of ventricular tachycardia?
Ventricular tachycardia (VT) is a wide complex tachycardia with an organized, uniform sawtooth pattern on the ECG. There are no P-waves visible.
What are the causes of ventricular tachycardia?
Ischemia, MI, anatomic cardiac disease. Quinidine, tricyclics, phenothiazines. Long QT syndromes. The dysrhythmia originates from an ectopic focus in the myocardium or from the AV node.
What are the ECG characteristics of ventricular tachycardia?
The slowness of the conduction produces the slower and wider complexes on the ECG. The rate varies between 160-240 beats per minute.
What is the ECG appearance of torsade de pointes VT?
Torsade is a form of VT in which the morphology varies with an undulating amplitude, making it seem to "twist around a point." Associated with hypomagnesemia.
What is the treatment of torsade de pointes VT?
Magnesium is most useful for torsade.
What is the presentation of ventricular tachycardia?
VT lasting more than 30 seconds is referred to as sustained VT. Symptoms include lightheadedness, hypotension, congestive heart failure, syncope, and death.
What is the treatment for unstable ventricular tachycardia?
Requires immediate synchronized cardioversion if hypotension, chest pain, altered mental, or CHF. A lower dose of electricity, starting at 100 J, can be used at first for monomorphic VT. Synchronized.
What is the management of ventricular tachycardia without a pulse?
Immediate synchronized cardioversion. VT without serious hemodynamic compromise can be treated with amiodarone. If there is no response, then lidocaine or electrical cardioversion is used.
What is the long-term therapy for ventricular tachycardia?
Long-term therapy is most effective with beta-blockers. VT producing sudden death or VT that is sustained through initial drug therapy requires placement of an implantable cardiac defibrillator.
What is pulseless electrical activity?
Loss of pulse with some type of electrical activity. Caused by hypovolemia, tamponade, tension pneumothorax, PE, MI, hypoxia, hypothermia, potassium disorders, acidosis, tricyclics, digoxin, beta-blockers, Ca-blockers.
What is the treatment for pulseless electrical activity?
CPR while determining the cause. Epinephrine. Atropine if there is bradycardia. Bicarbonate is useful if a known acidosis caused the arrest or in a prolonged resuscitation, or hyperkalemia.
What are the atrial dysrhythmias?
Atrial fibrillation, flutter, supraventricular tachycardia. Ectopic focus in atrium or re-entry at AV. Atrial dysrhythmias have normal conduction in ventricles. QRS normal and narrow. A normal P is not present.
What are the causes of supraventricular tachycardia?
Re-entrant around AV node. A-fib is most commonly caused by HTN, enlarged atria. Hyperthyroidism, pheochromocytoma, caffeine, theophylline, alcohol, cocaine, digoxin, pericarditis, PE, surgery, trauma, ischemia.
What percentage of cardiac output is attributable to the atria?
In normal heart, only 10% of cardiac output is from atria. In a person with a dilated or postinfarction heart, or in significant valvular disease, the contribution is 30-40%, in which case more severe symptoms arise.
What are the symptoms of supraventricular tachycardia?
Asymptomatic to palpitations, lightheadedness, hypotension, disorientation, CHF, syncope. Rate-related symptoms are unlikely if the heart rate is <150.
How is supraventricular tachycardia diagnosed?
ECG and may need a 24-72 hour Holter monitor to detect brief paroxysms of the dysrhythmia not seen on the ECG.
What is the origin of narrow complex tachycardias?
Narrow complex tachycardia is always atrial in origin (QRS <0.12). Wide complex tachycardia can be either atrial or ventricular in origin.
What is the treatment for supraventricular tachycardia?
Hypotension, confusion, CHF, chest pain require synchronized cardioversion. If stable, use vagal maneuvers such as carotid sinus massage, Valsalva, ice water. If vagal maneuvers are not effective, give IV adenosine.
What drugs are used to slow the ventricular rate in supraventricular tachycardias?
For atrial fibrillation, atrial flutter, and in SVT after the failure of adenosine, slow the heart rate with calcium-channel blockers (such as diltiazem or verapamil), beta-blockers, or digoxin.
What are the contraindications for verapamil?
Severe left ventricular dysfunction and low EJECTION FRACTION. Beta-blockers are contraindicated in reactive airway disease.
What is the best treatment approach for atrial fibrillation?
Rate control to <100 with anticoagulation with warfarin to an INR of 2-3 is superior to converting into sinus rhythm. Return to a normal sinus rhythm can prevent in embolic stroke in 5% per year.
What are the medications that convert atrial fibrillation to sinus rhythm?
Procainamide, amiodarone, ibutilide, sotalol, quinidine, propafenone, and dofetilide can convert a minority to sinus. Elective cardioversions should be preceded and followed by several weeks of Coumadin.
What is the treatment of atrial fibrillation that cannot be converted with antiarrhythmics?
If medications can not convert to sinus, then attempt electrical cardioversion. Preceded and followed by several weeks of anticoagulation if AF present >48 h. Transesophageal echo can exclude a clot.
What is bradycardia?
Heart rate <60 bpm. Degeneration of the node. Serious bradycardias are Mobitz-type II second-degree block and 3rd-degree block. Caused by ischemic damage to AV node, myocarditis, amyloidosis, sarcoidosis.
What is the presentation of bradycardia?
Hypotension and decreased cardiac output. Mobitz-type I second-degree block is characterized by progressive P-R lengthening, whereas in Mobitz-type II, the P-R interval remains constant.
What is the treatment for asymptomatic bradycardia?
Asymptomatic bradycardia, first-degree block, Mobitz-type I (Wenckebach) second-degree block need no therapy.
What is the treatment for symptomatic bradycardia?
Symptomatic bradycardia is treated with atropine and pacemaker. Mobitz-type II second-degree block and third-degree block pacemaker. Dopamine or epinephrine if hypotension.
Which toxins cause miosis?
Clonidine, barbiturates, opiates, cholinergics, and pontine stroke.
Which toxins may cause mydriasis?
Sympathomimetics and anticholinergics.
Which toxins may cause dry skin?
Anticholinergics
Which toxins may cause profuse sweating?
Cholinergics, sympathomimetics.
What are the indications for gastric lavage?
Lavage is only useful within first hour after ingestion (usually not done). Contraindicated with acids or alkalis. If altered mental, lavage should be preceded by intubation. Large bore oropharyngeal hose (eg, an Ewald tube).
What are the indications for activated charcoal administration?
The main therapy for toxin ingestion is activated charcoal administration every 2-4 h to block absorption and accelerate the removal of toxins. Not effective for hydrocarbon or for metals such as iron.
What are the indications for whole bowel irrigation?
Large-volume pill ingestions in which the pills can be seen on x-ray. Gastric tube is placed and 1-2 L/h of GoLYTELY (polyethylene glycol) until the bowel movements run clear.
What medications should be given first to any patient with altered mental status or coma?
Naloxone, dextrose, and thiamine should be given first to any altered mental status or coma. Naloxone has no adverse effects and is immediately effective. Dextrose prevents permanent brain damage from hypoglycemia.
What are the substances or drugs that may require dialysis for removal?
Ethylene glycol poisoning, lithium overdose, methanol poisoning, aspirin overdose, theophylline overdose.
What amount of acetaminophen will cause serious toxicity?
140 mg per kg of acetaminophen causes serious toxicity. For a 70-kg person, 7-10 grams will cause toxicity. Fatalities can occur above 12-15 grams.
What is the presentation of acetaminophen overdose?
Nausea, vomiting from gastritis. At 24-48 h, there is subclinical elevation transaminases, bilirubin. At 48-72 h, liver damage, nausea, jaundice, abdominal pain, encephalopathy, renal failure, death.
What is the treatment of acetaminophen overdose?
Large acetaminophen ingestions warrant - acetyl cysteine. Nomogram based on drug level is necessary to determine risk of toxicity. Elevated AST is more common than ALT. Bilirubin, PT indicate hepatic necrosis.
What elimination measures are used for acetaminophen overdose?
Gastric emptying should not be used because it will delay NAC as a. Activated charcoal is given in repeated doses. When more than 24 hours have elapsed since ingestion, there is no therapy that can prevent toxicity.
What are the common causes of methanol ingestion?
Methanol paint thinner, Sterno, solvents, and windshield washer solution.
What is the most common source of ethylene glycol ingestion?
Ethylene glycol is most often found in antifreeze.
What is the pathophysiology of alcohol toxicity?
All alcohols are metabolized by alcohol dehydrogenase, which metabolizes methanol to formaldehyde and formic acid. Ethylene glycol is metabolized partially to oxalic acid and oxalate, which leads to kidney damage.
What is the presentation of ethanol, methanol, ethylene glycol, and isopropyl alcohol toxicity?
Methanol causes visual disturbances, including blindness. Ethylene glycol causes renal failure, oxalate crystals, stones. Isopropyl alcohol can only be distinguished by drug level or by non-anion gap acidosis.
What are the laboratory signs of isopropyl alcohol ingestion?
Isopropyl alcohol results in ketonuria and ketonemia with little to no metabolic acidosis (without evidence of an increased anion gap).
How is toxicity from the alcohols diagnosed?
Specific levels. Ethylene glycol has oxalate crystals in urine, increasing BUN/creatinine, hypocalcemia. Ethylene glycol and methanol cause metabolic acidosis with anion gap.
What is the treatment for ethylene glycol and methanol intoxication?
Fomepizole (Antizol) is an IV alcohol dehydrogenase inhibitor. Fomepizole inhibits the production of toxic metabolites without leading to intoxication.
What are the symptoms of carbon monoxide poisoning?
Dyspnea, tachypnea, and shortness of breath. Headache, nausea, and dizziness occur early. Confusion, syncope. Chest pain, arrhythmia, and hypotension.
What is the laboratory evaluation of carbon monoxide poisoning?
Carboxyhemoglobin levels. Metabolic acidosis is caused by the failure of carboxyhemoglobin to release oxygen to tissues. The pO2 will be normal. CPK is elevated. Pulse oximetry is not helpful.
What is the treatment of carbon monoxide poisoning?
100% oxygen administration. Hyperbaric oxygen in severe cases with CNS abnormalities or chest pain.
What are the common sources of acid ingestions?
Toilet, drain, swimming pool, and metal cleaners. Common alkali ingestions are lye, dishwasher detergents, hair relaxers, and oven cleaners.
What is the presentation of acid or alkali ingestion?
Oral pain, drooling, odynophagia, abdominal pain. Esophageal injury with subsequent stricture formation from acid or alkali ingestion. Gastric perforation. Alkali is more destructive than acid.
What is the treatment of acid or alkali ingestion?
Wash out the mouth with large volumes of cold water. Irrigate ocular exposures with saline or water. Do not induce emesis with either acids or alkaline ingestion because emesis can worsen the injury.
What conditions predispose to digoxin toxicity?
Poisoning is more common with renal failure. Hypokalemia predisposes to toxicity because potassium and digoxin bind to the same site on the sodium-potassium ATPase pump.
What is the presentation of digoxin toxicity?
Nausea, vomiting, diarrhea, anorexia, blurred vision, color vision, hallucinations. Bradycardia, PVCs, VT, arrhythmias. Paroxysmal atrial tachycardia is most common. HyperK from inhibition of Na+/K+ ATPase. Digoxin level.
What is the treatment of digoxin toxicity?
Repeated charcoal. Digoxin-antibodies (Digibind) for arrhythmias. Potassium correction. Phenytoin, lidocaine for ventricular arrhythmias. Pacemaker for bradycardia or block refractory to atropine.
What are the signs of opiate intoxication?
Death from acute respiratory acidosis. Analgesic and euphoric effects, pupillary constriction, constipation, bradycardia, hypothermia, hypotension. Opiates can be rapidly reversed by naloxone.
What is the mechanism of cocaine?
Cocaine blocks the re-uptake of norepinephrine at synapse.
What is the presentation of cocaine toxicity?
Alpha-adrenergic stimulation, HTN, hemorrhagic stroke, subarachnoid hemorrhage, MI, arrhythmia, seizures, death. Metabolic acidosis, rhabdomyolysis, hyperthermia. Pulmonary edema.
What is the treatment for cocaine toxicity?
Benzodiazepines control agitation. Alpha/beta agents, such as labetalol, or alpha-blockers, such phentolamine, for HTN. Pure beta-blockers, propranolol, should be avoided.
What are the signs of benzodiazepine overdose?
Somnolence, dysarthria, ataxia, stupor. Rarely cause death from respiratory depression. Most deaths from benzodiazepine are co-ingestion with ethanol or barbiturate. Flumazenil is not used because seizures.
What are the signs of barbiturate overdose?
Death from respiratory, CNS depression. Hypothermia, loss of tendon reflexes, corneal reflexes, coma, flat EEG. Withdrawal may result in seizures. Bicarbonate can increase the urinary excretion of phenobarbital.
What are the signs of hallucinogen ingestion?
Marijuana, LSD, mescaline, peyote, psilocybin can cause delirium, bizarre behavior, flushed skin, dry mouth, dilated pupils, urinary retention. PCP may cause seizures. Treatment is benzodiazepines.
What are the symptoms of lead toxicity?
Adults: Abdominal pain, anemia, renal disease, headache, memory loss. Children: Abdominal pain, anemia, lethargy, seizures, coma; mental retardation.
What are the laboratory signs of lead toxicity?
Blood lead levels >10 mcg/dL. "Lead lines" are densities seen at the metaphyseal plate of the bones of children. Anemia and azotemia.
What is the treatment of lead overdose?
Chelation with calcium EDTA, dimercaprol (BAL), penicillamine, or succimer.
What are the signs of mercury toxicity?
Interstitial pneumonitis; tremors, excitability, memory loss, delirium, insomnia; nausea, vomiting, pain, bleeding.
What is the treatment of mercury toxicity?
Organic mercury is in paints and cosmetics. Treatment is oral chelation with succimer or dimercaprol.
What is the presentation of salicylate overdose?
Nausea, vomiting, gastritis, tinnitus. Stimulate respiratory centers to hyperventilation; salicylates directly toxic to lungs, causing edema. Hyperthermia, confusion, coma, seizures, encephalopathy.
What are the laboratory abnormalities in salicylate overdose?
Salicylates interfere with Krebs cycle and lead to metabolic acidosis. Salicylates cause lactic acid production with metabolic acidosis, an elevated anion gap and compensatory respiratory alkalosis.
How is salicylate overdose diagnosed?
Aspirin level. Elevated anion gap metabolic acidosis. Respiratory alkalosis early. An elevated prothrombin time and hypoglycemia.
What is the treatment for salicylate overdose?
Within the first hour after ingestion, gastric decontamination may be attempted. Charcoal and increase alkalization of the urine along with aggressive fluid resuscitation.
What is the pathophysiology of tricyclic antidepressant overdose?
Tricyclic antidepressants have anticholinergic and sodium channel blocker effects. Cardiac and CNS toxicities.
What is the presentation of tricyclic antidepressant overdose?
Anticholinergic dry mouth, tachycardia, dilated pupils, and flushed skin. Dysrhythmia, widening QRS, resulting in VT, and first-degree conduction blocks; confusion, seizures.
How is tricyclic antidepressant overdose diagnosed?
Serum drug levels, ECG: sinus tachycardia; prolongation of PR, QRS and QT.
What is the treatment for tricyclic antidepressant overdose?
Charcoal is the primary treatment in the acute setting. Any sign of cardiac toxicity should lead to the immediate use of bicarbonate, which protects the heart.
How is head trauma imaged?
CT of head. Contrast is not necessary because blood does not enhance. Hemorrhage should be visible. Skull x-rays are not useful. A concussion is a loss of consciousness plus a negative CT.
What is the CT-scan appearance of subdural hematomas?
Subdural hematomas are crescent-shaped and epidural hematomas are lens-shaped.
What are the indications for cervical spine x-rays after head trauma?
Cervical spine x-rays should be obtained in head trauma if there are focal findings consistent with a cervical radiculopathy or if spinal tenderness is present.
What is the treatment for severe intracranial hemorrhage?
Lowering intracranial pressure with hyperventilation to pCO2 of 30-35, mannitol, elevation of head of bed. Surgical evacuation. Maintain the SBP 110-160. Slight degree of HTN maintains cerebral perfusion.
A 50-year-old woman with sudden loss of consciousness, severe headache, nuchal rigidity, photophobia, and a temperature of 38.5 C (101.3 F). What is the diagnosis?
Subarachnoid hemorrhage
What is subarachnoid hemorrhage?
Bleeding into caused by aneurysm. Around the circle of Willis. Association with polycystic kidney disease, Ehlers-Danlos syndrome. Occurs spontaneously. Head trauma is a rare cause.
What is the presentation of subarachnoid hemorrhage?
Sudden, severe headache, loss consciousness 50%. Focal neurologic symptoms 30% from compression of occulomotor nerve. Nuchal rigidity, photophobia, headache, papilledema. Seizures.
What are the long-term outcome of subarachnoid hemorrhage?
One-year mortality 50%, with half of the people dying immediately. Longer-term manifestations include focal deficits, seizures, rebleeding, hydrocephalus.
How is subarachnoid hemorrhage diagnosed?
CT of head without contrast has 95% sensitivity. If CT is normal and SAH is suspected, lumbar puncture is done. Lumbar puncture is most sensitive. The absence of red cells in CSF excludes SAH.
What is the treatment for subarachnoid hemorrhage?
Maintain SBP 110-160. Seizure prophylaxis with phenytoin. Nimodipine is a calcium-channel antagonist that lowers risk of blood vessel spasm.
What test should be done after subarachnoid hemorrhage has been diagnosed?
Angiography to determine the site of the bleeding. Surgical clipping of the AVM should occur before rebleeding develops.
What is the management of hydrocephalus after subarachnoid hemorrhage?
If hydrocephalus occurs, then shunting should be done.
What is the treatment for spinal headache after lumbar puncture?
A spinal headache is treated with a blood patch.
What are the signs of respiratory injury in burn injuries?
Soot in the mouth or nose, stridor, wheezing, altered mental status, burned nasal hairs, and burns involving closed spaces are signs of impending pulmonary and laryngeal edema.
What are the signs of carbon monoxide overdose?
Altered mental status, dyspnea, headache, and chest pain are clues to severe carbon monoxide poisoning.
What are the signs of pulmonary thermal injury?
Laryngeal edema can result in stridor, hoarseness, and dyspnea. Soot in the nose and mouth can imply impending airway compromise.
How is the extent of skin burn injury assessed?
"Rule of Nines". The head and arms are 9% each. Chest, back, and legs are 18% each. Patchy burns can be estimated by using one hand's width as 1% of body surface area burned.
How is the severity of burn injury assessed?
Carboxyhemoglobin elevated in severe burns. Severe burns are defined as combined second- and third-degree burns >20% in adults or >10% in the old or very young or third-degree burns >5% of body surface area.
What is the management of severe respiratory injury?
If there are signs of severe respiratory injury, intubate the patient. If the carboxyhemoglobin level is significantly elevated (>5-10%), give 100% oxygen.
How is the fluid resuscitation requirement determined for burns?
Fluid resuscitation over first 24 hours is 4 ml per % BSA/kg. Ringer's to provide half fluid in first 8 h, with 1/4 in second 8 h, and 1/4 in final 8 h (Parkland formula). Maintain a urine output >0.5-1 mL/kg/h.
What is the treatment for burns?
Stress ulcer prophylaxis with H2 blockers. Silver sulfadiazine. Do not break blisters. Escharotomy for circumferential burns. Skin grafting.
What are heat cramps?
Mild fluid and electrolyte depletion. Painful muscular contractions lasting a few minutes. Body temperature normal. Treatment is oral rehydration and salt replacement.
What is heat exhaustion?
Weakness; body temperature slightly elevated; headache, anxiety. The patient is still able to sweat. Treatment with oral fluid and electrolyte replacement; may need intravenous hydration.
What is heat stroke?
Loss of ability to lose heat, inability to sweat. T >40 C, confusion, disorientation, nausea, blurred vision, seizures; hemoconcentration, rhabdomyolysis, elevation BUN, creatinine, WBC. Anuria, DIC, lactic acidosis.
What is the treatment of heat stroke?
Treatment with intravenous fluid replacement and rapid cooling. The body should be sprayed with water. Chlorpromazine and diazepam to control shivering.
What is malignant hyperthermia?
Nonexertional idiosyncratic heat reaction of rhabdomyolysis caused by anesthetic agents such as halothane or succinylcholine. Treatment is dantrolene.
What is neuroleptic malignant syndrome?
Idiosyncratic reaction to phenothiazines or butyrophenones, such as haloperidol. Muscular rigidity and rhabdomyolysis.
What is the treatment of neuroleptic malignant syndrome?
Treatment is bromocriptine or dantrolene.
What is hypothermia?
Core body temperature below 35 C (normal 37 C). Severe hypothermia is a core temperature below 30 C. Hypothermia often occurs with alcohol intoxication.
What is the presentation of hypothermia?
Lethargy, confusion, and weakness. Death from arrhythmia. Ventricular fibrillation or ventricular tachycardia. J-wave elevation may mimic ST segment elevation.
What is the effect of ionizing radiation on the body?
Ionizing radiation destroys DNA molecules. Lethal and can cause cancer. Nonionizing radiation includes infrared, ultraviolet, microwave radiation, which are less destructive and causes burns.
What are the symptoms of radiation exposure?
Nausea and vomiting early; intestinal ulceration, bleeding, infection later. Injury to skin, salivary glands, respiratory epithelium, thyroid, bone marrow.
What is the treatment of radiation exposure?
Antiemetics, platelets and RBC transfusions. Colony-stimulating factors (G-CSF, GM-CSF) to restore marrow. Bone marrow transplantation.
What is the presentation of near drowning?
Agitation, coma, cyanosis, coughing, and pulmonary edema, tachypnea, tachycardia, and blood-tinged sputum. Rales and rhonchi. Hypothermia. Hypoxia, hypercarbia, metabolic acidosis, hyperkalemia, hemolysis.
What is the presentation of anaphylaxis?
Urticaria, dyspnea, stridor, tachycardia, hypotension, and hemodynamic collapse.
What is the treatment of anaphylaxis?
Antihistamine (diphenhydramine), epinephrine injections, intravenous fluids, H2 blockers, and systemic corticosteroids.
What is the bacteriology of cat and dog bites?
Infections are most often polymicrobial. Cat bites are associated with Pasteurella multocida; dog bites are associated with Pasteurella, Eikenella, and Capnocytophaga canimorsus.
What is the treatment of cat and dog bites?
Wound care. Prophylactic antibiotic is amoxicillin/clavulanate (clindamycin and either ciprofloxacin or trimethoprim/ sulfamethoxazole or azithromycin if penicillin allergic).
What is the bacteriology of human bites?
Human bites carry an infection rate of 15%. Anaerobic and aerobic bacteria. Hepatitis B and HIV can also be transmitted through bites.
What is the treatment for human bite wounds?
Irrigate wound; cultures are not useful. If the bite is <12 hours old, close loosely. Tetanus and hepatitis B, prophylaxis. 5 to 7 day course of antibiotics.
Which animals carry rabies?
Raccoons, rats, wild dogs, woodchucks, skunks, foxes. 100% fatal. Less than 10 persons per year are infected in the US.
What is the presentation of rabies?
Incubation period up to one year. Prodrome of 2-10 days of fever and paresthesias at the bite site. Aphasia, paralysis, hypersalivation, and myoclonus.
How is rabies diagnosed?
Viral cultures from saliva, CSF, or serum
What is the treatment for rabies infection?
Ribavirin. Prophylaxis with human rabies immunoglobin provides immediate passive immunity. Human diploid cell vaccine should be given at 0, 3, 7, 14, and 28 days (5 shots total).
How many deaths per year are caused by snake bites?
<5-10 deaths per year, with rattlesnakes accounting for almost all fatalities. Snake venom contains hemolysis toxin, cardiotoxin, neurotoxin, and proteolytic enzymes.
What is the treatment of snake bites?
Decrease the spread of venom with compression bandages. Antivenin may cause anaphylactic reactions. Hypotension is managed with fluids. Ventilatory support for laryngeal edema.
What are the causes of spinal cord compression?
Commonly caused by lymphoma, multiple myeloma, carcinomas of prostate, lung, breast, kidney, or colon; herniated disk, epidural abscess, hematoma, or trauma.
What is the presentation of spinal cord compression?
Sensory loss, lower extremity weakness; sphincter dysfunction or sexual dysfunction. History of cancer, fever, and bowel or bladder incontinence or retention.
How is spinal cord compression diagnosed?
Plain x-rays are abnormal. The diagnostic test of choice is an MRI of the spine.
What is the treatment for spinal cord compression?
High-dose dexamethasone immediately, then MRI. For lymphoma or multiple myeloma, radiation therapy should be started. Surgical decompression for a herniated disk, epidural abscess, or hematoma.
What is syringomyelia?
Cavitation of spinal cord. Communicating with CSF or noncommunicating. Associated with congenital Arnold Chiari; caused by trauma, tumors.
What are the signs of syringomyelia?
Impaired pain, temperature; intact sensation to light touch. Loss of pain and temperature occurs in a cape-like distribution. Sparing of tactile sensation, position, and vibratory sense. Areflexia.
What is subacute combined degeneration?
B12 deficiency. Distal paresthesias and weakness of extremities followed by spastic paresis and ataxia. Deficit of vibration and proprioception with pyramidal signs (plantar extension, hyperreflexia). Low B12.
What is the presentation of anterior spinal artery infarct?
Flaccid paralysis, spastic paresis. Loss pain, temperature; sparing vibration and position. Motor, sensory, autonomic is lost below the level of the infarction with retained vibration and position. Treatment is supportive.
What is the Brown-Sequard syndrome?
Hemisection of the cord with ipsilateral spastic paresis. Ipsilateral loss of joint position sense, tactile discrimination, and vibratory sensations. Contralateral loss of pain and temperature.
A 60-year-old woman with sudden onset of right upper extremity weakness, aphasia, pronator drift and right facial nerve palsy. What is the most likely cause?
Ischemic stroke
What is a stroke?
Sudden onset of a focal neurologic deficit.
What are the mechanisms of strokes?
Large artery thrombosis, small artery thrombosis (lacunar), embolic (cardiogenic or artery-to-artery), vascular dissection, systemic hypotension, intracranial bleeding.
What is the presentation of ischemic stroke?
Acute onset of a focal neurologic deficit.
What is the blood supply to the brain?
Two major systems: carotid (anterior) circulation, and vertebrobasilar (posterior) circulation. The anterior circulation include the anterior cerebral artery and middle cerebral artery.
What are the signs of occlusion of the anterior cerebral artery?
Contralateral weakness in arm and sensory loss in leg more than in the upper extremity. Urinary incontinence, confusion, behavioral disturbances. Lower extremity weakness exceeds upper extremity.
What are the signs of occlusions of the middle cerebral artery?
Contralateral hemiplegia, hemisensory loss, homonymous hemianopia, eyes deviated toward lesion. Dominant results in aphasia. Nondominant results preserved speech, comprehension with confusion, apraxia.
What parts of the brain are supplied by the posterior circulation?
Cerebellum, brain stem, occipital lobe of the cortex, and pons. The major blood vessels of the posterior circulation are the posterior cerebral artery, basilar artery, and vertebral arteries.
What are the signs of posterior cerebral artery occlusion?
Contralateral homonymous hemianopia, hallucinations, agnosias. Occlusion of the penetrating branches of the PCA can result in CN III palsy with contralateral hemiplegia (Weber syndrome) or CN III palsy.
What is the "locked- in syndrome"?
Occlusion of the paramedian branches of the basilar artery, presenting as quadriparesis with intact vertical eye movements.
What is Wallenberg syndrome?
Occlusion of posterior inferior cerebellar artery, which presents as ipsilateral facial sensory loss, contralateral body sensory loss, vertigo, ataxia, dysarthria, dysphagia, and Horner syndrome (ptosis).
What are the signs of occlusion of the major cerebellar arteries?
Vertigo, vomiting, nystagmus, and ipsilateral limb ataxia.
How is stroke diagnosed?
Noncontrast CT to distinguish between hemorrhagic and ischemic stroke. CT often negative for ischemia within the first 48 hours. Diffusion-weighted MRI is the most accurate test for cerebral ischemia.
What tests will find the embolic source after an acute ischemic stroke?
Echocardiogram, carotid duplex, and 24-hour Holter monitor. Evaluation for inherited hypercoagulability.
What is the treatment for ischemic stroke?
Tissue plasminogen activator if within 4.5 hours of symptom onset.
What are the contraindications to tissue plasminogen activator for ischemic stroke?
Stroke or head trauma within 3 mo, hemorrhage (GI, GU), surgery within 14 d, intracranial hemorrhage, arterial puncture within 7 d, BP > 185/110, anticoagulants; platelets <100,000, PT >15 sec).
What is the prognosis for patients who receive tissue plasminogen activator?
39% of all patients treated with tPA have a good functional outcome. Only 26% of untreated have a good outcome. Intracranial hemorrhage occurs in 5.8%. The fatality rate after intracranial hemorrhage is 45%.
What is the secondary prevention of ischemic stroke?
Started 24 hours after TPA. When patients have allergy to aspirin or continue to have recurrent cerebrovascular events on aspirin, dipyridamole may be added or the patient may be switched to clopidogrel.
When should heparin be given to patients with ischemic stroke?
Heparin in acute ischemic stroke is limited to patients with atrial fibrillation, basilar artery thrombosis, or stroke in evolution.
What is the treatment of subarachnoid hemorrhage?
Nimodipine to reduce the risk of ischemic stroke. Early surgical clipping of the aneurysm or embolization of the vessel. Aspirin is contraindicated.
What is the management of intracranial aneurysms that are found incidentally?
Unruptured aneurysms found incidentally should be repaired if they exceed 10 mm.
What are the indications for carotid endarterectomy?
Carotid endarterectomy for occlusion > 70% and the lesion is symptomatic. Endarterectomy may benefit those who are asymptomatic if there is >60% stenosis in men who are <60 years old.
What is a seizure?
Paroxysmal event caused by abnormally discharging central nervous system neurons. Epilepsy is a condition of recurrent seizures due to a chronic underlying process.
What are the causes of seizures?
Stroke, bleed, AV malformation; meningitis, abscess, encephalitis, trauma, vasculitis, hypoNa, hypoCa, hypoMg, hypoglycemia, hypoxia, drug overdose/withdrawal, idiopathic, neoplasm, psychiatric.
What is the presentation of a seizure?
Paroxysmal, involuntary, abnormal movement or change of consciousness. Sudden onset, with or without an aura. Disorientation, sleepiness, muscle aches. Incontinence, tongue biting, headache.
How is syncope different from a seizure?
Patients with syncope will not complain of significant postictal symptoms. Syncope patients will recover consciousness within several minutes of the event, and will not have incontinence or tongue biting.
How are seizures classified?
Partial or generalized and complex or simple.
What are partial seizures?
Partial seizures affect discrete portions of the brain, such as causing involuntary jerking of a finger or hand.
What is a simple partial seizure?
Consciousness is maintained for the duration of a simple seizure, and the seizure affects a discrete portion of brain. When there is a change in consciousness, the seizure is termed a complex partial seizure.
What are generalized seizures?
Arise from both cerebral hemisphere. Generalized tonic-clonic seizures are tonic contraction of muscles throughout the body followed by intermittent relaxation (clonic phase).
What are absence seizures?
Sudden, brief loss of consciousness without loss of postural tone. The ECG shows a generalized, symmetric 3-Hz spike-and-wave discharge pattern.
What are atonic seizures?
Atonic seizures are characterized by sudden loss of postural tone, lasting 1 to 2 seconds.
What are myoclonic seizures?
Myoclonic seizures are characterized by a sudden, brief muscle contraction.
What is status epilepticus?
Recurrent or continuous seizures lasting at least 5-30 min.
How are seizures diagnosed?
Electroencephalogram, serum electrolytes, glucose, toxicology, and arterial blood gas to exclude hypoxia. A CT scan or MRI of the head is indicated to exclude a structural lesion as the cause of seizure.
What is the treatment for status epilepticus?
Secure airway, breathing, circulation. Evaluate and treat causes. Lorazepam, which potentiates GABA receptor. If seizure continues, phenytoin or fosphenytoin to add phenobarbital.
What are the side effects of phenytoin?
Diplopia, dizziness, ataxia. Gum hyperplasia, lymphadenopathy, hirsutism, and rash.
What are the side effects of phenobarbital?
Sedation, ataxia, and rash.
What drug should be given if patients continue to seize after administration of lorazepam, phenytoin and Phenobarbital?
Midazolam or propofol anesthesia.
When should anticonvulsant therapy be started in patients with a first-time seizure?
Anticonvulsants should be started only if the patients has an abnormal neurologic exam, presented with status epilepticus, has a strong family history of seizure, or has an abnormal EEG.
What is the treatment for primary generalized tonic-clonic seizures?
Valproic acid. Valproic acid increases the availability of GABA. Lamotrigine, carbamazepine, or phenytoin are alternatives.
What are the side effects of valproic acid?
Side effects include ataxia, tremor, hepatotoxicity, thrombocytopenia, gastrointestinal irritation, and hyponatremia.
What is the mechanism of action of lamotrigine?
Lamotrigine decreases glutamate release. Side effects: Stevens-Johnson syndrome.
What is the treatment of absence seizures?
Ethosuximide. Valproic acid is an acceptable option.
What is the treatment for myoclonic and atonic seizures?
Valproic acid.
What is the treatment for partial seizures?
Carbamazepine and phenytoin are first-line therapy. Valproic acid, lamotrigine, and levetiracetam are alternatives.
What is vertigo?
False sensation of movement.
What are the causes of vertigo?
Mèniére, labyrinthitis, positional vertigo, traumatic, perilymphatic fistula, cervical vertigo. Brain stem disease, AV malformations, tumor, multiple sclerosis, drug overdose, vertebrobasilar migraine.
What are the characteristics of central vertigo?
No tinnitus or hearing loss is present. There are is often diplopia, cortical blindness, dysarthria, extremity weakness or numbness. Nystagmus is pure, vertical, does not suppress with fixation, and multidirectional.
What are the characteristics of peripheral vertigo?
Tinnitus and hearing loss. Absence of diplopia, cortical blindness, dysarthria, extremity weakness or numbness. Nystagmus is mixed, horizontal, suppresses with fixation, and unidirectional.
What are the signs of Mèniére disease?
Tinnitus, hearing loss, episodic vertigo of 1 to 8 h. Symptoms wax and wane as the endolymphatic pressure rises and falls. Most common causes of Mèniére disease are syphilis and head trauma.
What are the signs of labyrinthitis?
Sudden onset of severe vertigo that lasts for several days with hearing loss and tinnitus. Occurs with an upper respiratory tract infection.
What are the signs of benign paroxysmal positional vertigo?
Peripheral vertigo that characteristically is exacerbated by head movement or change in head position. Episodes occur in clusters that persist for several days. The vertigo usually lasts 10 to 60 seconds.
What are the causes of perilymphatic fistula?
Form of peripheral vertigo related temporally to head trauma (slap to the ear) or extreme barotrauma during air flight, scuba diving, or vigorous Valsalva maneuver.
What are the causes of central vertigo?
Caused by cerebellar or brain-stem tumor, bleed, or ischemia. Drug toxicity or overdoses. In young patient with unexplained central vertigo, consider multiple sclerosis.
What is the treatment of peripheral vertigo?
Meclizine or diazepam.
What is the treatment of Mèniére disease?
Low-salt diet and diuretics. Surgical decompression.
What is the treatment of benign paroxysmal positional vertigo?
Positional maneuvers that attempt to move the otolith out of the circular canals (e.g., Dix Hallpike and Barany maneuvers).
What is the treatment of vertigo caused by labyrinthitis?
Meclizine and diazepam.
28-year-old woman with throbbing, right-side headache; nausea; worsened by walking, loud noise, bright light. What is the diagnosis?
Migraine
What are the primary headache syndromes?
Migraine, cluster, and tension headache. Migraine affects 15% of the population.
What are the secondary causes of headache?
Intracranial hemorrhage, brain tumor, meningitis, temporal arteritis, and glaucoma.
What type of headache is characterized by fever and nuchal rigidity?
Meningitis.
"The worst headache of my life" or a "thunderclap" at onset with nuchal rigidity without fever, suggests what type of headache?
Intracranial hemorrhage.
What are the clinical symptoms of brain tumors?
Deep, dull, aching pain that disturbs sleep. Vomiting before the onset of headache, or a history of headache induced by coughing, lifting or bending is typical of posterior fossa brain tumors.
What are the characteristics of temporal arteritis?
Unilateral pounding headache with visual changes; dull and boring with lancinating pains. Polymyalgia rheumatica, jaw claudication, fever, weight loss, scalp tenderness; elderly.
What are the laboratory abnormalities associated with temporal arteritis?
Increased sedimentation rate; diagnosed with biopsy of the temporal artery. Initiate therapy with steroid immediately.
What cause of headache syndrome is associated with eye pain?
Glaucoma
What are the characteristics of migraine headaches?
Benign and recurrent syndrome of headache, nausea, vomiting, and focal neurologic deficits. Pulsatile, throbbing, unilateral, aggravated by movement. Photophobia, phonophobia.
What are the triggers of migraine?
Alcohol, chocolate, cheeses, monosodium glutamate, hunger, or irregular sleep patterns.
What is migraine without aura?
Migraine without a preceding focal neurologic deficit. Migraine with aura is a migraine headache that is accompanied by a preceding motor, sensory, or visual symptoms. Focal neurologic symptoms.
What is the pathognomonic aura for migraine?
Scintillating scotoma. Only 20% of migraine has aura. Stars, sparks, and flashes of light. Migraine equivalent is defined as focal neurologic symptoms without headache.
What are complicated migraines?
Migraines with severe neurologic deficits that persist after the resolution of pain.
What are basilar migraines?
Migraine headaches associated with vertigo, diplopia, ataxia, or dysarthria.
What are the characteristics of tension-type headaches?
Tension-type headaches are tight, band-like headaches are bilateral.
What are the characteristics of cluster headaches?
Excruciating, unilateral, periorbital headache without warning 30 min-3 hrs; 1-3 times a day for 4-week period. Red eye, lacrimation, nasal stuffiness, nausea, sensitivity to alcohol. Horner syndrome (ptosis).
How is migraine headache diagnosed?
Severe, sudden onset of a first-time headache with signs of an underlying cause should have a CT scan of the head.
What is the treatment of migraine headache?
Abortive sumatriptan, which is a serotonin receptor agonist. Dihydroergotamine is an alternative. The triptans are contraindicated in cardiovascular disease. Almotriptan, naratriptan, zolmitriptan, and eletriptan.
What is the prophylactic treatment for migraine?
Prophylactic treatment for migraine should be initiated when acute migraine headaches occur more than three to four times per month. Propranolol, timolol, valproic acid, and topiramate. Verapamil and tricyclics.
What is the treatment of cluster headaches?
The most effective treatment is 100% oxygen. Cluster headaches are treated with prednisone, lithium, ergotamine, and verapamil. Sumatriptan.
What is pseudotumor cerebri?
Idiopathic increase in intracranial pressure. 8-10 times more common in women. Association with obesity, chronic lung disease, Addison disease, oral contraceptives, tetracycline, and vitamin A toxicity.
What is the presentation of pseudotumor cerebri?
Headache, diplopia, and 6th cranial nerve (abducens) palsy. Papilledema, blind spot on visual field testing. The CT and MRI are normal, CSF is normal except for an increase in pressure.
What is the treatment for pseudotumor cerebri?
Weight loss, discontinue oral contraceptives, acetazolamide, furosemide, prednisone. Repeated lumbar punctures; placement of ventriculoperitoneal shunt.
What is trigeminal neuralgia?
Idiopathic, sudden, severe, sharp pain side of the mouth and progressing to ear, eye, or nostril. Triggered by touch or movement; pain lasts a few seconds. Compression of the trigeminal nerve root by a vessel, MS, tumor.
What is the treatment of trigeminal neuralgia?
Carbamazepine. Phenytoin, baclofen, or gabapentin. Surgery or alcohol injection into the affected nerve.
16-year-old boy with leg weakness. 3 weeks ago had diarrhea. Bilateral lower-extremity weakness and a loss of reflexes. What is the diagnosis?
Guillain-Barre syndrome
What is Guillain-Barre syndrome?
Acute, often severe polyradiculopathy caused by autoimmune destruction of myelin by an immune response.
What is the presentation of Guillain-Barre syndrome?
Sudden weakness begins lower extremities and moves upward. Loss of reflexes. Legs are more affected than arms and face. Pain, tingling. Loss of large sensory fibers, loss of reflexes and proprioception.
What autonomic instabilities are associated with Guillain-Barre syndrome?
Profuse sweating, postural hypotension, labile blood pressure, cardiac dysrhythmia occurs in severe GBS.
What is the etiology of Guillain-Barre syndrome?
75% have history of infection 1-3 weeks prior; respiratory or GI © jejuni). GBS may be preceded by HSV, CMV, EBV. Influenza vaccine associated with one case per million. HIV, SLE, lymphoma.
How is Guillain-Barre syndrome diagnosed?
A lumbar puncture has an elevated protein without an associated rise in the cell count. Electromyography will show demyelination of the peripheral nerves.
What is the treatment for Guillain-Barre syndrome?
Intravenous immunoglobulin or plasmapheresis; monitor the vital capacity and initiate early respiratory support if respiratory weakness.
A 32-year-old woman with double vision, difficulty chewing. Symptoms improve following sleep. Snarling appearance when asked to smile, nasal tone in voice; weakness in the upper extremities. What is the diagnosis?
Myasthenia gravis.
What is myasthenia gravis?
Disease of the neuromuscular junction characterized by weakness and fatigability. Acetylcholine-receptor antibodies leads to a decreased acetylcholine receptors at the postsynaptic membrane.
What is the presentation of myasthenia gravis?
Weakness, fatigability. Initially, diplopia, ptosis, dysphagia. Speech nasal; "snarling." Asymmetric proximal muscle weakness. Normal reflexes, pupillary responses, sensory. Impaired respiration.
What is Eaton-Lambert myasthenic syndrome?
Eaton-Lambert myasthenic syndrome is characterized by weakness with increasing muscle strength on repetitive contraction; caused by a malignancy, especially small-cell carcinoma of the lung.
What are the signs of botulism?
Botulism may cause myasthenic-like weakness, but pupils are usually dilated and repetitive nerve stimulation shows an incremental increase in muscular fiber contraction (opposite of myasthenia).
How is myasthenia gravis diagnosed?
Initial test for myasthenia is acetylcholine-receptor Ab, 80-90% positive. Most accurate test is EMG. Edrophonium (Tension) test is sensitive but not specific. CT performed to detect a thymoma;10%.
What is the treatment of myasthenia gravis?
Anticholinesterase (pyridostigmine, neostigmine). If glucocorticoids fail, azathioprine used. Mycophenolate. Plasmapheresis, IV Ig rapidly improve myasthenic crisis with respiratory failure.
What are the indications for thymectomy in myasthenia gravis?
Thymectomy is indicated in postpubertal patients and in those younger than 60 before initiation of immunosuppressive therapy. Aminoglycosides exacerbate myasthenia gravis.
What is amyotrophic lateral sclerosis?
Idiopathic weakness with upper and lower motor neuron loss, CN palsies, respiratory involvement; preserved bowel, bladder sensory, cognitive, sexual.
What are the signs of amyotrophic lateral sclerosis?
Cranial nerve or bulbar palsies result in dysphagia, difficulty chewing, decreased gag, dysarthria; respiratory weakness; aspiration pneumonia is most common cause of death. Weak cough; head ptosis.
What is the presentation of amyotrophic lateral sclerosis?
Fully mentally alert patient loses nearly all motor control while still being able to think and perceive.
What are the upper motor neuron manifestations of amyotrophic lateral sclerosis?
Weakness with spasticity and hyperreflexia. Lower motor neuron manifestations are weakness with muscle wasting, atrophy, fasciculations. Upper and lower motor neuron weakness is unique to ALS.
What is the confirmatory test for amyotrophic lateral sclerosis?
Confirmatory test is the electromyogram. Cerebrospinal fluid and MRI are normal.
What is the treatment of amyotrophic lateral sclerosis?
Riluzole shows progression by inhibiting glutamate release. Spasticity is treated with baclofen and tizanidine. Death in 3-5 years.
What is the presentation of multiple sclerosis?
Weakness, numbness, tingling, or unsteadiness of a limb. Urinary urgency or retention, blurry vision, double vision. Symptoms persist for weeks; may resolve spontaneously.
How is multiple sclerosis diagnosed?
MRI of the brain is the most accurate test (sensitivity 95%); demyelinated plaques in the cerebrum and spine. Enhancement with gadolinium. Mild pleocytosis (< 50 cells/mcL), total protein mildly elevated.
What is the treatment for the spasticity of multiple sclerosis?
Baclofen; tizanidine and diazepam for nocturnal spasticity. Pain caused by trigeminal neuralgia and dysesthesias responds to carbamazepine, gabapentin, phenytoin, pregabalin, tricyclic.
What is the treatment of bladder hyperactivity in multiple sclerosis?
Bladder hyperactivity is treated with oxybutynin; urinary retention is treated with bethanechol.
What is dementia?
Loss of memory with impairment of any other cognitive function (aphasia, agnosia, apraxia, executive) sufficient to interfere with social or occupational functioning.
What are the reversible causes of dementia?
Hypothyroidism, B12 deficiency, hepatic or uremic encephalopathy, vasculitis, syphilis, abscess, tumor, anticholinergics, sleep apnea, trauma, subdural hematoma, normal pressure hydrocephalus, depression.
What are the irreversible causes of dementia?
Progressive multifocal leukoencephalopathy, Alzheimer, Lewy bodies, Pick disease, multi-infarct dementia, Binswanger disease, Creutzfeldt-Jakob disease. Alzheimer accounts for 80%.
What is the prevalence of dementia?
1-5% between ages 65-69; 45% by age 100. Only 5% of Alzheimer disease is inherited.
What is the presentation of frontotemporal dementia?
Pick disease presents with personality changes early, with relative sparing of visuospatial function. Social, interpersonal, emotional abnormalities. Frontotemporal dementia lacks insight.
What is the presentation of dementia with Lewy bodies?
Dementia with Lewy bodies is characterized by fluctuating cognitive impairment. Dementia caused by Parkinson disease should be accompanied by recurrent visual hallucinations are also characteristic.
What are the characteristics of dementia caused by Creutzfeldt-Jakob disease?
Short, aggressive course over weeks. Dementia, myoclonus, ataxia, and 14-3-3 protein in CSF. Variant CJD is bovine spongiform encephalopathy from ingestion of prions from cattle.
What are the characteristics of multi-infarct dementia?
Stepwise progression associated with discrete cerebrovascular events. Binswanger disease involves the subcortical white matter and presents with a slowly progressive course.
What are the characteristics of normal pressure hydrocephalus?
Prominent gait abnormalities before the onset of cognitive impairment, urinary incontinence.
What is the diagnostic evaluation for dementia?
Rule out reversible causes of the dementia. CBC, electrolytes, calcium, creatinine, LFTs, glucose, TSH, v B12, RPR, HIV. MRI or CT with contrast.
What is the treatment for dementia?
Memantine is a disease-modifying drug used in advanced disease either alone or with a cholinesterase inhibitor. Memantine is neuroprotective and reduces the rate of progression.
A 35-year-old man with spontaneous movements, grimacing with grunting. Father died at the age of 41 from dementia. What is the diagnosis?
Huntington disease
What is the cause of Huntington disease?
Degenerative brain disorder. HD gene contains a CAG trinucleotide repeat that codes for huntingtin. Abnormal cleavage of huntingtin protein interferes with nuclear mechanisms, cell death; autosomal dominant.
What is the presentation of Huntington disease?
Chorea in 4th-5th decade. Irritability, anger, paranoia, depression. Antisocial. Fidgeting that progresses to sudden movements of the trunk or limbs. Choreic gait. Memory is preserved until late.
How is Huntington disease diagnosed?
Genetic testing. CT scanning shows cerebral atrophy or atrophy of caudate nucleus.
What is the treatment for Huntington disease?
Haloperidol or clozapine control behavioral changes. Death occurs 15-20 years after diagnosis. Children should receive genetic testing.)
58-year-old man with resting tremor; slow moving. Stooped forward, small steps. Cogwheel rigidity in arms; positive Myerson sign. What is the diagnosis?
Parkinson disease
What is Parkinson disease?
Neurologic syndrome resulting from the deficiency of the neurotransmitter dopamine; degenerative, vascular, or inflammatory changes in the basal ganglia.
What are the causes of Parkinson disease?
Neuroleptics (haloperidol, chlorpromazine), metoclopramide, alpha-methyldopa, reserpine. MPTP, carbon monoxide, cyanide, manganese. Encephalitis. Basal ganglia lesions.
What is the presentation of Parkinson disease?
Bradykinesia (mask facies, reduction of automatic movements), cogwheel rigidity, postural instability, resting tremor. Myerson sign is inability to resist blinking when trapped on the glabella.
What is Shy-Drager syndrome?
Parkinsonism with prominent orthostatic hypotension.
How is Shy-Drager syndrome diagnosed?
Diagnosis of Parkinson disease is clinical. Identify any secondary causes of Parkinsonism that are reversible. There is no diagnostic test that can identify Parkinson disease.
What is the treatment for Parkinson disease?
Parkinson disease is caused by low dopaminergic and excessive cholinergic tone on the basal ganglia. Medical treatment increases dopaminergic tone or decreases cholinergic tone on the basal ganglia.
What medications are used to stimulate dopamine receptors in patients with Parkinson disease?
Carbidopa/levodopa, dopamine agonists, indirectly increase the amount of dopamine (COMT inhibitors, selegiline, amantadine).
Which medications block acetylcholine stimulation of the basal ganglia?
Benztropine and trihexyphenidyl.
What direct-acting dopamine agonists are used to treat Parkinson disease?
Pramipexole or ropinirole alone as initial therapy or with levodopa/carbidopa. Other dopamine agonists are bromocriptine, pergolide, tolcapone, entacapone.
How does the efficacy of pramipexole and ropinirole compare to levodopa for the treatment of Parkinson disease?
Dopamine agonists are less efficacious than levodopa. Dopamine agonists have less dyskinetic side effects.
What is the mechanism of the catechol-O-methyl transferase inhibitors?
COMT inhibitors decrease the metabolism of the levodopa. They are an adjunct to levodopa for the treatment of Parkinson disease.
What is the role of selegiline in Parkinson disease?
Selegiline can be used in those with a declining or fluctuating response to levodopa. Selegiline offers mild symptomatic benefit in early disease. Rasagiline is a newer version.
What is benign essential tremor?
Idiopathic disorder of isolated tremor of the hands, head. Worsened by caffeine or beta agonists. Interferes with ability to write. Improvement with the use of alcohol.
What is the treatment of benign essential tremor?
Propranolol. Alternatives are primidone, alprazolam, and clozapine. If no medical therapy is effective, thalamotomy is indicated.
What is restless leg syndrome?
Idiopathic sensation of creeping and crawling dysesthesia within legs; involuntary movements during sleep; familial; exacerbated by sleep deprivation, caffeine, pregnancy.
What is the treatment of restless leg syndrome?
Treatment is with dopamine agonists, such as pramipexole or ropinirole; levodopa/carbidopa, benzodiazepines.
What is pemphigus vulgaris?
Autoimmune disease of allergy to own skin. Pemphigus vulgaris is most often idiopathic, but ACE inhibitors or penicillamine can cause it.
What is the presentation of pemphigus vulgaris?
Vulgaris occurs in thirties and forties, whereas bullous pemphigoid occurs in those in their seventies and eighties. Pemphigus vulgaris is a potentially life-threatening disease. Vulgaris has oral erosions.
What is Nikolsky sign?
Removal of skin by a little pressure from the examiner's finger, pulling skin off like a sheet in pemphigus vulgaris, staph scalded skin syndrome, toxic epidermal necrolysis. Lesions of pemphigus vulgaris are painful.
How is pemphigus vulgaris diagnosed?
Biopsy the skin with immunofluorescent stains.
What is the treatment for pemphigus vulgaris?
Systemic glucocorticoids, such as prednisone; azathioprine, mycophenolate, or cyclophosphamide. Rituximab and IVIg are also effective.
What is pemphigus foliaceus?
Blistering disease often associated with other autoimmune diseases, or it can be induced by ACE inhibitors or NSAIDs.
What is the presentation of pemphigus foliaceus?
Foliaceus is much more superficial, and intact bullae are not seen because they break. There are no oral lesions. Treatment of pemphigus foliaceus is steroids.
What is bullous pemphigoid?
Pemphigoid is twice as common as pemphigus vulgaris and occurs in elderly persons in their seventies and eighties. May be induced with sulfa drugs, furosemide; penicillamine.
How is bullous pemphigoid diagnosed?
Biopsy with immunofluorescent antibodies.
What is the treatment of bullous pemphigoid?
Systemic steroids, such as prednisone; topical steroids only if no oral lesions.
What is porphyria cutanea tarda?
Deficiency of uroporphyrinogen decarboxylase results in accumulation of porphyrins, which leads to photosensitivity; HIV, alcoholism, liver disease, hepatitis C, oral contraceptives.
What is the presentation of porphyria cutanea tarda?
Fragile, nonhealing blisters on sun-exposed backs of the hands and the face. Hyperpigmentation of the skin and hypertrichosis of face.
How is porphyria cutanea tarda diagnosed?
Uroporphyrins are elevated two to five times above the coproporphyrins.
What is the treatment for porphyria cutanea tarda?
Stop alcohol and discontinue estrogen, and iron. Chloroquine increases the excretion of porphyrins.
What is acute urticaria?
Hypersensitivity reaction with IgE, mast cell activation; wheals, hives, without hypotension.
What are the causes of acute urticaria?
Allergic reaction to insect bites, foods, emotions, aspirin, NSAIDs, morphine, codeine, penicillin, phenytoin, quinolones, ACEI. peanuts, shellfish, tomatoes, strawberries, latex.
What is the presentation of acute urticaria?
Lasts <6 weeks and self-limited: onset of wheals and hives within 30 min, lasts for <24 h. Itching. Chronic urticaria lasts >6 weeks and is associated with pressure on the skin, cold, or vibration.
What is the treatment of acute urticaria?
H1 antihistamines. Severe, urticaria is treated with diphenhydramine, hydroxyzine, or cyproheptadine. H2 antihistamines, systemic steroids. Chronic therapy is with nonsedating antihistamines, such as loratadine.
What is a morbilliform rash?
Milder hypersensitivity reaction than urticaria. Typical type of drug reaction and lymphocyte mediated.
What is the presentation of morbilliform rash?
Usually caused by medications allergy, such as penicillin, sulfa drugs, allopurinol, or phenytoin. Generalized, maculopapular eruption that blanches with pressure.
What is the treatment of morbilliform rash?
Antihistamines and steroids.
What is erythema multiforme?
Caused by penicillins, phenytoin, NSAIDs, sulfa drugs. Majority of cases follow infection with herpes simplex or Mycoplasma.
What is the presentation of erythema multiforme?
Target-like lesions that occur on the palms and soles. Bullae are not found, and it does not involve mucous membranes. Treatment is antihistamines.
What is Stevens-Johnson syndrome?
Stevens-Johnson syndrome is erythema multiforme major. SJS and toxic epidermal necrolysis are considered to be degrees of same disorder. Penicillin, sulfa drugs, NSAIDs, phenytoin or phenobarbital.
What is the presentation of Stevens-Johnson syndrome?
SJS involves <10-15% of the total body surface area; the overall mortality rate is <5-10%. Mucous-membrane involvement of oral and conjunctivae, respiratory tract.
What is the treatment for Stevens-Johnson syndrome?
Admission to a burn unit, withdrawal of offending drug, mechanical ventilation. Death occurs from infection, dehydration, malnutrition. No benefit from steroids.
What is toxic epidermal necrolysis?
Toxic epidermal necrolysis is the most serious cutaneous hypersensitivity reaction. Mortality 40-50%.
What is the presentation of toxic epidermal necrolysis?
Skin sloughing involving a body surface area (BSA) 30 to 100%. Nikolsky sign, and the skin easily sloughs off. TEN is drug induced.
How is toxic epidermal necrolysis diagnosed?
Skin biopsy reveal full thickness epidermal necrosis in which the epidermis separates from the dermis. A skin biopsy is usually not necessary.
What is the treatment for toxic epidermal necrolysis?
Topical anesthetics, cover denuded skin with saline or Burow solution compresses. Sepsis is the most common cause of death. Systemic steroids are not effective.
What is a fixed drug reaction?
Recurrent localized allergic reaction to aspirin, NSAIDs, tetracycline, barbiturates. Round, sharply demarcated, and leave a hyperpigmented spot. Treated with topical steroids.
What is erythema nodosum?
Localized inflammatory condition, occurring after infections inflammatory conditions, pregnancy, strep infections, coccidioidomycoses, histoplasmosis, sarcoidosis, IBD, syphilis, hepatitis, Yersinia.
What is the presentation of erythema nodosum?
Multiple painful, red, raised nodules on anterior lower extremities; tender, do not ulcerate, last 6 weeks. ASLO titers can reveal a streptococcal infection.
What is the treatment for erythema nodosum?
Analgesics and NSAIDs. Potassium iodide solution; usually self-limiting.
What are the superficial fungal infections?
Tinea pedis, cruris, corporis, versicolor, capitis, and onychomycosis
What is the appearance of tinea versicolor on KOH?
Malassezia furfur organism appears as "spaghetti and meatballs" on KOH. The most accurate test is fungus culture, but this is not practical because dermatophytes take up to 6 weeks to grow.
What is the treatment for onychomycosis or hair infection (tinea capitis)?
Terbinafine or itraconazole for 6 weeks for fingernails and 12 weeks for toenails. Terbinafine is hepatotoxic. Griseofulvin must be used for 6-12 months and has much less antifungal efficacy than terbinafine.
What is the treatment of dermatophyte infection of the hair?
Griseofulvin for 6-8 weeks for tinea capitis. Terbinafine for 2-4 weeks.
What is the treatment of fungal infections of the skin that do not involve the hair or nails?
Topical ketoconazole, clotrimazole, econazole, terbinafine, miconazole, sertaconazole, sulconazole, tolnaftate, or naftifine.
What is tinea versicolor?
Tan to pink lesions. KOH and fungal culture reveals dermatophytes. Topical selenium sulfide every 2 to 3 weeks or oral therapy with itraconazole or fluconazole.
What are the most common bacterial organisms that cause skin infections?
Staphylococcus and Streptococcus.
What antibiotics are used to treat Staphylococcus?
Dicloxacillin, cephalexin, cefadroxil, cefazolin, nafcillin, oxacillin. If allergic to penicillin was a rash, then cephalosporins can be safely used. <5% cross-reactivity between penicillin and cephalosporins.
What is the treatment of staphylococcal skin infections if the patient is allergic to penicillin?
Cephalosporins cannot be used if anaphylaxis. Alternatives for skin are macrolides, or extended fluoroquinolones (levofloxacin, gatifloxacin, moxifloxacin). Ciprofloxacin will not cover skin.
What is impetigo?
Superficial bacterial infection of the skin; usually caused by Staphylococcus, but is sometimes caused by Streptococcus pyogenes.
What is the presentation of impetigo?
Weeping, oozing, honey colored crusts; warm, humid conditions. Contagious and autoinoculable. Impetigo may cause glomerulonephritis, but not fever.
What is the treatment of impetigo?
Topical mupirocin or retapamulin; antistaphylococcal oral antibiotics, such as dicloxacillin or cephalexin (Keflex).
What is erysipelas?
Dermis and epidermis infection by group A Streptococcus (pyogenes). Fever, chills, and bacteremia; involves the face, giving a bright-red, swollen appearance.
What is the treatment for erysipelas?
Oral dicloxacillin or intravenous nafcillin or cefazolin.
What is cellulitis?
Bacterial infection of the dermis and subcutaneous tissues with Staphylococcus and Streptococcus.
What is the presentation of cellulitis?
Redness, swelling, and warmth and tenderness of the skin; no oozing, crusting, weeping, or draining.
What is the treatment for cellulitis?
Dicloxacillin or intravenous oxacillin, nafcillin, or cefazolin.
What are folliculitis, furuncles, and carbuncles?
Folliculitis, furuncles, and carbuncles represent three different degrees of severity of staphylococcal infections occurring around hair follicle. Folliculitis can be caused by Pseudomonas from a whirlpool or hot tub.
What is the presentation of a folliculus?
Folliculitis starts as a superficial infection around a hair follicle progressing to a small collection of infected material known as a furuncle. Several furuncles merge into a single carbuncle, which is a skin abscess.
What is the treatment for folliculitis?
Warm compresses; mupirocin. Furuncles and carbuncles require systemic antistaphylococcal antibiotics. Carbuncles require dicloxacillin or cefadroxil or IV nafcillin and surgical drainage.
What is necrotizing fasciitis?
Severe, life-threatening infection that starts as a cellulitis that dissects into the fascial planes of the skin. Streptococcus and Clostridium produce a toxin. Diabetes increases risk of fasciitis.
What is the presentation of necrotizing fasciitis?
High fever, portal of entry into the skin, pain, bullae, palpable crepitus.
How is necrotizing fasciitis diagnosed?
Elevated creatine phosphokinase; x-ray, CT scan or MRI show air in the tissue or necrosis.
What is the treatment for necrotizing fasciitis?
Surgery. beta-Lactam/beta-lactamase combination medications, such as ampicillin/sulbactam (Unasyn), ticarcillin/clavulanate (Timentin), or piperacillin/tazobactam (Zosyn).
What is paronychia?
Infection under the skin surrounding a nail.
What is the treatment of paronychia?
Treatment: drainage, dicloxacillin, cefadroxil or cephalexin orally, or oxacillin, nafcillin, or cefazolin intravenously.
What is herpes simplex?
Infection of the genitals, multiple, painful vesicles. Most accurate diagnostic test is a viral culture, which will grow in 24 to 48 hours. Serology is not useful.
What is the treatment for herpes simplex?
Acyclovir, famciclovir, or valacyclovir. Topical acyclovir has little efficacy in primary lesions and will not prevent recurrent lesions. Acyclovir-resistant herpes is treated with foscarnet.
What are the complications of varicella?
Pneumonia, hepatitis, dissemination. Dermatomal herpes zoster occur more frequently in elderly and with immune defects. The vesicles are 2 to 3 mm in size on an erythematous base.
How is varicella diagnosed?
Tzanck prep and viral culture
What is the treatment for varicella?
If immunocompromised child or primary infection in adult, then acyclovir, valacyclovir, or famciclovir.
What is the treatment of zoster?
Acyclovir decreases risk of postherpetic neuralgia. Steroids in elderly with zoster. Postherpetic neuralgia is treated with gabapentin.
What is human papilloma virus?
Responsible for sexually transmitted anogenital and oral warts (condylomata acuminata). Lesions, are translucent, white, or flesh, and are located on skin and mucosal surfaces.
What is the treatment for condyloma acuminata?
Cryotherapy with liquid nitrogen, laser removal, and trichloroacetic acid or podophyllin (teratogenic). Imiquimod is a local immunostimulant that takes 14 weeks to result in sloughing off of the wart.
What is the presentation of primary syphilis?
The chancre of primary syphilis is an ulceration with heaped-up, indurated edges that is painless. Causative organism is Treponema pallidum.
What is secondary syphilis?
Generalized copper-colored, maculopapular rash that is more intense on the palms and soles of the feet. The other manifestations of secondary syphilis are mucous patch, alopecia areata, and condylomata lata.
How is primary syphilis diagnosed?
Initial test darkfield. False-neg rate 25% for VDRL/RPR. Serologic tests need several weeks to become positive, and only 75% sensitive in primary. For secondary syphilis, VDRL/RPR tests are 100% sensitive.
What is the treatment of syphilis?
Primary and secondary syphilis are treated with a single, intramuscular dose of penicillin; penicillin allergic patients are treated with doxycycline orally for 2 weeks.
What is scabies?
Vesicular eruptions resulting from Sarcoptes scabiei (hominis) mites burrowing into the skin.
What is the presentation of scabies?
Web spaces of hands, feet, pruritic lesions around penis, breasts. Itching, burrows and excoriations around small pruritic vesicles. Spares head. Patients with HIV have severe crusting, known as Norwegian scabies.
How is scabies diagnosed?
Scraping out the organism after mineral oil is applied to a burrow (not routinely done).
What is the treatment for scabies?
Permethrin. Lindane has equal efficacy, but greater toxicity. Norwegian scabies can be treated with ivermectin orally.
What is pediculosis?
Pediculosis (lice and crabs from the genus Pediculus) tends to involve the head. Both P. humanus (lice) and P. pubis (crabs) can be sexually transmitted.
What is the treatment for pediculosis?
Over-the-counter pyrethrins, Lindane.
What is the cause of Lyme disease?
Borrelia burgdorferi spirochete, transmitted by the deer tick (Ixodes scapularis).
What is the presentation of Lyme disease?
Erythematous with central clearing at least 5 cm in diameter. 7 to 10 days after the tick bite.
How is Lyme disease diagnosed?
This rash is more important than serologic testing for diagnosis.
What is the treatment for Lyme disease?
Doxycycline, amoxicillin, or cefuroxime. Without therapy, two-thirds develop joint disease. Some develop neurologic or cardiac disorders.
What is toxic shock syndrome?
Systemic reaction to a toxin produced by Staphylococci in a tampon, vaginal packing, nasal packing, retained sutures, or other surgical material.
What is the clinical presentation of toxic shock syndrome?
Presence of 3 or more or of the following: fever >102, SBP <90, desquamative rash, vomiting, involvement of mucous membranes of the eyes, mouth, or genitals, elevated bilirubin, platelets <100,000.
What is the treatment for toxic shock syndrome?
Remove the source of the infection; fluid resuscitation, dopamine, and clindamycin plus vancomycin.
What is staphylococcal scalded skin syndrome?
Occurs in infants and young children and in the immunocompromised. Mediated by a toxin from Staphylococcus, loss of the superficial layers of the epidermis in sheets.
What is the treatment for staphylococcal scalded skin syndrome?
Managed in a burn unit and nafcillin.
What is anthrax?
Bacillus anthracis cutaneous infection acquired from contact with infected livestock; woolsorters, bioterrorism.
What is the presentation of anthrax?
Papule appears that later becomes inflamed and develops with central necrosis, which is black in color.
How is anthrax diagnosed?
Gram stain and culture.
What is the treatment of anthrax?
In lesions due to occupational exposure, penicillin V or doxycycline. Bioterrorism: ciprofloxacin.
What are the characteristics of benign skin lesions?
Benign lesions do not grow in size and have smooth, regular borders with a diameter usually <1 cm. Homogenous in color.
What is seborrheic keratosis?
Benign hyperpigmented lesions in the elderly with a "stuck on" appearance; most common on the face shoulders, chest, and back.
What is the treatment for seborrheic keratosis?
Liquid nitrogen or curettage.
What are actinic keratoses?
Precancerous lesion on sun-exposed areas of the body in older persons. 1:1,000 progresses to squamous cell cancer. Light in color. Removed with cryotherapy, 5FU, imiquimod.
What is the presentation of melanoma?
Grow in size, irregular borders shape, inconsistent color.
What is lentigo maligna melanoma?
Lentigo maligna melanoma arises on sun-exposed body parts in the elderly. Acral-lentiginous melanoma arises on the palms, soles of feet, and nail beds.
What is the treatment for melanoma?
Melanoma is removed by excision. Interferon reduces the recurrence rates of melanoma.
What is squamous cell carcinoma?
Develops on sun-exposed skin surfaces in elderly patients. Ulceration. Metastases are rare.
What is basal cell carcinoma?
Sixty-five to eighty percent of all skin cancers are basal cell. Shiny or "pearly" appearance. Rate of metastases is <0.1%.
What is the treatment of basal cell carcinoma?
Mohs microsurgery. 5-FU for superficial lesions.
What is Kaposi sarcoma?
Purplish lesions found on the skin predominantly of patients with HIV and CD4 counts <100/mm3. Human herpes virus 8 is the causative organism.
What is the treatment for Kaposi sarcoma?
Antiretroviral therapy to raise the CD4 count. Cryotherapy liposomal doxorubicin hydrochloride or vinblastine.
What is the presentation of psoriasis?
Silvery scales on extensor surfaces. Local or extensive. Nail pitting. Koebner phenomenon.
What is the treatment for psoriasis?
Salicylic acid, coal tar or anthralin derivatives. Biologic agents, such alefacept, efalizumab, etanercept, and infliximab, which are monoclonal antibodies.
What is atopic dermatitis?
Pruritic disorder with high IgE levels. Red, itchy plaques appear on the flexor surfaces, cheeks, lichenification.
What is the treatment for atopic dermatitis?
Emollients, avoid hot water and drying soaps; wear cotton clothes. Topical steroids, antihistamines, coal tars, phototherapy. Antistaphylococcal antibiotics. Topical doxepin.
What is seborrheic dermatitis?
Oversecretion of sebaceous material and a hypersensitivity reaction to a superficial fungus, Pityrosporum ovale. Scaly, greasy, flaky skin is found on a red base on the scalp, eyebrows, and in the nasolabial fold.
What is the treatment for seborrheic dermatitis?
Low-potency hydrocortisone, or topical antifungals in the form of shampoos, such as ketoconazole or sulfide.
What is contact dermatitis?
Hypersensitivity reaction to soaps, detergents, latex, sunscreens, or neomycin, jewelry, nickel.
What is the presentation of contact dermatitis?
Linear, vesicles, particularly when it is from poison ivy.
What is the treatment of contact dermatitis?
Antihistamines and topical steroids.
What is pityriasis rosea?
Pruritic eruption that begins with a "herald patch". Mild, self-limited, and usually resolves in 8 weeks without scarring.
What is the presentation of pityriasis rosea?
Salmon colored patches that are similar to secondary syphilis, except spares the palms and soles and has a herald patch. Lesions on the back appear in a pattern like a Christmas tree. VDRL negative.
What is alopecia areata?
Autoimmune disease in which antibodies attack the hair follicles.
What is telogen effluvium?
Loss of hair in response to physiologic stress, such as cancer or malnutrition.
What is the treatment for acne?
Topical clindamycin or erythromycin. Benzoyl peroxide. Retinoids: tazarotene, tretinoin, and adapalene. Severe cystic acne is treated with minocycline, tetracycline, clindamycin, oral isotretinoin (teratogenic).
What are the steps in assessing the technical quality of a chest x-ray?
Determine if rotated by determining if trachea and spinous apophysis are midway between the clavicles. Ten ribs visible. Underexposed film appear too white. Overexposure is too dark.
What are the chest x-ray signs of chronic obstructive pulmonary disease and emphysema?
Hyperinflation of the lung causes darkening of the lung fields; flat the diaphragm and tubular-shaped heart. Increased anterior/posterior diameter, or "barrel chest," bullae are thin, white lines.
What are the chest x-ray signs of pneumonia?
Whitening of lobe. "Silhouette" sign: border between the affected lobe and a structure is obscured. Lower lobe pneumonia causes a silhouette of diaphragm. Right middle-lobe pneumonia obscures right heart border.
What are the signs of interstitial pneumonia?
Disease of the lung in the interstitial space produces a fine, lacy appearance visible in most of the lobes.
What disorders cause interstitial infiltrates?
Pneumocystis pneumonia, Mycoplasma, viruses, Chlamydia, pulmonary fibrosis caused by silicosis, asbestosis, mercury poisoning, berylliosis, idiopathic pulmonary fibrosis.
What are the chest x-ray signs of congestive heart failure?
Fluid overload causes the blood vessels toward the apices become fuller, which is pulmonary vascular congestion, or "cephalization" of flow. Cardiomegaly, pleural effusions, and Kerley B lines.
What are Kerley B lines?
Kerley B lines are small, horizontal lines at the bases that represent fluid in the interlobular septa.
What is the proper position of the tip of an endotracheal tube on a chest x-ray?
1 to 2 cm above the carina.
What is the correct position of a central line on a chest x-ray?
Tip at junction of the superior vena cava and the right atrium, at right main stem bronchus. The tip of the line should not be inside the atrium because it can irritate the heart and provoke an arrhythmia.
What conditions can cause air under the diaphragm on a chest x-ray?
Perforation of esophagus or ulcer. In the upright position. Air is also visible after surgery or laparoscopy.
What are the indications for high resolution CT scan?
Symptomatic patients with a normal chest x-ray to detect metastatic lesions, solitary nodules, bullae, bronchiectasis, and diffuse parenchymal disease. CT is used as a guide to determine the site of biopsy.
What conditions are detected by abdominal x-rays?
Abdominal x-rays are useful only in the detection of an abdominal obstruction, such as an ileus or a volvulus.
What is the best method of detecting mass lesions of the abdomen?
Mass lesions in all abdominal organs are best detected with CT scanning or an MRI of the abdomen. Polyps are detected by colonoscopy. Ascites is visualized by ultrasound or CT scanning.
What is the best test for detecting inflammatory bowel disease, diverticulitis, or colon cancer?
Inflammatory bowel disease, diverticulosis, and cancer are detected by either endoscopy or barium studies of the bowel.
What are the best tests for detecting kidney stones?
Kidney stones are best detected by sonography or CT scanning.
What are the best tests for detecting gallstones?
Only 10 to 15% of gallstones can be detected on an abdominal film; the best tests for gallstones are sonography and CT scanning.
What is the role of sonography in detecting abdominal lithiasis?
Sonograms should be used first for evaluation of the biliary tract for cholelithiasis. Nephrolithiasis is visible by sonography, although there is less accuracy because retroperitoneal location.
What is the role of sonography in evaluating the abdomen?
Sonography is useful in the evaluation of masses in the liver, spleen, pancreas, pelvis, and for evaluating the presence of ascites. CT has a greater sensitivity and specificity for the abdomen and pelvis.
What is endoscopic retrograde cholangiopancreatography?
Contrast is injected to visualize the biliary tract and pancreatic structures for detecting strictures, stones, and neoplasms. ERCP can remove stones and dilate strictures and biopsy.
What is capsule endoscopy?
Allows direct visualization of the small bowel by swallowing a camera. If a patient has serious bleeding and both upper and lower endoscopy do not reveal the source, capsule endoscopy is indicated.
What is HIDA scanning?
Nuclear scan useful only in the detection of acute cholecystitis. Useful in patients in whom the diagnosis of cholecystitis is not clear. An abnormal or positive test is lack of visualization of the gallbladder.
What is positron emission tomography?
Useful to determine if lesions are malignant or benign. Cancer is associated with increased uptake of fluorodeoxyglucose. Used after chemotherapy to assess the presence of residual cancer.
What is the most accurate test for evaluating central nervous system lesions?
Magnetic resonance imaging is superior for the detection of stroke, cancer, multiple sclerosis, infections, and in the evaluation of the posterior fossa, such as the cerebellum and brainstem.
What is the sensitivity of MRI in detecting ischemic strokes?
>90% within the 24 h after onset, whereas the CT needs 3 days before >90% of ischemic strokes are visible. CT is superior to MRI in detection of blood. CT is the best for trauma and to exclude hemorrhagic stroke.
What are the indications for using contrast during a head CT scan?
Contrast is indicated for the detection of cancers and infection. When an abscess or neoplastic process is present, extravasation of contrast is visible as "ring"-enhancing lesion.
What tests are useful for diagnosing osteomyelitis?
X-ray is done first. Plain x-rays lack sensitivity; the specificity for osteomyelitis is excellent. Earliest finding of osteomyelitis is elevation of the periosteum. The MRI has high sensitivity (95%) and specificity (95%).
What is diabetic retinopathy?
Caused by damage to the endothelial lining of small blood vessels, leading to occlusion. The earliest form is nonproliferative retinopathy, characterized by dilation of veins, microaneurysms, edema, hemorrhages.
What is proliferative retinopathy?
Proliferative retinopathy progresses rapidly to blindness. "Neovascularization," or new blood vessel formation leads to new vessel formation. Intravitreal hemorrhages. Treatment is laser photocoagulation.
What are the predisposing factors to retinal detachment?
Spontaneous, but may result from trauma. Predisposing factors are myopia and surgical extraction of cataracts; proliferative retinopathy, retinal vein occlusion, and age-related macular degeneration.
What is the presentation of retinal detachment?
Blurry vision developing in one eye without pain or redness. Floaters, flashes, "curtain coming down."
What are the causes of central retinal artery occlusion?
Carotid artery embolic disease, temporal arteritis, cardiac thrombi or myxoma, or thrombophilia, such as factor V Leiden mutation.
What is the presentation of retinal artery occlusion?
Sudden, painless, unilateral loss of vision. No redness. Pale retina, with diminished perfusion and a "cherry- red" spot at the fovea.
What is the treatment of central retinal artery occlusion?
Lay the patient flat and give oxygen; ocular massage in an attempt to unobstruct the vessel. Acetazolamide and thrombolytics.
What is the presentation of central retinal vein occlusion?
Sudden loss of vision without pain, redness, or abnormality in pupillary dilation. Ocular examination by funduscopy reveals disk swelling, venous dilation, tortuosity, and retinal hemorrhages.
What medications can cause acute angle-closure glaucoma?
Precipitated by anticholinergic medications, such as ipratropium bromide or tricyclic antidepressants.
How is open-angle glaucoma diagnosed?
Cup-to-disk ratio of >0.3; confirmed by an elevation in intraocular pressure by tonometry. This disorder accounts for >90% of cases of glaucoma; asymptomatic.
What medications are useful for decreasing the production of the aqueous humor in glaucoma?
Beta-blockers (timolol, betaxolol, levobunolol), alpha-adrenergic agonists (apraclonidine, brimonidine), and carbonic anhydrase inhibitors (dorzolamide and brinzolamide).
What treatments increase the outflow of aqueous humor in the treatment of glaucoma?
Prostaglandin analogs, such as latanoprost, travoprost, bimatoprost. Can change the color of the eyes and darken eyelid. Pilocarpine is a miotic agent that increases outflow of aqueous humor. Laser trabeculoplasty or surgical trabeculectomy.
What is the presentation of closed-angle glaucoma?
Eye is red, painful, hard, fixed midpoint pupil. The cornea has a hazy cloudiness, diminished visual acuity. Precipitated by anticholinergic medications.
What is the treatment of closed-angle glaucoma?
IV acetazolamide, urea, osmotic diuretics (mannitol or glycerol). Pilocarpine can be used to open the canal of Schlemm; beta-blockers decrease humor production. Laser trabeculoplasty.
What are the causes of infectious conjunctivitis?
Bacteria, viruses, and fungi.
What is the presentation of bacterial conjunctivitis?
Unilateral, purulent discharge. Crusting overnight. Less itching compared with viral conjunctivitis. Red eye, normally reactive pupil, normal ocular pressure, and no visual impairment.
What is the presentation of viral conjunctivitis?
Viral conjunctivitis is more often bilateral in nature, with much more severe ocular itching and enlarged preauricular adenopathy. The eyes are also red, the pupil reacts normally, and there is no photophobia.
What is the treatment of bacterial conjunctivitis?
Erythromycin ointment, sulfacetamide drops, or topical fluoroquinolones.
What is the treatment of viral conjunctivitis?
Topical antihistamine and decongestants.
What is subconjunctival hemorrhage?
Most commonly caused by trauma. The hematoma stops at the limbus, which is the junction between the conjunctiva and the cornea. There is no impairment of vision.
What is keratitis?
Any infection or inflammation of the cornea.
What is the presentation of herpes simplex keratitis?
Severe pain in the eye and a foreign body sensation under the eyelid.
How is herpes simplex keratitis diagnosed?
Characteristic dendritic pattern over the cornea on fluorescein staining of the eye under a blue light.
What is the treatment for herpes simplex keratitis?
Oral acyclovir, famciclovir, or valacyclovir and topical trifluridine 1% solution or idoxuridine.
What is periorbital cellulitis?
Cellulitis caused by Staphylococcus aureus or Streptococcus infecting the dermis and subcutaneous tissues surrounding the eye.
What is the treatment for periorbital cellulitis?
Antistaphylococcal penicillins, such as oxacillin or nafcillin. If there is an allergic rash to penicillins, then first-generation cephalosporins can be used (cefazolin).
What is uveitis?
Uveitis is inflammation of the iris, ciliary body, and choroid. Uveitis is caused by psoriasis, sarcoidosis, syphilis, reactive arthritis, and inflammatory bowel disease.
What is the presentation of uveitis?
Painful, red eye with photophobia. Pain also occurs when shining a light in the unaffected eye.
How is uveitis diagnosed?
Slit lamp examination. Inflammation of the iris, ciliary body, and choroid.
What is the treatment for uveitis?
Topical or systemic steroids.