• 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

1/1837

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;

1837 Cards in this Set

  • Front
  • Back
Common congenital malformations: Name 7 common congenital malformations
(1) Heart Defects (congenital rubella) (2) Hypospadias (3) Cleft lip with or without cleft palate (4) congenital hip dislocation (5) spina bifida (6) anencephaly (7) pyloric stenosis (associated with polyhydramnios & projectile vomiting)
Common congenital malformations: Name 2 neural tube defects (NTDs)
Spina bifida and anencephaly
Common congenital malformations: Name 2 diagnostic associations with NTDs during gestation
increased serum and amniotic fluid levels of a-fetoprotein
Common congenital malformations: How can the incidence of NTDs be reduced?
Folate ingestion during pregnancy
Congenital Heart Disease: What kind of shunts cause early cyanosis?
R-to-L "blue babies"
Congenital Heart Disease: What kind of shunts cause late cyanosis?
L-to-R "blue kids"
Congenital Heart Disease: Name 3 common R-to-L shunts.
The 3 T's: Tetralogy of Fallot (most common cause of early cyanosis), Transposition of the great vessels, truncus arteriosus
Congenital Heart Disease: What might you see children with a R-to-L shunt do to relieve discomfort?
Squat to increase venous return
Congenital Heart Disease: Name 3 common L-to-R shunts.
VSD, ASD, PDA
Congenital Heart Disease: Rank them in order of frequency
VSD > ASD > PDA
Congenital Heart Disease: What are the auscultatory signs of an Atrial Septal Defect?
Loud S1; wide, fixed split S2
Congenital Heart Disease: How do you treat PDA?
Indomethacin
Congenital Heart Disease: Name 3 sequelae of L-to-R shunts.
1) Increased pulmonary resistance due to arteriolar thickening. 2) Progressive pulmonary HTN 3) R-to-L shunt (Eisenmenger's)
Eisenmenger's Syndrome: What are signs/symptoms of Eisenmenger's?
Late cyanosis, clubbing, polycythemia
Tetrology of Fallot: What is the Tetrology?
(1) Pulmonary Stenosis (2) RVH (3) Overriding aorta (4) VSD (mnemonic: PROVe)
Tetrology of Fallot: What is the direction of the shunt in the tetralogy?
right-to-left across the VSD
Tetrology of Fallot: What is the x-ray finding on the tetralogy?
Boot-shaped heart due to RVH
Tetrology of Fallot: What is the embryological cause of the tetralogy?
Anterosuperior displacement of the infundibular septum.
Transposition of great vessels: The transposition occurs because of a failure of the --------------- --------- to -------
failure of the aorticopulmonary septum to spiral
Transposition of great vessels: In transposition of the great vessels, the aorta leaves ------------ and the pulmonary trunk leaves the ------------
aorta leaves the RV (anterior) and the pulmonary trunk leaves the LV (posterior)
Transposition of great vessels: How might transposition of the great vessels, be compatible with life?
If a shunt (e.g. VSD, PDA or patent foramen ovale) exists, blood from the systemic and pulmonary circulations can mix. Clinicians use PGE to keep PDA open.
Transposition of great vessels: Transposition of the great vessels is a common congenital heart disease in offspring of mothers with what disease?
Diabetes
Coarctation of aorta: Where is the aortic stenosis in the infantile type of coarctation?
Preductal (proximal to insertion of ductus arteriosus) - INfantile (in close to heart)
Coarctation of aorta: Where is the aortic stenosis in the adult type of coarctation?
Postductal (distal to ductus arteriosus). - ADult (Distal to Ductus)
Coarctation of aorta: Name 3 symptoms in adult type coarctation?
(1) Notching of the ribs (2) HTN in uper extremities (3) Weak pulses in uper extremities
Coarctation of aorta: What is the Male-to-Female ratio in coarctation?
3-to-1
Coarctation of aorta: What pulses should you check on physical exam?
Femoral
Patent ductus arteriosus: Which direction is the shunt in a neonate with PDA?
Left-to-right
Patent ductus arteriosus: What change occurs to the heart in PDA?
RVH and failure
Patent ductus arteriosus: What is the auscultatory finding in PDA?
Continuous, "machine-like" murmur.
Patent ductus arteriosus: How is patency maintained in PDA?
prostaglandin (PGE) synthesis and low Oxygen tension.
Patent ductus arteriosus: What drug is used to close a PDA?
Indomethacin
Patent ductus arteriosus: How is a PDA kept open?
porstaglandin--PGE
Patent ductus arteriosus: Why would you want to keep a PDA open?
To sustain life in conditions such as transposition of the great vessels.
Autosomal trisomies: What is the most common chromosomal disorder and cause of congenital mental retardation?
Down Syndrome (trisomy 21), 1:700
Autosomal trisomies: In Down syndrome, 4% of cases are due to ------- translocation, and 1% are due to -------- mosaicism
Robertsonian; Down
Autosomal trisomies: 95% of Down syndrome cases occur because of --------- --------- of homologous chromosomes
meiotic nondisjunction
Autosomal trisomies: What is the type and cause of the most common congenital heart malformation in Down syndrome?
Septum-primum-type ASD due to endocardial cushion defects
Autosomal trisomies: Name 8 findings in trisomy 21
(1) mental retardation (2) flat facial profile (3) prominent epicanthal folds (4) simian crease (5) duodenal atresia (6) congenital heart disease (7) Alzheimer's in affected >35 yrs old (8) Increased risk of ALL
Autosomal trisomies: Name a risk factor for Down Syndrome
Advanced maternal age (1:500 < 20 yo; 1:25 > 45 yo)
Autosomal trisomies: What is the name of the syndrome associated with trisomy 18?
Edwards' syndrome (Election age = 18)
Autosomal trisomies: Name 7 findings in trisomy 18
(1) severe mental retardation (2) rocker bottom feet (3) low-set ears (4) microngathia (5) congenital heart dz (6) clenched hands (flexion of fingers) (7) prominent occiput
Autosomal trisomies: What is the life expectancy in trisomy 18?
Death usually w/in 1 year of birth
Autosomal trisomies: What chromosomal anomaly is found in Patau's syndrome?
trisomy 13 (1:6000)
Autosomal trisomies: Name 7 findings in Patau's
(1) severe mental retardation (2) microphthamlia (3) microcephaly (4) cleft lip/palate (5) abnormal forebrain structures (6) polydactyly (7) congenital heart dz
Autosomal trisomies: What is the life expectancy in Patau's?
Death usually w/in 1 year of birth
Genetic gender disorders: What is the chromosomal anomaly in Klinefelter's syndrome?
XXY (1:850) - male
Genetic gender disorders: Name 5 characteristics of Klinefelter's syndrome
(1) Testicular atrophy (2) Enuchoid body shape (3) Tall, long extremities (4) Gynecomastia (5) Female hair distribution
Genetic gender disorders: What is a Barr body and is it found kin Klinefelter's syndrome?
It is the inactivated X chromosome. Yes.
Genetic gender disorders: What is the chromosomal anomaly in Turner's syndrome?
Female XO; (1:3000)
Genetic gender disorders: Name 4 characteristics of Turner's syndrome
(1) Short stature (2) Ovarian dysgenesis (streak ovary) (3) Webbing of next (4) Coarctation of the aorta
Genetic gender disorders: Turner's syndrome is the most common cause of primary ------------
amenorrhea
Genetic gender disorders: Is there a Barr body in Turner's syndrome?
No
Genetic gender disorders: Double Y males have what genotype?
XYY (1:1000) - male
Genetic gender disorders: What is the double Y phenotype?
Normal phenotype, very tall, severe acne, antisocial behavior (seein in 1-2% of XYY males; seen increasingly among prison inmates)
Muscular dystrophies: How are muscular dystrophies diagnosed?
Elevated CPK and muscle biopsy
Muscular dystrophies: What is the gene involved in muscular dystrophy?
Dystrophin
Muscular dystrophies: What is the mutation in Duchenne's MD?
Frame-shift mutation - deletion of dystrophin gene - accelerated muscle breakdown.
Muscular dystrophies: What is the age of onset in Duchenne's MD?
Before 5 years
Muscular dystrophies: In what muscles does weakness begin with Duchenne's MD?
Pelvic girdle muscles and progresses superiorly
Muscular dystrophies: Name 3 diagnostic characteristics of Duchenne MD.
(1) Pseduohypertrophy of calf muscles due to fibrofatty replacement of muscle (2) Cardiac myopathy (3) Use of Gowers' maneuver, requiring assistance of uper extremities to stand up - indicates proximal lower limb weakness
Muscular dystrophies: What distinguishes Becker's from Duchenne's MD?
Less severe mutation in Becker's. Less debilitating.
Pseudohermaphroditism: What is pseudohermaphroditism?
Disagreement between the phenotypic (external genitalia) and gonadal (testes vs. ovaries) sex.
Pseudohermaphroditism: A female pseudohermaphrodite has ------- present, but external genitalia are -------- or -----------.
ovaries; virilized or ambiguous
Pseudohermaphroditism: What causes female pseudohermaphroditism?
Excessive and inapropraite exposure to androgenic steroids during early gestation (i.e.,congenital adrenal hyperplasia or exogenous administration of androgens suring pregnancy).
Pseudohermaphroditism: What are the phenotypes in female and male pseudohermaphroditism?
female - XX; male - XY
Pseudohermaphroditism: A male pseudohermaphrodite has ------- present, but external genitalia are --------- or ---------.
testes; female or ambiguous
Pseudohermaphroditism: What is the most common form of male pseudohermaphroditism?
Testicular feminization (androgen insensitivity), which results from a mutation in the androgen receptor gene (x-linked); blind-end vagina
Pseudohermaphroditism: What determines gender identity?
External genitalia and sex of upbringing.
True Hermaphrodite (46,XX or 47, XXY): What is a true hermaphrodite?
Both ovary and testicular tissue present; ambiguous genitalia.
True Hermaphrodite (46,XX or 47, XXY): What are the genotypic possibilities for a true hermaphrodite?
46,XX or 47,XXY
Testicular Feminization syndrome (46, XY): What is the molecular defect in testicular feminization syndrome?
Defect in DHT receptor resulting in normal-apearing female
Testicular Feminization syndrome (46, XY): What genitalia are present in testicular feminization syndrome?
female genitalia with rudimentary vagina; uterus and uterine tubes generally absent; develop testes (often found in labia majora; surgically removed to prevent malignancy)
Testicular Feminization syndrome (46, XY): What are the genotype and horomone levels in testicular feminization syndrome?
46, XY; levels of testosterone, estrogen and LH are all high
5a-reductase deficiency: What is the biochemical consequence of 5a-reductase deficiency
Unable to convert testosterone to DHT.
5a-reductase deficiency: Describe genital development in 5a-reductase deficiency.
Ambiguous genitalia until puberty, when increased testosterone causes masculinization of genitalia.
5a-reductase deficiency: What are the hormone levels in 5a-reductase deficiency.
Testosterone/estrogen levels are normal; LH is normal or elevated.
Cri-du-chat syndrome: What is the mutation in Cri-du-chat syndrome?
Congenital deletion of short arm of chromosome 5 (46,XX or XY, 5p-)
Cri-du-chat syndrome: What are the findings in Cri-du-chat
Microcephaly, severe mental retardation, high-pitched crying/mewing, epicanthal folds, cardiac abnormalities.
Cri-du-chat syndrome: What does Cri-du-chat mean in english, which might help remember the symptoms?
Cry of the cat.
Fragile X syndrome: What is the 2nd most common cause of genetic mental retardation?
Fragile X syndrome
Fragile X syndrome: What genetic change is involved in Fragile X syndrome?
x-linked defect affecting the methylation and expression of the FMR1 gene.
Fragile X syndrome: Name 4 physical features associated with Fragile X syndrome?
(1) macro-orchidism (enlarged testes) (2) Long face with a large jaw (3) large everted ears (4) autism
Cystic Fibrosis (CF): What is the genetic defect in cystic fibrosis?
Autosomal-recessive defect in CFTR gene on chromosome 7
Cystic Fibrosis (CF): What is the molecular defect in CF?
Defective Cl- channel
Cystic Fibrosis (CF): What are the consequences of the defect in CF?
The defective chloride channel causes secretion of abnormally thick mucus that plugs lungs, pancreas and liver, which leads to recurrent pulmonary infections (pseudomonas species and S. aureus), chronic bronchitis, bronchiectasis, pancreatic insufficiency (malabsorption and steatorrhea), meconium ileus in newborns.
Cystic Fibrosis (CF): Name a diagnostic test for CF.
Increased concentration of Cl- ions in sweat test.
Cystic Fibrosis (CF): Why does CF cause infertility in males?
Absent vas deferens.
Cystic Fibrosis (CF): What vitamins are often deficient in CF?
Fat-soluble - A,D,E,K
Most common lethal genetic disease of Caucasians.
Cystic Fibrosis
Cystic Fibrosis (CF): What is the treatment for CF?
N-acetylcysteine to loosen mucous plugs.
Autosomal-dominant diseases: A patient presents with flank pain, hematuria, hypertension, progressive renal failure and has a mutation in one of his copies of APKD1 (chromosome 16); what is the diagnosis?
Adult polycystic kidney disease. N.B. the juvenile form is recessive.
Autosomal-dominant diseases: Name 2 other characteristics of adult polycystic kidney disease.
(1) always bilateral, (2) massive enlargement of the kidneys due to multiple large cysts.
Autosomal-dominant diseases: What sequelae are associated with adult polycystic kidney disease?
(1) polycystic liver disease (2) berry aneurysms (3) mitral valve prolapse
Autosomal-dominant diseases: What disease involves elevated LDL owing to defective or absent LDL receptor?
Familial hypercholesterolemia (type IIA)
Autosomal-dominant diseases: What are the average cholesterol levels in heterozygotes and homozygotes with familial hypercholesterolemia (tyype IIA)?
heterozygotes (1:500) - 300 mg/dL homozygotes - 700+ mg/dL
Autosomal-dominant diseases: What are 3 sequelae of familial hypercholesterolemia (type IIA)?
(1) severe atherosclerotic disease early in life (2) tendon xanthomas (classically in the Achilles tendon) (3) MI may develop before age 20
Autosomal-dominant diseases: What is the genetic defect in Marfan's syndrome?
Fibrillin gene mutation leads to connective tissue disorders.
Autosomal-dominant diseases: What are the skeletal abnormalities in Marfan's?
tall with long exteremities, hyperextensive joints, and long, tapering fingers and toes.
Autosomal-dominant diseases: What are the cardiovascular risks in Marfan's?
cystic medial necrosis of aorta leads to aortic incompetence and dissecting aortic aneurysms; flopy mitral valve.
Autosomal-dominant diseases: What are the ocular risks in Marfan's?
Subluxation of lenses.
Autosomal-dominant diseases: What is another name for Neurofibromatosis type 1?
von Recklinghausen's disease
Autosomal-dominant diseases: What chromosome is affected in NF type 1?
Long arm of chromosome 17 (17 letters in von Recklinghasen's)
Autosomal-dominant diseases: What are 5 classic characteristics of people with NF type 1?
(1) café-au-lait spots (2) neural tumors (3) Lisch nodules (pigmented iris hamartomas) (4) skeletal disorders (e.g., scoliosis) (5) Increased tumor susceptibility
Autosomal-dominant diseases: What disease is associated with bilateral acoustic neuroma?
Neurofibromatosis type 2
Autosomal-dominant diseases: What gene is affected in neurofibromatosis type 2?
NF2 gene on chromosome 22; (type 2 = 22)
Autosomal-dominant diseases: What is the genetic mutation in Von-Hippel-Lindau disease?
Deletion of VHL gene (tumor supressor) on chromosome 3p. (Von Hipel-Lindau = 3 words for chromosome 3)
Autosomal-dominant diseases: What are the clinical findings in Von-Hippel-Lindau disease?
hemiangioblastomas of retina/cerebellum/medulla; about half of affected individuals develop multiple bilateral renal cell carcinomas and other tumors.
Autosomal-dominant diseases: What is the diagnosis in a 20-50 year old person who presents with depression, progressive dementia, choreiform movements, caudate atrophy, and decreased levels of GABA and Ach in the brain?
Huntington's disease
Autosomal-dominant diseases: What chromosome holds the gene involved in Huntington's?
Chromosome 4; triplet repeat disorder ("Hunting 4 food")
Autosomal-dominant diseases: What disease is responsible for a colon that ecomes covered with adenomatous polyps after puberty?
Familial adenomatous polyposis (FAP)
Autosomal-dominant diseases: What is the progression in Familial adenomatous polyposis if colon not resected?
Colon cancer
Autosomal-dominant diseases: What is the genetic defect in Familial adenomatous polyposise?
deletion on chromosome 5 - 5 letters in polyp
Autosomal-dominant diseases: What are 3 characteristics of hereditary spherocytosis?
(1) spheroid erythrocytes (2) hemolytic anemia (3) increased MCHC
Autosomal-dominant diseases: What is the cure for hereditary spherocytosis?
splenectomy
Autosomal-dominant diseases: What is the genetic defect in achondroplasia?
Autosomal dominant cell-signaling defect of fibroblast growth factor (FGF) receptor 3.
Autosomal-dominant diseases: What are the physical characteristics of achondroplasia?
Dwarfism, short limbs, but head and trunk are normal size.
X-linked or AR:: Cystic fibrosis
AR
X-linked or AR:: albinism
AR
X-linked or AR:: Fragile X
X-linked
X-linked or AR:: Duchenne's muscular dystrophy
X-linked
X-linked or AR:: hemophilia A and B
X-linked
X-linked or AR:: a1-antitrypsin deficiency
AR
X-linked or AR:: Fabry's
X-linked
X-linked or AR:: G6PD deficiency
X-linked
X-linked or AR:: Hunter's syndrome
X-linked
X-linked or AR:: Phenylketonuria (PKU)
AR
X-linked or AR:: thalassemias
AR
X-linked or AR:: sickle cell anemias
AR
X-linked or AR:: ocular albinism
X-linked
X-linked or AR:: glycogen storage diseases
AR
X-linked or AR:: mucopolysaccharidoses (except Hunter's)
AR
X-linked or AR:: sphingolipidoses (except Fabry's)
AR
X-linked or AR:: infant polycystic kidney
AR
X-linked or AR:: Lesch-Nyhan syndrome
X-linked
X-linked or AR:: Bruton's agammaglobulinemia
X-linked
X-linked or AR:: hemochromatosis
AR
X-linked or AR:: Wiskott-Aldrich syndrome
X-linked
X-linked or AR:: What are femal carriers of x-linked recessive disorders rarely affected?
Inactivation of X chromosomes in each cell
Neural Tube Defects (NTD): Name two associations with neural tube defects during pregnancy.
(1) Low folic acid intake (2) Elevated a-fetoprotein in amniotic fluid
Neural Tube Defects (NTD): What NTD is associated with failure of bony spinal canal to close, but no structural herniation?
Spina bifida occulta
Neural Tube Defects (NTD): Which NTD is involved with herniation of meninges through spinal canal defect?
Meningocele
Neural Tube Defects (NTD): What is the NTD that involves herniation of meninges AND spinal cord through spinal canal defect?
Meningomyelocele
Neural Tube Defects (NTD): Which NTD is usually seen at lower vertebral levels?
Spina bifida occulta
Neural Tube Defects (NTD): Take a look at the figures in the book for good visualization of the NTDs.
0
Fetal Alcohol Syndrome: Who is at greatest risk of FAS?
Newborns of mothers who consumed significant amounts of alcohol (teratogen) during pregnancy (highest risk at 3-8 weeks).
Fetal Alcohol Syndrome: Name 5 congenital abnormalities in FAS?
(1) pre and postnatal developmental retardation (2) microcephaly (3) facial abnormalities (4) limb dislocation (5) heart and lung fistulas
Fetal Alcohol Syndrome: How does FAS rank among congenital malformations in the U.S.?
It's number 1
Fetal Alcohol Syndrome: What is the presumed mechanism of FAS?
Inhibition of cell migration?
Fetal Alcohol Syndrome: AUTHOR
Neha Pathak
NEOPLASTIC PROGRESSION: when cells increase in number it is called?
hyperplasia
NEOPLASTIC PROGRESSION: what are two enzymes that allow neoplastic cells to invade th ebasement membrane?
collagenase and hydrolase
NEOPLASTIC PROGRESSION: when cells have an increased nulear to cytoplasmic ratio but have not invaded the basement membrane, it is called?
carcinoma in situ
_ -PLASIA DEFINITIONS: squamous _________ occurs in the trachea and bronchi of smokers
metaplasia
_ -PLASIA DEFINITIONS: what is it called when a cell has reversible preneoplastic changes with abnormal shape and size?
dysplasia
_ -PLASIA DEFINITIONS: abnormal cells that lack differentiation are called?
anaplastic cells
TUMOR STAGE VS. GRADE: staging of a tumor takes into account what factors?
TMN size of tumor, node involvment, metastases
TUMOR STAGE VS. GRADE: histolgy of a tumor is used to determine?
grade
TUMOR STAGE VS. GRADE: what has more prognostic value? Grade or stage?
stage
TUMOR NOMENCLATURE: the term _________ implies epithelial origin, whereas _________ implies mesenchymal origin
carcinoma, sarcoma
TUMOR NOMENCLATURE: a tumor of blood vessels is called what when it is benign? When it is malignant?
hemangioma, angiosarcoma
TUMOR NOMENCLATURE: what is a benign tumor of skeletal muscle called?
rhabdomyoma
DISEASES ASSOCIATED WITH NEOPLASMS: Kaposi's sarcoma and aggressive malignant lymphomas are associated with what disease
AIDS
DISEASES ASSOCIATED WITH NEOPLASMS: Actinic Keratosis predispose to what disease?
squamous cell carcinoma of skin
DISEASES ASSOCIATED WITH NEOPLASMS: down syndrome is associated with what neoplasm?
ALL
ONCOGENES: c-myc oncogenes are associated with what neoplasm?
burkitt's lymphoma
ONCOGENES: breast, ovarian, and gastic carcinomas all have this oncogene mutation in common?
erb B2
ONCOGENES: MEN II and III are associated with what oncogenic mutation?
ret
TUMOR SUPRESSOR GENES: Name the associated tumor witht the following tumor supressor gene mutations: Rb, APC, WT1
retinoma blastoma, colorectal cancer, wilms tumor
TUMOR SUPRESSOR GENES: p53 is on what chromosome
17p
TUMOR SUPRESSOR GENES: osteosarcoma is associated with what tumor supressor gene mutation
Rb
TUMOR MARKERS: TRAP is a tumor marker for what neoplasm
hairy cell leukemia
TUMOR MARKERS: what tumor marker is elevated in hyatidiform moles
beta-HCG
TUMOR MARKERS: ovarian tumors are associated with what tumor marker
CA-125
ONCOGENIC VIRUSES: the virus associated with burkitts and nasopharyngeal cancer is?
ebv
ONCOGENIC VIRUSES: hpv is responsible for what cancers?
cervical, penile, and anal carcinoma
ONCOGENIC VIRUSES: what virus is responsible for kaposis sarcoma?
hhv8
CHEMICAL CARCINOGENS: aflatoxin, vinyl chloride and ccl4 damage what organ?
liver
CHEMICAL CARCINOGENS: nitrosamines cause cancer in what organs?
esophagus or stomach
CHEMICAL CARCINOGENS: asbestos causes what types of cancer?
bronchogenic and mesothelioma
LOCAL EFFECTS OF TUMORS: When the following are obstructed, what can occur: bronchus? Biliary tree? Left colon?
pneumonia, jaundice, constipation
LOCAL EFFECTS OF TUMORS: tumor mass in the brain can cause the following
seizure, increased intracranial pressure, mass effect and herniation
LOCAL EFFECTS OF TUMORS: compression of the recurrent laryngeal nerve can cause what?
hoarseness
PROSTATIC ADENOCARCINOMA: what lobe/zone is most often involved in prostatic cancer?
posterior lobe, peripheral zone
PROSTATIC ADENOCARCINOMA: what is a common site of metastases for prostate cancer?
bone
PROSTATIC ADENOCARCINOMA: what are useful tumor markers for prostate CA
psa, and prostatic acid phosphatase
SKIN CANCER: this type of skin cancer has palisading nuclei
basal cell carcinoma
SKIN CANCER: dyplastic nevi are a precursor fot this type of cancer
melanoma
SKIN CANCER: this type of skin cancer is associated with keratin pearls
squamous cell carcinoma of skin
PRIMARY BONE TUMORS: the translocation 11;22 is associated with this cancer of the bone that occurs most commonly in young boys
ewing's sarcoma
PRIMARY BONE TUMORS: this tumor is characterized by a "double bubble" or "soap bubble" appearance
benign giant cell tumor
PRIMARY BONE TUMORS: this is the most common benign bone tumor, usually in men younger than 25, with a rare transformation to malignancy
osteochondroma
PRIMARY BONE TUMORS: AUTHOR
Gabe Pitta
Primary Brain Tumors-Adult peak incidence: Most common adult brain tumor?
Glioblastoma multiforme (grade IV astrocytoma) See pseudopalisading tumor cells, central necrosis and hemorrhage.
Primary Brain Tumors-Adult peak incidence: Second most common adult tumor?
Meningioma, occurring in convexities of hemispheres and parasagittal region. See psammoma bodies.
Primary Brain Tumors-Adult peak incidence: Benign, slow-growing tumor in frontal lobes?
Oligodendroglioma, look for fried egg apearance, often calcified.
Primary Brain Tumors-Adult peak incidence: Third most common tumor, often localized to CN8?
Schwannoma, see it in acoustic Schwannoma. Antoni A (compact) and Antoni B (loose) patterns.
Primary Brain Tumors-Adult peak incidence: Most common forms are prolactin-secreting?
Pituitary Adenoma, which derives from Rathke's Pouch and can produce secondary bitemporal hemianopsia and hypopituitarism
Primary Brain Tumors-Child Peak Incidence: Highly malignant cerebellar tumor?
Medulloblastoma, can compress 4th ventricle and cause hydrocephalus. See rosettes or pseudorosettes.
Primary Brain Tumors-Child Peak Incidence: Cerebellar tumor associated with von Hippel-Landau syndrome?
Hemangioblastoma, see foamy cells and high vascularity, can produce excess EPO-->polycythemia.
Primary Brain Tumors-Child Peak Incidence: Commonly found in fourth ventricle, causing hydrocephalus?
Ependyomomas, which have perivascular rosettes and rod-shaped blepharoblasts near nucleus on exam.
Primary Brain Tumors-Child Peak Incidence: Diffusely infiltrating glioma, usually found in posterior fossa?
Low-grade Astrocytoma
Primary Brain Tumors-Child Peak Incidence: Benign tumor often confused with pituitary adenoma?
Craniopharyngioma, which also is derived from Rathke's pouch, also presents with bitemporal hemianopsia, also calcified.
Common sites that receive metastatic tissue: To Brain?
Lung, Breast, Skin (melanoma), Kidney (renal cell carcinoma), GI. Lots of Bad Stuff Kills Glia. 50% of brain tumors are due to metastases.
Common sites that receive metastatic tissue: To liver?
Colon>Stomach>Pancreas>Breast>Lung. Cancer Sometimes Penetrates Benign Liver. Metastases much more common than primary liver tumors.
Common sites that receive metastatic tissue: To bone?
Breast, lung, thyroid, testes, kidney, prostate.=> "BLT with a Kosher Pickle."
Common sites that receive metastatic tissue: Most common organ receiving metastases?
Adrenal medulla, then cortex.
Common sites that receive metastatic tissue: Most common organ sending metastases?
Lung>Breast/Stomach
Paraneoplastic effects of tumors: Cushing's Syndrome
ACTH/ACTH-like peptide from small-cell lung CA
Paraneoplastic effects of tumors: SIADH
ADH/ANP from small cell lung CA and intracranial neoplasms.
Paraneoplastic effects of tumors: Hypercalcemia
PTH-related peptide/TGF-alpha/TNF-Alpha/IL-2 from squamous cell lung CA, renal cell CA, Breast CA, multiple myeloma, bone metastasis.
Paraneoplastic effects of tumors: Polycythemia
EPO from renal cell CA.
Paraneoplastic effects of tumors: Lambert-Eaton Syndrome
Antibodies against presynaptic Ca2+ channels at NMJ, generated by Thymomas and bronchogenic CA.
Paraneoplastic effects of tumors: Gout
Hyperuricemia due to excess nucleic acid turnover, found in various neoplasms.
Cancer Epidemiology: Male incidence?
Prostate(32%)>Lung(16%)>Colon and Rectum (12%)
Cancer Epidemiology: Male Mortality?
Lung(33%)>Prostate(13%)
Cancer Epidemiology: Female incidence?
Breast (32%)>Lung(13%)>Colorectal(13%)
Cancer Epidemiology: Female Mortality?
Lung(23%)>Breast(18%)
HEMATOLOGIC PATHOLOGY: Anemia
0
HEMATOLOGIC PATHOLOGY: Anemia presenting with increased TIBC, decreased ferritin, decreased serum iron.
Mycrocytic, Hypochromic, usually secondary to iron deficiency. Also seen with Thalassemia, lead poisoning
HEMATOLOGIC PATHOLOGY: Anemia in patients with B12/Folate deficiency
Macrocytic (MCV>100), presents with PMN hypersegmentation on blood smear. B12 deficiency presents with neuro effects, folate deficiency much easily reached through bad diet. Also seen in patients taking drugs blocking DNA synthesis (sulfa drugs, AZT)
HEMATOLOGIC PATHOLOGY: Anemia in patients with normocytic, normochromic smears?
Hemorrhagic, autoimmune hemolytic, enzyme defects (G6PD), bone marrow dz, anemia of chronic dz. Decreased serum haptoglobin and increased LDH indicate RBC hemolysis. Direct Coomb's test can show an immune-mediated process.
HEMATOLOGIC PATHOLOGY: Anemia in patients with a decreased TIBC, decreased serum iron, and decreased iron saturation?
Anemia of chronic disease
HEMATOLOGIC PATHOLOGY: Anemia in patients with an increased TIBC, decreased serum iron, and normal iron saturation?
Iron-deficiency anemia.
HEMATOLOGIC PATHOLOGY: Patient with normal TIBC, increased serum iron, and 100% iron saturation (normal is 20-50%)
Iron overload (hemosiderosis)
Aplastic Anemia: Patient presentation of Aplastic Anemia?
Patients with pancytopenia characterized by severe anemia, neutropenia, and thrombocytopenia caused by failure or destruction of multipotent myeloid stem cells.
Aplastic Anemia: What causes aplastic anemia?
Radiation, benzene, chloramphenicol, alkylating agents, antimetabolites, viral agents, Fanconi's anemia, idiopathic causes.
Aplastic Anemia: Common Sx?
Fatigue, malaise, pallor, purpura, mucosal bleeding, petechiae, infection.
Aplastic Anemia: How do the blood smear and bone marrow aspirate appear?
Blood smear: pancytopenia with normal cell morphology. Aspirate:hypocellular marrow with fatty infiltration.
Aplastic Anemia: What's the treatment of aplastic anemia?
Withdraw offending agent, BMT, RBC/platelet transfusion, G-CSF or GM-CSF.
Hereditary Spherocytosis: How do patients with hereditary psherocytosis present (smear and labs)?
Smear shows microcytic RBCs that are small and round with no central pallor. The patients will have normal Hgb and MCV.
Hereditary Spherocytosis: What's the cause of hereditary spherocytosis?
Intravascular hemolysis due to spectrin defect.
Hereditary Spherocytosis: How is it confirmed and tested?
Will be Coombs' negative (unlike warm antibody hemolysis), can perform osmotic fragility test.
Hereditary Spherocytosis: What are some associated diseases?
Gallstones, splenomegaly, anemia, and jaundice.
Blood Dyscrasias-Sickle Cell Anemia: What is the HbS mutation?
Glutamate-->Valine in a Beta chain. Heterozygotes are usually malaria-resistant. *% of Af-Ams carry the HbS trait, 0.2% have the disease.
Blood Dyscrasias-Sickle Cell Anemia: What can precipitate sickling?
Low O2, dehydration.
Blood Dyscrasias-Sickle Cell Anemia: What are the common complications of HbS homozygotes?
aplastic crises from parvovirus infection, autosplenectomy, risk of encapsulated organism infection, salmonella osteomyelitis, vaso-occlusive pain crises, and splenic sequestration crises.
Blood Dyscrasias-Sickle Cell Anemia: What is the HbC defect?
Different Beta-chain mutation, the patients with HbC or HbSC have milder dz than HbSS patients.
Blood Dyscrasias-Sickle Cell Anemia: What are the treatment options?
Hydroxyurea (increased HbF) and bone marrow transplantation.
Blood Dyscrasias-Alpha-Thalassemia: What populations have the most prevalence?
Mediterranean populations. (thalaSEAmia)
Blood Dyscrasias-Alpha-Thalassemia: What is the etiology?
Underproduction of the alpha-globin chain, with no compensatory increase of any other chains. HbH has Beta-4 tetramers and lacks 3 alpha-globin genes.
Blood Dyscrasias-Alpha-Thalassemia: What is the presentation of a patient lacking all four alpha-globin genes?
hydrops fetalis and intrauterine fetal death.
Blood Dyscrasias-Beta-Thalassemia: Patient Presentation of Beta-Thalassemia major?
severe anemia requiring blood transfusion. Cardiac failure is often due to secondary hemochromatosis.
Blood Dyscrasias-Beta-Thalassemia: Etiology of Beta-Thalassemia major?
Beta chain is absent, fetal Hgb production is compensatorily increased but is inadequate.
Blood Dyscrasias-Beta-Thalassemia: Etiology of Beta-thalassemia minor?
The Beta chain is underproduced, again see fetal HgB compensatory increase. HbS/Beta-thalassemia heterozygote has mild to moderate disease.
DIC: What is the definition of DIC?
Activation of the coagulation cascade leading to microthrombi and global consumption of platelets, fibrin, and coagulation factors.
DIC: What causes DIC?
Obstetric complications (most common cause), gram-negative sepsis, transfusion, trauma, malignancy, acute pancreatitis, nephrotic syndrome.
DIC: What are the lab findings in DIC?
Increased PT, increased PTT, incrased fibrin split products (D-dimers), decreased platelet count.
Bleeding Disorders: What disorder type presents with mucous membrane bleeding, petechiae, purpura, and prolonged bleeding time?
Platelet abnormalities (microhemorrhages), which can be caused by ITP (antiplatelet antibodies and increased megakaryocytes), TTP (schistocytes), drugs, and DIC (increased fibrin split products).
Bleeding Disorders: What disorder type presents with hemarthroses (bleeding into joints), easy brusing, prolonged PT and/or PTT?
Coagulopathies (macrohemorrhages), which can be caused by Hemophilia A/B and von Willebrand's disease (most common)
Bleeding Disorders: What is the deficiency in Hemophilia A?
Factor VIII
Bleeding Disorders: What is the deficiency in Hemophilia B?
Factor IX
Bleeding Disorders: What is the deficiency in vW dz?
Deficiency of von Willebrand's antigen.
Bleeding Disorders: Which factors does the PT measure?
Extrinsic: Factors II, V, VII, and X.
Bleeding Disorders: Which factors does the PTT measure?
Intrinsic: All factors except VII and XIII.
Hemorrhagic disorders: What presents with increased bleeding time, normal platelet count, and normal PT/PTT and platelet count?
Qualitative platelet defects, can be Bernard Soulier disease (defect in adhesion) or Glanzmann's thrombasthenia (defect in aggregation). Can also see these labs in vascular bleeding.
Hemorrhagic disorders: What presents with decreased platelet count, increased bleeding time, and normal PT/PTT?
Thrombocytopenia
Hemorrhagic disorders: Which diseases present with increased PTT and normal PT, platelet count, and bleeding time?
Hemophilia A and B
Hemorrhagic disorders: What presents with a high PTT, normal PT, normal platelets, and increased bleeding time?
vW disease
Hemorrhagic disorders: What presents with increased Bleeding time/Pt/PTT and decreased platelet count?
DIC
Lymphomas: Hodgkin's Versus NHL: Which one presents with Reed-Sternberg cells?
Hodgkin's
Lymphomas: Hodgkin's Versus NHL: Which one is associated with HIV and immunosuppression?
NHL
Lymphomas: Hodgkin's Versus NHL: Which one involves multiple, peripheral nodes, with common extranodal involvement?
NHL
Lymphomas: Hodgkin's Versus NHL: Which one is localized to a single group of nodes, with contiguous spread and rare extranodal involvement?
HL
Lymphomas: Hodgkin's Versus NHL: Which one presents with constitutional signs/symptoms: low-grade fever, night sweats, weight loss?
HL (NHL has few signs/symptoms)
Lymphomas: Hodgkin's Versus NHL: Which one presents with mediastinal lymphadenopathy?
HL
Lymphomas: Hodgkin's Versus NHL: Which one involves mostly the B cells (except lymphoblastic origin)?
NHL
Lymphomas: Hodgkin's Versus NHL: Which one has hypergammaglobulinemia?
neither. Multiple Myeloma has hypergammaglobulinemia, where the excess B cells are in the resting state.
Lymphomas: Hodgkin's Versus NHL: Which one has a 50% association with EBV?
HL
Lymphomas: Hodgkin's Versus NHL: Which one has bimodal age distribution?
HL (NHL has peak incidence at 20-40 years old)
Lymphomas: Hodgkin's Versus NHL: Which one has more common male presentation?
HL
Hodgkin's: What factors denote a good prognosis?
Increased lymphocytes, decreased RS cells.
Hodgkin's: Which HL type has the best prognosis?
Nodular sclerosing (65-75%), which has least RS cells and lots of lymphocytes. Lymphocyte-predominant LH also has excellent prognosis.
Hodgkin's: Which HL type is the most common?
Nodular sclerosing, affecting women more than men and primarily young adults.
Hodgkin's: What is the prognosis of mixed cellular HL?
Intermediate. There are lots of lymphocytes but also lots of RS cells.
Hodgkin's: Which HL type has the worst prognosis?
Lymphocyte-depleted, which affects older males with disseminated disease.
Hodgkin's: Which HL type has the most RS cells?
Mixed cellular.
NHL: Which NHL type has only B cells?
Small Lymphocytic lymphoma, follicular lymphoma, Burkitt's
NHL: Which NHL type has a mix of B cells and T cells?
Diffuse large cell , occurring mostly in elderly but sometimes in children.
NHL: Which NHL type has only T cells?
Lymphoblastic Lymphoma, which has immature T cells. It is a very aggressive T-cell lymphoma.
NHL: Which type is associated with a t(8;14) c-myc gene mutation?
Burkitt's Lymphoma, occurring mostly in children. Has a high-grade "starry sky" apearance.
NHL: Which type is associated with a t(14;18) mutation and overexpression of bcl-2?
Follicular lymphoma, which is difficult to cure but has an indolent course.
NHL: Which type is associated with EBV infection and is endemic in africa?
Burkitt's
NHL: Which is the most common childhood NHL?
Lymphoblastic Lymphoma, which also presents with ALL and a mediastinal mass.
NHL: Which is the most common adult version of NHL?
Follicular lymphoma.
NHL: Which types occur in adults?
Small lymphocytic lymphoma, follicular lymphoma.
NHL: Which types occur in children?
Lymphoblastic lymphoma, Burkitt's lymphoma.
NHL: Which one has a distribution of 80% adults and 20% children?
Diffuse large cell lymphoma, which ALSO has an 80% B cells 20% T cell distribution.
NHL: Which low-grade NHL type presents like CLL?
Small Lymphocytic lymphoma.
NHL: AUTHOR
John Peoples
Leukemias: Peripheral and bone marrow characteristics
Increased circulating leukocytes, bone marrow infiltrates of leukemic cells
Leukemias: Consequences of marrrow failure
Anemia (dec. RBC's), infections (dec. WBC's), hemorrhage (dec. platelets)
Leukemias: Common organs of infiltration
Liver, spleen, lymph nodes
Leukemias: Characteristics of acute leukemias
Blasts predominate, children or elderly, short or drastic course
Leukemias: ALL characteristics (4)
Lympholasts (pre-B or pre-T), children, most responsive to therapy, associated with Down's Syndrome
Leukemias: AML characteristics
Myeloblasts, adults, auer rods
Leukemias: Characteristics of chronic leukemias
More mature cells, midlife age range, longer, less devastating course
Leukemias: CLL characteristics - cells
Lymphocytes, non-Ab producing B cells, increased smudge cells on peripheral blood smear
Leukemias: CLL - population
older adults
Leukemias: CLL - presentation and course
lymphadenopathy, hepatosplenomegaly, few sx., indolent course
Leukemias: CLL is similar to?
very similar to SLL (small lymphocytic lymphoma)
Leukemias: CLL is associated with what type of anemia?
warm antibody autoimmune hemolytic anemia
Leukemias: CML cell characteristics
Myeloid stem cells, increased neutrophils and metamyelocytes
Leukemias: CML translocation?
Ph Chromosome, t(9;22), bcr-abl
Leukemias: CML acute complications?
blast crisis (AML)
Chromosomal translocations - the following disorder is associated with what translocation?: CML
Ph chromosome, t(9;22), bcr-abl
Chromosomal translocations - the following disorder is associated with what translocation?: Burkitt's lymphoma
t(8;14), c-myc activation
Chromosomal translocations - the following disorder is associated with what translocation?: Follicular lymphomas
t(14;18), bcl-2 activation
Chromosomal translocations - the following disorder is associated with what translocation?: AML- M3 type
t(15;17), responsive to all-trans retinoic acid (ATRA)
Chromosomal translocations - the following disorder is associated with what translocation?: Ewing's sarcoma
t(11;22)
Chromosomal translocations - the following disorder is associated with what translocation?: Mantle cell lymphoma
t(11;14)
Lymphomas and Leukemias: What are the chronic leukemias associated with T-lymphoblasts?
Sezary Syndrome, CLL-T (both L2)
Lymphomas and Leukemias: What are the acute leukemias associated with T-lymphoblasts?
ALL-T (L2), ALL-null (L1), ALL-common(L1)
Lymphomas and Leukemias: What are the chronic leukemias associated with B-lymphoblasts?
CLL-B (L3)
Lymphomas and Leukemias: What are the acute leukemias associated with B-lymphoblasts?
ALL-B (L3)
Lymphomas and Leukemias: What are the chronic leukemias associated with monoblasts?
Chonic monocytic (M5), chronic myelomonocytic (M4)
Lymphomas and Leukemias: What are the acute leukemias associated with monoblasts?
Acute monocytic (M5), acute myelomonocytic (M5)
Lymphomas and Leukemias: What are the chronic leukemias associated with myeloblasts?
CML (M1, 2 and 3), Polycythemia rubra vera (M1), myelofibrosis (M1)
Lymphomas and Leukemias: What are the acute leukemias associated with myeloblasts?
AML (M2 and M1), Promyelocytic (M1)
Lymphomas and Leukemias: What are the chronic leukemias associated with eos-myeloblasts?
Eosinophilic (rare)
Lymphomas and Leukemias: What are the chronic leukemias associated with normoblasts?
Chronic erythroid (M6, rare)
Lymphomas and Leukemias: What are the acute leukemias associated with normoblasts?
acute erythroleukemia (M6)
Lymphomas and Leukemias: What are the chronic leukemias associated with megakaryoblasts?
Idiopathic thrombocytopenia (M7)
Lymphomas and Leukemias: What are the acute leukemias associated wwith megakaryoblasts?
acute megakaryocytic leukemias (M7)
Multiple Myeloma: What type of cell proliferates in MM, and what is its histologic appearance
Monoclonal plasma cell, fried egg apearance
Multiple Myeloma: Where does MM arise?
bone marrow
Multiple Myeloma: The 2 most common ab's, in order, are:
IgG (55%), IgA (25%)
Multiple Myeloma: Common symptoms are:
lytic bone lesions and hypercalcemia, renal insifficiency, increased suscpetibility to infection, anemia
Multiple Myeloma: This disease is associated with:
primary amyloidosis
Multiple Myeloma: Ig light chains are also called:
Bence Jones proteins
Multiple Myeloma: 3 key diagnostic features:
lytic bone lesions on x-ray, M-spike on serum protein electrophoresis, Bence-Jones proteins in urine
Multiple Myeloma: Red blood cell appearance on peripheral smear:
Rouleaux formation (poker chips)
Multiple Myeloma: What 2 differences are seen in Waldenstrom's macroglobulinemia?
M-spike is IgM, no lytic lesions
Achalasia: Achalasia results from:
failure of relaxation of lower esophogeal sphincter due to loss of the myenteric plexus (Aeurbach's). [A-chalasia = absence of relaxation]
Achalasia: Achalasia causes:
progressive dysphagia
Achalasia: Diagnosis is made by:
barium swallow showing "bird beak" dilated esophagus with distal stenosis.
Achalasia: Secondary achalasia may result from this protozoan disease:
Chagas disease (treponema cruzi)
Achalasia: Associated with increased risk of:
esophageal carcinoma
Barrett's esophagus: The distal esophageal colummnar epithelium is replaced with….
Gastric columnar epithelium
Barrett's esophagus: Barrett's results from:
Reflux
Barrett's esophagus: Barrett's can progress to:
adenocarcinoma
Esophageal Cancer: Risk factors are (ABCDEF):
achalasia, barrett's esophagus, corrosive esophagitis/cigarettes, diverticuli (e.g. zenker's), esophogeal web/EtOH, familial
Esophageal Cancer: The most common esophageal cancer in the world is:
squamous cell carcinoma
Esophageal Cancer: The most common esophageal cancer in the US is:
equal between squamous cell and adenocarcinoma (decreased cigarette use).
Esophageal Cancer: Esophageal Adenocarcinoma is found where?
lower third
Esophageal Cancer: Esophageal squamous cell carcinoma is found where?
uper 2/3
Congenital Pyloric Stenosis: What does this cause in infant?
Projectile vomiting at aproximately 2 weeks of age
Congenital Pyloric Stenosis: Treatment entails:
surgery
Malabsorption syndromes: Autoantibodies are seen to what in Celiac Sprue?
Gluten (gliaden) in wheat and other grains
Malabsorption syndromes: The histologic appearance of celiac sprue:
villous flattening, lymphocytic infiltrate
Malabsorption syndromes: Tropical sprue is due to what type of agent?
Infectious, treated with Antibiotics
Malabsorption syndromes: What are you infected with if you have Whipple's Disease?
Tropheryma whipelli
Malabsorption syndromes: Histologic cell type seen in Whipple's disease:
PAS+ macrophages
Malabsorption syndromes: What is the most common disaccharide deficiency leading to malabsorption?
lactase => milk intolerance
Chronic Gastritis: Fundal gastritis (type A) is characterized by 4 A's, what are they?
Autoimmune disorder with Autoantibodies to parietal cells, pernicious Anemia, Achlorydia
Chronic Gastritis: Antral gastritis (Type B) is caused by:
a Bug, H. Pylori infection
Chronic Gastritis: Both forms of gastritis carry an increased risk of what?
Gastric carinoma
Peptic Ulcer Disease: 2 etiologic agents underlying gastric ulcers are:
H. pylori (70%) and NSAID's
Peptic Ulcer Disease: Degeneration of what leads to gastric ulcers?
Gastric mucosal protection v. gastric acid
Peptic Ulcer Disease: Does the pain of a gastric ulcer increase or decrease with meals?
Increases => weight loss
Peptic Ulcer Disease: One etiologic agent underlies duodenal ulcers - what is it?
H. pylori (100%)
Peptic Ulcer Disease: 2 pathogenic mechanisms are thought to lead to development of duodenal ulcer - what are they?
Increased gastric acid secretion and decreased mucosal protection
Peptic Ulcer Disease: What tends to hypertrophy with duodenal ulcers?
Bruenner's glands
Peptic Ulcer Disease: The margins of a doudenal ulcer are:
clean and punched out
Peptic Ulcer Disease: The 4 common complications of of duodenal ulcers are:
bleeding, penetration, perforation, and obstruction (not necessarily pre-cancerous)
Peptic Ulcer Disease: Does the pain of a duodenal ulcer increase or decrease with meals?
Decreases => weight gain
Peptic Ulcer Disease: How is H.pylori infection treated?
triple therapy (metronidazole, bismuth salicylate, amoxocillin or tetracycline) with or without proton pump inhibitor
Peptic Ulcer Disease: Does smoking effect peptic ulcer development?
Yes, it is 2x as common in smokers
IBD: The possible etiology of Crohn's disease is:
Infectious
IBD: The location and lesion type of Crohn's disease is:
Skip lesions in any portion of GI tract, usually terminal ileum, SI, and colon. Rectal sparing.
IBD: Gross Morphological features (6) of Crohn's disease are:
Transmural inflammation, cobblestone mucosa, creeping fat, bowel wall tickening (string sign on x-ray), linear ulcers, and fissure. (FAT OLD CRONE SKIpING DOWN A COBBLESTONE ROAD)
IBD: The microscopic morphological features of Crohn's disease are:
non-caseating granulomas
IBD: The four major complications of Crohn's disease are:
Strictures, fistulas, perianal diease, malabsorption leading to nutritional depletion.
IBD: Two major extraintestinal manifestations associated with Chron's disease are:
migratory polyarthritis and erythema nodosum
IBD: The possible etiology of UC is:
autoimmune
IBD: Location and lesion type of UC is:
continuous lesions of colon with rectal involvement
IBD: The two gross morphological features of UC are:
mucosal inflammation, friable mucosal pseudopolyps with freely hanging mesentery.
IBD: The microscopic morphological features of UC are:
crypt abcesses, ulcers
IBD: 3 severe complications of UC are:
severe stenosis, toxic megacolon, and colorectal carcinoma
IBD: Two major extraintestinal manifestations associated with UC are:
pyoderma gangrenosum, sclerosing cholangitis
Diverticular Disease: Diverticuli are blind pouches found in the:
alimentary tract
Diverticular Disease: Diverticuli are lined by (3 layers):
Mucosa, muscularis, and serosa
Diverticular Disease: The 4 GI tract locations of diverticuli are:
esophagus, stomach, duodenum and colon
Diverticular Disease: Why are most diverticuli termed false?
They lack or have an attenuated muscularis mucosa.
Diverticular Disease: Percent of older patients likely to get diverticulosis (many diverticula):
50% of patients >60
Diverticular Disease: The reasons for increasing number of diverticula with age are:
Increased intraluminal pressure and focal weaknes in the colonic wall.
Diverticular Disease: What type of diet is diverticulosis associated with?
low-fiber
Diverticular Disease: Sx of diverticulosis?
usually asymptomatic, sometimes vague discomfort.
Diverticular Disease: Diverticulitis commonly presents with pain where?
LLQ
Diverticular Disease: Complications of diverticulitis include:
perforation, peritonitis, abcess formation, bowel stenosis
Intussusception and Volvulus: What is intussuception?
telecoping of 1 bowel segment into distal segment.
Intussusception and Volvulus: Complication of intussusception is:
compromised blood suply.
Intussusception and Volvulus: What is volvulus?
Twisting of portion of the bowel around its mesentery.
Intussusception and Volvulus: What volvulus lead to?
obstruction.
Stomach Cancer: What is the most common type of stomach cancer?
adenocarcinoma
Stomach Cancer: Is this cancer aggresive?
Yes, it has early aggressive local spread and node/liver mets
Stomach Cancer: Stomach Cancer is associated with what 3 etiologic factors?
dietary nitrosamines, achlorhydria, chronic gastritis
Stomach Cancer: What is stomach cancer termed when it is diffusely infiltrative with thickened and rigid appearance?
Linitis plastica
Stomach Cancer: What does Virchow's node signify?
involvement of supraclavicular node by stomach mets
Stomach Cancer: What is Krukenberg's tumor?
bilateral stomach cancer mets to the ovary
Stomach Cancer: What are characteristics of Krukenberg's tumor?
Abundant mucus, "signet ring" cells
Hirschprung's Disease: What is Hirschprung's diease?
congential megacolon
Hirschprung's Disease: What is missing?
enteric nerve plexus (both Auerbach's and Meissner's). Seen on biopsy
Hirschprung's Disease: What is the cause of this disease?
failure of neural crest migration.
Hirschprung's Disease: How does this disease present?
Chronic constipation early in life.
Hirschprung's Disease: Which part of the colon is dilated?
That part proximal to the aganglionic segment - aganglionic portion is constricted.
Colorectal cancer risk factors: The risk factors are:
colorectal villous adenoma, IBD, low-fiber diet, age, FAP, HNPCC, personal and family hx. of colon cancer.
Colorectal cancer risk factors: What is Peutz-Jeghers, and does it lead to colorectal cancer?
It is a benign polyposis syndrome which is not a risk factor.
Colorectal cancer risk factors: Who schould be screened for colorectal cancer and how?
People over age 50, screen with stool occult blood test.
Cirrhosis/Portal HT: Cirrho in Greek means:
tawny yellow
Cirrhosis/Portal HT: Cirrhosis is:
diffuse fibrosis of the liver with destruction of norml architecture, nodular regeneration.
Cirrhosis/Portal HT: Cause of micronodular cirrhosis (nodules <3mm, uniform in size) is:
metabolic insult
Cirrhosis/Portal HT: Causes of macronodular cirrhosis (nodules >3mm, varied in size) are?
Significant liver injury leading to hepatic necrosis (post-infectious, drug-induced hepatitis)
Cirrhosis/Portal HT: Cirrhosis is assocated with what type of cancer?
hepatocellular carcinoma
Cirrhosis/Portal HT: The effects of portal hypertension are:
esophageal varices (hemetemesis, melena), peptic ulceration (melena), splenomegaly, caput medusae, ascites, hemmorhoids, and testicular atrophy
Cirrhosis/Portal HT: Portal hypertension may be relieved by what method?
Portacaval shunt between splenic vein ad left renal vein.
Cirrhosis/Portal HT: What are the effects of liver cell failure?
Coma, scleral icterus, fetor hepaticus (breath smells like a freshly opened corpse), spider nevi, gynecomastia, jaundice, loss of sexual hair, asterixis, bleeding tendency, anemia, ankle edema
Alcoholic Hepatitis: Hepatocytes in alcoholic hepatitis are:
swollen and necrotic
Alcoholic Hepatitis: Other histologic changes seen in aloholic hepatitis are:
neutrophil infiltration, mallory bodies (hyaline), fatty change, and sclerosis areound central vein
Alcoholic Hepatitis: SGOT (AST):SGPT (ALT) ratio is:
usually more than 1.5 (A Scotch and Tonic: AST elevated)
Budd Chiari Syndrome: What is Budd-Chiari Syndrome?
Occlusion of the IVC or hepatic veins with centrilobular congestion and necrosis, leading to congestive liver disease
Budd Chiari Syndrome: The features of congestive liver disease are:
hepatomegaly, ascites, abdominal pain, and eventual liver failure
Budd Chiari Syndrome: Budd-Chiari Syndrome is associated with what 3 conditions:
polycythemia vera, pregnancy, hepatocellular carcinoma
Wilson's Disease: Wilson's disease is?
Coper accumulation, especially in liver, brain and cornea
Wilson's Disease: It is due to what?
failure of coper to enter circulation in the form of ceruloplasmin
Wilson's Disease: What is another name for Wilson's Disease?
Hepatolenticular degeneration
Wilson's Disease: What are the symptoms of Wilson's Disease? (A,B,C-6,D)
Asterixis, basal ganglia degeneration (parkinsonian symptoms), Ceruloplasmin decrease, cirrhosis, corneal deposits (kayser-fleischer rings), coper accumulation, carcinoma (hepatocellular), choreiform movements, Dementia
Hemochromatosis: What is hemosiderosis?
Deposit of iron (hemosiderin) - this causes hemochromatosis
Hemochromatosis: The traid of sx. are?
micronodular cirrhosis, pancreatic fibrosis, and skin pigmentation ("bronze" diabetes)
Hemochromatosis: Possible complications include?
CHF, increased risk of hepatocellular carcinoma
Hemochromatosis: What is primary form?
inherited, Autosomal recessive
Hemochromatosis: What is secondary form due to?
chronic transfusion therapy
Hemochromatosis: What are ferritin, iron, TIBC and transferrin saturation levels?
Ferritin increased, iron increased, TIBC decreased => transferrin saturation is increased
Hemochromatosis: How much iron may the body contain?
As much as 50g, enough to set off airport metal detectors
Hemochromatosis: How is this condition treated?
Phlebotomy, deferoxamine
Hemochromatosis: AUTHOR
Tzevan Poon
Jaundice: bacteria convert conjugated bilirubin into what? what happens to some of this?
urobilinogen --> some which is reabsorbed, some which is excreted. (urobilinogen that's reabsorbed is converted to urobilin= yellow pigment in urine)
Jaundice: which is soluble in water/urine: conjugated or unconjugated bilirubin?
conjugted of course!
Jaundice: check out the 3 jaundice types: what are they? ***cover up the various parts of the chart in the text and test yourself.
heptocellular vs. obstructive vs. hemolytic jaundice
Heriditary hyper-bilirubinemias: What is the biochemical disorder in GILBERTS SYNDROME?
mild ↓ in UDP-glucuronyl transferase (this is key enzyme in conjugation of bilirubin)
Heriditary hyper-bilirubinemias: what are the symptoms?
asymptomatic
Heriditary hyper-bilirubinemias: what type of bilirubin is seen upon the lab findings? Do you have do you have major hemolysis?
unconjugated bilirubin is elevated w/o overt hemolysis
Heriditary hyper-bilirubinemias: What is the syndrome called when you have an ABSENCE of UDP-glc-ur-transferase?
Crigler-Najjar (CN) syndrome type 1 (type 2 is LESS severe)
Heriditary hyper-bilirubinemias: when does it present in life? What is the prognosis?
presents early in life: pts die in a few years.
Heriditary hyper-bilirubinemias: name 3 findings of C-N syndrome?
jaundice, kernicterus, ↑ unconjugated bilirubin
Heriditary hyper-bilirubinemias: type 2 is more sever or less severe? What do you treat it with?
less severe than type 1; treat type 2 with phenobarbital.
Heriditary hyper-bilirubinemias: what do you treat type 1 with (name 2)?
plasmapheresis + phototherapy (breaks down unconj. Bilirubin)
Heriditary hyper-bilirubinemias: What is Dubin Johnson syndrome?
↑↑ hyperbilirubin (conjugated) due to defective liver EXCRETION
Heriditary hyper-bilirubinemias: what does the liver look on gross exam?
black liver grossly
Heriditary hyper-bilirubinemias: what is the name of the less severe syndrome akin to Dubin Johnson?
Rotor's syndrome: also no black liver.
Heriditary hyper-bilirubinemias: see drawing: it's nice.
0
Primary sclerosing cholangitis: what is the pathophysiology
segmental inflammation and fibrosis of bile ducts: unaffected parts are dilated.
Primary sclerosing cholangitis: what test do you do to see this stricture/dilating/beading pattern?
ERCP
Primary sclerosing cholangitis: what disease is this associated with?
Ulcerative Colitis
Primary sclerosing cholangitis: What can prim. Scl. Cholangitis lead to?
2° biliary cirrhosis/sclerosis
Biliary sclerosis : what is the cause of Primary Biliary Sclerosis (PBS)? (compare the name/disease of PBS vs. PSC: primary sclerosing cholangitis)
autoimmune disorder with antimitochondrial antibodies
Biliary sclerosis : name 3 signs associated with PBS:
1) severe obstructive jaundice 2) itching 3) hypercholesterolemia (xanthoma)
Biliary sclerosis : What is the cause of SBS: secondary biliary sclerosis?
EXTRA-hepatic biliary obstruction → ↑ pressure in INTRA-hepatic ducts → ↑ injury/sclerosis
Biliary sclerosis : what is 2° biliary sclerosis associated with (name 3 things)?
ascending cholangitis (bacterial infection), bile stasis, and bile lakes
Heptocellular carcinoma = hepatoma: in liver cancers, is this common?
yes, #1 most common!
Heptocellular carcinoma = hepatoma: there is an ↑ incidence of hepatoma due to what diseases/exposures (name 7)
1) hep B, 2) hep C, 3) Wilson's disease, 4) hemochromatosis, 5) a-antitrypsin def 6) EtOH cirrhosis 6) carcinogens (e.g. aflatoxin B1)
Heptocellular carcinoma = hepatoma: How do hepatomas spread? How does this differ with renal cell ca. spread?
They BOTH commonly spread hematogenously!
Heptocellular carcinoma = hepatoma: What is major lab finding with hepatocelluar ca?
↑ AFP (alpha fetoprotein)
Reye's syndrome = rare!!! But…: What is this syndrome?
Hepato-encephalopathy: fatal: occurs in childhood
Reye's syndrome = rare!!! But…: name 3 findings:
1) fatty liver w/ microvesicular fatty change 2) hypoglycemia, 3) coma
Reye's syndrome = rare!!! But…: is this associated with any outside factors/triggering factors?
yes: 1) viral infection (esp. VZV or influenza B) 2) salicylates!!!
Reye's syndrome = rare!!! But…: if baby/child has fever what do you use in place of aspirin?
use acetaminophen (but with caution): i.e. acetaminophen OD hepato-toxicity
Gallstones: name 3 types of stones and tell me how they show up on CT scan:
1) cholesterol stone (radiolucent but 10-20% opaque due to calcification) 2) mixed stone = cholesterol + pigment (radiolucent) 3) pigment stone (radio-opaque)
Gallstones: Which is the most common type of gall stone?
mixed stone
Gallstones: name at least 5 out 9 risk factors for Cholesterol stones: is there a mnemonic?
1) obesity 2) Crohn's D 3) Cystic fibrosis 4) elderly 5) clofibrate 6) estrogens 7) multiparity 8) rapid weight loss 9) native American origin mnemonic = 4 F's fat, female….
Gallstones: name 4 risk factors for pigment stones:
1) pts with chronic RBC hemolysis 2) alcoholic cirrhosis 3) elderly 4) biliary infection
Gallstones: What is a good way to diagnosis stones?
ultrasound
Gallstones: What is CHARCOT'S TRIAD
1) epigastric/RUQ pain 2)fever 3) jaundice
Acute pancreatitis: this is what our US attorney general had. : pathophysiology mechanism?
activation and thus autodigestion by pancreatic enzymes
Acute pancreatitis: this is what our US attorney general had. : Causes? (good mnemonic)
Get Smashed (EtOH will can lead to AP) = 1) Gallstones 2) EtOH 3) Trauma 4) Steroids 5) Mumps 6) autoimmune disease 7) scorpion sting of all things! 8) hyperlipidemia 9) drugs
Acute pancreatitis: this is what our US attorney general had. : clinically presents with?
intense epigastric pain radiating to back
Acute pancreatitis: this is what our US attorney general had. : labs(name 2): which of the two has a higher specificity?
↑ amylase + ↑lipase (higher specificity) "li is hi"
Acute pancreatitis: this is what our US attorney general had. : Name 5 nasty complications that can result from AP?
1) DIC 2) ARDS 3) diffuse fat necrosis 4) hypocalcemia 5) pseudocyst formation
Acute pancreatitis: this is what our US attorney general had. : if you have CHRONIC pancreatitis: what do we think of your life habits?
Chronic Pancr. Is strongly associated w/ alcoholism
Pancreatic cancer: what is a common and fatal pancreatic cancer?
pancreatic adenocarcinoma
Pancreatic cancer: what is the prognosis?
6 months: very aggressive: usually has mets when pt. presents
Pancreatic cancer: where are tumors most commonly located? And what does this lead to?
pancreatic head → obstructive jaundice.
Pancreatic cancer: PATHOLOGY-RESPIRATORY SECTION
0
COPD = also known as OLD: obstructive lung disease: why is it called obstructive?
obstruction of AIR FLOW → air traping in lungs
COPD = also known as OLD: obstructive lung disease: what is the major PFT finding?
FEV1 / FVC ration is ↓ (hallmark finding)
COPD = also known as OLD: obstructive lung disease: name the 4 types of COPD
1) Chronic Bronchitis (Blue Bloater) 2) emphysema (pink puffer) 3) asthma 4) bronchietasis
COPD = also known as OLD: obstructive lung disease: what is the definition of Chronic Bronchitis
productive cough for >3 consecutive months in two or more years.
COPD = also known as OLD: obstructive lung disease: what do you expect on lung histology?
hypertrophy of mucus-secreting glands in the bronchioles (Reid index of >50%)
COPD = also known as OLD: obstructive lung disease: leading cause is smoking: what are the physical findings for Chronic Bronchitis? (name 3)
1)cyanosis 2) wheezing 3) crackles
COPD = also known as OLD: obstructive lung disease: what is the pathophysiological mechanism of EMPHYSEMA?
destruction of fibrous septa/alveolar walls → enlargement of air space and ↓ total surface area for gas exchange
COPD = also known as OLD: obstructive lung disease: if the cause is smoking, what kind of emphysema would you see on histo slide?
centri-acinar emphysema
COPD = also known as OLD: obstructive lung disease: what else can cause emphysema: (also may work synergistically with smoking): What kind of findings do you see in lung histo and name another organ affected?
alpha 1-antitrypsin deficiency → panacinar emphysema + liver cirrhosis
COPD = also known as OLD: obstructive lung disease: what causes the emphysema in this disorder?
↑ elastase activity to damage lung tissue.
COPD = also known as OLD: obstructive lung disease: name 4 findings of emphysema (in general)
1) dyspnea; 2) ↓ breath sounds 3) tachycardia 4) ↓ I/E ratio
COPD = also known as OLD: obstructive lung disease: What is mechanism of asthma
BRONCHIAL hypersensitivity/hyperresponsiveness → REVERSIBLE bronchoconstriction
COPD = also known as OLD: obstructive lung disease: name 3 common triggers
1) viral URI 2) allergens 3) stress!!
COPD = also known as OLD: obstructive lung disease: name 7 findings
1) cough 2) wheezing 3) dyspnea 4) hypoxemia 5)↓ I/E ratio 6) tachypnea 7) pulsus paradoxus
COPD = also known as OLD: obstructive lung disease: BRONCHIECTASIS: what is its mechanism??
chronic necrotizing infection of BRONCHI → dilated airways, purulent sputum, recurrent infections, hemoptysis (see Robbins for a good discussion of this)
COPD = also known as OLD: obstructive lung disease: what disorders is bronchietasis commonly associated with?
1) bronchial obstruction 2) cystic fibrosis 3) poor ciliary motility
Restrictive Lung Disease (RLD): What are classic PFT findings for RLD?
↓ VC ↓TLC ; FEV1/FVC ration > 80%
Restrictive Lung Disease (RLD): Name the two MAJOR types of RLD
1) poor breathing mechanics (EXTRA-pulmonary) 2) Interstitial lung diseases
Restrictive Lung Disease (RLD): cover up various parts of the text/table/outline to test yourself on the following: 1) 2 types of poor breathing mechanics 2) 8 types of interstitial disease that give you a restrictive picture.
0
Lung Physical Findings: cover up parts of the very good table to test yourself on various findings.
0
Lung Physical Findings: Obstructive vs. Restrictive findings
0
Lung Physical Findings: Name 3 lung volumes that are increased in COPD
↑TLC, ↑FRC, ↑RV
Lung Physical Findings: What 2 values are BOTH reduced in COPD and RLD?
1) FEV1 and 2) FVC (think FEV1/FVC ratio) NOTE! in COPD, FEV1 is more dramatically reduced and thus the FEV1/FVC ratio is ↓
Asbestosis = long latency = think shipbuilders and plumbers: what is the main pathology resulting from asbestosis?
DIFFUSE, interstitial fibrosis caused by inhaled asbestos Fibers.
Asbestosis = long latency = think shipbuilders and plumbers: What cancers are increased in pts with asbestosis?
1) pleural mesothelioma 2) bronchogenic carcinoma (BC)
Asbestosis = long latency = think shipbuilders and plumbers: Major finding in lung?
Ferruginous bodies: asbestos fibers coated with hemosiderin also 2) ivory white pleural plaques
Neonatal respiratory distress syndrome: What is the main cause
surfactant deficiency --> to increased surface tension --> alveolar collapse
Neonatal respiratory distress syndrome: surfactant is made by which cells? After when?
type 2 pneumocytes after 35th gestational week
Neonatal respiratory distress syndrome: what do you measure? Where do you get this fluid?
lecithin-to-sphingomyelin ratio in the amniotic fluid = measure of lung maturation <1.5 in neonatal distress syndrome
Neonatal respiratory distress syndrome: what is surfactant made of (chemical name)
dipalmitoyl phosph-tidyl-choline (DP-PTC)
Neonatal respiratory distress syndrome: treatment for poor maturation of lungs
1) before birth = maternal steroids 2) after= artificial surfactant
Karta-gener's syndrome: what is this?
immotile cilia due to dynein arm defect
Karta-gener's syndrome: results in what in female and male? (4 things)
1) sterility (in male sperm also immotile) 2)bronchietasis 3)recurrent sinusitis (bacteria & particles not pushed out) 4) associated with situs inversus (e.g. dextro-cardia)
Lung Cancer: name the 3 main classes of cancers that affect parts of the lung
1) bronchogenic carcinoma (with different subtypes) 2) carcinoid tumor 3) metastasis
Lung Cancer: list the 5 types of major bronchogenic carcinomas
CENTRAL 1) squamous cell ca 2)small cell ca PERIPHERAL 3) adenocarcinoma 4) bronchoalveolar ca 5) large cell ca
Lung Cancer: mnemonic: what is meant by SPHERE of symptoms?
S= superior vena cava syndrome; P= pancoast tumor; H= Horner's syndrome; E= Endocrine (paraneoplastic); Recurrent laryngeal / hoarseness; E = Effusions (pleural OR pericardial)
Lung Cancer: What can a CARCINOID tumor cause?
Carcinoid Syndrome = flushing, diarrhea, wheezing, and salivation
Lung Cancer: Metastases to lung is very common, LUNG cancer also prone to metastasize to what other parts?
1) brain (epilepsy) 2) bone (fracture) 3)liver (jaundice + hepatomegaly)
Pancoast tumor: What is it?
it's a carcinoma of the apex of lung
Pancoast tumor: what may it affect?
may affect CSP= cervical sympathetic plexus causing Horner's syndrome
Pancoast tumor: what is Horner's syndrome?
P.A.M. is Horny = Ptosis, Anhydrosis, Miosis
Pneumonia: 1st AID breaks it down into TYPE/ ORGANISM/ CHARACTERISTICS = cover parts of this table and fill in the blanks
0
Pneumonia: Lobar Pneumonia; Organism(s) and characteristics
Pneumococcus; intraalveolar exudate -> consolidation, may involve entire lung
Pneumonia: Bronchopneumonia: Organism and characteristics
S aureus, H flu, Klebsiella, S pyogenes; Acute inflammatory infiltrates from bronchioles into adjacent alveoli; patchy distribution involving >= 1 lobes
Pneumonia: Interstitial pneumonia; Organism(s) and characteristics
Viruses, mycoplasma, legionella; diffuse patchy inflammation localized to interstitial areas at alvolar walls; distribution involving >= 1 lobes
Pneumonia: PATHOLOGY - NEUROLOGIC
0
Degenerative diseases: name two diseases that affect the CEREBRAL CORTEX
1) Alzheimer's disease 2) Pick's disease
Degenerative diseases: name two diseases that affect BASAL GANGLIA & BRAIN STEM:
1) Huntington's disease 2) Parkinson disease
Degenerative diseases: name 5 diseases that affect SPINOCEREBELLAR MOTOR NEURON
1) Olivo-ponto-cerebellar atrophy 2) Friedreich's ataxia 3)ALS = amyotrophic lateral sclerosis 4)Werdnig-Hoffmann disease 5) Polio
Degenerative diseases: what is the most common cause of elderly dementia?
Alzheimer's
Degenerative diseases: name two pathological findings characteristic of Alzheimer's
1) senile plaques = intracytoplasmic inclusion bodies 2) neurofibrillary tangles (abnormally phosphorylated tau protein)
Degenerative diseases: what is the 2nd most common cause of dementia in elderly?
multi-infarct dementia in elderly\\
Degenerative diseases: How can Alzheimer also affect intracranial vasculature?
amyloid angiopathy --> intracranial hemorrhage
Degenerative diseases: The familial form of Alzheimer's is associated with what chromosomes (name 4) and (name the allele's name in 2 of the 4)
chromosome 1, 14, 19 (APO-E4 allele), 21 (p-Ap gene)
Degenerative diseases: What is pathognemomic with Pick's disease upon histology?
Pick bodies = intra-cytoplasmic inclusion bodies
Degenerative diseases: What cortical areas does Pick's disease affect
Frontal and temporal lobes (remember; sharp, atrophic apearance of gross specimen)
Degenerative diseases: What is the inheritance pattern of Huntington's
Auto Dominant
Degenerative diseases: What are some symptoms?
1) chorea 2) dementia
Degenerative diseases: Huntington's is due to atrophy of _____ what? Loss of ___ ergic neurons.
atrophy of caudate nucleus = loss of GABA-nergic neurons
Degenerative diseases: what is the genetic abnormality of Huntington's?
1) Chr. 4 - expansion of CAG repeats
Degenerative diseases: Parkinson disease is associated with what pathology findings?
1) Lewy bodies 2) depigmentation of substantia nigra (loss of dopaminergic neurons)
Degenerative diseases: rare cases of Parkinson's have been linked to what contaminant of certain illicit drugs?
MPTP = contaminant to street drug
Degenerative diseases: Parkinson's can make you feel in a "TRAP" = ?
T = tremor at rest; R = cogwheel rigidity; A = akinesia ; Postural instability
Degenerative diseases: ALS = Lou Gehrig's disease (the Iron Horse of the Yankees, hero of Joe DiMaggio) = what signs is ALS associated with?
Both UMN and LMN deficits
Degenerative diseases: Werdnig-Hoffman disease - presents as birth as?
1) flopy baby syndrome 2) note tongue fasciculations as well (also seen in ALS)
Degenerative diseases: for Polio, what kind of signs to you see?
predominantly LMN deficits.
Intracranial Hemorrhage/Hemotomas: name 4 types of cranial related hemorrhages (think of layers that could possibly be involved)
1) epidural hemorrhage 2) subdural h 3) subarachnoid h 4) parenchymal h
Intracranial Hemorrhage/Hemotomas: what is a common site of epidural hematoma/hemorrhage
1) rupture of MMA: middle meningeal artery, often 2ndary to fracture of temporal bone
Intracranial Hemorrhage/Hemotomas: what does CT show for epidural h?
biconcave disc NOT crossing suture lines
Intracranial Hemorrhage/Hemotomas: what else do you see?
lucid interval
Intracranial Hemorrhage/Hemotomas: How do subdural h. most often occur?
rupture of bridging veins (think, elderly being jolted in roller coaster)
Intracranial Hemorrhage/Hemotomas: this is venous bleeding so how would this influence symptomatic findings?
venous = less pressure = delayed onset of symptoms
Intracranial Hemorrhage/Hemotomas: what types of people do you see subdural h.?
1) elderly, 2) alcoholics 3) blunt trauma/sudden change in velocity injuries
Intracranial Hemorrhage/Hemotomas: for SUBDURALs what do you see upon CT?
1) crescent-shaped hemorrhage instead 2) YES IT does cross the suture lines
Intracranial Hemorrhage/Hemotomas: subarachnoid hemorrhage is often seen where (name 2 types
rupture of 1) aneurysm (usually Berry aneurysm) or an 2)AVM
Intracranial Hemorrhage/Hemotomas: symptoms?
the worst headache of my life
Intracranial Hemorrhage/Hemotomas: since its subarachnoid, what do you see on what test?
bloody or xanthochromic CSF on spinal tap
Intracranial Hemorrhage/Hemotomas: name 4 risk factors for parenchymal hematoma
1) HTN 2) amyloid angiopathy 3) diabetes mellitus 4) tumor
Intracranial Hemorrhage/Hemotomas: AUTHOR
Hilcias Duran
Berry aneurysms: most common site for Berry aneurysms
bifurcation of the anterior communicating artery (Circle of Willis)
Berry aneurysms: berry aneurysms are associated with
adult polycystic disease, Ehlers-Danlos s., and marfan's
Berry aneurysms: Classic triad of Multiple Sclerosis
SIN: Scanning speech, intention tremor, nystagmus (affect woman 20-30s)
Berry aneurysms: Prevalence of MS
increase prevalence w/ increased distance from the equator
Berry aneurysms: Clinical s/s of MS
periventricular plaques, preservation of axons, loss of oligodentrocytes, reactive astrocystic gliosis, increased protein in csf(IgG) in CSF, relapsing course, optic neuritis, MLF syndrome, hemiparesis, bladder/bowel incontinence)
Berry aneurysms: Progressive multifocal leukoencephalopathy (PML) is associated w/
JC virus and seen in 2-4 % of AIDS pts (reactivation or latent infect)
Berry aneurysms: pathogenesis of Guillian-Barre syndrome
Inflammation and demyelination of peripheral nerves and motor fibers of the ventral roots (sensory effec less severe than motor)
Berry aneurysms: s/s of Guillian-Barre syndrome
symmetrical ascending muscle weakness beginning in the distal lower extremities. Autonomic fxn may be severely affected (eg. Cardiac irregularities, HTN, or hypoTN) Findings: inc. csf protein w/ normal cell count ("albumino-cytologic dissociation") elevated protein may lead to papilledema
Berry aneurysms: association between G-B syndrome and…
herpesvirus or camplobacter jejuni infection, inoculations, and stress but no definitive link to a pathogen
Poliomyelitis: organism causing polio and mechanism of action
poliovirus, transmitted via fecal oral route and enters blood stream then into CNS where it causes destruction of the anterior horn of S.C. leading in turn to LMN destruction
Poliomyelitis: s/s of polio
malaise, HA, fever, nausea, abd. Pain, sore throats, signs of LMN signs
Poliomyelitis: finding of polio
csf w/ lymphocytic pleocytosis w/ slight elevation of protein, virus recovered from stool or throat
Aphasia : describe Broca's (expressive) aphasia
confluent aphasia w/ intact comprehension broca's is broken speech
Aphasia : describe Wernicke's (aphasia)
Fluent aphasia w/ impaired concentration Wernick's ="What?" W area located in superior temperal gyrus
seizures : describe two types of partial seizures
1.simple partial (awareness is intact)--motor, sensory, autonomic, psychic 2.Complex partial (impaired awareness)
seizures : describe types of Generalized seizure ( diffuse)
1.absence- blank stare (petit mal) 2.myoclonic- quick repetitive jerks 3.tonic-clonic- alternating stiffening and mvmt (grand mal) 4. Tonic- stiffening 5.atonic-"drop" seizures
seizures : what are the causes of seizures in children?
genetic, infection, trauma, congenital, metabolic
seizures : adults?
tumors, trauma, stroke, infection
seizures : elderly?
stroke, tumor, trauma, metabolic, infection
Horner's syndrome: what is Horner's syndrome?
sympathectomy of face (lesion above T1) 1.Ptosis 2.anhidrosis and flushing of affected side 3.miosis [PAM is horny]
Horner's syndrome: Horner's syndrome is associated w/ what type of tumor?
pancoast tumor
syringomyelia: what is syringomyelia?
enlargement of the central canal of the S.C. Crossing fibers of spinothalamic tract are damaged.
syringomyelia: what are the s/s of syringomyelia?
b/l loss of pain and temp sensation in the uper extremities w/ the touch sensation. Most common at C8-T1
syringomyelia: commonly seen in what pts?
pts w/ Arnold-Chiari malformation
Tabes dorsalis : what infection is tabes dorsalis associated w/?
teriary syphilis
Tabes dorsalis : what happens in the CNS?
degeneration of dorsal columns and dorsal root
Tabes dorsalis : dorsalis also is associated with what s/s?
charcot's joint, shooting (lightning) pain, Argyll Robertson pupils, and absence of DTRs
Osteoarthritis : what occurs in OA?
mechanical wear and tear of the joints, leads to destruction of the articular cartilage, subchondral bone formation, sclerosis, osteophytes, eburnation, Herberden's nodes (DIP), and Bouchard's nodes (PIP)
Osteoarthritis : classical presentation of OA?
pain in weight bearing joints after use (worse at the end of the day0 improves w/ rest. No systemic symptoms
Rheumatoid Arthritis : what happens in in RA?
autoimmune inflammatory proccess which affects synovial joints, w/ pannus formation in joints (MCP, PIP), S-Q rheumatoid nodules, ulnar deviation,subluxation
Rheumatoid Arthritis : classical presentation of RA?
female, 80% pts Rf positive (anti-IgG Ab) morning stiffness improving w/ use, symmetrical joint involvement, and systemic symptoms (fever, fatigue, pleurtis, pericarditis)
Rheumatoid Arthritis : AUTHOR
Jason Portnof
Sjogren's Syndrome: What is the classic triad of Sjogren’s syndrome?
dry eyes (conjunctivitis, xeropthalmia), dry mouth (dysphagia, xerostomia), arthritis
Sjogren's Syndrome: What is SICCA syndrome?
dry eyes, dry mouth, nasal and vaginal dryness, chronic bronchitis, reflux esophagitis
Sjogren's Syndrome: T/F Sjogren’s syndrome predominantly affects males between 40 and 60 years of age.
False. Females between 40 and 60 y.o are predominantly affected in Sjogren’s syndrome.
Gout: T/F Gout is the precipitation of monosodium urate crystals into joints due to hyperuricemia.
TRUE
Gout: What is the favored manifestation of gout?
Podagra. (painful MTP joint)
Gout: The crystals of gout are:
needle shaped and negatively bifringent
Pseudogout: T/F Pseudogout is the precipitation of calcium pyrophosphate crystals within the joint space.
TRUE
Pseudogout: The crystals of psuedogout are:
basophilic, rhomboid crystals
Pseudogout: Psudogout usually affects:
Large joints (classically the knee)
Pseudogout: T/F Both gout and psuedogout predominantly affect men.
False. Gout is more common in men. Pseudogout affects both sexes equally, most pts are >50 years old.
Pseudogout: T/F The treatment for both pseudogout and gout is allopurinol, probenecid, colchicines, and NSAIDS.
False. That is the tx for gout. However, there is no tx for pseudogout.
Celiac sprue: Celiac sprue is the autoimmune-mediated intolerance of what substance, leading to steatorrhea?
gliadin (wheat)
Celiac sprue: T/F Findings in Celiac sprue include blunting of villi, neutrophils in the lamina propria, and abnormal D-xylose test.
False. lymphocytes, not neutrophils are found in the lamina propria.
Systemic Lupus Erythematosus: What drugs can produce a SLE-like syndrome that is commonly reversible?
HIp: Hydralazine, INH, Phenytoin, Procainamide
Systemic Lupus Erythematosus: SLE causes LSE. What is LSE?
Libman Sacks Endocarditis. Valvular vegetations found on both sides of valve (Mitral Valve Stenosis) and do not embolize.
Systemic Lupus Erythematosus: T/F 90% Systemic lupus erythematousus pts are female between ages 14 and 45. SLE is most common and severe in black females.
Both statements are true
Systemic Lupus Erythematosus: What are 4 lab tests in SLE? Which test, when positive is predictive of a poor prognosis?
1. ANA (antinuclear antibodies)- sensitive, not specific for SLE, 2. anti-ds DNA (antibodies to double stranded DNA)- very specific, poor prognosis, 3. Anti-Smith antibodies (anti-Sm)- very specific, but not prognostic, 4. Antihistone antibodies- drug induced lupus
Systemic Lupus Erythematosus: In what organ are wire loop lesions found in SLE?
kidney (with immune complex deposition and nephrotic syndrome)
Systemic Lupus Erythematosus: What causes death commonly in SLE?
Renal failures and infections
Systemic Lupus Erythematosus: What are symptoms of SLE?
fever, fatigue, wt loss, joint pain, malar rash, photosensitivity. Also: pleuritis, pericarditis, nonbacterial verrucous endocarditis, Raynaud’s phenomenon
Sarcoidosis: What disease is characterized by gamaglobulinemia, rheumatoid arthritis, ace increase, interstitial fibrosis, and noncaseating granulomas?
Sarcoidosis
Seronegative spondyloarthropathies: Name 2 seronegative spondyloarthropathies.
ankylosing spondylitis, reiters’ syndrome
Seronegative spondyloarthropathies: What is a seronegative spondyloarthropathy?
Arthritis without rheumatoid factor (no anti-IgG Ab). Strong association with HLA-B27 (gene that codes for HLA MHC1)
Seronegative spondyloarthropathies: T/F seronegative spondyloarthropahties occur more often in males
TRUE
Seronegative spondyloarthropathies: What is the classic triad for Reiter’s syndrome?
Urethritis, conjunctivitis and anterior uveitis, arthritis. “Can’t see, can’t pee, can’t climb a tree”
Seronegative spondyloarthropathies: T/F reiter’s syndrome occurs commonly post respiratory or chlamydia infections.
False. Post-GI or chlamydia infections
Seronegative spondyloarthropathies: Describe ankylosing spondylitis
Chronic inflammatory disease of spine and sacroiliac joints. Ankylosis (stiff spine), uveitis and aortic regurg.
Scleroderma: T/F Crest syndrome is wider-spread than diffuse scleroderma.
False. Diffuse scleroderma has widespread skin involvement, rapid progression, early visceral involvement. CREST syndrome has limited skin involvement, often confined to fingers and face. More benign clinical course.
Scleroderma: T/F Crest syndrome occurs in male pts that have allergic rxn to toothpaste.
False. CREST= calcinosis, raynaud’s phenomenon, esophageal dysmotility, sclerodactyly, telangiectasia.
Scleroderma: What antibodies are associated with scleroderma (progressive systemic sclerosis- PSS)?
"diffuse scleroderma- anti-Scl-70 antibody, Crest syndrome- anticentromere antibody
Skin disorders: T/F Pemphigus vulgaris is described as an autoimmune disorder with IgG antibody against epidural basement membrane (linear immunofluorescence). Affects skin but spares oral mucosa.
False. That description is for Bullous pemphigoid (less severe disease than Pemphigus vulgaris). Pemphigus vulgaris is a potentially fatal autoimmune skin disorder. Intradermal bullae involve the oral mucosa and skin. Findings include acantholysis, IgG ab against epidermal cell surface.
Skin disorders: T/F Dermatitis is a group of inflammatory pruritic skin disorders. The etiology is allergy (usually type IV hypersensitivity), chemical injury, or infection.
TRUE
Skin disorders: What HLA groups are associated with psoriasis?
HLA-B27, HLA-13, HLA-17
Skin disorders: What is psoriasis? Where is it most commonly found?
Psoriasis is nonpruritic chronic inflammation of the skin, particularly on the knees and elbows.
Goodpasture's: T/F Goodpasture’s syndrome affects the lungs and liver.
False. Lungs and Kidneys. (Pulmonary hemorrhagesà hemoptysis, renal lesionsà hematuria, anemia, crescentic glomerulonephritis)
Goodpasture's: What type of Abs are associated with Goodpastures?
anti-glomerular basement membrane abs produce linear staining on immunofluorescence.
Goodpasture's: Who commonly gets Goodpastures?
Men between 20-40 years.
Cushing's syndrome: What are etiologies of Cushing’s syndrome? Is ACTH always elevated?
increased cortisol due to:, 1. Cushing’s disease (primary pituitary adenoma) increased ACTH, 2. Primary adrenal (hyperplasia/neoplasia) decreased ACTH, 3. Ectopic ACTH production (ie- small cell lung ca) increased ACTH, 4. Iatrogenic, decreased ACTH
Cushing's syndrome: Describe the clinical picture of Cushings.
HTN, wt gain, moon facies, truncal obesity, buffalo hump, hyperglycemia (insulin resistance), skin change (thinning, striae), osteoporosis, immune supression
Hyperaldosteronism: T/F Conn’s syndrome is secondary hyperaldosteronism.
False. Conn’s syndrome is primary hyperaldosteronism, caused by an aldosterone-secreting tumor. Results in HTN, hypokalemia, metabolic alkalosis, low plasma renin.
Hyperaldosteronism: Which hyperaldosteronism is associated with high plasma renin?
Secondary hyperaldosteronism. It is due to renal artery stenosis, chronic renal failure, CHF, cirrhosis, or nephrotic syndrome. Kidney perception of low intravascular volume results in an overactive renin-angiotensin sysem.
Hyperaldosteronism: What is the tx for hyperaldosteronism?
Spironolactone, a diuretic that works by acting as a aldosterone antagonist.
Addison's Disease: What characterizes addison’s disease? (which hormones are elevated or deficient)? Is it associated with HTN or hypotension?
Primary deficiency of aldosterone and cortisol due to adrenal atrophy, causing hypotension and skin hyperpigmentation. Adrenal atrophy, absence of hormone production, involves all 3 cortical divisions.
Addison's Disease: T/F In primary insufficiency decreased pituitary ACTH production is characterized by skin hyperpigmentation.
False: increased ACTH causes MSH activity & hyperpigmentation
Tumors of adrenal medulla: T/F Neuroblastoma is the most common tumor of adrenal medulla in adults.
False. Pheochromocytoma is the most common tumor of adrenal medulla in adults. It is derived from chromaffin cells (arise from neural crest). It is associated with neurofibromatosis MEN types II and III.
Tumors of adrenal medulla: Where does neuroblastoma occur?
Neuroblastoma is the most common tumor of adrenal medulla in children. It can occur anywhere along the sympathetic chain.
Tumors of adrenal medulla: AUTHOR
Jason Lee
Pheochromocytoma: secrete combination of two molecules
epinephrine and norepinephrine
Pheochromocytoma: epidemiology (rule of 10's)
10% malignant, 10% bilateral, 10% extra-adrenal, 10% calcify, 10% kids, 10% familial
Pheochromocytoma: symptoms - 5 P's
(elevated blood) Pressure, Pain (headache), Perspiration, Palpitations, Pallor/diaphoresis --> relapsing and remittant
Pheochromocytoma: elevations in two lab values
urinary VMA and serum catecholamines
Pheochromocytoma: association with two other endocrine diseases
MEN II and III
Pheochromocytoma: treatment
alpha-antagonists
Multiple Endocrine Neoplasia: type I (Wermer's syndrome) - 3 P's
Pancreas, Pituitary, and Parathyroid tumors
Multiple Endocrine Neoplasia: type II (Sipple's syndrome)
medullary carcinoma of thyroid, pheochromocytoma, parathyroid tumor, or adenoma
Multiple Endocrine Neoplasia: type III
medullary carcinoma of thyroid, pheochromocytoma, oral/intestinal ganglioneuromatosis
Hypothyroidism and hyperthyroidism: myxedema is prominent in which one
hypothyroidism
Hypothyroidism and hyperthyroidism: chest pain, palpitations, arrhythmias
hyperthyroidism
Hypothyroidism and hyperthyroidism: TSH is (increased/decreased) in primary hyperthyroidism? In primary hypothyroidism?
hyper - decreased, hypo - increased
Hypothyroidism and hyperthyroidism: Graves' disease involves autoantibodies with what mechanism of action?
stimulation of TSH receptors
Hypothyroidism and hyperthyroidism: three symptoms of Graves'
ophthalmopathy, pretibial myxedema, diffuse goiter
Hypothyroidism and hyperthyroidism: Graves' is a type __ hypersensitivity
type II
Hashimoto's thyroiditis: thyroid is (enlarged/not enlarged) and (tender/nontender)
enlarged, nontender
Hashimoto's thyroiditis: autoimmune antibodies directed against ---
microsomes
Hashimoto's thyroiditis: histology shows (type of cell) infiltrate
lymphocytes (with germinal centers)
Subacute thyroiditis (de Quervain's): self-limited (hyper/hypo)thyroidism following ---
hypothyroidism following flu-like illness
Subacute thyroiditis (de Quervain's): symptoms include
jaw pain, tender thyroid gland, early hyperthyroidism
Thyroid cancer: most common, good prognosis, "ground glass" nuclei, psammoma bodies
papillary carcinoma
Thyroid cancer: poor prognosis, uniform follicles
follicular carcinoma
Thyroid cancer: calcitonin producing (C cells), sheets of cells
medullary carcinoma (MEN II and III)
Thyroid cancer: older patients, horrible prognosis
undifferentiated/anaplastic
Cretinism: caused by a lack of dietary --- or defect in --- formation
iodine (endemic), T4 (sporadic)
Cretinism: symptoms include
pot-belly, paleness, puffy face, protuberant tongue, protruding umbilicus
Acromegaly: caused by excess --- in adults
growth hormone
Acromegaly: symptoms include
large furrowed tongue, deep voice, large hands and feet, coarse facial features
Acromegaly: in children, leads to ---
gigantism
Diabetes mellitus: acute symptoms common to both types
polydipsia, polyuria, polyphagia, weight loss
Diabetes mellitus: match: (DKA/hyperosmolar coma) with (type I/type II)
DKA=type I, hyperosmolar coma=type II
Diabetes mellitus: effects (increased/decreased) of insulin deficiency and glucagon excess on 1) glucose uptake, 2) protein catabolism, 3) lipolysis
1) decreased, 2) increased, 3) increased
Diabetes mellitus: increased plasma free fatty acids leads to
ketogenesis
Diabetes mellitus: hyperglycemia has what effect on blood volume and electrolytes
decreased volume (osmotic diuresis), electrolyte depletion
Diabetes mellitus: three chronic manifestations
retinopathy (hemorrhage, exudate, microaneurysm), nephropathy (nodular sclerosis), neuropathy (sensory, motor, autonomic)
Diabetes mellitus: sorbitol accumulation leads to what sequelae in the eye
cataracts, glaucoma
Diabetes mellitus: three tests - what are they? which one measures long-term glucose control?
fasting glucose, glucose tolerance test, HbA1c
Diabetes mellitus: AUTHOR
Jacob Pugsley
Type 1 - juvenile onset: % of diabetes
0.15
Type 1 - juvenile onset: Insulin necessary in treatment
Always
Type 1 - juvenile onset: Age
<30
Type 1 - juvenile onset: Association with obesity
No
Type 1 - juvenile onset: Genetic predisposition
weak, polygenic
Type 1 - juvenile onset: Association with HLA system
Yes (HLA-DR3 & 4)
Type 1 - juvenile onset: Glucose intolerance
Severe
Type 1 - juvenile onset: Ketoacidosis
Common
Type 1 - juvenile onset: B-cell numbers in the islets
decreased
Type 1 - juvenile onset: Serum insulin levels
decreased
Type 1 - juvenile onset: Classic symptoms of polyuria, polydipsia, thirst, weight loss
Common
Type 1 - juvenile onset: Theorized cause
viral or immune destruction of B cells
Type 2 - adult onset: % of diabetes
0.85
Type 2 - adult onset: Insulin necessary in treatment
Sometimes
Type 2 - adult onset: Age
>40
Type 2 - adult onset: Association with obesity
Yes
Type 2 - adult onset: Genetic predisposition
Strong, polygenic
Type 2 - adult onset: Association with HLA system
No
Type 2 - adult onset: Glucose intolerance
mild to moderate
Type 2 - adult onset: Ketoacidosis
Rare
Type 2 - adult onset: B-cell numbers in the islets
Variable
Type 2 - adult onset: Serum insulin levels
Variable
Type 2 - adult onset: Classic symptoms of polyuria, polydipsia, thirst, weight loss
Sometimes
Type 2 - adult onset: Theorized cause
increased resistance to insulin
Diabetic Ketoacidosis (type 1): What precipitates this
Increase in insulin requirements from increase in stress (e.g. infection)
Diabetic Ketoacidosis (type 1): Ketone bodies from where
Excess fat breakdown, increase ketogenesis from increased free fatty acids which are made into ketone bodies
Diabetic Ketoacidosis (type 1): Signs/Symptoms
Kussmaul respirations (rapid/deep breathing), hyperthermia, nausea/vomiting, abdominal pain, psychosis/dementia, dehydration, fruity breath odor
Diabetic Ketoacidosis (type 1): Labs
Hyperglycemia, high H+, low HCO3- (anion gap metabolic acidosis), high blood ketone levels, leukocytosis
Diabetic Ketoacidosis (type 1): Complications
Life threatening mucormycosis, Rhizopus infection, cerebral edema, cardiac arrhythimias, heart failure
Diabetic Ketoacidosis (type 1): Treatment
Fluids, insulin, and potassium; glucose if necessary to prevent hypoglycemia.
Diabetes insipidus: summary
intensive thirst, polyuria, inability to concentrate urine with fluid constriction owing to lack of ADH (central DI) or to lack of renal response to ADH (nephrogenic DI). Caused by lithium or demeclocycline
Diabetes insipidus: Findings
Urine specific gravity < 1.006; serum osmolality > 290 mOsm/L
Diabetes insipidus: Treatment
adequate fluid intake; Central DI - intranasal desmopressin (ADH analog); nephrogenic DI - hydrochlorothiazide, indomethacin, or amiloride
SIADH: what does it stand for
Syndrome of inapropriate ADH
SIADH: finds
1) excess water retention 2) hyponatremia [may lead to seizures - correct slowly] 3) urine osmolarity > serum osmolarity
SIADH: causes
1) Ectopic ADH [small cell lung cancer] 2) CNS disorders/head trauma 3) pulmonary disease 4) Drugs
Hyperparathyroidism: What causes primary hyperparathyroidism?
Usually an adenoma
Hyperparathyroidism: What are the findings of primary disease?
hypercalcemia, hypercalciuria, hypophosphatemia, high parathyroid hormone, high cAMP in urine; often asymptomatic, may present with weakness and constipation
Hyperparathyroidism: what is osteitis fibrosa cystica? (von Recklinghausen's syndrome)
cystic bone spaces filled with non-neoplastic fibrous tissue
Hyperparathyroidism: What causes secondary hyperparathyroidism?
low serum Ca++, most often chronic renal disease.
Hyperparathyroidism: What are the findings of secondary hyperparathyroidism?
hypocalcemia, hyperphosphatemia, high parathyroid hormone
Hypoparathyroidism: findings
hypocalcemia, tetany.
Hypoparathyroidism: causes
accidental surgical excision (thyroid surgery) or DiGeorge syndrome
Hypoparathyroidism: note: pseudohypoparathyroidism
autosomal recessive kidney unresponsiveness to PTH. Hypocalcemia, shortened 4th/5th digits, short stature
Carcinoid syndrome: frequency
rare
Carcinoid syndrome: cause
carcinoid tumors (neuroendocrine cells), especially those of the small bowel; they secrete high levels of serotonin (5-HT) that does not get metabolized by the liver due to liver metastases.
Carcinoid syndrome: symptoms
recurrent diarhea, cutaneous flushing, asthmatic wheezing, and right-sided valvular disease.
Carcinoid syndrome: most common tumor of what?
apendix
Carcinoid syndrome: lab
high 5-HIAA in urine
Carcinoid syndrome: Rule of 1/3s:
1/3 metastasize; 1/3 present with second malignancy; 1/3 multiple
Carcinoid syndrome: Treatment?
Octreotide
Carcinoid syndrome: What is this medication?
synthetic octapeptide analog of somatostatin (growth hormone-inhibiting hormone)
Zollinger-Ellison syndrome: What kind of tumor causes this?
Gastrin secreting tumor (usually) of the pancreas
Zollinger-Ellison syndrome: what does it cause?
recurrent ulcers
Zollinger-Ellison syndrome: may be associated with what?
MEN type I
Osteoporosis: what is this?
reduction in bone mass in spite of normal bone mineralization
Osteoporosis: what characterizes type 1?
Postmenopausal (10-15 years after menopause); increased bone reabsorption due to low estrogen levels. Treated with estrogen replacement
Osteoporosis: what characterizes type 2?
Senile osteoporosis - affects men and women > 70 y/o
Osteoporosis: what races are affected most?
whites>blacks>asians
Osteoporosis: symptoms of vertebral crush fractures
acute back pain, loss of height, kyphosis
Osteoporosis: other
distal radius (Colles') fracures, vertebral wedge fracures.
Osteoporosis: see picure
0
Osteoporosis: PATHOLOGY - REPRODUCTIVE
(end of endocrine)
Benign prostatic hyperplasia: who gets BPH?
common in men > 50 y/o
Benign prostatic hyperplasia: what might cause it?
may be due to age-related increase in estradiol with possible sensitization of the prostate to the growth promoting effects of DHT.
Benign prostatic hyperplasia: findings
nodular enlargement of the periurethral (lateral and middle) lobes of the prostate gland, compressing the urethra to a vertical slit.
Benign prostatic hyperplasia: symptoms
increased frequency of urination, nocturia, difficulty in starting and stoping the stream of urine, dysuria.
Benign prostatic hyperplasia: complications
may lead to distention and hypertrophy of the bladder, hydronephrosis, UTIs
Benign prostatic hyperplasia: premalignant lesion?
No
Hydatidiform mole: What is it?
pathologic ovum (empty egg - ovum with no DNA) resulting in a cystic swelling of chorionic villi and proliferation of chorionic epithelium (trophoblast).
Hydatidiform mole: Most common precursor of what?
choriocarcinoma
Hydatidiform mole: what happens to B-hCG?
elevated
Hydatidiform mole: findings
honeycombed uterus, cluster of grapes apearance
Hydatidiform mole: genotype
complete mole is 46,xx and is completely paternal in origin (no maternal chromosomes); no associated fetus. PARTial mole is made up of 3 or more PARTS (triploid or tetraploid)
Pregnancy-induced hypertension (preeclampsia-eclampsia): what is the triad?
hypertension, proteinuria, edema
Pregnancy-induced hypertension (preeclampsia-eclampsia): what makes it eclampsia?
addition of seizures to the triad
Pregnancy-induced hypertension (preeclampsia-eclampsia): what % of pregnant women?
0.07
Pregnancy-induced hypertension (preeclampsia-eclampsia): when does it present?
20 weeks gestation to 6 weeks postpartum
Pregnancy-induced hypertension (preeclampsia-eclampsia): what increases the likelihood?
preexisting hypertension, diabetes, chronic renal disease, autoimmune disorders
Pregnancy-induced hypertension (preeclampsia-eclampsia): what is HELLP syndrome?
Hemolysis, Elevated LFTs, Low Platelets
Pregnancy-induced hypertension (preeclampsia-eclampsia): Clinical features
headache, blurred vision, abdominal pain, edema of face and extremeties, altered mentation, hyperreflexia
Pregnancy-induced hypertension (preeclampsia-eclampsia): Lab findings
thrombocytopenia, hyperuricemia
Pregnancy-induced hypertension (preeclampsia-eclampsia): Treatment
Delivery of fetus as soon as viable. Otherwise, bedrest, salt restriction, monitoring and treatment of hypertension
Pregnancy-induced hypertension (preeclampsia-eclampsia): Treatment for eclampsia (a medical emergency)
IV Magnesium sulfate, diazepam
Pregnancy complications: Abruptio placentae
premature separation of the placenta. Painful uterine bleeding (usually 3rd trimester). Fetal death. May be associated with DIC
Pregnancy complications: Placenta accreta
defective decidual layer allows placenta to attach directly to myometrium. Predisposed by prior C-section or inflamation. May have massive hemorrhage after delivery.
Pregnancy complications: Placenta previa
attachment of the placenta to lower uterine segment. May occlude cervical os. Painless bleeding in any trimester.
Pregnancy complications: Ectopic pregnancy
most often in fallopian tubes, predisposed by salpingitis (PID).
Cervical Pathology: Dysplasia and carcinoma in situ
Disordered epithelial growth; begins at basal layer and extends outward. Classified as CIN 1, CIN 2, or CIN 3 (carcinoma in situ), depending on extent of dysplasia. Associated with HPV. May progress to invasive carcinoma.
Cervical Pathology: Invasive carcinoma
Often squamous cell carcinoma. Pap smears can catch cervical dysplasia (koilocytes) before it progresses to invasive carcinoma.
Cervical Pathology: AUTHOR
Gabi Rizzuto
Uterine pathology: most common tumor in female (hint - has increased incidence in blacks)? Do they progress to malignancy?
leiomyoma; no.
Uterine pathology: endometriosis - most common site?
ovary
Uterine pathology: endometriosis in the myometrium = ?
adenomyosis
Uterine pathology: most common gyn malignancy? (hint - causes vaginal bleeding in a postmenopausal women)
endometrial carcinoma
Uterine pathology: Leiomyosarcoma: does it arise de novo or from leiomyoma?
de novo
PCOS (Stein-Leventhal syndrome): Major clinical manifestations include: amenorrhea, infertility, obesity, and ----?
hirsutism
Ovarian cysts: the four types of ovarian cysts
follicular, corpus luteum, theca-lutein, chocolate
Ovarian cysts: blood containing cyst from ovarian endometriosis
chocolate cyst
Ovarian cysts: cyst that is associated with choriocarcinoma and moles
theca-lutein cyst
Ovarian cysts: cyst defined by hemorrhage into persistent corpus luteum
corpus luteum cyst
Ovarian cysts: cyst defined by distention of unruptured graafian follicle
follicular cyst
Ovarian germ cell tumors : that which is analagous to male seminoma
dysgerminoma
Ovarian germ cell tumors : produces alpha-fetoprotein
yolk sac tumor
Ovarian germ cell tumors : associated with elevated hCG
choriocarcinoma
Ovarian germ cell tumors : monodermal teratoma made up of only thyroid tissue
struma ovarii
Ovarian germ cell tumors : most common germ cell tumor of ovary (90%)
teratoma
Ovarian germ cell tumors : True or False: immature teratomas are benign
False. Mature ("dermoid cyst") ones are benign, immature ones are aggressively malignant
Ovarian non-germ cell tumors: bilateral non germ cell tumor with fallopian tube like epithelium
serous cystadenoma
Ovarian non-germ cell tumors: Pseudomyxoma peritonei (intraperitoneal accumulation of mucinous material) may be a consequence of which ovarian non germ cell tumor?
mucinous cystadenocarcinoma
Ovarian non-germ cell tumors: tumor resembling bladder epithelium
brenner tumor
Ovarian non-germ cell tumors: triad of ovarian fibroma, ascites, and hydrothorax = ?
Meigs' syndrome
Ovarian non-germ cell tumors: Pathology term for the small follicles filled with eosinophilic secretions that are seen in granulosa cell tumors
Call-Exner bodies
Breast disease: Which one of the following does NOT put an individual at increased risk for breast disease? Gender, age, early 1st menarche, late 1st pregnancy, late menopause, family history, fibroadenoma/non-hyperplastic cysts
fibroadenoma/hyperplastic cysts
Breast disease: Eczematous patches on the nipple w/ corresponding large cell's with clear halo on histology are indicative of --?
Paget's disease & most likely an underlying ductal carcinoma
Breast disease: Fibrosis, cystic, sclerosing, & epithelial hyperplasia are all histologic subtypes of what breast disease?
Fibrocystic disease (note: no increase risk of developing carcinoma)
Breast disease: Most common breast tumor in women < 25 years of age? is it malignant or a precursor to malignancy?
Fibroadenoma. No, no.
Breast disease: A benign tumor of the breast that presents with nipple discharge
Intraductal papilloma
Breast disease: Are malignant breast tumors more common pre or post menopause
post
Hypertension: True of false: 50% is classified as secondary HTN (usually resulting from renal disease)
false. (90% essential/primary, 10% secondary)
Hypertension: "read the questioner's mind": HTN predisposes individuals to this disease (the one John Ritter died of)
aortic dissection
Hypertension: Pathology changes associated with HTN
hyaline thickening & atherosclerosis
Arteriosclerosis: This awful term refers to a stiffening of the arteries that invovles the media. Particularly likely to occur at the radial & ulnar arteries.
Monckeberg arteriosclerosis
Atherosclerosis: True or false: atherosclerosis is a disease of small sized arteries
false. affects elastic, large & medium muscular arteries.
Atherosclerosis: Earliest sign of atherosclerotic disease
fatty streak
Atherosclerosis: most likely location
abdominal aorta. (then coronary artery, popliteal artery, and carotid artery)
Ischemic heart disease: Type of angina resulting from coronary artery spasm
Prinzmetal's variant
Ischemic heart disease: This coronary artery branch is most commonly implicated in myocardial infarction
LAD (left anterior descending)
Ischemic heart disease: most common cause of sudden cardiac death
(lethal) arrhythmia
Infarcts: red vs. pale: Solid tissues like the heart, brain, kidney and spleen have only a single blood supply (not so good collaterals). Therefore infarcts are more likely to be --?
pale
Infarcts: red vs. pale: 2 instances where red infarct is likely
(1) reperfusion (2) loose tissues with good collaterals - like the lungs or intestine
Evolution of MI: Rank the following vessels from most to least commonly occluded: RCA, LAD, circumflex
LAD>RCA>circumflex
Evolution of MI: Histologic changes on day 1 of an MI?
pallor of infarcted area; coagulative necrosis
Evolution of MI: days 2-4?
dilated vessels (hyperemia); neutrophil invasion; extensive coagulative necrosis
Evolution of MI: days 5-10?
yellow-brown softening of infarcted region; macrophages present; granulation tissue begins to grow in
Evolution of MI: after 7 weeks?
infarct is gray-white; scar complete
Diagnosis of MI: True or false: ECG is not diagnostic during the first 6 hours following an MI
False; it is the gold standard within this time period
Diagnosis of MI: What is the test of choice within the first 24 hours?
CK-MB
Diagnosis of MI: This enzyme is elevated from 4 hours up to 10 days after an MI and is the most specific protein marker
cardiac troponin I
Diagnosis of MI: on ecg, transmural infarction causes ______
ST elevation, Q wave changes
MI complications: Most common (90% of patients)
arryhthmias, esp. 2 days after infarct
MI complications: automimmune phenomen several weeks post-MI that results in fibrinous pericarditis
Dressler's syndrome
MI complications: high risk of mortality
cardiogenic shock (large infarcts)
MI complications: seen about a week after the infarction
rupture of ventricular wall, septum, or papillary muscle
Cardiomyopathies: Most common
dilated (congestive) cardiomyopathy; heart looks like a ballon on X-ray
Cardiomyopathies: True or False: substance abuse is a common cause of dilated cardiomyopathy
True; cocaine and alcohol especially
Cardiomyopathies: These two infectious diseases are associated with dilated myopathy
coxsackievirus B and Chagas' disease
Cardiomyopathies: True or false: hypertrophic cardiomyopathy causes systolic dysfunction
False; dilated myopathy causes systolic dysfunction, hypertrophic causes diastolic
Cardiomyopathies: Half of hypertrophic myopathies are inherited as an _________ trait (x-linked, dominant, etc.)
autosomal dominant; major cause of sudden death in young athletes
Cardiomyopathies: On echo in hypertrophic disease, the LV thickens and the chamber looks how?
like a banana
Cardiomyopathies: These "-osis" diseases are major causes of restrictive/obliterative cardiomyopathy
sarcoidosis, amyloidosis, hemochromatosis, endocardial fibroelastosis, endomyocardial (Loffler's) fibrosis….also, scleroderma but it's not an -osis
Heart murmurs: Name two causes of holosystolic murmurs
1) VSD, 2) mitral regurg, and 3) tricuspid regurg
Heart murmurs: Widened pulse pressure seen with this diastolic murmur
aortic regurg
Heart murmurs: Describe the murmur associated with the most common valvular lesion
Mitral prolapse; late systolic murmur following mid-systolic click
Heart murmurs: True or false: aortic stenosis causes a decrescendo-crescendo murmur following an ejection click
False; ejection click is followed by a crescendo-decrescendo systolic murmur
Heart murmurs: cause of a continuous murmur loudest at time of S2?
patent ductus artieriosis
Heart murmurs: opening snap followed by late diastolic rumbling?
mitral stenosis
Cardiac tumors: most common heart tumor?
metastasis
Cardiac tumors: primary cardiac tumor in 1) adults and 2) children
adults=myxoma (almost always in left atrium); children=rhabdomyoma
CHF: fun gross pathologic term for changes in liver with CHF?
nutmeg
CHF: what are "heart failure cells"?
hemosiderin-laden macrophages in lung
CHF: dyspnea on exertion, pulmonary edema, and paroxysmal nocturnal dyspnea are symptoms of?
left heart failure
CHF: patient says "I have to sleep upright." the clinical term for this is?
orthopnea
embolus types: most pulmonary emboli arise from?
DVT
embolus types: True or false: Amniotic fluid can lead to DIC
TRUE
deep vein thrombosis: what are the component of virchow's triad?
stasis, hypercoagulability, endothelial damage
cardiac tamponade: what is pulsus paradoxus?
greater than 10 mmHg drop in systolic on inspiration
cardiac tamponade: what is electrical alternans?
characteristic of tamponade on ECG in which QRS complex height varies beat-to-beat
cardiac tamponade: AUTHOR
James Rosoff / Flora Waples-Trefil
Bacterial Endocarditis: What valve is usually involved in endocarditis?
Mitral
Bacterial Endocarditis: What valve indicated drug use if it is involved?
Tricuspid
Bacterial Endocarditis: Can endocarditis be non-bacterial?
Yes. It can be secondary to metastasis, renal failure (maranctic or thrombotic), fungal
Bacterial Endocarditis: What type of endocarditis does s. Aureus cause?
rapid onset, high virulence, tends to occur secondary to infection elsewhere
Bacterial Endocarditis: What do the vegetations look like
Large.
Bacterial Endocarditis: What type of endocarditis does s. viridians cause?
subacute. Tends to have smaller vegetations.
Bacterial Endocarditis: What predisposes you to s.viridins endocarditis?
tends to occur on previously damaged valves, so rheumatic fever. It is commonly seen after dental work.
Bacterial Endocarditis: What are the 8 sings of endocarditis?
JR=NO FAME Janeway lesions, Roth's spots, Nail-bed hemorrhages, Osler's nodes, Fever, Anemia, Murmur (new), Emboli
Bacterial Endocarditis: What do Janeway Lesions look like?
multiple small flat erythematous lesions on palms and soles
Bacterial Endocarditis: What are roth spots?
round white spots on the retina surrounded by hemorrhage.
Bacterial Endocarditis: What are osler's nodes?
Tender raised lesions on the fingers and toes.
Bacterial Endocarditis: What is the etiology of these lesions?
Bacterial vegetations fliping off the heart valve and lodging in the periphery.
Rheumatic Fever/Rheumatic Heart Disease: What type of bacteria causes rheumatic fever?
Group A beta-hemolytic strep
Rheumatic Fever/Rheumatic Heart Disease: when does it occur?
Children 5-15 years, four weeks after a bacterial infection
Rheumatic Fever/Rheumatic Heart Disease: Is the bacteria responsible for the symptoms?
No. RF is an autoimmune reaction of a cross-reactive protein that is found in the initial bacterial infection.
Rheumatic Fever/Rheumatic Heart Disease: What are the non cardiac clinical signs of rheumatic fever?
FEVERSS - Fever, Erythema marginatum, valve damage, Elevated ESR, Red-hot joints (migratory polyarthritis), Subcutaneous nodules, and St. Vitus dance (chorea)
Rheumatic Fever/Rheumatic Heart Disease: What hear valves are effected?
Mitral (most frequent), Aortic, Tricuspid (5%) - high pressure valves mainly.
Rheumatic Fever/Rheumatic Heart Disease: What is an Aschoff body?
classic histological sign of RF, found in the myocardium, contained fibrinoid material, fragmented collages, surrounded by giant cells.
Rheumatic Fever/Rheumatic Heart Disease: What are the cardiac signs of RF?
Verrucious vegetations on the valve, pancarditis, possible pericardial effusions and myocarditis (most common cause of death)
Pericarditis: Name four causes of serous pericarditis.
RAIL - Rheumatic Arthritis, Infection, Lupus, and Uremia
Pericarditis: What is serous pericarditis?
straw colored, protein rich exudates - non-purulent, and acute
Pericarditis: What are three causes of fibirnous exudates?
MI, Rheumatic fever, and Uremia
Pericarditis: Cloudy pericardial exudates indicated what?
Bacterial infection
Pericarditis: What are two causes of Hemorrhagic pericarditis?
malignancy and TB
Pericarditis: What is hemorrhagic pericarditis?
Bloody and inflammatory exudates
Pericarditis: What are the clinical signs of pericardial exudates?
pericardial pain, friction rub, decreased heart sounds, ST elevation throughout, and pulses paradoxes (like cardiac tamponade)
Pericarditis: What are the long term sequela of pericarditis?
chronic adhesive or constrictive pericarditis
Pericarditis: What is constiricit pericadritis
Fibrous scarring in the pericardium obliterates the space and constrict the right side of the heart (because it is less able to withstand the pressure)
Pericarditis: What types of pericaditis lead to this?
TB and pyrogenic staph infections
Syphilitic Heart Disease: What part of the heart does syphilis damage?
The vaso vasorum of the aorta
Syphilitic Heart Disease: What does this lead to?
Dilation of the aorta and valve ring
Syphilitic Heart Disease: What clinical results does this have?
it can cause an aortic aneurysm or valvular incompetence
Syphilitic Heart Disease: What parts of the aorta are effected
ascending and arch
Syphilitic Heart Disease: What is the appearance or the aorta?
Called a "tree-bark" apearance.
Buerger's Disease: Buerger's disease is also known as?
smoker's disease, thromboangiitis obliterans
Buerger's Disease: What are the Sx (5)?
intermittent claudication, superficial nodular phlebitis, cold sensitivity, severe pain in affected part, may lead to gangrene
Buerger's Disease: What is the Tx?
Quit smokng
Takayasu's arteritits: Also known as ?
pulseless disease
Takayasu's arteritits: What is it?
thickening of aortic arch and/or proximal great vessels
Takayasu's arteritits: ESR elevated?
yes
Takayasu's arteritits: Primarily affects whom?
Young Asian Females
Takayasu's arteritits: Mnemomic
FAN MY SKIN On Wednesday
Takayasu's arteritits: Sx (7)
Fever, Arthritis, Night Sweats, Myalgia, Ocular Disturbances, Weak pulses in uper extremities (FAN MY SKIN On Wednesday)
Temporal Arteritis: Also known as?
Giant Cell Arteritis
Temporal Arteritis: Affects what vessels?
medium and small arteries (usually carotid branches)
Temporal Arteritis: ESR elevated?
yes
Temporal Arteritis: Primarily affects whom?
Elderly Females
Temporal Arteritis: Sx (3)
Unilateral Headache, Jaw claudication, impaired vision
Temporal Arteritis: Many patients are also affected with this syndrome
Polymalgia Rheumatica
Temporal Arteritis: Tx (?)
Steroids
Polyarteritis Nodosa: How would you describe the inflammation in this disease?
necrotizing, caused by immune complex deposition
Polyarteritis Nodosa: In which vessels?
small to medium-sized muscular arteries, renal and visceral vessels
Polyarteritis Nodosa: ESR elevated?
yes
Polyarteritis Nodosa: Sx (7) … and they are SO specific
fever,weight loss,malaise,abdominal pain,headache ,myalgia,hypertension
Polyarteritis Nodosa: Other findings
cotton-wool spots, microaneurysms, pericarditis, myocarditis, palpable purpura
Polyarteritis Nodosa: What viral infection is found in 30% of PN patients?
Hepatitis B
Polyarteritis Nodosa: what antibody is associated with this disease?
P-ANCA
Polyarteritis Nodosa: What two ways can you remember the assoc Ab?
P-ANCA --> PAN (PolyArteritis Nodosa)
Polyarteritis Nodosa: What does P-ANCA refer to?
Perinuclear pattern of Antineutrophil Cytoplasmic Antibodies
Polyarteritis Nodosa: Primarily affects whom?
Young males
Polyarteritis Nodosa: Tx
Corticosteroids, Azathioprine, Cyclophosphamide
Wegener's Granulomatosis: What are three major findings (hint: they are necrotizing)
Focal Necrotizing Vasculitis, Necrotizing granulomas of the lung and uper airway, Necrotizing glomerulonephritis
Wegener's Granulomatosis: Sx (7)
Perforated nasal septum, Chronic Sinusitis, Otitis Media, Mastoiditis, Cough , Dyspnea, Hemoptysis
Wegener's Granulomatosis: What antibody is associated with this disease
C-ANCA
Wegener's Granulomatosis: What should you look for on CXR?
large nodular densities
Wegener's Granulomatosis: Tx
Corticosteroids, Azathioprine, Methotrexate
Kawasaki Disease: Who gets it?
infants, kids
Kawasaki Disease: What is the disease course?
acute, self-limiting
Kawasaki Disease: What vessels are involved?
small and medium sized
Kawasaki Disease: What does CRASH stand for
Conjunctivitis, Rash (truncal), Aneurysms of Coronary Arteries, Strawberry tongue, changes in lips/oral mucosa, Hands and Feet show induration and desquamation
Kawasaki Disease: Glomerular Pathology
275
Kawasaki Disease: What is prerenal azotemia?
selective rise in BUN during early renal failure
Kawasaki Disease: Define segmental.
involving part of the glomerular tuft
Kawasaki Disease: Nephritic Syndrome
0
Kawasaki Disease: hematuria, hypertension, oliguria, azotemia = ?
Nephritic Syndrome, "I" = inflammation
Kawasaki Disease: LM findings in post-strep glomerulonephritis?
lumpy bumpy and hypercellular w/ neutrophils
Kawasaki Disease: EM findings in post-strep glomerulonephritis?
subepithelial humps (from deposition of cationic antigen)
Kawasaki Disease: IF findings in post-strep glomerulonephritis?
granular
Kawasaki Disease: Goodpasture's is what type of hypersensistivity?
type II (IgG binding directly to BM)
Kawasaki Disease: Goodpasture's IF?
linear
Kawasaki Disease: Symptoms of Goodpasture's?
hemoptysis, hematuria
Kawasaki Disease: Membranoproliferative glomerulonephritis EM?
subendothelial humps; "tram track"
Kawasaki Disease: Course of crescentic glomerulonephritis?
rapid (the Red Crescent is the Islamic Red Cross, an organization that provides relief during emergencies, like crescentic glomerulonephritis)
Kawasaki Disease: Course of Membranoproliferative glomerulonephritis?
slow
Kawasaki Disease: IgA nephropathy IF and EM?
mesangial deposits of IgA (Mes"A"ngium)
Kawasaki Disease: Which can be post infectious, IgA nephropathy or Goodpasture's?
IgA nephropathy
Kawasaki Disease: Nephrotic Syndrome
0
Kawasaki Disease: Signs of nephrotic syndrome?
massive proteinuria, hypoalbuminemia, generalized edema, hyperlipidemia (think "erotic" = an erection(edema) and ejaculate(massive proteinuria)
Kawasaki Disease: LM of membranous glomerulonephritis?
diffuse capillary and basement membrane thickening
Kawasaki Disease: IF of membranous glomerulonephritis?
granular
Kawasaki Disease: EM of membranous glomerulonephritis?
spike and dome ("if you have a spike in your dome, you must be insane in the "membran"e," -Chirag)
Kawasaki Disease: Minimal change disease EM?
foot process effacement
Kawasaki Disease: Most common cause of childhood nephrotic syndrome?
minimal change disease
Kawasaki Disease: Focal segmental glomerular sclerosis LM?
segmental sclerosis and hyalinosis
Kawasaki Disease: Diabetic nephropathy LM?
Kimmelstiel-Wilson lesions
Kawasaki Disease: SLE (5 patterns of renal involvement) LM?
wire-loop apearance w/ extensive granular BM depsits in membranous glomerulonephritis pattern
Kidney Stones: What is the most common kidney stone?
Calcium (with either oxalate of phosphate)
Kidney Stones: What kidney stone is associated with a bacterial infection?
Ammonium magnesium phosphate (struivte) stones are associated with urase positive bugs that make ammonium
Kidney Stones: What are the two most common stone-forming bacteria?
proteus vulgaris and Staph.
Kidney Stones: What stones are associated with gout?
Uric acid stones
Kidney Stones: What else can cause uric acid stones?
Any disease with high cell turnover (remember that uric acid is a by-product of DNA formation), so leukemia and myeloproliferative disease
Kidney Stones: What stones are associated with cystineuria?
cystine stones
Kidney Stones: What two stones are radiolucent?
Uric acid and cystine (the two purely metabolic causes)
Kidney Stones: What are the complications of stones?
Hydronephrosis and pyelonephritis
Kidney Stones: What causes calcium stones?
Anything that increases calcium in the blood, so high PTH, malignancy (bone breakdown, PTH production), vitamin D overdose.
Renal Cell Carcinoma: Is this a common renal cancer?
Yes, the most common
Renal Cell Carcinoma: Who is prone to this?
males, ages 50-70, smokers, gene deletions on chromosome 3
Renal Cell Carcinoma: This is a cancer of what cell type?
renal tubule cells, histologically they apear clear.
Renal Cell Carcinoma: What are the clinical signs of this cancer?
flank pain, fever, hematuria, palpable mass, secondary polycythemia
Renal Cell Carcinoma: How does it spread?
It invades the renal veins and IVC, to spread hematogenously
Renal Cell Carcinoma: What hormones can it produce?
ACTH, prolactin, Parathyroid-like hormone, gonadotropins and renin.
Wilm's Tumor: Who gets this cancer?
Children 2-4 years
Wilm's Tumor: Is it common?
The most common renal cancer in children
Wilm's Tumor: What is the chromosomal abnormality that leads to this?
deletion of tumor supressor gene WT1 on chromosome 11
Wilm's Tumor: What tetrad is it associated with?
WAGR - Wilm's, Anirida (lack of an iris), Genitourinary malformation, and Retardation
Wilm's Tumor: How does it present?
huge palpable mass and hemihypertrophy
Wilm's Tumor: What is the histological appearance?
mixed - with stromal, mesenchymal, tubular, glomerular and fibrous elements
Transitional Cell Carcinoma: Where does this cancer occur?
Urinary spaces (tract, calyces, pelvis, bladder)
Transitional Cell Carcinoma: Does it recur?
yes, often
Transitional Cell Carcinoma: How does it spread?
Local invasion
Transitional Cell Carcinoma: What can predispose you to it?
Pee SAC toxins - Phenacetin, Smoking, Aniline dye (benezenes), and Cyclophsophimide.
Transitional Cell Carcinoma: How does it present?
Hematuria
Acid-Base Physiology: What is the primary disturbance in Metabolic acidosis?
a decrease in bicarbonate
Acid-Base Physiology: What is the compensation?
A drop in CO2 by hyperventilation
Acid-Base Physiology: What are common causes?
diabetic ketoacidosis (production of ketone acids), diarrhea (loss of GI bicarb), salisylate overdose, acetazoleamide (diuretic) OD, lactic acidosis, renal failure (can't excrete organic acids), ethylene glycol ingestion
Acid-Base Physiology: What is the primary disturbance in respiratory acidosis?
A build-up in CO2
Acid-Base Physiology: What is the compensation?
Increased bicarb reabosrobtion from the kidney
Acid-Base Physiology: What are some common causes?
COPD, airway obstruction, opiates and sedatives, guillan-barr or ALS,
Acid-Base Physiology: What is the primary disturbance in metabolic alkalosis?
increased bicarbonate
Acid-Base Physiology: What is the compensation?
Increased CO2 by decreased respiration
Acid-Base Physiology: What are some common causes?
Vomiting, hyperaldosteronism (increased H+ secretion), loop or thiazide diuretics (volume contraction)
Acid-Base Physiology: What is the primary disturbance in respiratory alkalosis?
A drop in CO2
Acid-Base Physiology: What is the compensation?
increased excretion of bicarb by the kidney
Acid-Base Physiology: What are some common causes?
hyperventilation, high altitude, pneumonia and pulmonary embolus (hypoxemia causes hyperventilation_
Acid-Base Physiology: What is the Henderson Haselbach equation?
pH = pKa + Log (HCO3-)/(.03*pCO2)
Acid Base Nomogram: Is there an awesome graph on page 277 that you should know?
Yes
Acid Base Nomogram: basically four cases
0
Acid Base Nomogram: Low pH, low pCO2 (low HCO3-)
metabolic acidosis
Acid Base Nomogram: Low pH, high pCO2 (high HCO3-)
chronic respiratory acidosis
Acid Base Nomogram: High pH, low pC02 (low HC03-)
acute respiratory alkalosis
Acid Base Nomogram: High pH, high pCO2 (high HCO3-)
metabolic alkalosis
Acidosis/Alkalosis: Check arterial pH
0
Acidosis/Alkalosis: pH < 7.4
acidosis
Acidosis/Alkalosis: pH > 7.4
alkalosis
acidosis: P (CO2) > 40
Respiratory acidosis
acidosis: P (CO2) > 40, Hypoventilation or Hyperventilation
HYPO
acidosis: Causes
Acute lung disesae, chronic lung disease, Drugs (opioids, narcotics, sedatives), Weakening of Resp. muscles
acidosis: P (CO2) < 40
0
acidosis: What should you do next?
Check Anion Gap
acidosis: What is the normal anion gap?
8-12 mEq/L
acidosis: Increased Anion Gap - 4 Causes
Renal Failure, lactic acidosis, ketoacidosis, aspirin ingestions
acidosis: Normal anion gap metabolic acidosis (4)
Diarrhea, sniffin glue, renal tubular acidosis, hyperchloremia
acidosis: pH > 7.4
alkalosis
acidosis: pCO2 < 40
Respiratory alkalosis
acidosis: respiratory acidosis Causes (2)
HYPERventilation, aspirin ingestion (early)
acidosis: pCO2 > 40
metabolic alkalosis w/ compensation
acidosis: metabolic alkalosis Causes (4)
vomiting, diuretic use, antacids, hyperaldosteronism
Anion gap acidosis: How do you calculate it
Na - Cl - HCO3
Anion gap acidosis: What is normal?
8-12 mEq/L
Anion gap acidosis, MUDPILES: M
Methanol
Anion gap acidosis, MUDPILES: U
Uremia
Anion gap acidosis, MUDPILES: D
DKA
Anion gap acidosis, MUDPILES: P
Paraldehyde or Phenformin
Anion gap acidosis, MUDPILES: I
Iron tablets or INH
Anion gap acidosis, MUDPILES: L
Lactic Acidosis
Anion gap acidosis, MUDPILES: E
Ethanol, Ethylene Glycol
Anion gap acidosis, MUDPILES: S
Salicylates
Acid Base Compensations: Metabolic acidosis
pCO2 = 1.5(HCO3) + 8 +/- 2
Acid Base Compensations: Metabolic alkalosis
pCO2 increases 0.7 mm Hg per 1 mEq/L HCO3 increase
Acid Base Compensations: Respiratory acidosis (acute)
HCO3 increases by 1 mEq/L for every 10 mmHg increase of pCO2
Acid Base Compensations: Respiratory acidosis (chronic)
HCO3 increases by 3.5 mEq/L for every 10 mmHg increase of pCO2
Acid Base Compensations: Respiratory alkalosis (acute)
HCO3 decreases by 2 mEq/L for every 10 mmHg decrease of pCO2
Acid Base Compensations: Respiratory alkalosis (chronic)
HCO3 decreases by 5 mEq/L for every 10 mmHg decrease of pCO2
Acute Pyelonephritis: Affects _______ of kidney
cortex
Acute Pyelonephritis: Spares ____________
glomeruli/vessels
Acute Pyelonephritis: _______ _______ in urine are pathognomonic
WBC casts
Chronic Pyelonephritis: Tubules may contain __________ casts
Eosinophilic
Chronic Pyelonephritis: Coarse ________, _________ scarring
Asymmetric corticomedullary
Diffuse Cortical Necrosis: infarction of _________ of both kidneys
cortex (cortices)
Diffuse Cortical Necrosis: due to combination of _________ and _________
vasospasm and DIC
Diffuse Cortical Necrosis: associated with _________ and _________
obstetric catastrophes and septic shock
Acute Tubular Necrosis: Most common cause of
renal failure
Acute Tubular Necrosis: reversible or irreversible
reversible
Acute Tubular Necrosis: fatal or non-fatal
fatal, if left untreated
Acute Tubular Necrosis: associated with (3)
renal ischemia (shock), crush injury (myoglobulinuria), toxins
Acute Tubular Necrosis: Death occurs most often during initial oliguric phase; T or F
TRUE
Renal Papillary Necrosis: Associated with (3)
Diabetes mellitus, Acute pyelonephritis, Chronic phenacetin use
Acute Renal Failure: Definition
abrupt decline in renal function over a period of days
Acute Renal Failure: Will see increase in _______ & _______
BUN & Creatinine
Acute Renal Failure: Prerenal is due to decreased ______
RBF
Acute Renal Failure: Renal is due to
ATN, Ischemia, toxins
Acute Renal Failure: Post renal is due to
outflow obstruction
Acute Renal Failure: 3 causes of above
renal stones, BPH, neoplasia
Acute Renal Failure: bilateral or unilateral
bilateral
Pre or post renal failure: Urine osmolality > 500
prerenal
Pre or post renal failure: Urine osmolality < 350
renal or postrenal
Pre or post renal failure: urine Na < 10
prerenal
Pre or post renal failure: urine Na > 40
postrenal
Pre or post renal failure: urine Na > 20
renal
Pre or post renal failure: FE(Na) >4%
postrenal
Pre or post renal failure: FE(Na) <1%
prerenal
Pre or post renal failure: FE(Na) >2%
renal
Pre or post renal failure: BUN/Cr ratio <15
renal
Pre or post renal failure: BUN/Cr ratio >15
postrenal
Pre or post renal failure: BUN/Cr ratio > 20
prerenal
Pre or post renal failure: Chronic renal failure due to (2)
diabetes, hypertension
Pre or post renal failure: Consequences of Renal Failure (8) p.279
Why?
Pre or post renal failure: Anemia
lack of EPO
Pre or post renal failure: Renal osteodystrophy
lack of active Vit D
Pre or post renal failure: Arrythmia
hyperkalemia
Pre or post renal failure: Metabolic acidosis
decreased acid secretion
Pre or post renal failure: Uremia
increase in BUN and Cr
Pre or post renal failure: CHF/Pulmonary Edema
retention of Na and H20
Pre or post renal failure: Chronic Pyelonephritis
0
Pre or post renal failure: Hypertension
0
Pre or post renal failure: AUTHOR
Aaron Goldberg
Alcoholism: tremor, tachcardia, hypertension, malaise, nausea, delirium tremens are symtpoms of what?
alcohol withdrawal
Alcoholism: when do you get symptoms of alcohol withdrawal?
in case of physiological tolerance and dependence when intake is interrupted
Alcoholism: what is disulfiram and how does it work?
disulfiram is a pharmacological treatment of alcoholism - negatively conditions patient against EtOH
Alcoholism: a good possible referral to sustain EtOH abstinence in alcoholics
Alcoholics Anonymous and other peer groups
Alcoholism: 3 mechanisms of EtOH action
1) interpolates into membranes --> toxic effects, partic. in brain 2) alcohol dehydrogenase converts EtOH to acetaldehyde, forms adducts with proteins and nucleic acids, converted to acetate, Ac-CoA, FA synthesis, fatty liver 3) increased NADH/NAD ratio
Complications of alcoholism: Condition in which these are seen: hepatitis and cirrhosis, pancreatitis, dilated cardiomyopathy, peripheral neuropathy, cerebellar degeneration, Wernicke-Korsakoff syndrome, testicular atrophy and hyperestrinism, and Mallory-Weiss syndrome
alcoholism
Complications of alcoholism: name histological type of cirrhosis in alcoholism
micronodular cirrhosis
Complications of alcoholism: accompanying symptoms of alcoholic cirrhosis
jaundice, hypoalbuminemia, coagulation factor deficiences, portal hypertension
Complications of alcoholism: list clinical findings in alcoholic cirrhosis besides jaundice
peripheral edema and ascites, encephalopathy, neurologic manifestations (asterixis, flaping tremor of hands)
Complications of alcoholism: What is the cause of Wernicke-Korsakoff syndrome?
thiamine (B1) deficiency, particularly in alcoholics
Complications of alcoholism: what is the presenting triad of Wernicke's encephalopathy?
psychosis, ophthalmoplegia, and ataxia
Complications of alcoholism: distinguishing features of Korsakoff's from Wernicke's
in Korsakoff, also *memory loss*, confabulation,confusion.
Complications of alcoholism: is Korsakoff's syndrome reversible?
No.
Complications of alcoholism: Tx for Wernicke-Korsakoff syndrome
IV Vitamin B1 (thiamine)
Complications of alcoholism: What is Mallory-Weiss syndrome?
longitudinal lacerations at the gastroesophageal junction caused by excessive vomiting (for ex., in alcoholism) with failure of LES relaxation that could lead to fatal hematemesis
Argyll Robertson pupil: name for a pupil that constricts with accomodation but is not reactive to light, and what is it pathognomonic for?
Argyll Robertson pupil. Pathognomonic for 3' syphilis. "Prostitute's pupil" accomodates but does not react.
Amyloidosis: detect what using apple green biorefringence on Conco red stain?
amyloid
Amyloidosis: most common cause of amyloidosis?
primary light chain deposition seen with multiple myeloma.
Amyloidosis: another cause of primary amyloidosis?
Waldenstrom's macroglobulinemia.
Amyloidosis: Secondary amyloidosis can cause what in the kidney?
nephrotic syndrome
Amyloidosis: disease associated with beta amyloid deposition in cerebral cortex
Alzheimer's
Amyloidosis: islet cell amyloid deposition characteristic of what
diabetes mellitus type 2
Aschoff body: "-------- body" found in rheumatic heart disease
Aschoff body (granuloma with giant cells)
Aschoff body: "------- cells" found in rheumatic heart disease
Anitschkow's cells (activated histiocytes) "Aschoff and Anitschow" -- two RHussians with RHeumatic heart disease
Auer bodies (rods): peroxidase-positive cytoplasmic inclusions in granulocytes and myeloblasts, name and primary association
Auer rods; primarily seen in acute promyelocytic leukemia (M3)
Auer bodies (rods): treatment of AML M3 can release what, leading to what?
release Auer rods, leading to DIC
Casts: RBC casts indicate
glomerular inflammation (nephritic syndromes), ischemia, or malignant hypertension
Casts: how to determine if hematuria/pyuria is of renal origin
presence of casts
Casts: WBC casts indicate
inflammation in renal interstitium, tubules, and glomeruli
Casts: casts often seen in normal urine
hyaline casts
Casts: casts seen in chronic renal failure
waxy casts
Erythrocyte sedimentation rate: ESR elevated in
dramatically in infection, malignancy, connective tissue disease; also in pregnancy, inflammatory disease, and anemia
Erythrocyte sedimentation rate: ESR lowered in
sickle cell anemia, CHF, and polycythemia
Erythrocyte sedimentation rate: Should ESR be used for asymptomatic screening
No
Erythrocyte sedimentation rate: ESR used to monitor course of -----
temporal arteritis and polymyalgia rheumatica
Erythrocyte sedimentation rate: ESR is expensive and specific. T/F
false. Cheap and nonspecific.
Ghon complex: indicates TB granulomas with lobar or perihilar lymph node involvement
Ghon focus and lymph node involvement, Ghon complex.
Ghon complex: Does a Ghon complex reflect primary or secondary TB?
primary.
Hyperlipidemia signs: plaque in blood vessel wall
atheromata
Hyperlipidemia signs: plaque or nodule composed of lipid-laden histiocytes in skin, especially eyelids
xanthoma
Hyperlipidemia signs: lipid deposit in tendon, especially Achilles
tendinous xanthoma
Hyperlipidemia signs: lipid deposit in cornea, nonspecific (arcus senilis)
corneal arcus
Psammoma bodies: laminated, concentric, calcific spherules in tumor
Psammoma bodies
Psammoma bodies: Psammoma bodies seen in:
1. papillary adenocarcinoma of thyroid 2. serous papillary cystadenoma of ovary 3. meningioma 4. malignant mesothelioma "PSaMMoma" Papillary (thyroid), Serous (ovary), Meningioma, Mesothelioma
RBC forms: Biconcave RBC
normal
RBC forms: Spherocytes
hereditary spherocytosis, autoimmmune hemolysis
RBC forms: Elliptocyte
hereditary elliptocytosis
RBC forms: Macro-ovalocyte
megaloblastic anemia, marrow failure
RBC forms: Helmet cell, schistocyte
DIC, traumatic hemolysis
RBC forms: Sickle cell
obviously sickle cell anemia
RBC forms: Teardrop cell
myeloid metaplasia with myelofibroblasts
RBC forms: Acanthocyte
spiny apearance in abetalipoproteinemia
RBC forms: Target cell
Thalassemia, liver disease, HbC
RBC forms: Poikilocyte
Nonuniform shapes in TTP/HUS, microvascular damage, DIC
RBC forms: Burr cell
TTP/HUS
HLA subtypes!: HLA B27
Psoriasis, Ankylosing spondylitis, Inflammatory bowel disease, Reiter's syndrome ("PAIR")
HLA subtypes!: HLA 13,17
Psoriasis
HLA subtypes!: HLA DR2
Multiple sclerosis, hay fever
HLA subtypes!: HLA DR3, DR4
Diabetes mellitus type I
HLA subtypes!: HLA DR5
Pernicious anemia -- B12 deficiency
HLA subtypes!: HLA DR7
Steroid-responsive nephrotic syndrome
Reed-Sternberg cells: Distinctive giant cell in blood; binucleate or bilobed with 2 halves as mirror images ("owl's eyes"); name and disease association
Reed-Sternberg cells, necessary but not sufficient for dx of Hodgkin's disease. Variants include lacunar cells in nodular sclerosis variant.
Reed-Sternberg cells: How many types of Hodgkin's disease?
4 - see also p. 241
Reed-Sternberg cells: Only variant of Hodgkin's seen more in women than men
nodular sclerosis
Reed-Sternberg cells: prognosis of nodular sclerosis variant of Hodgkin's
excellent
Virchow's (sentinel) node: firm supraclavicular lymph node, often on left side, easily palpable (ie, by medical students), also known as "jugular gland" - name and association
Virchow's node - presumptive evidence of malignant visceral neoplasm (classically stomach)
Peripheral blood smears: most common cause of microcytic hypochromic anemia
iron deficiency
Peripheral blood smears: ferritin levels in microcytic hypochromic anemia
low
Peripheral blood smears: serum iron-binding capacity in microcytic hypochromic anemia
elevated
Peripheral blood smears: lead poisoning gives what appearance on peripheral smear
microcytic hypochromic
Peripheral blood smears: causes of megaloblastic anemia
folate or B12 deficiency
Peripheral blood smears: 5-7 lobes (hypersegmented) PMNs in
megaloblastic anemia
Peripheral blood smears: large RBCs (MCV >100) in
megaloblastic anemia
Peripheral blood smears: do you give folate to pt. deficient in B12?
Never
Peripheral blood smears: autoimmune disease that causes B12 deficiency; what does it deplete?
pernicious anemia, depletes intrinsic factor
Peripheral blood smears: where is B12 absorbed in gut?
terminal ileum, with intrinsic factor from stomach
Peripheral blood smears: Target cells mnemonic
HALT (Hemoglobin C disease, Asplenia, Liver disease, Thalassemia)
Peripheral blood smears: Hemoglobin SS in what disease?
sickle cell anemia
Peripheral blood smears: molecular change B-globin GLU->VAL at #6
HbS
Peripheral blood smears: 8% of US african-americans carry what Hb
HbS
Peripheral blood smears: conditions in which HbSS cells sickle
hypoxia, dehydration, increased blood viscosity
Peripheral blood smears: vaso-occlusive crises with chest pain in young african-americanl; consider
sickle cell anemia
Peripheral blood smears: aplastic crises in sickle cell anemia associated with which virus
B19 virus
Peripheral blood smears: manifestation of sickle cell anemia in spleen
splenic sequestration crisis
Peripheral blood smears: increased risk of CVA in sickle cell anemia?
yes
Enzyme markers: AST or ALT used for MI?
AST only
Enzyme markers: AST or ALT more elevated in viral hepatitis
ALT>AST in viral hepatitis ("virALT")
Enzyme markers: AST or ALT more elevated in alcholic hepatitis
AST>ALT in alcoholic hepatitis ("ToAST")
Enzyme markers: Amylase elevation
acute pancreatitis, mumps
Enzyme markers: Low ceruloplasmin
Wilson's disease
Enzyme markers: Creatine phosphokinase elevated in
muscle disorders (Duchenne's muscular dystrophy), and MI
Enzyme markers: subtype of CPK elevated in MI
CPK-MB
Enzyme markers: GGT stands for and elevated in
gamma glutamyl transferase, various liver diseases
Enzyme markers: Lipase elevated in
acute pancreatitis
Enzyme markers: Alkaline phosphatase elevated in
Bone disease (Paget's disease of bone), obstructive liver disease (hepatocellular carcinoma)
Immunohistochemical stains: Vimentin stains what
connective tissue
Immunohistochemical stains: Desmin stains what
muscle
Immunohistochemical stains: Cytokeratin stains what
epithelial cells
Immunohistochemical stains: Glial fibrillary acid proteins (GFAP) stains what
neuroglia
Immunohistochemical stains: Neurofilament stains what
neurons
Immunohistochemical stains: AUTHOR
Shailen Sehgal
Enzyme Kinetics: _______ is elevated in a myocardial infarction. (AST, ALT, or both)
AST
Enzyme Kinetics: In viral hepatitis, is ALT or AST more elevated?
ALT
Enzyme Kinetics: In alcoholic hepatitis, is ALT or AST more elevated?
AST
Enzyme Kinetics: ________ can be elevated in mumps or acute pancreatitis, while _________ elevation is more specific for acute pancreatitis.
amylase, lipase
Enzyme Kinetics: What enzyme is elevated in Duchenne's muscular dystrophy, and after a myocardial infarction?
CPK
Enzyme Kinetics: _______ can be elevated in Paget's disease and hepatocellular carcinoma.
alkaline phosphatase
Enzyme Kinetics: Elevated GGT is seen in various diseases of which organ?
liver
Enzyme Kinetics: True or False? In a myocardial infarction, serum LDH-1>LDH-2
TRUE
Enzyme Kinetics: Decreased serum ceruloplasmin is seen in _______
Wilson's Disease
Immunohistochemical Stain Corresponding cell type: (Match the stain with the corresponding cell type)
0
Immunohistochemical Stain Corresponding cell type: Vimentin
connective tissue
Immunohistochemical Stain Corresponding cell type: Cytokeratin
Epithelial cells
Immunohistochemical Stain Corresponding cell type: Neurofilaments
neurons
Immunohistochemical Stain Corresponding cell type: Glial Fibrillary acid proteins (GFAP)
neuroglia
Immunohistochemical Stain Corresponding cell type: Desmin
muscle
Pharmacokinetics: amount of drug in body/_______ = Vd
plasma drug concentration (note: Vd is Volume of Distribution)
Pharmacokinetics: rate of elimination of drug/[plasma drug] = ?
CL (Clearance)
Pharmacokinetics: (.7)(Vd)/CL = ?
T 1/2
Pharmacokinetics: A drug infused at a constant rate reaches about 94% of steady state after _______ t 1/2s.
4
Dosage Calculations: A loading dose is calculated using this formula.
(Cp)(Vd)/F (note: Cp = target plasma concentration, and F = bioavailability)
Dosage Calculations: A maintenance dose is calculated using this formula.
(Cp)(CL)/F
Elimination of Drugs: Rate of elimination is proportional to _______ ______ in 1st order elimination.
drug concentration
Elimination of Drugs: In the case of EtOH, which is elimated by _____ order elimination, a constant amount of drug is eliminated per unit time.
zero
Phase I vs. Phase II metabolism: Phase ____ (I or II) reactions yield slightly polar metabolites that are often _____ (active or inactive)
I, active
Phase I vs. Phase II metabolism: Phase ____ (I or II) reactions yield very polar metabolites that are often _____ (active or inactive) and are excreted by the _______.
II, inactive, kidney
Phase I vs. Phase II metabolism: Phase II reactions are often of this type.
conjugation
Phase I vs. Phase II metabolism: Cytochrome P-450 is involved in _____ phase (I or II) reactions.
I
Drug Development: A patent lasts for _____ years after filing for application.
20
Drug Development: How many phases are there in drug development?
4
Drug Development: Drugs are first tested in patients in phase _____ of clinical testing, pharmacokinetic safety is determined in phase ______ of clinical testing, double blind tests are done in phase ____ and post-market surveillance is done in phase _____.
2,1,3,4
Pharmacodynamics: In a dose response curve, a competitive antagonist shifts the curve _____, while a non-competitive antagonist shifts the curve ______.
right, down
Pharmacodynamics: AUTHOR
HiralShah
Pharmacodynamics (continued): What pharmacologic relationship would determine the existence of spare receptors?
EC50 < Kd
Pharmacodynamics (continued): What does it mean if EC50 and Kd are equal?
The system does not have spare receptors
Pharmacodynamics (continued): A partial agonist acts on the same receptor system as a full agonist? T/F
TRUE
Pharmacodynamics (continued): What's the main difference between a partial agonist and a full agonist?
A partial agonist has a lower maximal efficacy.
Pharmacodynamics (continued): Is a partial agonist less potent than a full agonist?
Not necessarily. It can be less, more or equally potent as a full agonist.
Antimicrobial Tx -- Mechanism of Action: The penicillin type drugs work by blocking ------ synthesis, specifically by inhibiting this molecule from cross-linking?
blocks bacterial cell wall synthesis by inhibition of peptidoglycan synthesis.
Antimicrobial Tx -- Mechanism of Action: Which other drugs (aside from penicillin) have this same mechanism of action?
Imipenem, aztreonam and cephalosporins
Antimicrobial Tx -- Mechanism of Action: Bacitracin, vancomycin and cycloserine block the synthesis of this molecule, preventing cell wall synthesis
peptidoglycans
Antimicrobial Tx -- Mechanism of Action: These drugs block the 50s ribosomal subunit
clindamycin, chloramphenicol, erythromycin, lincomycin, linezolid, streptogramins "Buy AT 30, CELL at 50"
Antimicrobial Tx -- Mechanism of Action: These drugs block the 30s ribosomal subunit
Aminoglycosides and tetracyclines "Buy AT 30, CELL at 50"
Antimicrobial Tx -- Mechanism of Action: These drugs block nucleotide synthesis by interfering with the folate pathway
Sulfonamides (e.g. Bactrim), trimethoprim
Antimicrobial Tx -- Mechanism of Action: These drugs block DNA topoisomerases
Quinolones (e.g. Cipro)
Antimicrobial Tx -- Mechanism of Action: Which drug blocks mRNA synthesis
rifampin
Antimicrobial Tx -- Mechanism of Action: Which are the bacteriacidal Abx
Penicillin, cephalosporin, vancomycin, aminoglycosides, fluoroquinolones, metronidazole
Antimicrobial Tx -- Mechanism of Action: These drugs disrupt the bacterial/fungal cell membranes
polymyxins
Antimicrobial Tx -- Mechanism of Action: These specific disrupt fungal cell membranes
amphotericin B, nystatin, fluconazole/azoles (FAN the fungal cell membranes)
Antimicrobial Tx -- Mechanism of Action: What is the mechanism of action of Pentamidine
Unknown
Penicillin: Which is the IV form and which is the oral form
G = IV, V=oral
Penicillin: Which of these is not a mechanism of penicillin action: (1) binds penicillin-binding protein, (2) blocks peptidoglycan synthesis, (3) blocks transpeptidase catalyzed cross-linking of cell wall and (4) activates autolytic enzymes
Penicillin does not block peptioglycan synthesis, bacitracin, vancomycin and cycloserine do that
Penicillin: T or F: penicillin is effective against gram pos and gram neg rods
False: penicillin is used to treat common streptococci (but not staph), meningococci, gram pos bacilli and spirochetes (i.e. syphilis, treponema). Not used to treat gram neg rods.
Penicillin: What should you watch out for when giving penicillin?
Hypersensitivity rxn (urticaria,severe pruritus) and hemolytic anemia
Methicillin, nafcillin, dicloxacillin: These drugs are used mainly for what type of infection
Staphlococcal infection (hence very narrow spectrum)
Methicillin, nafcillin, dicloxacillin: T or F: these drugs have the same mechanism of action as penicillin
TRUE
Methicillin, nafcillin, dicloxacillin: Are these drugs penicillinase resistant? If so why?
Bulkier R group makes these drugs resistant to penicillinase
Methicillin, nafcillin, dicloxacillin: What should you watch out for when giving these drugs?
Hypersensitivity rxn (urticaria,severe pruritus); methicillin can cuase interstitial nephritis
Ampicillin and amoxicillin: T or F: these drugs have the same mechanism of action as penicillin
TRUE
Ampicillin and amoxicillin: Which has greater oral bioavailability?
amOxicillin (O for Oral)
Ampicillin and amoxicillin: What do you use these for?
Ampicillin/amoxicillin HELPS to kill enterococci (H. influenzae, E. coli, Listeria monocytogenes, Proteus mirabilis, Salmonella)
Ampicillin and amoxicillin: Can penicillinase effect these drugs efficacy?
Yes, they are penicillinase sensitive
Ampicillin and amoxicillin: Why not give these drugs with a penicillinase inhibitor. Name one.
clavulanic acid
Ampicillin and amoxicillin: What should you watch out for when giving these drugs?
Hypersensitivity rxn (ampicillin rash), pseudomembranous colitis
Carbenicillin, piperacillin, ticarcillin: Why are these considered to have an extended spectrum?
Because they are effective against pseudomonas and other gram neg rods (enterobacter and some species of klebsiella)
Carbenicillin, piperacillin, ticarcillin: What should you watch out for when giving these drugs?
Hypersensitivity rxn
Carbenicillin, piperacillin, ticarcillin: Why does concomitant administration with clavulanic acid increase the efficacy of these drugs?
Because they are penicillinase sensitive. (only piperacillin and ticarcillin)
Cephalosporins: What is the mechanism of action of Cephalosporins?
inhibit cell wall synthesis
Cephalosporins: How are they similar/different from penicillin?
both have a beta-lactam ring structure but cephalosporins are less susceptible to penicillinases
Cephalosporins: What are the main similarities/difference between 1st and 2nd generation cephalosporins?
2nd gen has extensive gram neg coverage but weaker gram pos coverage
Cephalosporins: 1st gen covers what bugs?
gram positives (staph and strep), Proteus mirabilis, E. coli, Klebsiella (PEcK)
Cephalosporins: 2nd gen covers what bugs?
gram positives (staph and strep) though less so, H. influenzae, Enterobacter aerogenes, Neisseria, Proteus mirabilis, E. coli, Klebsiella (HEN PEcK)
Cephalosporins: What can 3rd generation drugs do that 1st and 2nd generation can't?
Cross the blood brain barrier
Cephalosporins: What are some other benefits of 3rd gen?
better activity against gram neg bugs resistant to beta-lactam drugs. Ceftazidime for Pseudomonas and ceftriaxone for N. gonorrhea
Cephalosporins: What are the benefits of 4th gen (e.g. Cefipime)?
increased activity against Pseudomonas, gram pos organisms and more beta-lactamase resistant (i.e. 4th gen combines 1st gen and 3rd gen characteristics into super drug)
Cephalosporins: What drugs should you avoid taking with cephalosporins?
Aminoglycosides (increases nephrotoxicity) and ethanol (causes a disulfiram-like rxn -- headache, nausea, flushing, hypotension)
Aztreonam: When would you use aztreonam?
Only to treat Klebsiella, Pseudomonas and Serratia sp.
Aztreonam: Is it beta-lactamase resistant?
Yes, this is one of the huge benefits of the drug, and it is not cross-reactive with PCN!
Aztreonam: Which population of pt. is this drug good for?
The PCN-allergic patient that can't take aminoglycosides b/c of renal insufficiency
Aztreonam: Are there any toxicity issues with this drug?
Not really. Generally well tolerated with occasional GI upset. Vertigo, Headache and rare hepatotoxicity have been reported.
Imipenem/cilastatin: What is imipenem?
broad spectrum beta-lactamase-resistant abx
Imipenem/cilastatin: What do you always administer it with and why?
cilastatin -- it decreases inactivation of imipenem in renal tubules
Imipenem/cilastatin: What do you use it for?
Gram pos cocci, gram neg rods and anaerobes (broad spectrum)
Imipenem/cilastatin: What bug is it the drug of choice for?
Enterobacter
Imipenem/cilastatin: What are its side-effects
GI distress, skin rash, seizures at high conc.
Vancomycin: Is it bactericidal or bacteriastatic and why?
Bactericidal because it blocks cross linkage and elongation of peptidoglycan by binding D-ala D-ala protion of cell wall.
Vancomycin: How does resistance to Vanco occur?
D-ala D-ala is replaced with D-ala D-lactate which vanco does not block
Vancomycin: What is it used for?
Used for serious infection that is resistant to other drugs (e.g. gram pos multi-drug resistant organisms like S. aureus and C. difficile, methicillin resistant staph (MRSA))
Vancomycin: What are the important toxicities of vanco?
generally NOT many problems except, Nephrotoxicity, Ototoxicity and Thrombophlebitis
Vancomycin: What can happen with rapid infusion of vanco?
Red man's syndrome. Diffuse flushing which can be controlled by pretreatment with anti-histamines and with slow infusion rate
Protein Synthesis Inhibitors: Which drugs target bacterial protein synthesis by blocking the 30S unit vs 50S unit?
Buy AT 30, CELL at 50
Protein Synthesis Inhibitors: What does AT stand for?
A = Aminoglycosides (streptomycin, gentamicin, tobramycin an damikacin. And T = Tetracyclines
Protein Synthesis Inhibitors: What does CELL stand for?
C = Chloramphenicol, E= Erythromycin, L= Lincomycin and L= cLindamycin
Protein Synthesis Inhibitors: Which of the above are bactericidal?
Only the aminoglycosides are, the rest are bacteriostatic
Aminoglycosides: Name some aminoglycosides?
Gentamicin, neomycin, amikacin, tobramycin and streptomycin
Aminoglycosides: How do these drugs work?
They inhibit formation of the initiation complex in mRNA translation
Aminoglycosides: Why are they ineffective against anaerobes?
They require oxygen for uptake into bacteria
Aminoglycosides: When would you use aminoglycosides?
against severe gram-negative rod infections
Aminoglycosides: What drugs can you use aminoglycosides with for synergy?
the drugs that inhibit cell wall synthesis (e.g. penicillin and cephalosporins -- the beta-lactam antibiotics). Presumably this allows the drug to get in with out reliance on oxygen transport
Aminoglycosides: What drug in this class is commonly used for bowel surgery?
Neomycin
Aminoglycosides: What are the two major toxicities?
Nephrotoxicity (esp. when used with cephalosporins) and Ototoxicity (esp. when used with loop diuretics). amiNOglycosides
Tetracyclines: Name some tetracylcines
Tetracycline, doxycycline, demeclocycline, minocycline
Tetracyclines: How does it work?
Blocks t-RNA attachment to 30S subunit
Tetracyclines: Which tetracycline can you use in patients with renal failure and why?
Can use doxycycline because its elimination is fecal
Tetracyclines: Should you take these drugs with a glass of milk?
NO, because it intereferes with absorption in the gut as does antacids and iron-containing preparations
Tetracyclines: What are tetracyclines used for?
VACUUM your Bed Room -- Vibrio cholerae, Acne, Chlamydia, Ureaplasma, Urealyticum, Mycoplasma pneumoniae, Borrelia burgdorferi, Rickettsia, tularemia
Tetracyclines: What are the common toxicities
GI distress, teeth discoloration, inhibition of bone growth in children, Fanconi's syndrome and photosensitivity
Macrolides: Name some macrolides?
Erythromycin, azithromycin, clarithromycin
Macrolides: How do these drugs work?
inhibit protein synthesis
Macrolides: What are they used for?
URIs, pneumonias, STDs -- gram pos cocci in patients that are allergic to PNC --- Mycoplasm, Legionella, Chlamydia, Neisseria.
Macrolides: Pneumonic for macrolide use?
Eryc's Niple is at his Mid Clavicular Line (Eryc is brand name for erythromycin). Mycoplasm, Legionella, Chlamydia, Neisseria.
Macrolides: What are the major toxicities?
GI discomfort, acute cholestatic hepatitis, eosinophilia, skin rashes
Macrolides: What is the most common cause for non-compliance to macrolides?
GI discomfort
Chloramphenicol: How does this drug work?
inhibits 50S peptidyltransferase
Chloramphenicol: Main use?
Meningitis (H. influenzae, N. meningitides, S. pneumo). Used conservatively b/c of toxicity
Chloramphenicol: What are the main toxicities?
Anemia and aplastic anemia (both dose dependent), gray baby syndrome (in premes b/c they lack UDP-glucoronyl transferase)
Clindamycin: How does it work?
blocks peptide bond formation at 50S
Clindamycin: When do you use it?
Anaerobic infections (e.g. Bacteroides fragilis and C.perfringens)
Clindamycin: Toxicities?
Pseudomembranous colitis, fever, diarrhea
Sulfonamides: Name some sulfonamides
Sulfamethoxazole (SMX), sulfisoxazole, triple sulfa and sulfadiazine
Sulfonamides: How does it work?
Inhibits bacterial folic acid synthesis from PABA by blocking dihydropteroate synthase.
Sulfonamides: What are its uses?
Gram-positive, gram-negative, Nocardia, Chlamydia. Triple sulfas and SMX for simple UTIs
Sulfonamides: Toxicities?
hypersensitivity rxn, hemolysis if G6PD deficient, nephorotoxicity (tubulointerstitial nephritis), kernicterus in infants, displace other drugs from albumin (e.g. warfarin)
Trimethoprim: How does it work?
inhibits folic acid pathway by blocking dihydrofolate reductase which humans have as well
Trimethoprim: What are its uses?
used in combo with Sulfamethoxazole (TMP-SMX) causing a sequential block of folate synthesis. Used for recurrent UTIs, Shigella, Salmonella, and prophylaxis for PCP in AIDS patients
Trimethoprim: Toxicities?
Megaloblastic anemia, pancytopenia (may be alleviated with suplemental folinic acid)
Fluoroquinolones: What the most famous floroquinolone?
Ciprfloxacin (treatment for Anthrax)
Fluoroquinolones: How does it work?
inhibits DNA gyrase (topoisomerase II)
Fluoroquinolones: What are its uses?
Gram neg rods or urinary and GI tract (incl. pseudomonas), Neisseria, some gram pos sp
Fluoroquinolones: What population is contraindicated for use?
pregnancy and children
Fluoroquinolones: What are its toxicities?
GI upset, superinfection, skin rashes, headache, dizziness and tendonitis and tendon rupture in adults. FluoroquinoLONES hurt attachment to BONES.
Metronidazole: How does it work?
forms toxic metabolites in the bacteria. Bactericidal.
Metronidazole: What are its uses?
anti-protozoal: Giardia, Entamoeba, Trichomonas, Gardnerella vaginalis, anaerobes (bacteroides, clostridium)
Metronidazole: What is the role of Metronidazole in H. pylori infection?
Used as part of triple therapy: bismuth, amoxicillin and metronidazole
Metronidazole: Main toxicity?
disulfiram-like (antabuse) reaction to alcohol and headache
Metronidazole: Which drug do you use to treat anaerobic infections above the diaphram and below the diaphram
anaerobes above diaphram: Clindamycin, and anaerobes below diaphram: metronidazole
Polymyxins: How does it work?
disrupts osmotic properties of bacteria, acts like a detergent
Polymyxins: What is it used for?
resistant gram negative infections
Polymyxins: Toxicities?
neurotoxicity, ATN
Isoniazid: How does it work?
decreases synthesis of mycolic acid
Isoniazid: What is it used for?
MTB (mycobacterium tuberculosis). The only agent used as solo prophylaxis against TB
Isoniazid: Toxicities?
Hemolysis if G6PD deficient, neurotoxicity, hepatotoxicitiy, drug induced SLE. INH, Injures Neurons and Hepatocytes
Isoniazid: What vitamin prevents neurotoxicity
Vitamin B6 (pyridoxine)
Isoniazid: Why are toxicities particularly important to monitor in patients taking INH?
INH half-lives are different in fast versus slow acetylators!
Rifampin: How does it work?
inhibits DNA-dependent RNA polymerase
Rifampin: What is it used for?
MTB, meningococcal prophylaxis
Rifampin: Toxicities?
Minor hepatotoxicity and increases P-450
Rifampin: How can it be used for leprosy?
rifampin delays resistance to dapsone when used for leprosy
Rifampin: What would happen if you used rifampin alone?
get rapid resistance
Rifampin: What does it do to bodily fluids?
makes them red/orange in color
Rifampin: What are the 4 R's of Rifampin
RNA polymerase inhibitor, Revs up microsomal p-450, Red/Orange body fluids, Resistance is rapid
Anti-TB Drugs: What are the anti-TB drugs?
Rifampin, Ethambutol, Streptomycin, Pyrazinamide, Isoniazid (INH) -- RESPIre
Anti-TB Drugs: What do you use for TB prophylaxis?
INH
Anti-TB Drugs: What toxicity is common to all?
hepatotoxicity
Anti-TB Drugs: AUTHOR
Michael Shino
Resistance mechanisms for various antibiotics: Most common resistance mechanism for penicillins / cephalosporins.
Beta-lactamase cleavage of beta-lactam ring.
Resistance mechanisms for various antibiotics: Most common resistance mechanism for aminoglycosides.
Modification via acetylation, adenylation, or phosphorylation.
Resistance mechanisms for various antibiotics: Most common resistance mechanism for vancomycin.
Terminal D-ala of cell wall component replaced with D-lac; decrease affinity.
Resistance mechanisms for various antibiotics: Most common resistance mechanism for Chlorampenicol.
Modification via acetylation.
Resistance mechanisms for various antibiotics: Most common resistance mechanism for macrolides.
Methylation of rRNA near erythromycin's ribosome-binding site.
Resistance mechanisms for various antibiotics: Most common resistance mechanism for tetracycline.
Decrease uptake or increase transport out of cell.
Resistance mechanisms for various antibiotics: Most common resistance mechanism for sulfonamides.
Altered enzyme (bacterial dihydropteroate synthetase), decrease uptake, or increase PABA synthesis.
Nonsurgical antimicrobial prophylaxis: Drug of choice for meningococcal infection.
Rifampin (drug of choice), minocycline.
Nonsurgical antimicrobial prophylaxis: Drug of choice for gonorrhea.
Cefriaxone.
Nonsurgical antimicrobial prophylaxis: Drug of choice for syphilis.
Benzathine penicillin G.
Nonsurgical antimicrobial prophylaxis: Drug of choice for history of recurrent UTIs.
TMP-SMX.
Nonsurgical antimicrobial prophylaxis: Drug of choice for Pneumocystis carinii pneumonia.
TMP-SMX (drug of choice), aerosolized pentamindine.
Anti-fungal therapy: Mechanism of action of the anti-fungal therapy polyenes.
Form artificial pores in the cytoplasmic membrane.
Anti-fungal therapy: Mechanism of action of the anti-fungal therapies terbinafine and azoles.
Terbinafine blocks the conversion of squalene to lanosterol. Azoles block the conversion of lanosterol to ergosterol.
Anti-fungal therapy: Mechanism of action of the anti-fungal therapy flucytosine.
Blocks the production of purines from the precurors.
Anti-fungal therapy: Mechanism of action of the anti-fungal therapy griseofulvin.
Disrupts microtubles.
Amphotericin B: Mechanism of action of Amphotericin B.
Binds ergosterol (unique to fungi); forms membrane pores that allow leakage of electrolytes and disrupt homeostasis. "Amphotericin 'tears' holes in the fungal membrane by forming pores."
Amphotericin B: Clinical uses of Amphotericin B.
Used for a wide spectrum of sytemic mycoses. Cryptococcus, Blastomyces, Coccidioides, Aspergillus, Histoplasma, Candida, Mucor (systemic mycoses). Intrathecally for fungal meningitis; does not cross blood-brain barrier.
Amphotericin B: Symptoms of Amphotericin B toxicity.
Fever/chills ("shake and bake"), hypotension, nephrotoxicity, arrhythmias ("amphoterrible").
Nystatin: Mechanism of action of Nystatin.
Binds to ergosterol, disrupting fungal membranes.
Nystatin: Clinical use of Nystatin.
Swish and swallow for oral candidiasis (thrush).
Fluconazole, ketoconazole, clotrimazole, miconazole, itraconazole, voriconazole.: Mechanism of action for fluconazole, ketoconazole, clotrimazole, miconazole, itraconazole, voriconazole.
Inhibits fungal steroid (ergosterol) synthesis.
Fluconazole, ketoconazole, clotrimazole, miconazole, itraconazole, voriconazole.: Clinical uses of fluconazole, ketoconazole, clotrimazole, miconazole, itraconazole, voriconazole.
Systemic mycoses. Fluconazole for cryptococcal meningitis in AIDS patients and candidal infections of all types (i.e., yeast infections). Ketoconazole for Blastomyces, coccidioides, Histoplasma, Candida albicans; hypercortisolism.
Fluconazole, ketoconazole, clotrimazole, miconazole, itraconazole, voriconazole.: Symptoms of fluconazole, ketoconazole, clotrimazole, miconazole, itraconazole, voriconazole toxicity.
Hormone synthesis inhibition (gynecomastia), liver dysfunction (inhibits cytochrome P-450), fever, chills.
Flucytosine: Mechanism of action of Flucytosine.
Inhibits DNA synthesis byconversion to fluorouracil, which competes with uracil.
Flucytosine: Clinical uses of Flucytosine.
Used in sytemic fungal infections (e.g. Candida, Cryptococcus).
Flucytosine: Symptoms of Flucytosine toxicity.
Nausea, vomitting, diarrhea, bone marrow supression.
Caspofungin: Mechanism of action for Caspofungin.
Inhibits cell wall synthesis.
Caspofungin: Clinical use of Caspofungin.
Invasive aepergillosis.
Caspofungin: Symptoms of Caspofungin toxicity.
GI upset, flushing.
Terbinafine: Mechanism of action of Terbinafine.
Inhibits the fungal enzyme squalene epoxidase.
Terbinafine: Clinical use of Terbinafinel.
Used to treat dermatophytoses (especially onychomycosis).
Griseofulvin: Mechanism of action of Griseofulvin.
Interfers with microtubule function; disrupts mitosis. Deposits in keratin-contianing tissues (e.g. nails).
Griseofulvin: Clinical use of Griseofulvin.
Oral treatment of superficial infections; inhibits growth of dermatophytes (tinea, ringworm).
Griseofulvin: Symptoms of Griseofulvin toxicity.
Teratogenic, carcinogenic, confusion, headaches, increase warfarin metabolism.
Antiviral chemotherapy: Viral adsorption and penetration into the cell is blocked by ---------.
Gama-globulins (non-specific).
Antiviral chemotherapy: Uncoating of the virus after its penetration into the cell is blocked by --------.
Amantadine (influenza A).
Antiviral chemotherapy: Early viral protein synthesis is blocked by --------.
Fomivirsen (CMV).
Antiviral chemotherapy: Viral nuclei acid synthesis is blocked by --------.
Purine, pyrimidine analogs; reverse transcriptase inhibitors.
Antiviral chemotherapy: Late viral protein synthesis and processing is blocked by --------.
Methimazole (variola); protease inhibitors.
Antiviral chemotherapy: Packaging and assembly of new viron is blocked by --------.
Rifampin (vaccinia).
Amantadine: Mechanism of action of Amantadine.
Blocks viral penetration/uncoating; may buffer pH of endosome. Also causes the release of dopamine from intact nerve terminals. "Amantadine blocks influenza A and rubellA and causes problems with the cerebellA."
Amantadine: Clinical uses of Amantadine.
Prophylaxis for influenza A; Parkinson's disease.
Amantadine: Symptoms of Amantadine toxicity.
Ataxia, dizziness, slurred speech. (Rimantidine is a derivative with fewer CNS side effects.)
Zanamivir: Mechanism of action of Zanamivir.
Inhibits influenza neuraminidase.
Zanamivir: Clinical use of Zanamivir.
Both influenza A and B.
Ribavirin: Mechanism of action of Ribavirin.
Inhibits synthesis of guanine nucleotides by competitively inhibiting IMP dehydrogenase.
Ribavirin: Clinical use of Ribavirin.
RSV (respiratory syncytial virus).
Ribavirin: Symptoms of Ribavirin toxicity.
Hemolytic anemia. Severe teratogen.
Acyclovir: Mechanism of aciton of Acyclovir.
Perferentially inhibits viral DNA polymerase when phosphorylated by viral thymidine kinase.
Acyclovir: Clinical use of Acyclovir.
HSV, VZV, EBV. Mucocutaneous and genital herpes lesions. Prophylaxis in immunocompromised patients.
Acyclovir: Symptoms of Acyclovir toxicity.
Delirium, tremor, nephrotoxicity.
Ganciclovir (DHPG dihydroxy-2-propoxymethyl guanine): Mechanism of action of Ganciclovir.
Phosphorlation by viral kinase; perferentially inhibits CMV DNA polymerase.
Ganciclovir (DHPG dihydroxy-2-propoxymethyl guanine): Clinical use of Ganciclovir.
CMV, especially in immunocompromised patients.
Ganciclovir (DHPG dihydroxy-2-propoxymethyl guanine): Symptoms of Ganciclovir toxicity.
Leukopenia, neutropenia, thrombocytopenia, renal toxicity. More toxic to host enzymes than acyclovir.
Foscarnet: Mechanism of action of Foscarnet.
Viral DNA polymerase inhibitor that binds to the pyrophophate binding site of the enzyme. Does not require activation by viral kinase. "FOScarnet = pyroFOSphate analog."
Foscarnet: Clinical use of Foscarnet.
CMV retinitis in immunocompromised patients when ganciclovir fails.
Foscarnet: Symptoms of Foscarnet toxicity.
Nephrotoxicity.
HIV therapy: Saquinavir, ritonavir, indinavir, nelfinavir, amprenavir are example of this type of anti-HIV drug.
Protease inhibitor.
HIV therapy: Mechanism of action of protease inhibitors.
Inhibit assembly of new virus by blocking protease enzyme.
HIV therapy: Symptoms of protease inhibitor toxicity.
GI intolerance (nausea, diarrhea), hyperglycemia, lipid abnormalities, thrombocytopenia (indinavir).
HIV therapy: Reverse transcriptase inhibitors:
0
HIV therapy: Zidovudine (AZT), didanosine (ddI), zalcitabine (ddC), stavudine (d4T), lamivudine (3TC), and abacavir are examples of --------- reverse transcriptase inhibitors.
Nucleoside.
HIV therapy: Nevirapine, delavirdine, and efavirenz are examples of --------- reverse transcriptase inhibitors.
Non-nucleoside.
HIV therapy: Mechanism of action of reverse transcriptase inhibitors.
Preferentially inhibit reverse transcriptase of HIV; prevent incorporation of viral genome into host DNA.
HIV therapy: Symptoms of reverse transcriptase inhibitor toxicity.
Bone marrow supression (neutropenia, anemia), periphral neuropathy, lactic acidosis (nucleosides), rash (non-nucleosides), megaloblastic anemia (AZT).
HIV therapy: Highly active antiretroviral therapy (HAART) generally entails combination therapy with ---------- and -----------.
Protease inhibitors, reverse transcriptase inhibitors.
HIV therapy: When should HIV therapy be initiated?
When patients have low CD4 counts (<500 cells/mm3) or high viral load.
HIV therapy: -------- is used during pregnancy to reduce risk of fetal transmission.
AZT.
Interferons: Mechanism of action of Interferons.
Glycoproteins from human leukocytes that block various stages of viral RNA and DNA synthesis.
Interferons: Clinical use of Interferons.
Chronic hepatitis B and C, Kaposi's sarcoma.
Interferons: Symptoms of Interferon toxicity.
Neutropenia.
Antiparasitic drugs: Clinical uses of Ivermectin.
Onchocerciasis "rIVER blindness treated with IVERmectin".
Antiparasitic drugs: Clinical uses of Mebendazole / thiabendazole.
Nematode/roundworm (e.g., pinworm, whipworm) infections.
Antiparasitic drugs: Clinical uses of Pyrantel pamoate.
Giant roundworm (Ascaris), hookworm (Necator/Ancylostoma), pinworm (Enterobius).
Antiparasitic drugs: Clinical uses of Praziquantel.
Trematode/fluke (e.g., schistosomes, Paragonimus, Clonorchis) and cysticercosis.
Antiparasitic drugs: Clinical uss of Niclosamide
Cestode/tapeworm (e.g., Diphyllobothrium latum, Taenia species) infections except cysticercosis.
Antiparasitic drugs: Clinical uses of Pentavalent antimony.
Leishmaniasis.
Antiparasitic drugs: Clinical uses of Chloroquine, quinine, mefloquine, atovaquone, proguanil.
Malaria.
Antiparasitic drugs: Clinical uses of Primaquine.
Latent hypnozoite (liver) forms of malaria (Plasmodium vivax, P.ovale).
Antiparasitic drugs: Clinical uses of Metronidazole.
Giardiasis, amebic dysentery (Entamoeba histolytica), bacterial vaginitis (Gardnerella vaginalis), Trichomonas.
Antiparasitic drugs: Clinical uses of Pentamidine.
Pneumocystis carinii pneumonia prophylaxis.
Antiparasitic drugs: Clinical uses of Nifurtimox.
Chagas' disease, American trypanosomiasis (Trypanosoma cruzi).
Antiparasitic drugs: Clinical uses of Suramin.
African trypanosomiasis (sleeping sickness).
Pharmacology - CNS / Neurologic drugs: Parasympathetic preganglionic neurons release the neurotransmitter -------- which act on -------- receptors.
Ach, nicotinic.
Pharmacology - CNS / Neurologic drugs: Parasympathetic postganglionic neurons release the neurotransmitter -------- which act on ------- receptors.
Ach, muscarinic.
Pharmacology - CNS / Neurologic drugs: Sympathetic preganglionic neurons to sweat glands release the neurotransmitter ------- which act on ------- receptors.
Ach, nicotinic.
Pharmacology - CNS / Neurologic drugs: Sympathetic postganglionic neurons to sweat glands release the neurotransmitter ------- which act on ------- receptors.
Ach, muscarinic.
Pharmacology - CNS / Neurologic drugs: Sympathetic preganglionic neurons to glands, cardiac and smooth muscles release the neurotransmitter ------- which act on ------- receptors.
Ach, nicotinic.
Pharmacology - CNS / Neurologic drugs: Sympathetic postganglionic neurons to glands, cardiac and smooth muscles release the neurotransmitter ------- which act on ------- receptors.
NE, alpha and beta
Pharmacology - CNS / Neurologic drugs: Sympathetic preganglionic neurons to renal vascular smooth muscle release the neurotransmitter ------- which act on ------- receptors.
Ach, nicotinic.
Pharmacology - CNS / Neurologic drugs: Sympathetic postganglionic neurons to renal vascular smooth muscle release the neurotransmitter ------- which act on ------- receptors.
Dopamine, D1
Pharmacology - CNS / Neurologic drugs: Sympathetic preganglionic neurons to the adrenal medulla release the neurotransmitter ------- which act on ------- receptors.
Ach, nicotinic.
Pharmacology - CNS / Neurologic drugs: Sympathetic preganglionic neurons to the adrenal medulla synapse directly on ------- cells of the adrenal medulla.
Chromaffin.
Pharmacology - CNS / Neurologic drugs: Somatic neurons synapse directly on -------- muscle and release the neurotransmitter ------- which act on ------- receptors.
skeletal muscle, Ach, nicotinic.
Autonomic drugs: Cholinergic:
0
Autonomic drugs: Ach is synthesized from acetyl-CoA and choline by the enzyme ---------.
Choline acetyltransferase.
Autonomic drugs: The transport of choline into the nerve terminal can be inhibited by --------.
Hemicholinium.
Autonomic drugs: The release of transmitter from vesicles in the nerve ending require the entry of ------ into the neuron.
Calcium.
Autonomic drugs: --------- inhibits the release of the stored Ach.
Botulinum toxin.
Autonomic drugs: The action of Ach in the synapse is terminated by its metabolism to acetate and choline by the enzyme ---------.
Acetylcholinesterase.
Noradrenergic:: In the noradrenergic nerve terminal, tyrosine is hydroxylated to -------, which is decarboxylated to --------, which is finally hydroxylated to NE.
DOPA, dopamine.
Noradrenergic:: Dopamine is transported into vesicles for hydroxylation to NE. This transport can be blocked by the drug --------.
Reserpine.
Noradrenergic:: The action of NE and DA is terminated by --------- and ----------.
Reuptake, diffusion (different than for Ach).
Noradrenergic:: --------- inhibits the release of the stored NE.
Guanethidine.
Noradrenergic:: The --------- drugs promote catecholamine release.
Amphetamine.
Noradrenergic:: The drugs --------- and ---------- inhibit the reuptake of NE.
Cocaine, TCA.
Noradrenergic:: The release of NE from a sympathetic nerve ending is modulated by ---------, --------- and ---------.
NE, Ach, angiotensin II.
Noradrenergic:: NE inhibits its own release at the noradrenergic nerve terminal through --------- receptors.
Alpha 2.
Noradrenergic:: Angiotensin II --------- (inhibits / stimulates) the release of NE from the noradrenergic nerve terminal.
Stimulates.
Noradrenergic:: Ach inhibits the release of NE from the noradrenergic nerve terminal by binding to --------- receptors.
M1.
Direct agonists:: Clinical application and action of Bethanechol.
Postoperative and neurogenic ileus and urinary retention. / Activates bowel and bladder smooth muscle.
Direct agonists:: Clinical application and action of Carbachol and Pilocarpine.
Glaucoma. / Activates ciliary muscle of eye (open angle), pupillary sphincter (narrow angle).
Indirect agonists (anticholinesterases):: Clinical application / action of Neostigmine.
Postoperative and neurogenic ileus and urinary retention, myasthenia gravis, reversal of neuromuscular junction blockade (postoperative). / Increase endogenous Ach.
Indirect agonists (anticholinesterases):: Clinical application / action of Pyridostigmine.
Myasthenia gravis. / Increase Ach; increase strength.
Indirect agonists (anticholinesterases):: Clinical application / action of Edrophonium.
Diagnosis of myasthenia gravis (extremely short acting). / Increase endogenous Ach.
Indirect agonists (anticholinesterases):: Clinical application / action of Physostigmine.
Glaucoma (crosses blood-brain barrier) and atropine overdose. / Increase endogenous Ach.
Indirect agonists (anticholinesterases):: Clinical application / action of Echothiophate.
Glaucoma. / Increase endogenous Ach.
Indirect agonists (anticholinesterases):: Symptoms of cholinesterase inhibitor poisoning.
Diarrhea, Urination, Miosis, Bronchospasm, Bradycardia, Excitation of skeletal muscle and CNS, Lacrimation, Sweating, Salivation (also abdominal cramping). "DUMBBELSS".
Indirect agonists (anticholinesterases):: Cholinesterase inhibitor poisoning may be caused by ---------.
Parathion and other organophosphates.
Indirect agonists (anticholinesterases):: The cholinesterase regenerator ------- can be used as an antidote for cholinesterase inhibitor poisoning.
Pralidoxime.
Indirect agonists (anticholinesterases):: Mechanism of action of Pralidoxime.
Regenerates active cholinesterase, chemical antagonist, used to treat organophosphate exposure.
Cholinoreceptor blockers:: Clinical uses of the muscarinic antagonist Atropine.
Dilate pupils, decrease acid secretion in peptic ulcer disease, decrease urgency in mild cystitis, decrease GI motility, reduce airway secretions, and treat organophosphate poisoning. "Blocks SLUD: Salivation, Lacrimation, Urination, Defecation."
Cholinoreceptor blockers:: Side effects of Atropine.
Increase body temp, rapid pulse, dry mouth, dry/flushed skin, disorientation, mydriasis with cycloplegia, and constipation. "Atropine parasympathetic block side effects: Blind as bat, Red as a beet, Mad as a hatter, Hot as a hare, Dry as a bone."
Cholinoreceptor blockers:: Hexamethonium (ganglionic blocker) blocks -------- receptors.
Nicotinic.
Cholinoreceptor blockers:: AUTHOR
Tzivia Moreen
antimuscarinic drugs: "tropi" are anti-muscarinic
while vacationing in the tropics you lie on a beach and your muscles waste away!
antimuscarinic drugs: benztropine is used to treat
Parkinson's disease
antimuscarinic drugs: scopolamine is used to treat
motion sickness
antimuscarinic drugs: scopolamine is an antimuscarinic that does not convert to the mnemonic!
0
antimuscarinic drugs: name 2 antimuscarinic drugs that act on the CNS
benztropine, scopolamine
antimuscarinic drugs: name a muscarinic used to treat motion sickness
scopolamine
antimuscarinic drugs: name a muscarinic used to treat Parkinson's disease
benztropine
antimuscarinic drugs: mechanism of action of benztropine
antimuscarinic
antimuscarinic drugs: mechanism of action of scopolamine
antimuscarinic
antimuscarinic drugs: name three antimuscarinics that act on eye
atropine, homatropine, tropicamide
antimuscarinic drugs: the action of atropine is ______
produce mydriasis, cycloplegia
antimuscarinic drugs: mechanism of atropine is
antimuscarinic
antimuscarinic drugs: the action of homatropine is ______
produce mydriasis, cycloplegia
antimuscarinic drugs: mechanism of homatropine is
antimuscarinic
antimuscarinic drugs: the action of tropicamide is
produce mydriasis, cycloplegia
antimuscarinic drugs: mechanism of tropicamide is
antimuscarinic
antimuscarinic drugs: ipatropium is used to treat
asthma, COPD
antimuscarinic drugs: mechanism of ipatropium is
antimuscarinic
antimuscarinic drugs: name an antimuscarinic used to treat asthma and COPD
ipatropium
neuromuscular blocking drugs: neuromuscular blocking drugs are used for
muscle paralysis in surgery or mechanical ventilation
neuromuscular blocking drugs: name a depolarising neurmuscular blocking drug
succinylcholine
neuromuscular blocking drugs: name 6 nondepolarizing neuromuscular blocking drugs
tubocurarine
neuromuscular blocking drugs: *mnemonic -- the "cur" drugs are nondepolarizing neuromuscular blocking agents
atracurium
neuromuscular blocking drugs: is succinylcholine depolarizing or nondepolarizing?
depolarizing
neuromuscular blocking drugs: is tubocurarine depolarizing or nondepolarizing?
nondepolarizing
neuromuscular blocking drugs: is atracurium depolarizing or nondepolarizing?
nondepolarizing
neuromuscular blocking drugs: is mivacurium depolarizing or nondepolarizing?
nondepolarizing
neuromuscular blocking drugs: is pancuronium depolarizing or nondepolarizing?
nondepolarizing
neuromuscular blocking drugs: is vacuronium depolarizing or nondepolarizing?
nondepolarizing
neuromuscular blocking drugs: is rapacuronium depolarizing or nondepolarizing?
nondepolarizing
neuromuscular blocking drugs: what is tubocurarine used for
nondepolarizing neuromuscular blockade
neuromuscular blocking drugs: what agents are used to reverse neuromuscular blockade by succinylcholine?
cholinesterase inhibitors in phase II (ex -- neostigmine)
neuromuscular blocking drugs: what phase of succinylcholine neuomuscular bloackade is reversible?
phase II (repolarized but blocked)
neuromuscular blocking drugs: what agents are used to reverse pahse I neuromuscular blockade by succinylcholine?
phase I Succinylcholine neuromuscular blockade cannot be reversed
neuromuscular blocking drugs: what phase of succinylcholine neuomuscular bloackade is irreversible?
phase I Succinylcholine neuromuscular blockade cannot be reversed
neuromuscular blocking drugs: what is atracurium used for
nondepolarizing neuromuscular blockade
neuromuscular blocking drugs: what is the effect of cholinesterase inhibitors on succinylcholine neuromuscular blockade?
phase I: cholinesterase inhibitors potentiates the blockade phase II: cholinesterase inhibitors reverse the blockade
neuromuscular blocking drugs: what cholinesterase inhibitor is used to reverse phase II of succinylcholine neuromuscular blockade?
neostigmine
neuromuscular blocking drugs: what is mivacurium used for
nondepolarizing neuromuscular blockade
Dantrolene: what is dantrolene used for
treat malignant hyperthermia
Dantrolene: what causes malignant hyperthermia
use inhalation anesthetics and succinylcholine together
Dantrolene: what inhalation anesthetic DOES NOT cause malignanat hyperthermia?
N2O
Dantrolene: what is dantrolene used for
neuroleptic malignant syndrome
Dantrolene: what is neuroleptic malignant syndrome
a toxicity of antipsychotic drugs
Dantrolene: what drug is used to treat malignant hyperthermia
dantrolene
Dantrolene: what is the mechanism of dantrolene
prevents release of Ca++ from saarcoplasmic reticulum of skeletal muscle
Sympathomimetics: epinephrine, NE, isoproterenol, dopamine, and dobutamine are all________________
catecholamines
Sympathomimetics: catecholamines are_____________________
sympathomimetics
Sympathomimetics: name 5 catecholamines
EPI, NE, Isoproterenol, dopamine, dobutamine
Sympathomimetics: what receptors does epinephrine act on?
alpha-1, alpha-2, beta-1, beta-2 adrenergics
Sympathomimetics: what receptors does NE work on?
alpha-1, alpha-2, beta-1 adrenergics
Sympathomimetics: what receptors does isoproterenol work on?
beta-1 = beta-2 adrenergics
Sympathomimetics: what receptors does dopamine work on?
D1 = D2, D1 and D2 more than beta, beta more than alpha
Sympathomimetics: what receptors does dobutamine work on?
beta-1 > beta-2
Sympathomimetics: which catecholamines are agonists to alpha-adrenergic receptors
EPI, NE > dopamine
Sympathomimetics: which catecholamines are agonists to beta-1 adrenergic receptors
EPI, NE, Isoproterenol, dopamine, dobutamine
Sympathomimetics: which catecholamines are agonists to beta-2 adrenergic receptors
EPI, isoproterenol, dopamine and dobutamine (less)
Sympathomimetics: what is epinephrine used to treat?
anaphylaxis, open-angle glaucoma, asthma, hypotension
Sympathomimetics: what is norepinephrine used to treat?
hypotension (but decreases renal perfusion)
Sympathomimetics: what is isoproterenol used to treat?
AV block
Sympathomimetics: what is dopamine used to treat
shock with renal failure, heart failure
Sympathomimetics: what is dobutamine used to treat
shock, heart failure
Sympathomimetics: what catecholamine is used to treat anaphylaxis
epinephrine ("EPI-pen")
Sympathomimetics: what catecholamines are used to treat hypotension
EPI, NE
Sympathomimetics: what catecholamine is used to treat asthma
epinephrine
Sympathomimetics: what catecholamine is used to treat AV block
isoproterenol
Sympathomimetics: what catecholamines are used to treat shock
doapmine, dobutamine
Sympathomimetics: what is the action of amphetamine
indirect general adrenergic agonist, releases stored catecholamines
Sympathomimetics: what is the action of ephedrine
indirect general adrenergic agonist, releases stored catecholamines
Sympathomimetics: what is amphetamine used to treat
narcolepsy, obesity, attention deficit disorder
Sympathomimetics: what is ephedrine used to treat
nasal decongestion, urinary incontinence, hypotension
Sympathomimetics: name three sympathomimetic drugs used to treat hypotension
epinephrine, norepinephrin, ephedrine
Sympathomimetics: what is the action of phenylephrine
adrenergic agonist, alpha-1 > alpha-2
Sympathomimetics: what is the action of albuterol
adrenergic agonist, beta-2 >beta-1
Sympathomimetics: what is the action of terbutaline
adrenergic agonist, beta-2 >beta-2
Sympathomimetics: what is phenylephrine used for?
pupil dilator, vasoconstriction, nasal decongestion
Sympathomimetics: what sympathomimetics are used to treat nasal congestion
ephedrine, phenylephrine
Sympathomimetics: what is the mechanism of cocaine
indirect general adrenergic agonist, catecholamine uptake inhibitor
Sympathomimetics: what is the action of cocaine
vasoconstriction, local anesthesia
Sympathomimetics: what is the mechanism of clonidine
centrally acting alpha-adrenergic agonist, decreases central adrenergic outflow
Sympathomimetics: what drug has the same mechanism as amphetamine
ephedrine
Sympathomimetics: what is the mechanism of alpha-methyldopa
centrally acting alpha-adrenergic agonist, decreases central adrenergic outflow
Sympathomimetics: what drug has the same mechanism as clonidine
alpha-methyldopa
Sympathomimetics: what are clonidine and alpha-methyldopa used to treat
hypertension, especially in renal disease because they do not decreased blood flow to the kidney
Sympathomimetics: what sympathomimetic is used to treat urinary incontinence
ephedrine
Sympathomimetics: what sympathomimetic is used to treat attention deficit disorder
amphetamine
Sympathomimetics: what sympathomimetic is used to treat narcolepsy
amphetamine
alpha-blockers: name a nonselective irreversible alpha blocker
phenoxybenzamine
alpha-blockers: name a nonselective reversible alpha blocker
phentolamine
alpha-blockers: what is the mechanism of phenoxybenzamine
nonselective irreversible alpha blocker
alpha-blockers: what is the mechanism of phentolamine
nonselective reversible alpha blocker
alpha-blockers: what are phenoxybenzamine and phentolamine used for
pheochromocytoma
alpha-blockers: what are the side effects of nonselective alpha blockers
orthostatic hypotension, reflex tachycardia
alpha-blockers: name 3 alpha-1 selective adrenergic blockers
prazosin, terazosin, doxazosin
alpha-blockers: what is the mechanism of prazosin
alpha-1 selective adrenergic blocker
alpha-blockers: what is the mechanism of terazosin
alpha-1 selective adrenergic blocker
alpha-blockers: what is the mechanism of doxazosin
alpha-1 selective adrenergic blocker
alpha-blockers: what are alpha-1 selective adrenergic alpha blockers used for
hypertension, urinary retention in BPH
alpha-blockers: what are the side effects of alpha-1 blockers
orthostatic hypotension, dizziness, headache
alpha-blockers: what is prazosin used for?
hypertension, urinary retention in BPH
alpha-blockers: what drugs have the same action as prazosin
terazosin, doxazosin
alpha-blockers: what are the side effects of terazosin?
orthostatic hypotension, dizziness, headache
alpha-blockers: what selective alpha blockers cause orthostatic hypotension
phenoxybenzamine, phentolamine, terazosin, prazosin, doxazosin
alpha-blockers: name an alpha-2 selective adrenergic blocker
yohimbine
alpha-blockers: what is yohimbine used for
impotence (effectiveness controversial)
alpha-blockers: what alpha blockers are used to treat pheochromocytoma
phenoxybenzamine, phentolamine
beta-blockers ("lol"s): name some beta-blockers
propranolol, metoprolol, atenolol, nadolol, timolol, pindolol, esmolol, labetalol
beta-blockers ("lol"s): what is the mechanism of propanolol
selective beta-adrenergic blocker
beta-blockers ("lol"s): what is the mechanism of metoprolol
selective beta-adrenergic blocker
beta-blockers ("lol"s): what is the mechanism of esmolol
selective beta-adrenergic blocker
beta-blockers ("lol"s): what is the mechanism of pindolol
selective beta-adrenergic blocker
beta-blockers ("lol"s): what are beta-blockers used to treat
hypertension, angina, MI, SVT, CHF, glaucoma
beta-blockers ("lol"s): how do beta blockers treat hypertension
decrease cardiac output, decrease renin secretion
beta-blockers ("lol"s): how do beta blockers treat angina
decrease heart rate, decrease cardiac contractility, decreased O2 consumption
beta-blockers ("lol"s): why are beta blockers used to treat MI
decrease MI mortality
beta-blockers ("lol"s): which beta blockers are used to treat SVT
propanolol, esmolol
beta-blockers ("lol"s): how do propanolol and esmolol treat SVT
decrease AV conduction velocity
beta-blockers ("lol"s): how do beta blockers treat CHF
slow progression of chronic failure
beta-blockers ("lol"s): which beta blocker is used to treat glaucoma
timolol
beta-blockers ("lol"s): what is timolol used to treat glaucoma
decrease secretion of aqueous humor
beta-blockers ("lol"s): what are the toxic effects of beta blockers
impotence, exacerbation of asthma, caution in diabetes
beta-blockers ("lol"s): what are the cardiovascular toxic effects of beta blockers
bradychardia, AV block, CHF
beta-blockers ("lol"s): what are the CNS adverse effects of beta blockers
sedation, sleep alterations
beta-blockers ("lol"s): which beta blockers are beta-1 selective
acebutolol, betaxolol, esmolol, atenolol, metaprolol (A BEAM of beta-1 blockers)
beta-blockers ("lol"s): which beta-1 blocker is short-acting
esmolol
beta-blockers ("lol"s): which beta blockers are non-selective
propanolol, timolol, pindolol, nadolol, labetalol
beta-blockers ("lol"s): which beta blocker also blocks alpha receptors
labetalol (all others are spelled "olol")
glaucoma drugs: which alpha agonists are used to treat glaucoma
epinephrine, brimonidine
glaucoma drugs: which beta blockers are used to treat glaucoma
timolol, betxolol, carteolol
glaucoma drugs: which cholinomimetics are used to treat glaucoma
pilocarpine, carbachol, physostigmine, echothiophate
glaucoma drugs: which diuretics are used to treat glaucoma
acetazolamide, dorzolamide, brinzolamide
glaucoma drugs: which prostaglandin is used to treat glaucoma
latanoprost
glaucoma drugs: what classes of drugs are used to treat glaucoma
alpha agonists, beta blockers, cholinomimetics, diuretics, prostaglandins (*mnemonic -- treating glaucoma is easy as ABCD)
glaucoma drugs: what is the effect of epinephrine in glaucoma
increase outflow of aqueous humor
glaucoma drugs: what are the side effects of epinephrine treatment in glaucoma
mydriasis, stinging
glaucoma drugs: what glaucoma should epinephrine NOT be used for
closed-angle glaucoma
glaucoma drugs: what is the effect of brimonidine in glaucoma
decreased aqueous humor synthesis
glaucoma drugs: what are the side effects of brimonidine treatment in glaucoma
no pupillary or vision changes
glaucoma drugs: what is the effect of beta-blocker treatment in glaucoma
decrease aqueous humor secretion
glaucoma drugs: what are the side effects of beta blocker treatment in glauzoma
no pupillary or vision changes
glaucoma drugs: what is the effect of cholinomimetics in glaucoma
ciliary muscle contraction, opening of trabecular meshwork, increase outflow of aqueous humor
glaucoma drugs: what are the side effects of cholinomimetics in glaucoma
miosis, cyclospasm
glaucoma drugs: what is the effect of diuretic treatment in glaucoma
inhibition of carbonic anhydrase --> decrease HCO3 secretion --> decrease aqueous humor secretion
glaucoma drugs: what are the side effects of diuretics in glaucoma
no pupillary or vision changes
glaucoma drugs: what is the effect of prostaglandin (latanoprost) treatment in glaucoma
increase outflow of aqueous humor
glaucoma drugs: what is the side effect of prostaglandin treatment in glaucoma
darkens color of iris (browning)
glaucoma drugs: which drugs used to treat glaucoma increase outflow of aqueous humor
cholinomimetics, prostaglandin, epinephrine
glaucoma drugs: can you use epinephrine in closed-angle glaucoma
NO
glaucoma drugs: brimonidine is used to treat what eye disease
glaucoma
glaucoma drugs: what kind of drug is latanoprost
prostaglandin
glaucoma drugs: latanoprost is used to treat what eye disease
glaucoma
glaucoma drugs: which glaucoma drugs decrease aqueous secretion
beta blockers, diuretics
L-dopa/carbidopa: what does L-dopa stand for
levodopa
L-dopa/carbidopa: what is the mechanism of action of L-dopa/carbidopa
increase dopamine level in brain
L-dopa/carbidopa: what is L-dopa/carbidopa used to treat
Parkinson's disease
L-dopa/carbidopa: how is L-dopa different from dopamine
L-dopa can cross the blood-brain barrier, dopamine cannot
L-dopa/carbidopa: what happens to L-dopa after it crosses the BBB
converted to dopamine by dopa decarboxylase
L-dopa/carbidopa: what enzyme convertes L-dopa to dopamine
dopa decarboxylase
L-dopa/carbidopa: what is the function of carbidopa
peripheral decarboxylase inhibitor
L-dopa/carbidopa: why is carbidopa given with L-dopa
increase L-dopa availability in CNS by inhibiting decarboxylase in periphery, also limits peripheral side effects
L-dopa/carbidopa: what are the side effects of L-dopa.carbidopa treatment
arrhythmias, dyskinesias
L-dopa/carbidopa: why do patients taking L-dopa get arrhythmias
peripheral effects of dopamine
L-dopa/carbidopa: why do patients taking L-dopa get dyskinesias
excess dopamine stimulation in CNS
Parkinson's disease drugs: what drugs are used to treat Parkinson's disease
dopamine agonists, MAO inhibitors, antimuscarinics
Parkinson's disease drugs: specifically, which drugs are used to treat Parkinson's
Bromocriptine, Amantadine, Levodopa, Selegiline, Antimuscarinics (BALSA)
Parkinson's disease drugs: which dopamine agosts are used to treat Parkinson's
L-dopa/carbidopa, bromocriptine, pramipexole, ropinirole, amantadine
Parkinson's disease drugs: what is the action of bromocriptine in Parkinson's
ergot alkaloid, partial dopamine agonist
Parkinson's disease drugs: what is the action of amantadine in Parkinson's
enhances dopamine release
Parkinson's disease drugs: what MAOI is used to treat Parkinson's
selegiline
Parkinson's disease drugs: what is the mechanism of selegiline
selective MAO type B inhibitor
Parkinson's disease drugs: what antimuscarinic is used to treat Parkinson's
benztropine
Parkinson's disease drugs: what is the effect of benztropine in Parkinson's
improves tremor, rigidity, little effect on bradykinesia
Sumatriptan: what is sumatriptan used for
acute migraine, cluster headache attacks
Sumatriptan: what is the mechanism of sumatriptan
5-HT1D agonist
Sumatriptan: what is the half life of sumatriptan
less than 2 hours
Sumatriptan: what are the side effects of sumatriptan
chest discomfort, mild tingling
Sumatriptan: what are the contraindications for sumatriptan
patients with CAD or Prinzmetal's angina
Epilepsy drugs: which drugs are used for simple and complex partial seizures
phenytoin, carbamazapine, lamotrigine, gabapentin, topiramate, phenobarbital
Epilepsy drugs: what types of seizures is phenytoin indicated for
simple and complex partial, tonic-clonic, status epilepticus
Epilepsy drugs: what types of seizures is carbamazepine indicated for
simple and complex partial, tonic-clonic
Epilepsy drugs: what types of seizures is lamotrigine indicated for
simple and complex partial, tonic-clonic
Epilepsy drugs: what types of seizures is gabapentin indicated for
simple and complex partial, tonic-clonic
Epilepsy drugs: what types of seizures is topiramate indicated for
simple and complex partial
Epilepsy drugs: what types of seizures is phenobarbital indicated for
simple and complex partial, tonic-clonic
Epilepsy drugs: what drugs can be used for tonic-clonic seizures
phenytoin, carbamazapine, lamotrigine, gabapentin, phenobarbital, valproate
Epilepsy drugs: what drugs can be used for absence seizures
valproate, ethosuximide
Epilepsy drugs: what drugs can be used for status epilepticus
phenytoin, benzodiazapines (diazepam, lorazepam)
Epilepsy drugs: what types of seizure is valproate indicated for
tonic-clonic, absence
Epilepsy drugs: what types of seizure is ethosuximide inidcated for
absence
Epilepsy drugs: what type of seizure are benzodiazepines indicated for
status epilepticus
Epilepsy drugs: other than anti-seizure, what else is phenytoin used for
class 1B anti-arrhythmic
Epilepsy drugs: how should a patient taking carbamazepine be followed
monitor LFT's weekly
Epilepsy drugs: which seizure drugs have adjunct use
gabapentin, topiramate
Epilepsy drugs: which seizure drug is safest in pregnant women
phenobarbital
Epilepsy drugs: which seizure drug is used in Crigler-Najjar II
phenobarbital
Epilepsy drugs: what are the advantages of phenobarbital
can be used in pregnant women, Crigler Najjar II
Epilepsy drug toxicities: what are the side effects of benzodiazepines
sedation, tolerance, dependence
Epilepsy drug toxicities: what are the side effects of carbamazepine
diplopia, ataxia, CYP induction, blood dyscrasias, liver toxicity
Epilepsy drug toxicities: what are the side effects of ethosuximide
GI distress, lethargy, headache, urticaria, Stevens-Johnson syndrome
Epilepsy drug toxicities: what are the side effects of phenobarbital
sedation, CYP induction, tolerance, dependence
Epilepsy drug toxicities: what are the side effects of phenytoin
nystagmus, diplopia, ataxia, sedation, ginigival hyperplasia, hirsutism, anemias, teratogenic
Epilepsy drug toxicities: what are the side effects of valproate
GI distress, rare by fatal hepatotoxicity, neural tube defects (spina bifida)
Epilepsy drug toxicities: what are the side effects of lamotrigine
life-threatening rash, Stevens-Johnson syndrome
Epilepsy drug toxicities: what are the side effects of gabapentin
sedation, movement disorders
Epilepsy drug toxicities: what are the side effects of topiramate
sedation, mental dulling, kidney stones, weight loss
Epilepsy drug toxicities: which anti-epileptic drug is teratogenic
phenytoin
Epilepsy drug toxicities: which anti-epileptic drug can cause dependence
benzodiazepines, phenobarbital
Epilepsy drug toxicities: which anti-epileptic drug can cause neural tube defects
valproate
Epilepsy drug toxicities: which anti-epileptic drugs can cause GI distress
valproate, ethosuximide
Epilepsy drug toxicities: it is necessary to check LFT's with which anti-epileptic drugs
carbamazepine, valproate
Epilepsy drug toxicities: which anti-epileptic drugs cause CYP induction
phenobarbital, carbamazepine
Epilepsy drug toxicities: which anti-epileptic drugs can cause blood problems
carbamazepine, phenytoin
Epilepsy drug toxicities: which anti-epileptic drugs can cause Stevens-Johnson syndrome
lamotrigine, ethosuximide
Epilepsy drug toxicities: which anti-epileptic drugs can cause diplopia
carbamazepine, phenytoin
Phenytoin: what is the mechanism of phenytoin action
use-dependent blockade of Na+ channels
Phenytoin: what is the clinical application of phenytoin
grand mal seizures
Phenytoin: what are the toxicities of phenytoin
nystagmus, ataxia, diplopia, lethargy
Phenytoin: what are the chronic toxicities of phenytoin
gingival hyperplasia in children, peripheral neuropathy, hirsutism, megaloblastic anemia, malignant hyperthermia (rare)
Phenytoin: should pregnant women take phenytoin
NO -- teratogenic
Phenytoin: why does phenytoin cause megaloblastic anemia
causes decreased vitamin B-12
Barbiturates: name 4 barbiturates
phenobarbital, pentobarbital, thiopental, secobarbital
Barbiturates: what is the mechanism of barbiturate action
increase duration of Cl channel opening --> decreased neuron firing --> facilitate GABA-A action
Barbiturates: how do barbiturates facilitate GABA-A action
increase duration of Cl channel opening which decreases neuron firing (Barbidurate increases duration
Barbiturates: is barbiturate action on the CNS stimulatory or inhibitory
inhibitory
Barbiturates: what is the clinical application of barbiturates
sedative for anxiety, seizures, insomnia, anesthesia induction (thiopental)
Barbiturates: which barbiturate is used for anesthesia induction
thiopental
Barbiturates: what are the side effects of barbiturates
dependence, additive CNS depression effects with alcohol, respiratory or CV depression (death), drug interactions due to CYP induction
Barbiturates: what should you find out before giving a patient barbiturates
what other medications they take, because of CYP induction and many drug interactions
Barbiturates: what happens if you give barbiturates to a patient in alcohol-induced coma or DT's
they might DIE!! Because of additive effect of barbiturates and alcohol --> respiratory depression
Barbiturates: when are barbiturates contra-indicated
porphyria
Barbiturates: can barbiturates cause dependence
YES
Barbiturates: My friend Barb was very anxious so her doctor gave her barbiturates to increase the duration of the time she could speak in public without freaking out and having a seizure. She became so dependent on it that she recommended it to her friend Portia who couldn't take it because of porphyria. One day Barb drank too much alcohol and took her barbiturates and never woke up! THE END
clinical pharmacology made ridiculous. Period
Benzodiazepines: name a bunch of benzodiazepines
diazepam, lorazepam, triazolam, temazepam, oxazepam, midazolam, chlordiazepoxide (all have ZZZ in them)
Benzodiazepines: what is the mechanism of benzodiazepines
increase frequency of Cl channel opening --> facilitate GABA-A action (Frenzodiazepines increase frequency)
Benzodiazepines: which GABA receptors are facilitated by barbiturates and bezodiazepines
GABA-A
Benzodiazepines: what are the clinical applications of benzodiazepines
anxiety, spasticity, status epilepticus (diazepam), detoxification (alcohol withdrawal, DT's)
Benzodiazepines: which benzodiazepine can be used for status epilepticus
diazepam
Benzodiazepines: what drugs can be used to treat alcohol withdrawal
benzodiazepines
Benzodiazepines: which benzodiazepines are short-acting
TOM thumb: Triazolam, Oxazepam, Midazolam
Benzodiazepines: what are the toxic effects of benzos
dependence, additive CNS depression effects with alcohol
Benzodiazepines: how are benzos better than barbiturates
less respiratory depression and coma risk
Benzodiazepines: how do you treat benzo overdose
flumazenil
Benzodiazepines: what is flumzenil used for
benzo overdose
Benzodiazepines: how does flumazenil work
competitive antagonist at GABA receptor
Benzodiazepines: can a patient become benzodiazepine dependent
YES
Benzodiazepines: are barbiturates or benzodiazepines used for alcohol withdrawal
benzodiazepines
Antipsychotics (neuroleptics): what is another name for antipsychotics
neuroleptics
Antipsychotics (neuroleptics): name 4 antipsychotic drugs
thioridazine, haloperidol, fluphenazine, chlorpromazine
Antipsychotics (neuroleptics): how do you keep benzos straight from antipsychotics
Benzos help 3rd year Jon Kazam be less anxious around patients: Shazam Kazam! Without antipsychotics patients talk like a crazy 'zine (well, not perfect, but I'm working on it)
Antipsychotics (neuroleptics): what is the mechanism of most antipsychotics
block dopamine D2 receptors
Antipsychotics (neuroleptics): what is the clinical application of antipsychotics
schizophrenia, psychosis
Antipsychotics (neuroleptics): what are the side effects of antipsychotics
extrapyramidal side effects (EPS), sedation, endocrine, muscarinic blockade, alpha blockade, histamine blockade
Antipsychotics (neuroleptics): what is a long-term effect of antipsychotic use
tardive dyskinesia
Antipsychotics (neuroleptics): what is neuroleptic malignant syndrome
a side effect of antipsychotics; rigidity, autonomic instability, hyperpyrexia
Antipsychotics (neuroleptics): how do you treat neuroleptic malignant syndrome
dantrolene, dopamine agonists
Antipsychotics (neuroleptics): what is tardive dyskinesia
side effect of neuroleptics; stereotypic oral-facial movements, may be due to dopamine receptor sensitization
Antipsychotics (neuroleptics): what is the "rule of 4" with EPS side effects from antipsychotic drugs
evolution of EPS side effects: 4 hours -- acite dystonia, 4 days -- akinesia, 4 weeks -- akasthesia, 4 months -- tardvie dyskinesia
Antipsychotics (neuroleptics): is tardvie dyskinesia reversible
often irreversible
Antipsychotics (neuroleptics): what is fluphenazine used for
schizophrenia, psychosis
Atypical antipsychotics: name 3 atypical antipsychotics
clozapine, olanzapine, risperidone
Atypical antipsychotics: what type of antipsychotic is clozapine
atypical
Atypical antipsychotics: what type of antipsychotic is olanzapine
atypical
Atypical antipsychotics: what type of antipsychotic is risperidone
atypical
Atypical antipsychotics: what is the mechanism of atypical antipsychotics
block 5-HT2 and dopamine receptors
Atypical antipsychotics: what is the mechanism of clozapine
block 5-HT2 and dopamine receptors
Atypical antipsychotics: what is the mechanism of olanzapine
block 5-HT2 and dopamine receptors
Atypical antipsychotics: what is the mechanism of risperidone
block 5-HT2 and dopamine receptors
Atypical antipsychotics: what is the clinical application of clozapine
schizophrenia positive and negative symptoms
Atypical antipsychotics: what is the clinical application of olanzapine
schizophrenia positive and negative symptoms, OCD, anxiety disorder, depression
Atypical antipsychotics: what is the clinical application of risperidone
schizophrenia positive and negative symptoms
Atypical antipsychotics: how are atypical antipsychotics different from classic ones
atypicals treat positive and negative symptoms of schizophrenia, fewer extrapyramidal and anticholinergic side effects than classic antipsychotics
Atypical antipsychotics: which antipsychotics should be used to treat positive and negative symptoms of schizophrenia
atypical ones -- clozapine, olanzapine, risperidone
Atypical antipsychotics: which antipsychotics should be used for fewer side effects
atypical ones -- clozapine, olanzapine, risperidone
Atypical antipsychotics: what is a potential toxicity of clozapine
agranulocytosis
Atypical antipsychotics: which antipsychotic drug can cause agranulocytosis
clozapine
Atypical antipsychotics: what test must be done weekly on patients taking clozapine
WBC count because of potential agranulocytosis
Lithium: what is the mechanism of action of lithium
unknown; may be related to inhibition of phosphoinositol cascade
Lithium: what is the clinical application of lithium
mood stabilizer for bipolar disorder
Lithium: how does lithium help people with bipolar disorder
prevents relapse and acute manic episodes
Lithium: what are the side effects of lithium
tremor, hypothyroidism, polyuria, teratogenic
Lithium: is it OK for women taking lithium to get pregnant
NO -- teratogenic
Lithium: what does lithium cause polyuria
ADH antagonist --> nephrogenic diabetes insipidus
Lithium: AUTHOR
Hina Talib
Antidepressants: What do the following drugs inhibit: 1. MAO inhibitors, 2. Desipramine/maprotilline, 3. Mirtazapine and 4. Fluoxetine/trazodone?
1. MAO 2. NE reuptake 3. Alpha 2-R 4. 5HT reuptake
Antidepressants: All of the above actions are ------synaptic
PRE
List the Tricyclic Antidepressants: What are the three C's of their toxicity?
Convulsions, Coma, Cardiotoxicity (arrythmias). Also respiratory depression, hypyrexia.
List the Tricyclic Antidepressants: How about toxicity in the eldery?
confusion and hallucinations due to anticholinergic SE
List the Tricyclic Antidepressants: What is the mechanism of TCA?
block reuptake of NE and 5HT
List the Tricyclic Antidepressants: What is the clinical uses of TCAs?
Endogenous depresion. Bed wetting - imipramine. OCD- clomipramine.
List the Tricyclic Antidepressants: How are tertiary TCA's different than secondary in terms of side effects?
Amitriptyline (tertiary) has more anti-cholinergic effects than do secondary (nortriptyline). Desipramine is the least sedating.
List the Tricyclic Antidepressants: what are the SE of TCAs?
sedation, alpha blocking effects, atropine-like anti cholinergic side effects (tachycardia, urinary retention)
List the Tricyclic Antidepressants: Fluoxetine, sertraline, paroxetine, citalopram are what class of drugs?
pg 311 SSRI's for endogenous depression
List the Tricyclic Antidepressants: How long does it take an anti-depressant to have an effect?
2-3weeks
List the Tricyclic Antidepressants: How does the toxicity differ fromTCA's and what are they?
Fewer than TCA's. CNS stimulation - anxiety, insomnia, tremor, anorexia, nausea, and vomiting.
List the Tricyclic Antidepressants: What toxicity happens with SSRI's and MAO inhibitors given together?
Seratonin Syndrome! Hyperthermia, muscle rigidity, cardiovascular collapse
List the Tricyclic Antidepressants: What are heterocyclics?
pg 312 2nd and 3rd generation antidepressants with varied and mixed mechanisms of action. Used major depression.
List the Tricyclic Antidepressants: Examples of heterocyclics?
trazodone, buproprion, venlafaxine, mirtazapine, maprotiline
List the Tricyclic Antidepressants: Which one is used for smoking cessation?
Buproprion. Mechanism not known. Toxicity - stimulant effects, dry mouth, aggrevation of pyschosis
List the Tricyclic Antidepressants: Which one used in GAD?
Venlafaxine - inhibits 5HT and DA reuptake. Toxicity - stimulant effects
List the Tricyclic Antidepressants: which one blocks NE reuptake
maprotiline
List the Tricyclic Antidepressants: Which one increases release of NE and 5HT via alpha 2 antagonism?
mirtazapine. Also potent 5HT Rantagonist. Toxicity - sedation, increase serum cholesterol, increase apetite
List the Tricyclic Antidepressants: What is trazodone and it' SE?
primarily inhibits seratonin reuptake. Toxicity - sedation, nausea, priapism, postural hypotension
Give 2 examples of MAO: Mechanism and Clinical Uses?
non selevtive MAO inhibition. Atypical antidepressant, anxiety, hypochondriasis
Give 2 examples of MAO: What is the toxicity with tyramine ingestion (in foods) and meperidine?
Hypertensive crisis
Give 2 examples of MAO: Other toxicities?
CNS stimulation, contraindicated with SSRI's or B-agonists
What is the mechanims of selgiline (deprenyl)?: what is the clinical use and toxicity?
adjunctive agent to L-dopa for Parkinsons. May enhance adverse effects of L-dopa
General principles: What is the significance of drugs with decreased solubility in blood?
rapid induction and recovery times . Ie. N20
General principles: What is the significance of drugs with increased solubility in blood?
increased potency = I/ MAC. Ie. Halothane
Inhaled Anesthetics: list them
halothane, enflurane, isoflurane, sevoflurane, methoxyflurane, nitrous oxide
Inhaled Anesthetics: What is good about lower solubility?
the quicker the anesthetic response, and the quicker the recovery
Inhaled Anesthetics: What are these drug's effects?
myocardial depression, respiratory depression, nausea/emesis, increase cerebral blood flow
Inhaled Anesthetics: What toxicity mactches the following drugs 1. Halothane 2. Methoxyflurane 3. Enflurane 4. Rare
1. Hepatotoxcity 2. Nephrotoxicty 3. Proconvulsant 4. Malignant hyperthermia
IV anesthetics: What do barbituates, benzodiazepines, arylcyclohexylamines and narcotic analgesics have in common?
they are IV anesthetics
IV anesthetics: What the pharmacokinetics and uses of thiopental?
high lipid solubility, rapid entry into brain. Used for induction of anesthesia for short surgical procedures. Terminated by redistribution from brain. Decreased cerebral blood flow
IV anesthetics: Give an example of a benzo and what is this class's shortcoming?
midazolam used for endoscopy. Used with gaseous anesthetics and narcotics. May cause severe post-op respiratory depressio and amnesia
IV anesthetics: What does Ketamine (PCP analog and an arylcyclohexylamine) do?
dissociative anesthetic. Cardiovascular stimulant. Causes disorientation, hallucination, bad dreams. Increases cerebral blood flow.
IV anesthetics: How are narcotic analgesics used? Examples?
Morphone and fentanyl are used with CNS depressant during general anesthesia.
IV anesthetics: What is the advantage of propofol
used for rapid anesthesia induction and short procedures. Less post-op nausea than thiopental
Local anesthetics: Name some esters?
procaine, cocaine, tetracaine,
Local anesthetics: Name some amides?
lidocaine, bupivacaine, (amides have two I's in name!)
Local anesthetics: What is the mechanism and clinical use?
bind receptor and block Na channels. Tertiary amine local anesthetics penetrate membrane in uncharge form, then bind charged form. Use for minor surgical procedures, spinal anesthesia.
Local anesthetics: How do you decide to use ester or amides?
if allergic to esters, give amides
Local anesthetics: what is the toxicity
CNS excitation, severe cardiovascular toxicity (bupivacaine), hypertension, arrhythmias (cocaine)
Local anesthetics: In infected ________ tissue, anesthetics are charged and cannot penetrate membrane. Therefore, ______ anesthetics are needed.
acidic; more
Local anesthetics: What is the order of nerve blockade for size and myelination? Which factor predominates?
small diameter> large diameter. Myelinated fibers> unmyelinated fibers. Size factor predominates
Local anesthetics: what is the order of loss of sensation?
pain first, then temp, then touch, then pressure
Local anesthetics: Why would you give these drugs with vasoconstrictors?
to enhance local action
Opiod analgesics: List as many as you can.
morphine, fentanyl, codeine, heroin, methadone, meperidine, dextromethorphan
Opiod analgesics: Mechanism: They act as _____ for opiod receptors to modulate synaptic transmission
agonists
Opiod analgesics: which drugs act at the mu, delta, kappa receptors?
morphine enkephalin, dynorphin
Opiod analgesics: Clinical use?
pain, cough supression (dex), diarrhea (loperamide), acute pulmonary edema, methadone maintenance programs
Opiod analgesics: What are the major toxicities?
addiction, respiratory depression, constipation, miosis, additive CNS depression wth other drugs
Opiod analgesics: Tolerance does not develop to __________and ______
miosis and constipation
Opiod analgesics: How would you treat toxicity?
naloxone, naltrexone (opiod R antagonist)
Other NSAIDS: List three NSAIDS?
ibuprofen, naproxen, indomethacin
Other NSAIDS: What is their mechanism?
reversibly inhibit COX 1 and 2. Blocks PG synthesis
Other NSAIDS: What is their clinical use (3As)?
Antipyretic, analgesic, anti-inflammatory. Indomethacin is used to close a PDA.
Other NSAIDS: What are common toxicities?
renal damage, aplastic anemia, GI distress, ulcers
COX 2 Inhibitors: Where is cox2 found?
in inflammatory cells and mediates inflammation and pain
COX 2 Inhibitors: Why is cox2 inhibition better than cox1?
cox1 helps to maintain gastric mucosa, thus, should not have the corrosive effects of other NSAIDs on the GI lining (less incidence of ulcers and bleeding)
COX 2 Inhibitors: Clinical Use?
RA and osteoarthritis
Acetaminophen: What is its mechanism and where does it work?
reversibly inhibits cox, mostly in CNS. Inactivated peripherally.
Acetaminophen: What are its 2 As?
antipyretic, analgesic but NOT anti-inflammatory.
Acetaminophen: Overdose effects?
hepatic necrosis, acetaminophen metabolites depletes glutathine and forms toxic tissue adducts in the liver
cardiovascular therapy: Changes in CO affect two major pathways?
1. Carotid sinus firing, sympa discharge 2. Renal blood flow, renin-ang pathway
cardiovascular therapy: What is the effect of the following drugs: 1. Positive inotropic drugs 2. Beta blockers 3. Ace inhibitors 4. AII antagonists 5. Vasodilators and 6. Diuretics
1. Increases cardiac output. 2. Inhibit renin release. 3. Inhibit ACE 4. Inhibits effects of AngII including increasing the preload, increasing the afterload and remodelling. 5. Decrease the preload and afterload. 6. Decrease the preload and afterload
antihypertensive drugs: What are the adverse effects of these two diueretics: hydrochlorothiazide, loop diuretics
1. Hypokalemia, hyperlipidemia, hyperuricemia, lassitude, hypercalcemia, hyperglycemia 2. Hypokalemia, met alk, hypotension, ototoxicity
antihypertensive drugs: These are wahat class of drugs: clonidine, methyldopa, ganglionic blockers, reserpine, guanethidine, prazosin, beta blockers?
sympathoplegics
antihypertensive drugs: Adverse effects of clonidine?
dry mouth, sedation, severe rebound HTN
antihypertensive drugs: Adverse effects of methyldopa?
sedation, positive coombs test
antihypertensive drugs: Adverse effects of ganglionic blockers?
orthostatic HTN, blurred vision, constitpation, sexual dysfuncction
antihypertensive drugs: Adverse effects of reserpine?
sedation, depression, nasal stuffiness, diarrhea
antihypertensive drugs: adverse effects of beta blockers?
impotence, asthma, cardiovascular, cns
antihypertensive drugs: Adverse effects of guanethidine?
orthostatic and exercise Hypotension, sex dysfxn, diarrhea
antihypertensive drugs: Adverse effects of prazosin?
1st dose orthostatic hypotension, dizzy, headache
antihypertensive drugs: The following are what class: hydralazine, minoxidil, nifedipine, verapamil, nitroprusside
vasodilators
antihypertensive drugs: which one causes lupus like syndrome? Other toxicities?
hydralazine, nausea, headache, reflex tachycardia, angina, salt retention
antihypertensive drugs: adverse effets of minoxidil?
hypertrichosis (hair growth - think Rogaine with minoxidil!), pericardial effusion, reflex tachycardia, angina, salt retention
antihypertensive drugs: Side effects of nifedipine, verapamil?
dizziness, flushing, constipation, nausea
antihypertensive drugs: which one causes cynide toxicity?
nitroprusside
antihypertensive drugs: Adverse effects of ACE-I Captorpil? Think CAPTOPRIL
C: cough, A: angioedema, P: proteinuria, T: taste changes, O: hypOtension, P: pregnancy problems like fetal renal damage, R: rash, I: increased renin, L: lower angiotensin. Also hyperkalemia.
antihypertensive drugs: Losartan is a ----------- R-Inhibitor? With ____-toxicity and ____kalemia
angiotensin II, fetal renal, hyper
Hydralizine: Which two anti-htn drugs do you use with B blockers to prevent reflex tachycardia, diuretic to block salt retention?
hydralizine, minoxidil
Hydralizine: What is hydralizine's mechanims and clinical use?
increase cGMP --> smooth muscle relaxation. Vasodilates arteries > veins. Reduces afterload. Used for severe HTN or CHF
Calcium channel blockers, name three: Mechanism: block _____ chanels of cardiac and smooth muscles to reduce contractility
voltage dependednt L type Ca
Calcium channel blockers, name three: Rank their effects on vascular smooth muscle ad on the heart.
smooth muscle nifed> diltia > verapamil heart: vera> diltia> nifedepine
Calcium channel blockers, name three: What is the calcium channel blockers use?
HTN, angina, arrythmias (not nifedipine)
Calcium channel blockers, name three: ACE -I, name three
captopril, enalapril, lisinopril
Calcium channel blockers, name three: Mechanim considering bradykinin and renin release?
reduce lvels of ang II, prevent inactivation of bradykinin, renin release is increased to to loss of feedback inhibition
Calcium channel blockers, name three: what is the clinical use of these?
HTN, CHF, diabetic renal disease
Diuretics- site of action: What is the site of action of 1. Acetazolamide, 2. Osmotic agents, 3. Loop agents, 4. Thiazides, 5. Potassium sparing, 6. ADH antagonists
1. PCT 2. PCT, thin desc limb, CD 3. Thick ascending limb 4. Distal conv tubule 5. DCT a bit later 6. CD in inner medulla
Diuretics- site of action: How does mannitol an osmotic diuretic work?
increase tubular fluid osmolarity, producing increased urine flow
Diuretics- site of action: what is the use and toxicity?
Use: shock, drug overdose, decrease intracranial pressure. Toxicity - pulmonary edema, dehydration. Contraindicated in anuria, CHF
Acetazolamide: Is a ______inhibitor. Causes ______diuresis and _____ in total body HC03 stores.
Carbonic anhydrase, self-limited NaHCO3, reduction.
Acetazolamide: What electrolye disturbace does it treat? Does it cause?
treats met alk, causes in toxicity hyperchloremic met acidosis. ACIDazolamide caues ACIDosis.
Acetazolamide: Other toxicity?
neuropathy, NH3 toxicity, sulfa allergy
Acetazolamide: uses?
glaucoma, urinary alk, met alk, altitude sickeness
Furosemide: This sulfonamide loop diuretic inhibits _______cotransport
NA, K, 2CL
Furosemide: Furosemide also works by?
abolishes hypertonicit y of medulla, prevent concentration of urine. Increase Caexcertion. Loops Lose calcium
Furosemide: The three uses for this loop diuretic?
edematous states, htn, hypercalcemia
Furosemide: Toxicity using the OH DANG?
ototoxicity, hypokalemia, dehydration, allergy, nephritis interstitial, gout
Ethacrynic Acid: How is this drug different from furosemide? And how does that affect its use?
Although both have the same action, ethacrynic is a phenoxyacetic acid derivative not a sulfonamide. Therefore use this drug when you are allergic to sulfa.
Ethacrynic Acid: What drug can be used to treat acute gout?
ethacrynic acid
Ethacrynic Acid: AUTHOR
Stacy Ugras
Hydrochlorothiazide: Hydrochlorothiazide is a thiazide diuretic that inhibits the reabsorption of ----- in the ---- tubule
NaCl; early distal tubule
Hydrochlorothiazide: Does hydrochlorothiazide increase or decrease the excretion of calcium ion?
decrease
Hydrochlorothiazide: A toxic dose of hydrochlorathiazide will do what to the blood levels of these electrolites: potassium, sodium, glucose, lipid, uric acid, calcium
hypokalemic metabolic alkalosis, hyponatremia, hyperGlycemia, hyperLipidemia, hyperUricemia, hyperCalcemia (hyperGLUC)
K+-sparing diuretics: Spironolactone is a competitive antagonist to the --- receptor in the ---- tubule
aldosterone; cortical collecting tubule
K+-sparing diuretics: Name two K+-sparing diuretics that block Na+ channels in the cortical collecting duct
Triamterine and amiloride
K+-sparing diuretics: Besides causing hyperkalemia, a toxic dose of spironolactone will cause this endocrine effect
Gynecomastia (antiandrogen effect)
K+-sparing diuretics: Name three K+-sparing diuretics
Spironolactone, Triamterene, Amiloride (The K+ STAys.)
Diuretics: electrolye exchange: Diuretics are classified as carbonic anhydrase inhibitors, loop diuretics, thiazides, and K+-sparing diuretics. Which of these causes in increase in urine NaCl?
All of them!