• 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

1/328

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;

328 Cards in this Set

  • Front
  • Back
role of gut-associated lymphatic tissue (GALT)
protect against potential microbial pathogens; permit immunologic tolerance to both potentially immunogenc dietary substances & bacteria that normally reside in the lumen.
describe immune system via Peyer's Patches
M cells take up antigens; in germinal cells, B cells stimulated -> differentiate into IgA-secreting plasma cells
name "one-alphabet-named" cells and what they secrete in small intestine
I cell: CCK; K cell: GIP; S cell: secretin; D cells: somatostatin
role of CCK; where and when is it secreted?
stimulate pancreatic enzyme secretion, gallbladder secretion, satiety factor; decreaes gastric emptying; secreted in duodenum & jejunum in response to presence of lipids & partially digested proteins; activated by gastrin
role of GIP (gastric inhibitory peptide)
stimulate insulin reliease, inhibits gastrin & gastric acid release when lipids reach duodenum ; response to duodenal glucose;
what activates parietal cells, and what inhibits it?
activated by histamine, ACh, gastrin; decreased by somatostatin, GIP, prostaglandins
role of secretin; when?
stimulates pancreatic bicarbonate, bile acid, fluid secretion; decreases gastric acid secretion when lipids enter duodenum & when chyme pH is low
role of somatostatin; when?
inhibits gastrin & pepsinogen release; inhibits bile flow/ gallbladder contraction, inhibits pancreatic endocrine & exocrine secretions, increase intestinal fluid absorption & smooth m. contraction, inhibits intestinal secretion; secreted in response to acid, inhibited by vagus n. (pretty much inhibits all GI secretions, activates absorption)
role of cimetidine, rantidine, famotidine
reversible block of H2 receptors (affect of histamine); potent inhibitor of p450 system.
role of omeprazole, lansoprazole
irreversibly inhibit H+/ K+ ATPase pump
role of misoprostol
prostaglandin analogue
role of pirenzepine, propantheline
muscarinic antagonist; block the activation of ECL cells and parietal cells by vagus nerve
role of calcium carbonate, aluminum OH
antacid
role of infliximab
monoclonal antibody of TNF; anti-inflammatory
role of sulfasalazine
combo of antibiotics and anti-inflammatory;
role of 5ASA
anti-inflammatory
role of ondansetron
5HT3 antagonist; anti-emetic
role of metoclopramide
D2 receptor antagonist; anti-emetic; increase resting tone for LES - for diabetic & post-surgery gastroparesis; treatment for gastricstasis
role of ciprofloxacin
blocks bacterial DNA synthesis (antibiotics)
what's in saliva?
salt, water, mucins, IgA, DNAase, lysozyme, lipase, alpha-amylase
what's special about salivary gland innervation
stimulated by both sympathetic & parasympathetic activity
affect of flow rate for salivary content
low flow rate: hypotonic, rich in K+; high flow rate: isotonic; fast flow: sodium, chloride, HCO3 go up (not absorbed as much; HCO3 not secreted until activated)
mechanism of salivary ducts
absorb sodium, chloride; not water; secrete HCO3, K+
Sjogren syndrome
autoimmune disease; inhibit salivary secretions; xerostomia (dry mouth)
two types of peristalsis by esophagus
primary: initiated by swallowing; secondarY: elicited by distention to clear remaining fo od
phases of swallowing reflex; and regulation of LES relax'n
oral (voluntary), pharyngeal (involuntary), esophageal (involuntary); VIP and NO activate LES relax'n, ACh inhibit it
tx to esophageal varices
treat hypertension; surgical shunt to divert blood to systemic circulation
achalasia: ID
LES doesn't fully relax
achalasia: etiology, pathophysiology
possibly autoimmune attack-> degeneration of nerve ganglia in myenteric plexus
achalasia: dx
barium esophagogram (bird's peak); Manometry test to measure pressure in LES & reveal absence of peristalsis/ LES increased tone
achalasia: tx
myotomy of circular muscle over LES; channel blockers to relax LES; balloons to offer relief
sources of infectious esophagitis
herpes, CMV, candida; bacteria is rare
implication of eosinophil for esophagitis
allergy, asthma, infection
GERD: ID, pathophysiology, sx
failure of LES to prevent gastric regurgitation -> heartburn (pyrosis), inflammation; chest pain, which can radiate to neck
GERD: risk factors
hiatal hernia, obesity, pregnancy, smoking, alcohol, caffeine
GERD: dx, tx
endoscopy; PPI, histamine receptor antagonist, antacid
Barret's esophagus: tx
primary tx: surgical removal
two types of GI motility
segmental & peristaltic
Importance of UES
part of swallowing mechanism; prevents air entry (to maintain certain pressure level)
innervation of stomach relaxation
vagovagal reflex; swallowing -> relax
ileogastric reflex
distension in ileum -> decreases gastric motility
role of atropine
inhibits vagal stimulation of parietal cells
change in parietal cell for activation
tubulovesicles move to canaliculus
cellular reaction w/in parietal cell
H+/K+ ATPase on apical side; Na+/K+ ATPase on basolateral side; HCO3 to blood; Cl- to lumen; H+ and Cl- unite in lumen;
how is G cell activated via nerve innervation
via GRP from vagus n.
phases of gastric acid secretion
cephalic: afferent senses -> activate parietal & ECL (ACh), G cell (GRP), inhibit D cells;; gastric: food -> distension -> vagovagal reflex, partially digested protein -> G cells;; intestinal: AA in SI -> acid secretion
gastroduodenal mucosal defense
mucus-bicarbonate layer; microvascular system in submucosal layer -> defense/ repair, supply of nutrients/ oxygen, remove toxic; prostaglandins in mucosa: regulate release of mucus, bicarbonate, other; growth factors, proliferation
etiology & mechanism: vomiting
receptors in ear, overdistension of stomach/ duodenum, receptors in throat; reverse peristalsis via reflex by medullary centers
etiology & tx for hypertrophic pyloric stenosis
lack of inhibitory influences on pylorus or inadequate generation of NO; tumor, ulcer; surgical pyloromyotomy
etiologies of acute gastritis
NSAIDS, H. pylori, excessive alcohol consumption, radiation/ chemo
gastroparesis: ID, sx, etiology, tx
impaired/ delayed gastric emptying w/o evidence of obstruction; sx: satiety, nausea, vomiting of undigested/ partially digested food, bloating; etiology: diabetes, surgical, medical treatments; tx: small meals, low fat diets to inhibit CCK (CCK inhibits gastric movement)
GUD vs DUD
GUD: normal or low acid level; pain w/ meals; older paitent;; duodenal: acid level high, pain decrease w/ food, almost always H.pylori, "punched out" margins
side effect of cimetidine
increases gastric pH -> bacterial colonization -> pneumonia if inspirated into airway
mechanism of H.pylori; dx; tx
urease: produces ammonia from urea -> alkalize the surrounding; reduces somatostain, makes proteases & phospholipases to break down glycoprotein lipid complex of mucous gel; dx: urea breath test and serology tests for antibodies; tx: antibiotics, PPIs, H2 receptor blockers
mechanism of autoimmune gastritis (reduced parietal)
lower HCl -> higher gastrin and G cell (hyperplasia); pernicious anemia
ZES: classical sx, tx
increased gastrin secretion, PUD, diarrhea (due to inactive pancreatic enzymes due to high acidity); tx: whipple's, duodenotomy; PPIs, H2 blocker
what pancreatic enzymes released?
trypsin, chymotrypsin, carboxypeptidase, elastase, lipase, amylase, ribonuclease, deoxynuclease
activation/ role of duodenal enzymes
acidic chyme -> enterokinase -> trypsinogen to trypsin; CCK & vagal ACh stimulate acinar secretions; lipid presence in duodenum -> CCK release
mechanism to protect pancreas from auto-digest
acinar cells secrete trypsin inhibitor and ductal cells secrete HCO3- to keep the trypsin inactivated
regulation of pancreatic HCO3- release
response to secretin & ACh
phases of pancreatic secretion
cephalic: sensory-> ACh -> enzymes; gastric: distension -> ACh -> enzymes; intestinal: acid/ fat/ peptides, AA -> secretin/ CCK/ ACh -> enzymes + HCO3-
cellular reaction w/in pancreatic duct cell
Cl-HCO3- exchanger in apical side; CFTR release Cl to lumen in apical side; Na K ATPase exchanger on basolateral; H2O is split inside the cell; OH- used to make HCO3-, and H+ leave to blood
etiology of acute pancreatitis
GET SMASHED; gall stones, ethanol, trauma, steroids, mumps, autoimmune, scorpion stings, hyperlipidemia, ERCP, drugs
pathogenesis of acute pancreatitis
zymogens -> active form (esp. trypsinogen) in acinar cells -> autodigestion
acute pancreatitis: sx, dx, tx
epigastric pain -> radiate to back; nausea, anorexia, tachycardia, fever, peritonitis (leakage of pancreatic or bile fluids); dx: increased serum amylase & lipase, WBC; etiology would accopany liver enzymes & bilirubin; tx: depends on cause: EtOH cessation, gallbladder removal, etc.
etiology, pathophys: chronic pancreatitis
Mainly EtOH. Other (stone, trauma, CF, autoimmune); inflammation -> fibrosis & atrophy (irreversible);
chronic pancreatitis: sx, dx, tx
diabetes, steatorrhea, maldigestion, epigastric pain, weight loss; dx: serum, but more on imaging to reveal calcifications in pancreatic duct; tx: analgesics, surgical correction of duct obstruction, lipase supplement, EtOH cessation, diabetes management
CF: pathophysiology
CFTR defect -> decreased HCO3- & water secretion by ductal cells -> acinar secretions thicken -> obstruction -> maldigestion & steatorrhea
secretion/ absorption of intestines
net absorption of Na, Cl, water, K; secretion of HCO3-
absorption of sodium in intestines
sodium-glucose (SGLT1) & sodium-AA transport after meal in proximal; when bicarbonate in lumen -> sodium-proton exchange; ENaC: enhanced by aldosterone (mainly in colon), inhibited by CFTR
absorption of chloride in intestins
driven by sodium absorption, voltage-dependent chloride absorption; chloride-bicarbonate exchanger;
absorption/ secretion of potassium in intestines
proximal segments (SI) absorb passively, distine segments (LI) secrete passively (paracellular) & actively (transcellular);
mechanism of cholera & VIP
activates AC -> increase cAMP -> permanent GTP bound state -> opens CFTR to drive chloride out -> diarrhea
regulation of intestinal ion transport
aldosterone: stiumlate electrogenic sodium absorption in colon; cAMP, cGMP, calcium -> enhance anion secretion, inhibit Na Cl absorption; mineralocorticoids, glucocorticoids, somatostatin -> enhance absorption; antigen activates mast cells -> secrete fluid
basic digestion/ absorption of carb
salivary & pancreatic alpha amylase, SI brushborder disaccharides; glucose & galactose via SGLT1 (2ndarily active); fructose via GLUT5 (passive); GLUT2 on basolateral
basic digestion/ absorption of protein
gastric & pancreatic proteases, some by brush border enzymes; AA & peptides absorbed via PepT1 (due to sodium-proton exchanger) -> further digested inside enterocyte -> shipped out passively
basic digestion/ absorption of fat
lingual, gastric, pancreatic lipase; pancreatic lipase (secreted as active) w/ colipase, phospholipidase A2 -> enters as micelle (short, medium FAs enter paracellularly) -> form chylomicrons inside -> lymphatics
site of nutrient absorption
calcium, iron, folate: mostly duodenum; bile acids: mostly in ileum; carbs, protiens, lipids: small intestine, decreasingly as it progresses;
neonatal protein absorption
whole protein apical pinocytosis -> passive immunity from mother
pathophysiology of Hartnup's disease
tryptophan malabsorption; tryptophan appears in normal
pathophysiology of cystinuria
transporter for the dibasic amino acids cystine, lysine, arginine, and ornithine is absent in both the small intestine and the kidney
pathophysiology of abetalipoproteinmia
mutation in protein that catalyze formation of chylomicrons; steatorrhea
dx for steatorrhea
test for TAG or FFA in stool (qualitative); fecal fat from 72 hr -> titrate for fecal lipid (quantitative)
Describe schilling's test
To determine whether it's ileum, B12, or IF's problem; labeled B12 & collect it in urine to measure progress
calcium absorption
regulated by vitamin D; active transport (transcellular) in duodenum; passive (paracellular) in other areas of SI
Magnesium absorption & fn
active in ileum; passive in duo/jeju; cofator for neurotransmission & muscular contractions; proper secretion of parathyroid hormone
iron absorption
only in duodenum; ferric to ferrous in apical; ferous to ferric in basolateral; carried by transferrin in blood
pathophysiology, sx, dx, tx of hemochromatosis
too much iron accumulation in organs; liver (cirrhosis), pancreas (diabetes), arthritis, bronze pigmentation; not as much to women (menstrual flow out); dx: high serum iron, transferrin, ferritin, liver bx; tx: phlebotomy (blood removal)
celiac: pathophysiology; sx; dx; tx
autoimmune response to alpha-gliadin (resistant to degradation); absence of villi, malabsorption, diarrhea, joint pain, anemia,; dx: hypocalcemia, hypoalbuminemia, diagnostic antibodies (IgG antigliadin, IgA antigliadin, IgA tissue transglutaminase, IgA endomysial) , duodenal bx, confirmation of improvement after gluten-free
gastrocolic reflex
urge to defacate after gastric distention
bacterial overgrowth: pathophysiology, dx, tx
impaired motility, obstruction, fistula -> stasis -> overgrowth -> competition for nutrients, impaired micelle formation, diminished brush border enzymes, lactic acidosis; dx: hydrogen breath test after oral glucose take; tx: correction of anatomical abnormality & antibiotics
Hirschsprung's disase: etiology, pathophysiology, sx, dx, tx
neural crest cells' no caudal migration -> partial absence of ganglion cells in plexuses, no interstitial cells of cajal -> internal anal sphincter not relax, no coordinated peristaltic contractions -> chronic constipation early; dx: IHC stain to ID ganglion cells; tx: remove segment missing plexuses, connect gap
Meckel's diverticulum: ID, pathophys, dx
diverticulum in ileum of gastric or pancreatic tissue; failed involution (closure) of vitelline duct (w7) -> palpable abdominal mass, painless rectal bleeding; dx, tx: barium enema
Zenker's diverticulum: ID
false diverticulum at pharyngoesophageal junction
mechanisms of intestinal obstruction
hernia, adhesion, volvulus, intussusception (telescoping)
diarrhea: dx
osmotic: stops w/ fasting, increased osmotic gap in stool, lose more water than electrolytes, no fecal fat; secretory: little to no structural damage, no blood, pus, fat in stool; persist w/ fasting, isotonic stool
C. Difficile mechanism
after normal flora killed by antibiotics, C. Difficile becomes predominant -> release proteins A & B -> binds and cause epithelial cells to slough off -> mucosal ulceration & pseudomembranes
ID: colonic diverticulosis
most common cause for hematochezia; susceptible to rupture, bleeding; most are in left colon, but right sided more likely to bleed
ulcerative colitis - different from crohn's
only colon & rectum, mucosa & submucosa; increased risk for colon cancer; continuous ulcer, no stricture or granuloma
crohn's - different from UC
anywhere in GI, but ileum & colon (not rectum) likely, not continuous, not as risky for cancer, transmural, fistula, fat/ vitamin malabsorption, stricture
inflammatory bowel disease (IDS): pathophysiology, sx, tx
host interaction & intestinal microbiota, epithelial dysfunction & mucosal immune response; sx: diarrhea, abdominal pain, WBC count, joint pain; tx: 5-ASA, corticosteroids, antibiotics, probiotics, colectomy curable for U.C., not for Crohn's
prednisone
corticosteroids
ID: McBurney's point, Rovsing's sign, psoas sign, obturator sign; dx, and other possible ddx
mcburney's: 2/3 distance from umbilicus to right ASIS; rovsing's: push left, pain right; psoas: iliopsoas muscle movement (retroperitoneal) -> retrocecal orientation; obturator: in contact w/ obturator internus; dx: CT; ddx: ectopic pregnancy & diverticulitis
name different liver cells & function
stellate cells: contain lipid droplets, produce type 1, 3 collagen; kupffer cells: macrophages of liver; pit cells: natural killer cell activity; cholangiocytes lining bile duct
different zones and relation to nearby structures
zone 1: closest to portal, most oxygenated, affected first by viral hep; zone 3: next to central venule, susceptible to hypoxia, ischemia, has p450 -> sensitive to toxic injury of acetominophen & EtOH
liver functions
bile, plasma protein, fat soluble vitamins production; AA deamination; carb homeostasis, lipid metabolism, iron homeostasis, detox, conversion of thyroid hormones, vitamin D activation, insulin & glucagon degradation
AST, ALT, ALP,
AST: many tissues like RBC, skeletal m., cardiac m.; ALT: more spc. To liver; ALP: enzyme in cells lining biliary tracts of liver
circulation of bilirubin metabolism
heme -> UCB (not soluble) -> CB in liver (soluble) -> exit as bile -> via enteric bacteria become UCB -> UBG (colorless) -> reabsorbed or excreted as stercobilin in feces or via kidney as urobilin (yellow)
significance of unconjugated bilirubin
increase in heme production; liver function -> not as capable of conjugating
significance of conjugated bilirubin
obstruction -> CB not being sent to intestine; liver cell necrosis
complications of liver failure
hepatic encephalopathy: asso. w/ elevated ammonia level, brain edema, asterixis, confusion; hepatorenal syndrome: renal failure w/ no functional causes, elveated creatinine & urea; hepatopulmonary syndrome: hypoxemia, decreased oxygen saturation, increased dyspnea
ID: cirrhosis
irreversible fibrosis leading to portal hypertension; parenchymal nodule formation or regenerating hepatocytes surrounded by fibrosis; disrpution of entire liver
sx of portal hypertension
hepatic encephalopathy, skin spider angiomata, esophageal varices, splenomegaly, periumbilical caput medusae, ascites, hemorrhoids
pathophysiology of wilson's disease; sx
mutation -> copper not excreted in bile as much -> decreased ceruloplasmin -> Cu deposits in many tissues, such as liver, brain, eye; asterixis, basal ganglia degenration, corneal copper, cirrhosis, dementia
function of alpha-antitrypsion
inhibits proteases (elasttase, trypsin) & neutrophil elastase;
what is bad comorbidity of ALD?
chronic infection of hep C
disulfram
inhibits acetaldehyde dehydrogenase -> bad sx whenever drink alcohol
naltrexone
opioid receptor antagonist -> reduces cravings for EtOH
cirrhosis's role in creating portal hypertension
stellate cells -> type 1, 3 collagen -> fibrosis -> loss of fenestrations in sinusoids -> change in pressure
MELD calculation
creatinine, bilirubin, INR (prothrombin time)
causes of hepatitis
drug allergy, inflammation due to toxins, viruses; alcohol,
HAV: transmission, tendency, dx
via water, food; blood transmission rare; mild, asymptomatic, or acute, not chronic; dx: IgG -> already exposed/ vaccinated, IgM antibodies & fecal HAV shedding -> active
HBV: dx
idea: look for HBsAg or anti-HBcAg (IgG), antiHbs; HBeAg starts later & ends earlier; a
cute: HBsAg, HBeAg, IgM,
window phase: just IgM; ;
chronic: HBsAg, IgG, HBeAg
healthy chronic: HBsAg, IgG, but not HBeAg
cured: anti-HBcAg (IgG), anti-HBs
immunized: anti-HBs
HBV: transmission, tx
parenteral, sexual, perinatal; interferon alpha-2a
ribavirin
guanosine analog -> inhibits viral mRNA; for HepC
HCV: dx
rtPCR for HCV RNA; HCV-RNA & anti-HCV; anti-HCV means infection or recovery, not protective antibody;
HDV: transmission, dx
Parenteral transmission; IgM, IgG, HDV RNA, HDAg
superinfection of hep V
HBV + HDV; carrier of HBV exposed to HDV -> superinfection; elimination of HBV -> elimination of HDV
HEV: transmission, tendency, dx
fecall-oral; not chronic; dx: PCR for RNA, IgM, IgG
autoimmune hepatitis: dx, tx
dx: anti-nuclear, anti-smooth muscle, anti-actin; tx: immunosuppresants
acetaminophen-induced liver damage: pathophys
metabolism via cytc p450 -> reactive compound -> ROS; worsen by ethanol -> less degradation of cytc 450
tx for encephalopathy
lactulose: sugar only digested by colonic bacteria -> lower pH -> protonation of ammonia -> ammonium excertion; low protein diet to reduce ammonia production
cause of ascites
decreased degradation of aldosterone by liver -> more sodium, water retension
EtOH metabolism; pathophysiology
EtOH -> acetaldehyde (via alcohol dehydrogenase) -> acetate (via acetaldehyde dehydrogenase); increases NADH/NAD+ ratio; pyruvate -> lactate, OAA -> malate; no gluconeogensis, more FA synthesis; hypoglycemia, hepatic fatty change
fomepizole
inhibits alcohol dehydrogenase
role of estrogen in liver pathology
estrogen not broken down as much; gynecomastia; estrogen -> increased vascularity -> palmar erythemia, spider angiomas, portal hypertension
gallbladder: electrolyte control
concentrate bile, sodium-proton exchanger to acidify bile -> calcium salt precipitation not as likely
regulation of bile secretion
secretin: stimulates bicarb release into bile ducts; CCK & ACh stimulate release; somatostatin inhibits bicarb release into bile ducts
two types of cholelithiasis; causes; dx
cholesterol stones: not enough bile acids to keep cholesterol (estrogen, OCT, obesity) in miccelles, + hypomotility; unconjugated bilirubin (pigment stones): bacterial infection of biliary tract -> deconjugate bilirubin -> less soluble, complexes w/ calcium -> black stone; Murphy's sign, US
causes of obesity; risk factors
diet, basal metabolism, hormones, cytokines, adipokines, sleep, gut flora, activity; family history, gender, ethnicity, childhood obesity, lower socioeconomic status, sedentary lifestyle,
BMI
kg/ m^2
metabolic syndrome: criteria
abdominal obesity, elevated TAG, low HDL, hypertension, high fasting glucose;
changes in gastric bypass
gastric pouch smaller; duodenum and a lot of jejunum bypassed since food exits via outlet/ not pyloric sphincter; pancreatic and billiary secretions to the distal ileum; ghrelin lower; PYY higher; some malabsorption
mechanisms of complications of gastric bypass
rapid filling of hyperosmotic chyme -> diarrhea, emesis, diarrhea; reduced lipolytic enzymes -> steatorrhea; vitamin D defiency (fat soluble), iron, calcium, thiamine; most important: b12 & Fe; dumping syndrome; mild protein defiency; lactose intolerance
lifestyle for patients w/ gastric bypass
avoidance of simple & refined carb; eat small frequent meals;
body's adaptation after gastric bypass
villi hypertrophy (not plasia); management of nitrogen excretion; ghrelin suppressed
gut -> CNS control
paracrine/ endocrine signal -> ileum -> CNS (appetite/ satiety), ex. Leptin from adipocytes -> satiety (hypothalamus); stomach, intestine -> ghrelin -> hunger (hypothalamus); PYY (peptide YY) from L cells distal bowel, colon -> decrease ghrelin level; L cells of distal bowel -> GLP (glucagon like peptide) -> increase insulin secretion/ insulin sensitivity
name plasma lipoproteins & their density; their source
chylomicrons (lowest density, dietary); VLDL (liver), LDL, HDL
HDL's mechanism
pick up cholesterol & phospholipids via ABCA-1; LCAT inside HDL esterifies cholesterol; cholesterol taken directly by SR-B1 (scavenger receptor) to liver, or to chylomicrons & VLDL via cholesterol ester transfer protein (CETP)
Tangier disease: cause
defect in ABCA-1; cant take cholsterol/ phospholipids from periphery to HDL
mechanism of atherosclerosis
too much cholesterol in tissues -> don't take up -> longer duration & more concentration -> oxidized LDL -> macrophages take cholesterol esters -> foam cells
ezetimibe
blocks cholesterol uptake from gut by inhibitting SI transporter -> lower LDL
niacin
increases HDL
mechanism of cholestyramine, colestipol
bile resins; binds to bile acids in SI -> prevent absorption -> more LDL uptake by liver to make bile acid -> less LDL
role of gastric acid
denature protein, kill bacteria, activate pepsin
role of prostaglandin
stimulate mucus, bicarb secretion; increased mucosal blood flow; limit diffusion of acid to epithelium
common lab tests to check liver function
serum albumin & prothrombin time; half life of albumin: 20 days; prothrombin - 5~7 days of half life; bilirubin
which reactions occur in mitochondria?
beta oxidation, TCA, ox phos, heme synthesis, urea cycle, gluconeogenesis
which reactions occur in cytoplasm
glycolysis, FA synthesis, PPP shunt, protein synthesis, steroid synthesis
rate determining enzyme for glycolysis?
PFK-1
rate determining enzyme for gluconeogenesis?
F1,6 bisphosphatase
rate determining enzyme for TCA cycle
isocitrate dehydrogenase
rate determining enzyme for glycogen synthesis?
glycogen synthase
rate determining enzyme for glycogenolysis
glycogen phosporylase
rate determining enzyme for PPP shunt
glucose6phosphate dehydrogenase (G6PD)
rate determining enzyme for de novo pyrimidine synthesis; regulation
carbamoyl phosphate synthetase II (CPS2); activated by PRPP, inhibited by UTP
rate determining enzyme for de novo purine synthesis
glutamine-PRPP amidotransferase
rate determining enzyme for urea cycle; regulation
carbamoyl phosphate synthetase I (CPS1); activated by NAG (N-acetyl-glutamate), which is activated by arginine.
rate determining enzyme for FA synthesis
acetyl-CoA carboxylase (ACC)
rate determining enzyme for FA oxidation; regulation
carnithine acyltransferase I or CPT1; allosterically inhibited by malonyl-coA, which is product of ACC
rate determining enzyme for ketogenesis
HMG-CoA synthase
rate determining enzyme for cholesterol synthesis; regulation
HMG-CoA reductase; requires 2 NADPH molecules; release CoA (so irreversible); inhibited by cholesterol; insulin & thyroxine up-regulate it; glucagon, glucocorticoid downregulate; targetted by statin drugs
covalent regulation of glycogen synthesis
insulin activates phosphodiesterase activates glycogen synthase
covalent regulation of glycogen break down
glucagon/ epinephrine, via PKA, phosphorylates pohsphorylase kinase, which phosphorylates to activate glycogen phosphorylase
allosteric regulation to encourage glycogen synthesis
G6P activates glycogen synthase ; in liver, glucose inhibits glycogen phosphorylase; ATP inhibits glycogen phosphorylase. (think in terms of muscle energy)
allosteric regulation of glycogen break down
in muscle, AMP activates glycogen phosphorylase; calcium binds to calmodulin and activates phosphorylase kinase w/o PKA phosphorylation; in liver, calcium activates PKC, which phosphorylates to inactivates glycogen synthase
another name for vitamin C. Why is it needed for collagen synthesis?
ascorbic acid; hydroxylates proline & lysine; needed for hydrogen bond;
what is produced from PPP? Regulation of PPP.
NADPH produced; thus NADP+ needed. NADPH inhibits; insulin upregulates G6PD; irreversible.
Important role of NADPH from PPP for RBCs
used by glutathione reductase; anti-oxidant & DNA synthesis
Explain G6P dehydrogenase deficiency
No PPP; PPP only source of NADPH in RBC -> hemolytic anemia
what does oxidative portion of PPP produces? What can it become?
ribulose 5-phosphate; when NADPH high, convert to ribose 5-phospohate for nucleotide ; when NADPH low, fructose 6P or G3P for glycolysis.
when PPP product converts to glycolysis product, what's produced; what enzyme and cofactor?
ribulose 5-phosphate to F6P and G3P. Transketolase and transaldolase; TPP needed. (thiamine derivative)
citrate's role in allosteric regulation
inhibits citrate synthase; inhibits PFK1, activates acetyl-CoA carboxylase; citrate synthase inhibited by ATP
regulation of isocitrate dehydrogenase
its work = irreversible; activated by ADP & ca2+; inhibited by NADH, ATP; requires NAD+
regulation of alpha-ketoglutarate dehydrogenase
requires TPP, lipoid acid, FAD, NAD+, CoA; inhibited by NADH, ATP, succinyl CoA; activated by Ca2+
regulation of PDH. Distinguish allosteric & covalent regulation. What other cofactors required?
covalent: via PDH kinase, inhibted by ATP, acetyl CoA, NADH, activated by pyruvate; covalent: via PDH phosphatase, activated by Ca2+, Mg2+, ADP, NAD+; allosteric: acetyl coA & NADH inhibitors; req TPP, lipoic acid, coA, FAD, NAD+
what are irreversible enzymes for glycolysis?
GK/ HK; PFK-1; PK
gluconeogenesis, how to go from pyruvate to PEP? Regulation?
pyruvate carboxylase for pyruvate -> OAA; biotin, ATP, CO2 needed, activated by acetyl CoA; malate dehydrogenase for OAA -> malate, NADH needed; shuttle, back to OAA, NADH produced; PEPCK for OAA -> PEP, GTP required
gluconeogenesis, how to go from F1,6BP to F6P? Regulation?
F1,6 Biphosphatase; inhibited by AMP and F-2, 6-BP; activated by ATP; glucagon -> PKA -> phosphorylate PFK2/FBP2 (bifunctional); no glucagon -> not phosphorylated PKF2/FBP2;
gluconeogenesis, how to go from G6P to glucose? Regulation?
*** G6Phosphatase; compartmentalizaiton in ER.
defect in G-6-Phosphatase. Impact.
glycogenolysis and gluconeogenesis inhibited. -> severe hypoglycemia.
glucagon's stimulation of gluconeogenesis
allosteric: increase F-2,6phosphatase -> less F2,6BP; covalent: phosphorylate to inactivate PK; enzyme: increases txn of PEPCK gene
AMP's role in gluconeogenesis
drives glycolysis: inhibits fructose 1,6-biphosphatase, activates PFK-1
futile cycle control for gluconeogenesis, how is PK portion controlled?
enzyme synthesis for both: glucagon; alanine & ATP also inhibit PK
futile cycle control for gluconeogenesis, how is PFK-1 portion controlled?
allosteric for both: F-2,6-BP (controlled by glucagon, insulin)/ AMP/ ATP
futile cycle control for gluconeogenesis, how is G6Pase portion controlled?
enzyme synthesis for both: insulin/ glucagon; compartmentalization
How is PFK-1 regulated?
activated by AMP, F-2,6-BP; inhibited by citrate, ATP, low pH; Mg2+ req. glucagon/ insulin covalently regulates F-2,6-BP kinase/ase & PFK1
How is PK regulated?
PK inhibited by alanine, glucagon, ATP; activated by F-1,6-BP; activated by insulin; Mg 2+ req.
How is G6Pase regulated?
** G6Pase is compartmentalized in ER
two NADH shuttle for glycolysis to ETC, what organs, how many ATPs?
basically reduce first in cytoplasm, then oxidize in mitochondria; Called "G3P shuttle"; DHAP <-> G3P (you use NADH to make glycerol); NADH -> FADH2; 1.5 ATP/FADH2; brain, muscle;; called "malate-aspartate shuttle" malate <-> OAA; NADH; 2.5 ATP/ NADH2; heart, liver
how is HK and GK regulated?
HK inhibited by G6P. GK inhibited by F6P, stimulated by glucose; Mg2+ required.
How is PFK-2 regulated?
PFK-2/FBP-2 is bifunctional enzyme; insulin -> activate PFK-2 via dephosphorylation; glucagon/epinephrine activates F-2,6-biphosphotase
what happens when PK deficient?
hemolytic anemia
What does CPS2 stand for, what pathway, and regulation?
**carbamoyl phosphate synthetase II; de novo pyrimidine synthesis; activated by ATP, PRPP; inhibited by
In de novo purine synthesis, how is balance of nucleotide bases shown?
AMP synthesis requires GTP; GMP synthesis requires ATP; AMP, GMP inhibit their own synthesis from IMP; first step inhibited by GMP, IMP, AMP collectively, so if one of them in access, no go
what are the enzymes of purine salvage? Their regulation?
HGPRT and APRT; HGPRT inhibited by IMP, GMP; APRT inhibited by AMP;
With ALT, what are substrate and product?
alanine, pyruvate
With AST, what are substrate and product?
OAA, aspartate
from purine nucleotide cycle, ammonia and TCA intermediate can be generated. What stage and how?
prolonged fasting; ribose-5P + aspartate -> fumarate + ammonia
How are vitamin B1 and B12 used together in function?
N5-methyl THF -> THF only done with B12. Then homocysteine -> methionine.
fed state, what activates glycogen synthesis
G6P activates glycogen synthase
fed state, what activates PPP
elavated G6P increases PPP activity; FA synthesis -> req. NADPH
fed state, what activates glycolysis
insulin -> glycolysis; GK (liver) has high Km, receives lots of glucose, let them in (GLUT2);
fed state, what activates TCA
pyruvate activates PDH -> lots of acetyl CoA; AA -> lots of TCA intermediates
fed state, what activates FA syn
high acetyl CoA, NADPH -> activated ACC -> FA synthesis
fed state, why no gluconeogenesis
*low acetyl CoA inactivates pyruvate carboxylase
what happens in fed state in liver
protein synthesis, lipid synthesis, AA degredation for E production, increased glycolysis & TCA, increased PPP, increased glycogen synthesis
what happens in fed state in muscle?
glucose absorbed -> glycogen & TCA; AA absorbed -> protein
what happens in fed state in adipocyte?
glucose -> TCA, acetyl coA -> TAG; chylomicrons (from gut) -> TAG; VLDL (liver) -> TAG
what metabolic hormone receptors for muscle?
insulin and epinephrine; no glucagon; muscle doesn't synthesize glucose
during fasting, proteins -> AA. Explain three locations where AA's are converted to enter different pathways.
alanine -> pyruvate -> gluconeogenesis in liver; glutamine -> glutamate -> alpha-ketoglutarate & ammonia in kidney; ammonia combines w/ ketonic hydrogen to make ammonium (buffer for acidity); gut: absorb glutamine -> metabolize to alanine -> liver
time line of glucose homeostasis during fasting
first ~4 hrs: exogenous glucose, all tissues use glucose; 4~16 hrs: from glycogen, then hepatic gluconeogenesis; glucose used by all, but muscle & adipocytes use less; 16~day 2: more hepatic gluconeogenesis than glycogen, liver doesn't use glucose; adipocytes & muscle use even less glucose; d 2~24: hepatic and renal gluconeogensis, glucose & ketone bodies used by brain, rbcs, renal medulla, small amt by muscle; d24 and on: hepatic & renal gluconeogen, brain, rbcs, renal medulla, more ketone bodies than glucose
from glutamine, how is it introduced to TCA, and in what organ?
in kidney; glutamine -> deaminated to glutamate -> further deaminated & dehydrogenizd (NADH) to alpha-ketoglutarate
PKA activity; where does it happen w/ what hormones? Which pathways does it affect?
glucagon & epinephrine actvates PKA in the liver (no glucagon affect in muscle); PKA increases activity of glycogenolysis & gluconeogenesis; decreases activity of glycolysis & lipogenesis
fasting state, what happens in liver
glycogenolysis; gluconeogenesis; beta oxidation from FFA from adipocyte; AA -> pyruvate (gluconeogensis & acetyl coA) & TCA; acetyl coA -> ketone
fasting state, what happens in adipocyte
TAG -> FFA + glycerol; FFA -> TCA in the cell, and ship to liver; glycerol ship to liver
fasting state, what happens in muscle
FFA from adipocyte -> TCA; ketone bodies from liver -> TCA; protein -> ship AA to liver
what hormones during fasting?
cortisol, epinephrine, glucagon, little bit of insulin
How many energy products per TCA cycle? (from one acetyl coA)
3x NADH, 1xFADH2, 1xGTP
what enzyme is embedded in TCA cycle? What does it do?
succinate dehydrogenase; complex 2 in ETC; oxidizes FADH2
which enzymes produce what energy products in TCA cylce? Name the substrate & product of the enzyme
isocitrate dehydrogenase creates NADH (isocitrate -> oxalosuccinate); alpha-ketoglutarate dehydrogenase creates NADH (alpha-ketoglutarate -> succinyl coA); succinyl CoA synthase creates GTP (succinyl CoA -> succinate); succinate dehydrogenase creates FADH2 (succinate -> fumarate); malate dehydrogenase creates NADH (L-malate -> oxaloacetate)
talk about anaplerosis, open & closed cycle; in which tissues?
creation of TCA cycle itnermediates; high in liver & muscle where cycle is open; low in most (closed)
examples of anaplerosis; enzyme, what organ, regulation
pyruvate carboxylase for pyruvate -> OAA; most tissues; biotin, ATP, CO2 needed, activated by acetyl CoA;; glutamate dehydrogenase: glutamate -> alpha-ketoglutarate in liver; ribose-5P + aspartate -> fumarate + ammonia, part of pure purine synthesis;; aspartate -> OAA via AST;
which two AA's are ketogenic?
leucine & lysine
what happens when PDH deficient? Treatment?
excess pyruvate (to restore NAD+) -> lactic acid; congenital lactic acidosis; neurological defects; tx: high fat foods of ketogenic nutrients to enter TCA w/o using PDH, oral citrate supplement;
what other cuase of PDH deficiency besides congenital?
TPP deficiency in alcoholics
There are two types of GLUT we covered. Which GLUT are there and in which organs?
GLUT4 - major transporter in skeletal m.; GLUT2 - liver, pancreas
what's special about RBC?
converts 1,3BPG to 2,3BPG; fast turnover; very dependent on glycolysis & PPP
compare & contrast GK & HK
HK has lower Km and Vmax. Non-cooperative. Inhibited by G6P; GK has higher Km & Vmax (to prevent hyperglycemia). Activated by glucose, inhibited by F6P. Insulin stimulates gene expression of GK
what can pyruvate become?
lactate, acetyl coA, OAA, alanine
gluconeogenesis: sources of carbon in liver
alanine, glutamine most common; glycerol, lactate
what pathways does fructose go into?
non-insulin dependent entry; enter glycolysis as DHAP or G3P. Also can go to TAG synthesis as glycerol
what pathology of fructose can occur?
essential fructosuria: fructokinase problem; hereditary fructose intolerance: aldolase problem (accumulate as F1P); both: fructose in blood/ urine; low ATP -> inhibition of gluconeogensis/ glycogenolysis -> hypoglycemia; tx: remove fructose, sucrose, sorbitol from diet
what pathways does galactose go into?
non-insulin dependent entry; from UDP-galactose, can become lactose, UDP-glucose.
what pathology of galactose?
classic galactosemia: absence of galactose-1-P uridyltransferase -> can't become UDP-galactose; galactokinase deficiency: can't become galactose-1-P; both: too much galactose -> become galactitol: galactose appear in blood/ urine; galacitol in eye (cataract), failure to thrive; tx: no galactose & lactose in diet
general construction of lipoprotein
core TAG, cholesterol esters; shell of apoproteins, phospholipids, cholesterol
essential fatty acids. Pathology for deficiency.
linoleic & alpha-linolenic acid; scaly dermatitis.
destination of TAG's constituents during fasting: organs, pathways
beta-oxidation & TCA from nearly all tissues; ketone bodies in liver
what organ, part of the cell does fatty acid synthesis occur? What are required to run this?
in liver, mammary glands, adipocytes; cytosol; acetyl coA, ATP, NADPH req
transport of acetyl coA to cytosol for fatty acid synthesis. Name enzyme used. What happens to the other product? Its significance?
acetyl coA + OAA -> citrate +coA ----> citrate + ATP + coA -> acetyl coA + OAA; via ATP citrate lyase; OAA (from cytosol) converts to malate then to pyruvate, pyruvate enters mitochondria; malate -> pyruvate creates NADPH.
rate limitting enzyme for fatty acid synthesis; substrate/ product; regulation; what diabetes drug interferes with this pathway?
acetyl coA carboxylase (ACC): acetyl coA -> malonyl coA; rate limiting; activated by citrate; inactivated by palmitoyl coA (end product) & AMP kinase (muscle) (phosphorylates ACC); glucagon & epinephrine activates PKA which activates AMPK; opposite for insulin; metformin also activates AMPK.; AMP also inactivates ACC allosterically. (low E -> should go TCA -> ETC); biotin & ATP required.
What is the key enzyme for fatty acid synthesis; one after ACC. Regulation. End product.
fatty acid synthase (FAS). Requires NADPH, which comes from PPP & malate-> pyruvate conversion. End product: palmitate..
what are substrates and products of ketone body synthesis? How is it carried in blood? Anything to note about urine test?
substrates: acetyl coA, ketogenic AA's, fatty acyl coA (beta oxidation); products: acetone (deadend) + 3-hydroxybutyrate; soluble in blood; urine test doesn't detect beta-hydroxybutyrate.
which organs use ketone body? Why not liver?
heart, muscle, brain; liver missing enzyme (3-ketoacyl coA transferase)
ketone synthesis' rate limiting enzyme. Its substrate/ product.
HMG coA synthase; substrate: acetoaccetyl coA, product: HMG coA.
how is FFA transported into mitochondria for beta ox? Which proteins are important? Regulation
long FFA: carnithine transport; short, mediam: no carrier; carnitine palmitoyl-transferase I (CPT1) in outer mitochondrial membrane; carnitine palmitoyl transferase II (CPT2) in inner mitochondrial membrane; basic idea: CPT1 removes coA from fatty acyl, adds carnitine; CPT2 removes carnithine and adds coA to fatty acyl; CPT aka CAT (carnitine acyltransferase); malonyl coA inhibits CPT1 (thus rate limiting)
explain beta ox once inside mitochondria
fatty acyl coA -> fatty acyl coA (2 carbons shorter) + acetyl coA; NADH and FADH2 produced. ; last fatty acid (3C long) (propionyl coA) metabolized to succinyl coA (biotin, vitamin b12 req)
what enzyme for TAG breakdown? Regulation?
lipase; covalent regulation: activation via adrenaline/ glucagon, inactivation by insulin
one pathology for beta ox
medium-chain fatty acyl coA dehydrogenase deficiency; can't oxidize these FFAs. Dx: increased in urine; hypoglycemia (these tissues take in more glucose); tx: avoid fasting, carnitine suplpementation
role of branching in glycogen; what linkage is considered branching?
alpha 1->6; branching makes glycogen more soluble; accelerates rate of glycogen synthesis.
what is limiting enzyme for glycogen synthesis; what linkage created? What is substrate
glycogen synthase; creates alpha(1->4) linkage; substrate: UDP-glucose
how are alpha(1-6) and alpha(1-4) bonds lyased in glycogen? Which is rate limiting?
glycogen phosphorylase (rate limiting) breaks alpha(1->4) linkage; 4:4 transferase removes outer three of four residues for alpha(1-6) branch; 1:6 glucosidase removes the last one on alpha(1-6) branch.
name one glycogen storage disorder and mechanism. Treatment.
mcardle's disease: glycogen phosphorylase deficient, too much glycogen, diminished exercise tolerance. Tx: sucrose supplementation, aerobic exercise (non-glucose TCA cycle) w/ creatine & vitamin B6.
branched AA's
leucine, isoleucine, valine.
10 essential AA's; characteristic?
PVT TIM HALL; phenylalanine, valine, tryptophan, threonine, isoleucine, methionine, histidine, arginine, leucine, lysine; no acidic; all branched, basic AAs included.
what is coenzyme for aminotransferase?
pyridoxal phosphate (vitamin b6 derivative)
what two AA's not transaminated?
lysine, threonine
how is redox and transamination related?
deamination is oxidative, while amination is reductive; NADPH/NADP+ or NADH/NAD+ used
explain transport of ammonia. From where to where? What happens once arrives in the destination?
glutamine: non-toxic transport, from anywhere to liver, produces glutamate; forms alanine from muscle, shipped to liver, transaminated back to pyruvate. Ammonia then turns to urea.
what's special about branched chain AA? Connection to pathology and vitamin?
different enzymes. Oxidative decarboxylation via branched-chain alpha-ketoacid dehydrogenase (BKAD); require TPP. Subsequent step also requires biotin and vitamin b12. Defect in BKAD -> maple syrup urine disease; accumulation of branched alpha-ketoacid in urine -> sweet odor -> CNS defects
two one-carbon carriers; their sigficance
THF & SAM; THF derived from folic acid. Folic acid -> THF requires NADPH; conversion from N5-CH3-THF to THF requires b12; THF then aids in methionine production. THF also produced from bacteria; different DHFR enzyme, so inhibitor of bacteria -> antibiotics, inhibitor of human -> chemotherapy; SAM synthesized from ATP & methionine.
list some products from AA metabolism
catecholamines (from tyrosine, which is from phenylalanine); NO from arginine; histamine from histidine
Name most common error in AA metabolism & its mechanism
phenylketonuria (PKU); defect in conversion phenylalanine -> tyrosine; build up of phenylalanine, deficiency of tyrosine; urine: musty odor, elevated phenylalanine level, melanin deficiency (tyrosine) -> hypopigmentation.; tx: restrict phenylalanine, replace tyrosine.
what's an important pathology concerning pyrimidine synthesis? Its significance?
orotic aciduria; inability to convert orotic acid to UMP; megaloblastic anemia (like b12/ thymidine deficiency), but doesn't improve w/ vitamin supplementation; no hyperammonemia like OTC deficiency; tx: uridine supplement
connection between PPP & purine biosynthesis
PPP's product ribulose-5-P -> ribose-5-P (substrate of purine biosyn)
explain mycophenolic acid's role (purine synthesis)
reversible inhibitor of IMP dehydrogenase. Deprives T & B cells of key components of nucleic acid; immune suppressants; to prevent graft rejection
explain methotrexate's role (purine synthesis)
inhibit reduction of DHF -> THF. Slow down DNA replication; chemotherapy
explain allopurinol's purpose and mechanism
treatment option for gout; inhibits xanthine oxidase; xanthine more soluble than uric acid
explain adenosine deaminase deficiency
excess ATP/ dATP inhibits ribonucleotide reductase -> prevent DNA synthesis; inability to complete DNA synthesis in B & T cells -> Severe Combined Immunodeficiency Disease
explain Lesch-Nyhan syndrome
deficiency of HGPRT; can't salvage hypoxanthine/ guanine; elevated PRPP, decreased IMP, GMP -> elevated purine biosynthesis -> gout; tx: allopurinol
How is uric acid secretion modulated in kidney? Examples of the one that promotes reabsorption?
uricosuric: inhibit URAT1 -> secretion; antiuricosuric -> keep uric acid; ex: lactate, nicotinate, pyrazinoate
general cause of hyperuricemia & treatment
too much synthesis of uric acid or too few excretion of it; allopurinol to reduce synthesis; uricosurics to underexcretors; anti-inflammatory drugs for gout.
what are probenecid or sulfinpyrazone. Which mediator does it work w/?
uricosurics; URAT1.
explain cori cycle
lactate released by skeletal muscle & RBCs -> turned to glucose by liver -> glucose goes to muscle & RBCs; net loss of 4 ATPs/ cycle
EtOH's impact on biochem
Too much NADH -> pyruvate and OAA are both gluconeogenic intermediates, but now they're being diverted to make NAD+ -> decreased gluconeogenesis, TCA cycle inhibited, increase in ketone bodies
sources of two ammonia from urea cycle, starting nw/ glutamine
one from oxidative deamination of glutamate by glutamate dehydrogenase & another from transamination of OAA by AST
what is OTC deficiency? Tx?
one of urea cycle disorders. One of the enzymes missing. High ortic acid in blood/ urine. Hyperammonemia; restriction of protein intake;
dietary treatment to newborns of urea cycle disorder
IV dextrose
phenylbutyrate & benzoate's role
in urea cycle disorder: alternative route for nitrogen disposal;
describe hemoglobin; describe type A & F; describe difference.
4 polypeptide subunits; each binds heme moiety; cooperative binding; 2 forms: Tense (deoxy)/Relaxed(oxy); HbF: fetal, alpha & gamma, higher O2 affinity; HbA: alpha, beta; HbF has weaker binding of 2,3-BPG (which reduces O2 affinity)
explain bohr effect
decrease in pH, increase in temp -> reduces O2 affinity
role of 2,3-BPG
reduces O2 affinity; allows body to adjust to environment (anemia, altitude, hypoxia, doping)
saturation curves of myoglobin & hemoglobin -> advantage of sigmoid curve
not too adherent, not too loosely adherent for O2 = just right; too adherent -> can't give O2 to tissue; too weak -> gives O2 to any tissue, not the ones that are deoxygenated; this precision is required to accommodate the difference in partial pressure of oxygen from lung & peripheral tissues are different;
mechanism of CO poisoning
binds to iron, increases affinity for oxygen; unable to release O2; no O2 -> inhibition of complex 4 for ETC
what forces run the ETC; products of ETC.
pH gradient (hydrogen in intermembrane space) & electrical potential (negative in matrix); ATP, CO2, H2O
what are two mobile e- carriers in ETC?
CoQ & cytochrome c
steps and constituents of ETC.
1 (NADH dehydrogenase), 2(succinate dehydrogenase - FADH2) -> CoQ -> 3 -> cyt c -> 4; ATP synthase/ ATPase (F0, F1)
Describe ATP synthase of ETC
F0 (transmembrane); F1: cytosolic; F1 rotates, changing from open, loose, tight (catalytic) conformation; F0 pump H+ down the gradient to fuel ATP synthesis.
what is 2,4-dinitrophenol
synthetic uncoupler. Creates proton leak w/o ATP formation; releases heat
mechanism of brown fat; regulation
beta oxidation -> TCA -> ETC; uncoupling via UCP-1 -> heat.; regulated by norepinephrine & thyroid hormone.
describe function of rotenone.
binds complex I of ETC -> no reduction of CoQ from complex 1. reduced efficiency. Complex 2 still functions.
explain apoenzyme, holoenzyme, cofactor, coenzyme,
holoenzyme: enzyme w/ non-protein part as activator; apoenzyme: holoenzyme w/o its activator; cofactor: metal ion serving as activator; coenzyme: small organic factor like vitamin as activator
explain cosubstrate, prosthetic group, synthetase, synthase
cosubstrate: coenzyme transiently associated; prosthetic group: coenzyme permanently associated; synthetase: requires ATP; synthase: doesn't require ATP
describe carboxylase
adds 1 carbon w/ help of biotin
factors affecting enzyme rxn velocity
temp: increase T -> increased rxn, until it is too high -> denature; pH: can change ionization of AA residues, extreme change -> denature
explain steady state assumption
[ES] doesn't change; same rate of formation as breakdown.
explain the affect of competitive, un-competitive, noncompetitive inhibitors
competitive: increases Km; un-competitive: decreased V max and Km; non-competitive: decreased V max
what is collagen made of, what structure, constituents
glycoproteins; triple alpha-helix; 1/3: glycine, 1/3: proline, 1/3: hydroxylated proline & lysine
regulation of collagen synthesis; possible pathology w/o the cofactor
vitamin c to hydroxylate proline/ lysine -> no H bond; deficient -> scurvy's;
describe osteogenesis imperfecta: etiology, sx, dx
point mutation in COL1A1 or COL1A2 genes (type 1 collagen); affect glycine residue; heterozygous -> mixture of abnormal collagen; sx: blue-greyish sclera (blue choroid shown), weakened teeth due to lack of dentin, hearing impairment, brittle bone; dx: skin bx to check fibroblasts, genomic DNA from WBCs to see mutation
describe pamidronate for collagen synthesis
tx for osteogenesis imperfecta; anti-resorptive agent for calcium; inhibit osteoclast activity
describe difference between point mutation & nonsense mutation for osteogenesis imperfecta
point mutation -> mixture of bad collagen; stop codon: reduced normal, only normal is translated.
describe folic acid deficiency
neural tube defects; THF -> methionine -> purine & pyrimidine synthesis; path -> megaloblastic anemia w/o neurological sx;
Vitamin b12 deficiency (cobalamin)
THF for homocysteine -> methionine -> megablastic anemia; odd FA accumulation -> neurologic sx
thiamine (b1) deficiency
as TPP, coenzyme in formation/ degradation of alpha-ketols, PPP, PDC, BCAA, alpha-ketoglutarate dehydrogenase complex; path: beriberi, Wernicke-Korsakoff Syndrome; reduced ATP production and accumulation of ketoacids
vitamin c deficiency
collagen synthesis, iron absorption, norepinephrine synthesis; pathology: scurvy
vitamin d defiency
regulate calcium; rickets (bending bone)
statin drug: mechanism
targets HMG-CoA reductase as competitive inhibitors
to differentiate between b12 & folic acid deficiency; pathophysio for that sx
neuopathy only w/ b12 deficiency; propionyl coA stuck in membrane because b12 also needed for propionnyl coA -> succinoyl coA
exact reasons for megaloblastic anemia
increased homocysteine level, reduced emthionine level, impaired formation of THF -> inadequate conversion of deoxyuridylate -> thymidylate -> slow DNA synthesis & nuclear maturation
distinguish anaplerosis/ cataplerosis
anaplerosis: refilling intermediate to TCA; cataplerosis: removing intermediate for biosynthetic purpose (gluconeogenesis, FA synthesis, AA synthesis)
glycolysis: liver vs. muscle
liver: only during fed, gluconeogenesis in fast; muscle: glycolysis for E. fast and fed; PFK-2 regulation different, spc isoform; liver: PFK2 inhibited by cAMP-dependent phosphorylation when insulin is low; not in muscle; Exercise in muscle: ca2+ -> glycogen phosphorylase kinase, ADP/ ATP -> glycogen phosphorylase, AMP -> allosteric activate PFK1 -> more F1,6BP -> allosteric activate PK, Ca2+ -> activate PDH
how can much THF mask b12 deficiency? What metabolic reaction is involved?
while N5-methyl THF -> THF requires B12, when folic acid is high, folic acid -> directly THF by getting dehydrogenated by NADPH; THF is ultimately used to convert homocystine -> methionine
Purpose of DHFR
bacteria: make THF; humans: one of two enzymes to make THF
trimethoprim
inhibit bacterial DHFR
methotrexate
inhibit mammalian DHFR; selective for rapidly dividing cells; chemo
consequence of medium chain acyl co A dehydrogenase deficiency; tx
limited beta-oxidation -> not as efficient in deriving energy; prolonged fasting is dangerous; more carb/ protein based diet, avoid fasting; carnithin supplement, because these are used in these patients to excrete medium chain fat via urine
consequences of atkin's diet
low insulin, low glycogen -> gluconeogenesis, fat used by muscle, FA used by liver to make ketones