• 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

1/599

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;

599 Cards in this Set

  • Front
  • Back
Name the four components of the amino acid.
1. Amino group
2. Carboxyl group
3. Hydrogen atom
4. Unique R-group (side chain)
What type of chirality do amino acid enantiomers exhibit?
L-enantiomers, D-enantiomers do not exist naturally.
What is the CORN rule?
If the amino acid is oriented with the hydrogen atom sticking out towards you - if the groups can be read (clockwise) as CORN (CO group, R-group, and N group) - the the amino acid is L-amino.
What are zwitterions?
Electrically neutral molecules that contain one positive charge (of the amine group) and one negative charge (of the carboxyl group)
How does pH and H+ relate?
pH = -log[H+]
What is the equation of pKa?
pKa = log{([H+][A-])/[HA]}
What is the Henderson-Hasselbach equation?
pH = pKa + log([A-]/[HA])
Buffer solutions can buffer what range of pH's?
Above and below 1 pH of the pKa (+/- 1)
Define what the pI is?
The pH at which a molecule has no net charge.
How do you calculate the pI?
1. Figure out which pKa yields the neutral molecule.
2. Figure out which pKa yields the negative molecule.
3. Calculate the average of the two.
Name all the aliphatic amino acids (6 total).
Glycine, alanine, valine, leucine, isoleucine, proline
Which amino acid is achiral?
Glycine.
Which amino acid is typically seen contributing to sharp turns in amino acid structures?
Prolline residues (due to the cyclic structure which promotes turn and provides rigidity in structures)
Name all the aromatic amino acids (3 total).
Phenylalanine, Tyrosine, Tryptophan
Which amino acid occurs the least naturally?
Tryptophan (1.1%)
G, Gly
Glycine
V, Val
Valine
A, Ala
Alanine
L, Leu
Leucine
I, Ile
Isoleucine
P, Pro
Proline
F, Phe
Phenylalanine
W, Trp
Tryptophan
Y, Tyr
Tyrosine
S, Ser
Serine
T, Thr
Threonine
N, Asn
Asparagine
Q, Gln
Glutamine
C, Cys
Cysteine
M, Met
Methionine
D, Asp
Aspartate
E, Glu
Glutamate
H, His
Histidine
K, Lys
Lysine
R, Arg
Arginine
Which two amino acids contain hydroxyl groups?
Serine, Threonine
Name all the polar but uncharged amino acids (4 total)
Serine, threonine, asparagine, glutamine
Which two amino acids have amide groups?
Asparagine, glutamine [analogs of aspartate an glutamate]
What are the two sulfur-containing amino acids?
Cysteine and methionine
Which amino acid participates in forming disulfide bridges?
Cysteine
Which amino acids have a net negative charge at physiological pH?
Aspartate and glutamate
Which amino acids have a net positive charge at physiological pH?
Histidine, Lysine, Arginine
Which amino acids are hydrophilic?
Asparagine, Glutamine, Aspartate, Glutamate, Histidine, Lysine, Arginine [N, Q, D, E, H, K, R]
Which amino acids are hydrophobic?
Valine, Leucine, Isoleucine, Phenylalanine, Methionine
[V, L, I, F, M]
Which amino acids absorb UV light?
All aromatic amino acids, F, Y, and W
{phenylalanine, tyrosine, tryptophan}
Which three amino acids can be phosphorylated (contain -OH groups)?
Serine, threonine, Tyrosine [S, T, Y]
Which amino acids have TWO chiral centers?
Isoleucine and threonine
Which two amino acids form O-glycosidic bonds?
Serine and threonine
Which amino acid can form N-glycosidic bonds?
Asparagine
What are non-standard amino acids?
Amino acids not encoded by the genetic codes and not used for the synthesis of protein.
Which non-standard amino acids are critical to the structure of collagen?
3 and 4-Hydroxyproline and 5-hydroxylysine.
Which non-standard amino acid is critical to the structure of elastin?
Desmosine.
Explain the symptoms of scurvy?
In the lack of Vitamin C - collagen cannot appropriately form (due to dysfunctionality of hydroxyprolines) - thus collagen breaks down (bloody gums and spotting on the skin)
What is the crucial coenzyme required for functionality of hydroxprolines?
Vitamin C
What provides the flexibility of the peptide bonds?
Rotations around the C-Ca [psi bond] bond and the C-N [chi bond]
What provides the rigidity of the peptide bonds?
Planar unit [in order from N-C] between C_alpha, carbonyl, amino group, and C_alpha of next amino acid residue
What can limit the flexibility of the peptide bonds?
Steric hindrance of R-group (i.e. tryptophan, tyrosine, or histidine)
How do you write out an amino acid sequence?
From N-terminal to C-terminal
What determines the acid/base characterization of a peptide?
Ionizing groups located on the R-groups of each amino acid residue.
What determines the 3-D structures of proteins?
The primary structure (folding begins before the protein completes translation) - the interactions between the side chains determine the 3-D structure.
What are the two types of secondary structures?
Alpha-helix and Beta-pleated sheets
Where do the hydrogen bonds occur in a alpha helix?
Between the n and n+4 amino acid residue - between the oxygen of the carbonyl group (n residue) and the hydrogen off the amine (n+4 residue)
Is the alpha-helix left or right handed spiral?
Right-handed.
What is the overall dipole moment of a alpha helix?
Negative on the C-terminal side and positive on the N-terminal side.
Describe amphipathic alpha helices?
R-groups are hydrophobic on one side (exposed to lipids or other hydrophobic residues) and hydrophilic on the other side (exposed to water)
What are the three configurations of the Beta-pleated sheet? And how do you determine what configuration it is in?
Parallel (chains all travel in the same direction - i.e. N --> C or C-->N)
Antiparallel (alternating chains, one starts with C the other with N)
Mixed (mix of both antiparallel and parallel)
What contributes to the partial double bond characteristic of the carbonyl carbon and nitrogen atom?
The resonance between the free electrons on the nitrogen and the carbonyl double bond (this also allows for rigidity of planar peptide bond)
All proteins have a _____ structure, but not necessarily a _____ structure. An example of such a protein is ______.
Tertiary; quarternary; myoglobin.
What is the purpose of super-secondary structures?
Many times, the supersecondary structures (a.k.a motifs) provide important binding function (i.e. active sites of enzymes).
What supersecondary structures typically form the stable core of many proteins?
Beta-barrels and and saddles.
What type of loops occur in proteins (for supersecondary structures)? Right-handed or left-handed?
Right-handed.
What kind of COVALENT bonds are present in TERTIARY structures?
Disulfide bonds between cysteine residues.
Define the domain of a protein?
Unit of the tertiary structure that has a functional purpose (i.e. G3P binding domain of G3P dehydrogenase)
How are quarternary structures held together?
Non-covalent interactions only!
What are chaperones?
"Heat shock" proteins that assist in the proper folding of proteins.
Once denatured, how are proteins renatured?
As long as the primary amino acid sequence is still intact, renaturation can occur as long as the denaturing agent is gone.
How is amyloid plaque formed?
Proteolytic cleavage of amyloid (protein found on cell surfaces in the brain) into 40-42 amino acid residues that form a Beta-pleated sheet.
What type of amyloid plaque is formed in Alzheimers?
A-Beta (amyloid beta)
How does the "tau" protein play a role in alzheimer's?
Normal tau protein helps form microtubular structures; abnormal tau protein thus promote accumulation of neurfibrillary tangles in the brain.
How does prion disease progress?
Infective agent sets up a template to convert normal PrP to infective PrP.
What exactly is the change to create infectious PrP?
alpha-helices converted to Beta-sheets (resists proteolytic degradation by the body)
What are the two classification of proteins based on their folding patterns?
Globular and fibrous proteins.
Describe the structure of alpha-keratin?
R-handed alpha helices that combine into protofilaments and protofibrils cross-linked by disulfide bonds.
How does permanent waving at a salon linked to keratin?
Hair is made up of keratin - thus when applying heat to the hair the disulfide bonds are broken and then reformed in a different form (keeping hair curled).
All fibrous proteins are _______ in water.
insoluble.
Describe the structure of collagen?
Rich in glycine/proline and provide structural components to bone/teeth/cartilage/tendons/skin/blood vessels
What is the repetitive amino acid sequence in collagen?
Gly-X-Y.

X = proline
Y = hydroxyproline
Lack of vitamin C leads to....
Scurvy
Explain the clinical symptoms of scurvy?
Lack of vitamin C inhibits formation of hydroxyprolines which are critical to structure of collagen (thus blood vessels become fragile, wounds do not heal well, and skin easily hemorrhages).
What are the steps in collagen synthesis?
1. cleavage of signal peptides.
2. hydroxylation of prolines and lysine residues.
3. glycosylation of some hydroxylysine and asparagine residues.
4. association of three C propeptides
5. disulfide bridges form
What is tropocollagen?
Three left-handed alpha-chains twisted around each other in a right-handed manner (forms a larger helix)
Why is glycine critical to collagen structure?
Glycine is the smallest amino acid residue, thus allows for tight turning of collagen.
What causes the reduced muscular flexibility and agility in the elderly?
Additional covalent-crosslinks in tropocollagen
What is lathyrism?
Deformed bones, joint dislocations, aortic aneurysms (plant toxin inhibits lysyl oxidase which forms defective tropocollagen).
Why can lathyrism be confused with cupper-deficiency?
Cupper is required for lysyl oxidase - thus the abnormality of this enzyme will induce similar clinical symptoms.
Which type of collagen mutation is lethal?
Point mutation in Type II procollagen - leads to osteogenesis imperfecta
What is Ehler-danlos syndrome?
Errors in processing Type I, III, IV procollagen - hypermobility and skin hyperextensibility.
What role does desmosine play in elastin?
Links different elastin fibers (helps control the stretched/relaxed positions of elastin
What is the key structural difference (in terms of amino acid composition) between elastin and collagen?
Elastin LACKS hydroxyprolines (but has glycine/proline residues)
What role does alpha-antitrypsin play in emphysema?
This glycoprotein inhibits elastase (which breaks down elastin) - in deficiencies of this glycoprotein, elastase levels are high and break down the elasticity of lung tissue causing emphysema.
SDS-PAGE separates proteins according to _____.
Size.
Gel electrophoresis separates protein according to _____.
Charge.
Isoelectric focusing separates proteins according to ______.
pI value (the gel is in a pH scale)
Two-dimensional gel electrophoresis separates proteins according to _____ first then by ______.
Isoelectric focusing, SDS page
(pI, then size)
What is the first step of tissue preparation?
Fixation (the processes and structures of that cell must be preserved to give an accurate description).
Once fixated, what is the next step in tissue preparation?
Dehydration - removal of water from the sample and subsequent replacement with xylene.
What is the critical last step of tissue preparation?
Staining - allows for different areas of the cell to become prominent (most common staining, H&E)
What regions does hematoxylin stain?
Basic blue stain, labels acidic regions (i.e. DNA/RNA)
What regions does eosin stain?
Acidic pink stain, labels basic regions of the cell (i.e. cytoplasm or extracellular matrix)
What does the periodic acid-Schiff (PAS) reaction stain emphasize?
Carboyhydrates (glycogen/mucus/basement membrane)
Which of the following structures are associated with nuclear lamins [microfilaments, intermediate filaments, plasma membrane, rough ER]
Intermediate filaments
Which of the following is not found in lysosomes (nucleases, lipases, catalses, OR acid phosphatase)
Catalase
The electron microscopic appearance of plasma membrane is best described as?
Trilaminar structure.
What is the most abundant organelle in the cell (aside from the cytosol)?
Mitochondria
How do molecules/substances shift from the nucleus to the cytosol?
Via nuclear pores.
What is euchromatin?
Loosely packed DNA that is transcriptionally active.
What is heterochromatin?
Tightly packed DNA that is transcriptionally inactive.
What is unique about the cellular division of platelet-producing megakarocytes of the bone marrow?
Genetic material is replicated but without cell division.
Mitochondrial DNA differs from nuclear DNA in that it is _______ and replicates via _________.
Circular, fission/fusion.
The smooth endoplasmic reticulum is responsible for synthesis of _______ and ____ of compounds.
steroid synthesis and detoxification of compounds (liver)
The entry side of golgi apparatus is known as ____- side and the exit side is called the _____-side.
cis/trans
What is the main function of the golgi apparatus?
Post-translational modification and packaging of macromolecules/proteins.
The environment within a lysozome is (acidic/basic) and therefore the enzymes within are known as ___________.
acidic; acid hydrolases
What is the cellular explanation of Tay-sachs Disease?
Lysosomal storage disease - hexosaminidase is deficient and therefore there is an accumulation of gangliosides in the CNS.
Endosomes are divided into _____ and ______ endosomes based on their location.
early (by plasma membrane), late (by cell center)
What is the cellular explanation of familial hypercholesterolemia?
LDL-receptor is mutated and cannot accept LDL (receptor-mediated endocytosis) thus leading to high levels of LDL in the blood.
Catalase and oxidative enzymes are found in what cellular organelle?
Peroxisomes.
What allows the detoxification of hydrogen peroxide in the cell?
Catalase.
What is the cellular explanation of Zellwegger disease?
Peroxisomal enzymes cannot cross the preoxisomal membrane - thus leading to childhood death.
The energy stores of the cell are found in the _____ and are typically stored as ____ droplets and _____ deposits.
cytosol; lipid; glycogen
The cytoskeleton is divided into three types of fibers which are....
Actin filaments, intermediate filaments, and microtubules
Actin filaments provide what type of cellular movements?
Crawling, contraction/pinching of the two daughter cells during mitosis, also compose part of microvilli in intestine
Where is the highest concentration of actin located?
Beneath the plasma membrane in a region of cells known as the CORTEX.
What is the main function of intermediate filaments?
Great tensile/mechanical stress that distributes mechanical stress across the cell
Intermediate filaments are classified into four different categories, which are....
1. Keratin
2. Vimentin
3. Neurofilaments
4. Nuclear lamins
Where do microtubules originate from?
Centrosomes, the gamma-tubulin ring
Describe the structure of microtubules?
Heterodimers made up of alpha and beta tubulin.
Motor protiens move cargo based on the polarized structure of mirotubules that contain a ____ end and a _____ end.
minus; plus.
Kinesins move in the _____ direction and dyneins move in the _____ direction.
plus; minus
Cilia and flagella have a unique microtubuel structure which is....
9+2 arrangement of microtubules.
What allows the lipid bilayer to aggregate spontaneously?
Hydrophobic tails congregate together (to avoid water) while the hydrophilic heads face out towards either the extracellular area or internally to the cell.
Which of the following four major phospholipids contain a negative charge? (Phosphatidylcholine, phophatidylethanolamine, phosphatidylserine, sphingomeylin)
Phosphatidylserine (PS)
Which phospholipid is used for cellular signaling?
Phosphatidylinositol (PI)
What restricts the mobility/fluidity of the cell membrane?
Cholesterol
Which of the following phospholipids are found on the cytosolic leaflet of the membrane - and what does this indicate? [PI, PS, PC, PE, SM]
PS and PI are on the cytosolic leaflet (thus the inside is negatively charged)
What are the function of lipid rafts?
Clusters of sphingolipids and cholesterol - thickening of the plasma membrane that typically contain proteins for cell signaling.
What type of molecules can transmit through lipid bilayers?
Small, non-polar, lipid soluble, hydrophobic molecules.
What are the two types of membrane proteins?
Integral and peripheral proteins.
How are peripheral proteins attached?
Via non-covalent binding to integral membrane proteins.
Transmembrane proteins contain both ______ and ______ regions.
hydrophobic; hydrophilic
Which type of secondary protein structure is typically found in membrane proteins?
alpha-helices
Due to the rigidity of Beta-barrels - these structures are typically confined to ________.
Mitochondrial membranes.
Glycoproteins are contained only on the _____ surface of the cell.
extracellular
What is the least likely movement of the lipid bilayer?
Flip-flop (where the cytosolic layer becomes the extracellular layer)
If a co-factor is covalently linked to an enzyme it is a ______.
Prosthetic group.
If a co-factor is loosely bound to the enzyme it is a _____.
Co-substrate.
What is a co-factor?
Non-protein containing component of the enzyme.
What is an apoenzyme?
Protein-containing component of the enzyme.
What is the function of oxidoreductases?
NAD+, FAD+, dehydrogenases, or oxidases -- redox reactions!
What is the function of transferases?
Transfer functional groups (hexokinase/glucokinase transfer phosphate group)
What is the function of hydrolases?
Enzyme that breaks bonds via hydrolysis (H20) [e.g. peptidases]
What is the function of lyases?
Forming or breaking double bonds (decarboxylases) -- splits one compound into two.
What is the function of isomerase?
Isomerization of molecules (mutase or isomerase)
What is the function of ligase?
Joins substituents at the EXPENSE OF ATP (carboxylase)
How do enzymes function?
They lower the activation energy.
Does the free energy of the reactants or products change in a catalyzed reaction?
No.
What is an endergonic reaction?
The free energy of the products are higher than the free energy of the reactants [non-spontaneous, del G >0]
What is an exergonic reaction?
The free energy of the products is lower than the free energy of the reactants [spontaneous, del G <0]
What is a catalytic group?
Specific amino acid residues (typically far apart) that come together in the active site to directly interact with the substrate.
Identify the catalytic triad for chymotrypsin.
Histidine (57), Aspartate (102), Serine (195)
What is the purpose of transition state analogs?
Inhibit the forward reaction of an enzyme and also help understand enzyme-catalyzed reactions (research)
What is the vitamin derivative of thiamine?
B1
Vitamin B2 is associated with ...
Riboflavin
Vitamin B3 is associated with ...
Niacin
What is the vitamin derivative of pantothenic acid?
B5
Vitamin B6 is associated with ...
Pyridoxine
What is the vitamin derivative of cobalamin?
B12
Tetrahydrofolate is a cosubstrate that derives from....
Folic acid.
What is the function of biotin in teh body?
Prosthetic group that helps transfer CO2 [ATP-dependent]
What is the function of Thiamin pyrophosphate (TPP)?
Derived from B1, transfers aldehyde - acts as a prosthetic group to enzymes like pyruvate dehydrogenase
What is the two main differences between FAD and NAD?
FAD can be reduced to FADH2 while NAD can be reduced to NADH only.

FAD is a prosthetic group, while NAD is a cosubstrate.
Aside from FAD, what other coenzyme is produced from B2 (riboflavin)?
FMN
What is the main difference between NADH and NADPH?
NAD is utilized in catabolic processes, while NADP is utilized in anabolic processes.
How is NAD regenerated once reduced?
Via FAD.
What is the function of tetrahydrofolate (THF)?
Transfers one-carbon groups.
What is the purpose of cobalamin?
Transfers methyl groups.
Define what V-max is?
Maximum rate of the enzymatic reaction.
Define what K-m is?
The substrate concentration that yields 1/2(V-max)
What are the axes for the Michaelis-Mentel model graph?
x-axis is [S] ... mol/L
y-axis is v (reaction rate) ... mol/sec
What is the enzyme turnover number?
V-max
How can you calculate V-max without a graph?
V-max = k3 * [E_total]
How do you calculate the fraction of the enzyme occupied by the substrate?
Rearrange M-M equation:
v/Vmax = [S]/(K-m + [S])
What is unique about the pancreatic enzymes and their activation?
In a proenzyme (zymogen) form until activated by trypsin which was originally activated by enteropeptidase (released by intestines)
What is the main difference between competitive and non-competitive inhibitions?
Competitive inhibitions interacts with the active site while non-competitive inhibitions interacts with a non-active site to induce changes at the active site.
What changes occur in the Michaelis-Menten model with a competitive inhibitor?
Increase in K-m, V-max stays the same.
What changes occur in the Michaelis-Menten model with a non-competitive inhibitor?
Decrease in V-max, K-m stays the same.
How can you determine by what amount an inhibitor is changing a variable in the Michaelis-Menten equation?
1+([I]/K-i)
[I] = concentration of inhibitor
K-i is like the K-m, but specific to the inhibitor
How do insecticides function?
Irreversible inhibitors of acetylcholinesterase (parathion is metabolized to paraoxon that cannot be metabolized by insects) - and thus functions as an inhibitor.
How do monoamine oxidases inhibitors function?
Irreversible inhibitors of monoamine oxidases preventing degradation of norepinephrine and DOPA [since irreversible, takes 1-2 weeks for new enzymes to regenerate]
How is disulfiram used in treatment of alcoholism?
Disulfiram is an irreversible inhibitor of ALDH (the 2nd step in alcohol metabolism) - thus worsening the symptoms/effects of ethanol on the body [preventing excess use of EtOH]
How is methanol or ethylene glycol poisoning remedied?
K-m for ethanol is lower than that for methanol/ethylene glycol - so ethanol is given as a competitive inhibitor.
How do statin drugs serve to lower cholesterol?
Competitive inhibitor of HMG-CoA Reductase which prevents the formation of cholesterol.
How does lead poisoning occur?
As lead enters the circulation - creates covalent bonds non-competitively - preventing function of whatever enzyme it has chosen to bind to.
How does ACE inhibitors work?
Prevent the conversion of angiotensin I to II - which is a potent vasoconstrictor (so keeps the blood vessels open).
How does the reaction rate curve change for allosteric enzymes?
Sigmoidal because subunits work with each other to bind substrate.
How does allosteric regulation work?
Inhibitor or activator will bind to one subunit which will also induce a change in the other subunit towards a certain equilibrium.
Allosteric inhibitors preferentially bind to ____ state while activators preferentially bind to ____ state.
Tense (T), Relaxed (R)
If an allosteric enzyme is activated, the curve will shift to the _____ while inhibition will cause the curve to shift to the ______.
Left (lower concentration needed for higher rate); Right (vice versa).
How many different forms of creatine kinase are there?
CK-M, CK-B, CK-MB
In case of a heart attack, what levels would you expect to change and how.
CK-MB elevates (increased permeability of the heart tissue due to necrosis), and LDH1 will increase (predominantely found in the heart)
In a normal person, LDH (lactate dehydrogenase) exists in _____ forms, where the most dominant is _____.
Five; LDH2.
What is an isoenzyme?
Different structure, same function.
What is the difference between glucokinase and hexokinase?
Glucokinase has a higher K-m than hexokinase since it is found in the liver - and the heart/muscle requires more glucose for conversion to ATP.

Also - hexokinase is inhibited by the product (glucose 6-phosphate) while glucokinase is not.
Coat Protein I and II are responsible for shuttling molecules between what to organelles?
Golgi body and rough endoplasmic reticulum.
Clathrin-coated vesicles facilitate the movement between what two organelles?
Golgi body and cell membrane.
If the protein coats (vesicle formation) on molecules are temporary - why are they necessary?
For the appropriate signaling and shuttling to the right destination.
Through what mechanism do vesicles know where to go?
SNAREs - v-SNARES (on the vesicles ) that connect with the t-SNARES (on the target membranes)
What is the function of Rab proteins and dynamin?
Help with the fusion/formation of the vesicle to the membrane (GTP-binding proteins)
What are the two non-receptor mediated forms of endocytosis?
Pinocytosis and phagocytosis.
What is the main difference between pinocytosis and receptor-mediated endocytosis?
Pinocytosis is "cellular drinking" ingesting ions/waters while receptor-mediated endocytosis is "cellular eating" ingesting solutions of molecules (still liquid)
What is phagocytosis?
Ingestion of particulate matter (actual solids) - such as deadened remains of a cell.
What is the first step in LDL uptake by a cell?
Clathrin-pit and vesicle formation on plasma membrane.
What is the critical step in LDL uptake that allows for the dissociation of the receptor and ligand?
Reduction of pH from 7 to 5.
What is the final step of LDL uptake? (discounting for recycling of receptor to plasma membrane)
Degradation by lysozome.
What allows the pH to be so low in the lysosomes?
Action of vacuolar H+ ATP-ase
What are the three mechanisms by which material is delivered to lysosomes?
Intracellular receptors signaling autophagy (in aging cells/malformed cells, etc.) or endocytosis (pinocytosis, receptor-mediated endocytosis, phagocytosis)
What is the pathology behind I-cell disease?
Lysosomal storage disease has dysfunction with mannose 6-phosphate (indicates which enzymes to send to lysosome) - thus lysosomes cannot function.
What is the difference between constitutive and regulated exocytosis?
Constitutive is constantly occurring (for lipids/proteins to the membrane that are constantly degraded/shifted), but regulated is signal-triggered.
Where do proteins go first once formulated in the rough endoplasmic reticulum?
Cis-golgi apparatus.
How do proteins enter the mitochondria?
First unfolded in the cytosol, shuttled across the membrane, and then re-folded.
Myoglobin is primarily used for ______.
Oxygen storage.
Hemoglobin is primarily used for ______.
Oxygen transport.
Does myglobin contain a quarternary structure?
No, it is a single polypeptide unit that binds a single O2.
What is the quarternary structure of hemoglobin?
Two alpha and beta dimers that form alpha-beta complexes (so two heterodimers)
Where is heme synthesized?
Succinyl CoA of the citric acid cycle (and requires biotin during synthesis)
What does oxygen specifically bind to in hemoglobin?
Iron within the porphyrin ring (T-state shifts to the R-state)
How does cooperative binding occur?
As oxygen binds to each subunit - the T state (deoxy) shifts to R state (oxy) which promotes the equilibrium shift towards oxygenated state.
How is the different dissociation curve shapes explained between hemoglobin and myoglobin?
Hemoglobin (sigmoidal) due to cooperative binding, and myoglobin (hyperbolic) only one unit that has very high affinity for oxygen.
How does 2,3-BPG function in RBC regulation?
Allosterically inhibits RBC by binding/stabilizing the T-state of the hemoglobin - leads to increased unloading oxygen in tissues.
What effect does low pH have on RBC?
T-state favors protonation - so decreased pH promotes T-state and increased unloading of oxygen in tissues.
Where is 2,3-BPG synthesized?
From the glycolytic pathway (intermediate after G3P has formed and reacts)
What effect does carbon dioxide have on hemoglobin?
Inhibits hemoglobin via carbamoylation - also accounts for 20% of removal of CO2 from the body.
What is the main difference between HbF and HbA?
HbF has a much higher affinity for oxygen (small percentage remains as an adult) - and because of this the fetus can get oxygen from maternal blood.
How many variations of alpha-thalassemia are there?
Four - since there are four total alpha genes (two per gene)
Due to defect in the alpha or beta genes in thalassemia - what kind of quarternary structures will develop?
Beta tetramers or alpha tetramers (or homodimers form) - body attempts to compensate for defects in subunits.
What clinical symptoms will occur in thalassemia?
Anemia (in more severe forms there is heart failure due to unoxygenated heart)
What is the coagulated form of hemoglobin in the blood called?
Heinz body.
What is the point mutation in sickle cell anemia?
Glutamate changes to valine at beta-6.
What can be an indicator of hyperglycemia in diabetic patients?
Hba1c (amount of glycosylated hemoglobin)
What causes the sickle shape in sickle-cell anemia?
Promotion of clumping in the deoxy state of hemoglobin (polymerization)
What is pharmacodynamics?
Effect of the drug on the body.
What is pharmacokinetics?
Effect of the body on the drug.
What is the first-pass effect?
The drug is metabolized by the liver into non-functioning metabolites to be excreted before it is absorbed by the GI tract.
Your body will digest drugs to make them more ________ and thus promote ______.
hydrophilic; excretion.
What is the most important mechanism for absorption of a drug into the body?
Diffusion via cell membranes (ergo, lipophilic substances)
Unionized forms of a drug are (more/less) likely to pass through membranes.
More.
What is the Henderson-Hasselbalch equation?
pH = pKa + log ([deprotonated]/[protonated])
If phenobarbitol (weak acid) has a pKa of 7.4, and the stomach has a pH of 2.4 - is the drug readily absorbed or excreted?
Readily absorbed because the protonated form is in a much larger amount than deprotonated.
Acids prefer to be _____ while bases prefer to be ______.
protonated; deprotonated....always prefer to be neutral.
How can you avoid the first pass effect?
IV injection, anal suppositories, sublingual adminstration.
Molecules that cannot cross the blood brain barrier have what characteristics?
Too charged, too large, and not lipid soluble.
What are P-glycoproteins?
Efflux transporters that remove hydrophobic substances from the cell? - it is a protective mechanism against toxicity.
How do you treat a morphine overdose?
Since morphine is a weak base, it is not readily absorbed by the stomach - therefore, gastric lavage techniques are used.
Why is it bad to chew medications that are not meant to be chewed?
Can result in toxic "dumping" of medication into the GI - because it does not follow regular digestion processes.
What do agonists do?
Drug that activates a receptor
What do antagonists do?
Prevents an agonist from activating receptor.
How do inert binding sites affect the bioavailability of a drug?
Drugs can bind to endogenous substances without inducing a response - but since they are no longer a free drug - cannot create a response anymore.
How can a partial agonist become an antagonist?
If the receptors are already activating, the partial agonist will compete for the same ligands but provide lower functionality.
How does Viagra (sildenafil) work?
Nitroglycerine (NO) inhibits phosphodiesterase which works to deactivate cGMP (without this, cGMP stays active and promotes relaxation of smooth muscle)
What is the EC50 value?
Half of the dosage at which maximal responsive was seen.
What is LD50 value?
Half of the dosage at which the maximal lethal/adverse side effects were seen.
How do you calculate the therapeutic index and how does this indicate the safety of the medicine?
LD50/ED50 = therapeutic index

The higher the number, the safer (the lethal dose is far higher than the effective dose)
What is the IC50 value?
In competitive ANTAGONISTS, you want to see the effective inhibition of some process (the 50% mark of maximal inhibition)
What is the difference between competitive and non-competitive antagonists?
Non-competitive antagonists are irreversible and the therapeutic response cannot be determined by the plasma levels of the drug.
What determines the effective duration of an irreversible antagonist?
The turnover rate of the receptor.
If bound irreversibly to the receptor, how are irreversible antagonists eventually cleared?
Via the regeneration of receptors by the cell.
What is the difference between potency and efficacy?
Potency is the ability to induce an effect at a lower dose - while efficacy is the ability to reach the maximal effect.
What is an inverse agonist?
Binds to the receptor in the inactive form (preferentially) therefore keeping most receptors inactive.
What is the margin of safety?
MS = LD1/ED99 (the larger the better)
How do you calculate the half-life?
Either graphically by seeing time it takes to get to 1/2 of initial concentration OR 0.693/k
How long does it take to reach steady-state for a drug?
5 half-lives.
Calculate the loading dose.
D_l = (C_o * V_d)/F
Calculate the maintenance dose.
D_m = (C_ss * Cl)/F
How do you calculate the volume of distribution?
V_d = (F*D)/(C_o)
How do you calculate the clearance?
Cl = V_d * k
How do you determine the infusion rate?
Q = C_ss * Cl
If you have overdosed a patient how can you determine the new dose to reduce the concentration?
New dose = old dose * (C-target/C-old)
What is the purpose of calculating creatinine clearance?
Determines the excretory capabilities of the kidneys
How do you calculate the creatinine clearance?
C_Cr = [urine concentration of creatinine * urine flow]/(plasma concentration of creatinine)
If a person's creatinine clearance is low what does this imply?
Kidney function is less than normal - thus the drug you are administering is not being excreted appropriately - and has a prolonged effect (higher likelihood of drug toxicity).
Glucose + glucose = ?
Maltose
Glucose + fructose = ?
Sucrose
Glucose + galactose = ?
Lactose
Which GLUT receptors are high affinity?
Glut 1, 3
Which GLUT receptors are low affinity?
Glut 2.
Which GLUT receptor is hormone receptive?
Glut 4 (not found in liver)....well b/c livers secrete the hormones!
Glut 5 receptors are sensitive to ______.
Fructose.
What substance follows zero-order kinetics? And what IS zero-order kinetics?
Alcohol; substance is removed in equal parts consistently (i.e. 5 mg/hr instead of a reduced fraction every time)
What is the rate-limiting step in glycolysis?
Conversion of fructose 1-phosphate to fructose 1,6-bisphosphatase (by PFK-1)
What are the two energy-producing steps of glycolysis?
1. Pyruvate kinase (conversion of PEP to Pyruvate)
2. Phosphoglycerate kinase (conversion of 1,3-BPG to 3-PG)
What is the fate of Acetyl CoA?
TCA cycle or fatty acid synthesis.
How is NADH returned to NAD+?
Respiratory chain or lactic acid fermentation.
How does the NADH/NAD affect the rate of glycolysis?
If the ratio is higher, then glycolysis slows down (has to wait for replenishment of NAD+).
Why do two types of aldolases exist?
One breaks down Fructose 1.6 BP, while the other type converts Fructose 1-P to the intermediates of glycolysis?
What intermediate connects fructose with glucose metabolism?
Glyceraldehyde 3-Phosphate.
What is the energy production from glycolysis?
2 ATP and 2 NADH
What is the energy production from the conversion of pyruvate to Acetyl CoA?
2 NADH
How can hemolytic anemias be treated? And what is the purpose of this?
Folic acid (tetrahydrofolate) supplementation - which produces thymidilate synthase responsible for regeneration of cells - in this case, RBCs.
How does galactose metabolism link into glucose metabolism? And what is the important enzyme in this reaction?
Glucose-6 phosphate; UDP-galactose --> UDP-glucose
What is the significance of the Cori cycle?
Liver uptakes the excess lactate and converts it to glucose for re-use by muscles.
What is done with excess glucose?
Converted to sorbitol (which is eventually converted to fructose in the liver or sperm cells).
What is the characteristic glucose usage by tumors?
Very high demands, stimulate HIF-1 (hypoxia-induced factors) which increase glut receptors (thus promoting growth of tumor).
What are the clinical symptoms of an excessive fructose diet?
Gout and hyperuricemia....excess production of fructose 1-P limits inorganice phosphate use of ADP --> ATP (thus ADP ==> AMP)
Enzymatic defects in fructose metabolism has what effect on glycogen metabolism?
High amounts of fructose 1-P inhibits glycogen metabolism (thus fructose can't breakdown and neither can glycogen...leading to hypoglycemia)
What can cause lactose intolerance?
Enzymatic defect in beta-galactosidase or aldolase reductase (either genetic, damage-induced, or from intestinal removal)
What is excess galactose converted to - and what clinical problems does this present with?
Galactitol; causes cataracts.
How does fatty acid metabolism link to carbohydrate metabolism?
Triglycerides converted to glycerol 3-P in ADIPOSE TISSUE and then converted to dihydroxyacetone phosphate in the LIVER (either going through glycolytic or gluconeogenic pathways)
Is the process of gluconeogenesis an exact reversal of glycolytic steps? Why?
No; you have to overcome the three irreversible steps of glycolytic.
What is the analog of pyruvate kinase in glycolysis in gluconeogenesis?
Pyruvate carboxylase.
Why is there an additional step in gluconeogenesis?
Oxaloacetate (product of pyruvate carboxylase) has to be converted to PEP (via the additional PEP carboxylase)
Why is gluconeogenesis only limited to liver and kidneys?
Pyruvate carboxylase is only located in the mitochondria of these organs.
Is oxaloacetate permeable to the mitochondrial membrane? [in terms of gluconeogenesis]
No - has to be converted to malate - crosses into the cytosol to continue the steps of gluconeogenesis.
What is the "opposite" of a kinase? And what role does this enzyme play in gluconeogenesis?
Phosphatase; fructose bisphosphatase and glucose 6-phosphatase convert the molecule to glucose (final two steps of gluconeogenesis)
The functionality of gluconeogenesis is critical for the brain because....
The brain can only consume glucose as a source of energy
What are the energy requirements for gluconeogenesis? And what is the final product?
6 ATP and 2 NADH required, 2 mol pyruvate become 1 mol glucose.
How does insulin/glucagon play a role in regulation of glycolysis?
Insulin promotes glycolysis by activating a phosphatase for pyruvate kinase; glucagon promotes gluconeogenesis by activating a kinase for pyruvate kinase.
What is the most important regulatory molecule of glycolysis?
phosphofructokinase-1
How does ATP/AMP levels affect PFK-1?
High ATP levels inhibit PFK-1 while high AMP levels activate it.
How does PFK-2 regulate PFK-1?
Excess glucose converted to fructose in the liver where PFK-2 converts Fructose 6-P to Fructose 1,6-BP (which activates PFK-1)
What is the main enzyme regulated in gluconeogenesis and by what mechanism?
Fructose 1,6-bisphosphatase - inhibited by low levels of AMP in the body.
What bonds cause the "kinks" in glycogen?
alpha 1,6 - glycosidic bonds
What is the function of glycogenin?
enzyme that provides a scaffold/support for new glycogen synthesis.
By what mechanism is additional glucose molecules added to the initial chain of glycogen?
UDP-glucose catalyzed by glycogen synthase to form alpha 1,4 bonds.
What creates the alpha-1,6 glycosidic bonds in glycogen?
Glucosyl transferase.
Why is the branching of glycogen so important in animals?
Makes the compound soluble (if it were purely linear would not be soluble/functional in the body)
What is the function of glycogen phosphorylase? How is the regulation of this enzyme unique?
Breaks down glycogen to a limit dextrin, is ACTIVATED by phosphorylation.
What are the two debranching enzymes in the glycogen breakdown pathway?
glucosyl transferase and alpha-1,6 glucosidase.
What does a defect in alpha 1,4-glucosidase lead to?
Pompe's disease (cardiorespiratory failure/death by age 2) - due to massive increase in glycogen in lysosomes.
What is Von Gierke's disease?
Defect in glucose 6-phosphatase which leads to accumulation of glycogen in kidney/liver (hepatomegaly and severe hypoglycemia)
What will a defect in alpha 1,6-glucosidase cause?
Cori disease - milder course of Von Gierke's disease.
What is McArdle's disease?
Defect in glycogen phosphorylase which leads to a slight increase in glycogen (painful muscle cramps, but otherwise normal).
What is the only glycogen storage disease (that we have studied) that deals with the synthesis of glycogen?
Andersen's disease - defect in branching enzymes [abnormal glycogen formed leading to liver failure]
How does epinephrine glycogen degradation?
Epinephrine signals the cAMP cascade which eventually promotes kinase activity to phosphorylate glycogen phosphorylase (which promotes glycogen breakdown).
What are the two main purposes of the Pentose phosphate pathway (HMP)?
Production of NADPH and ribulose phosphate (for nucleotide biosynthesis)
What role does NADPH play in RBCs?
Co-factor of glutathione reductase.
The products of non-oxidative HMP can yield what type of molecules?
Intermediates of the glycolytic pathway.
What enzyme is required for the oxidative HMP pathway?
Glucose 6-phosphate dehydrogenase.
What clinical symptoms will G6PD deficiency cause?
Hemolysis/anemia during oxidative stress of RBCs.
What are the two main functions of the uronic acid pathway?
Formation of:
1. Glucoronate (for glucoronidation reactions)
2. Pentose phosphates (for utilziation in HMP pahtway)
What is the purpose of a loading dose?
To reach a therapeutic level of a drug faster (does NOT reach steady-state faster)
Give two examples of drugs that are NOT given orally due to first-pass effect?
Morphine and nitroglycerine.
What are the two main types of reactions involved with drug metabolism?
Phase I (polarizing reactions)
Phase II (conjugation)
What are the types of Phase I reactions?
Oxidation, reduction, and hydrolysis
What type of reactions are categorized under oxidation reactions?
N-Dealkylation, O-dealkylation, deamination, and desulfuration.
Give an example of an N-dealkylation reaction?
Imipramine --> Desipramine.
Give an example of an O-dealkylation reaction?
Codeine --> morphine
Give an example of a deamination reaction?
Amphetamine --> Phenylacetone

Amphetamine (Adderall!)
Give an example of a hydrolysis reaction?
Procaine --> Cocaine
Give an example of a desulfuration reaction?
Parathion to paraoxon
What are the two types of oxidation in Phase I reactions?
microsomal and non-microsomal
What are the two non-microsomal oxidation reactions discussed?
Alcohol dehydrogenase and aldehyde dehydrogenase (ADH and ALDH)
What are the two types of reduction reactions and give examples of each.
Azoreductions (prontosil)
Nitroreductions (chloramphenicol)
Prontosil and Levodopa are examples of....
Prodrugs.
What is the terminal oxidase to which the drug binds during microsomal oxidation?
cytochrome P450
What co-factor is required for microsomal oxidation reactions?
NADPH
How does malaoxon and paraoxon function as suicide synthesis?
Originally were malathion and parathion - then converted to malaoxon and paraoxon which inhibit acetylcholinesterase (this increased amount of acetylcholine causes paralysis/death)
What is the most frequent conjugation reaction - provide an example.
Glucoronidation (morphine)
Name all types of conjugation reactions and provide a drug example for each.
Acetylation (isozianid)
Methylation (norepinephrine)
Glucoronidation (morphine)
What substances induce P450 activity?
Phenobarbitol, rifampin, and tobacco smoke products (PAHs)
What substances inhibit P450?
Cimetidine and grapefruit juice.
While intravenous drug injections provide the most accuracy/tracking of drug concentrations - what is a very serious side effect/concern?
Higher risk of toxic effects.
What is the energy production of the TCA cycle (from one ACETYL CoA molecule)?
3 NADH, 1 FADH2, and 1 GTP
ATP homeostasis refers to....
constant levels of ATP under normal conditions
If delta G is negative the reaction is....
spontaneous/moves forward
If delta G is positive the reaction is...
reverse reaction will tend to occur
If detal G is zero the reaction is....
at equilibrium
What affects the delta G outside of the free energy of the products/reactants?
Pressure, initial concentrations of products/reactants, and pH
How does delta G relate to equilibrium constant?
delta G = -1.362 * ln(Keq)
Does the standard free energy of a reaction always have to be less than zero for a reaction to occur spontaneously?
No, if this reaction is coupled with another reaction with standard free energy less than zero - the OVERALL delta G will be <0.
Oxidation is...
Loss of electrons
Reduction is....
Gain of electrons
The more positive a reduction potential the more likely....
Electrons will go there
How do the reduction potential and standard free energy values relate?
The greater the reduction potential, the more spontaneous/negative standard free energy is.
Where is the energy in ATP located?
In the phosphoanhydride bonds (phosphate groups)
Which organ needs the most ATP?
Kidneys (think...all that active transport of molecules/ions/water)
How does shivering thermogenesis work?
Muscles contract, production of ATP increases, and this energy releases as heat.
How does non-shivering thermogenesis work?
Brown adipose tissue inhibits thermogenin - decreases efficiency of OxPhos and H+ gradient in mitochondria no longer used for ATP-synthase but rather heat production.
Why does FAD need to be covalently bonded to the molecule?
Because it can produce highly reactive radicals (semiquinones) which would otherwise damage the body.
List the four dehydrogenase enzymes involved in TCA cycle.
isocitrate dehydrogenase
alpha-ketoglutarate dehydrogenase
succinate dehydrogenase
malate dehydrogenase
The last three steps converting succinate to oxaloacetate is similar to what other metabolic process?
Fatty acid oxidation and oxidation of branched chain amino acids.
What are the three energy-driving reactions in TCA (delta G < 0)
1. Formation of citrate
2. Isocitrate --> alpha-ketoglutarate
3. Formation of succinyl CoA (high energy thioester bond)
What is the energy production from the conversion of pyruvate to acetyl CoA (just one molecule)?
1 NADH
What is the only dehydrogenase complex that resides in the inner mitochondrial membrane?
Succinate dehydrogenases, all other DHs are located in the matrix.
List all three alpha-ketoacid dehydrogenase complexes...
1. pyruvate dehydrogenase
2. alpha ketoglutarate dehydrogenase
3. branched chain alpha-ketoacid DH
What are the 5 cofactors of these alpha-ketoacid dehydrogenase complexes...
1. FAD (prosthetic group)
2. NAD
3. TPP (Thiamine) (prosthetic group)
4. Lipoate (prosthetic group)
5. Coenzyme A (co-substrate)
What clinical symptoms may result in thiamine deficiency (which can often occur in chronic alcoholics)?
Alpha ketoglutarate, pyruvate, and alpha keto-acid accumulation in the blood.
What are the two main purposes of the TCA cycle?
1. NADH/FADH2 production
2. Production of intermediates for gluconeogenesis, heme and AA syntehsis
Isocitrate dehydrogenase is the only dehydrogenase enzyme sensitive to what regulatory mechanism?
ADP stimulates the activity.
Malate dehydrogenase is ONLY sensitive to which regulatory mechanism?
NAD+ stimulates its activity.
What effect does calcium have in TCA cycle?
Up-regulates isocitrate DH, alpha ketoglutrate DH, and pyruvate DH.
Why are problems with glucose metabolism first detected in the CNS?
Because the brain relies heavily on oxidation of glucose and no other energy source - so it is the most sensitive organ.
Pyruvate dehydrogenase is regulated by...
PD Phosphatase and PD kinase
List the three components of the PD-complex.
E1 - TPP
E2 - lipoate
E3 - FAD
What is the only regulator of PD-phosphatase?
Activated by calcium.
What all regulates PD-kinase?
High levels of ADP and pyruvate INHIBIT kinase (so activate dehydrogenase).
NADH and Acetyl CoA activate kinase to inhibit dehydrogenase.
What does a PDC (pyruvate dehydrogenase complex) insufficiency lead to?
Severe lactic acidemia (in its severe form can lead to death).
How does arsenic poisoning manifest itself?
Covalently binds to lipoate on all alpha-ketoacid dehydrogenase complexes - thus removing the functionality of the coenzyme and subsequently the enzyme.
How do you obtain cytosolic Acetyl CoA?
Cleave citrate (which has traveled to cytosol) into Acetyl CoA and OAA [requires ATP]
Succinyl CoA is used as the precursor to....
heme.
What are the two TCA cycle intermediates that contribute to amino acid synthesis?
Oxaloacetate, alpha ketoglutarate.
What intermediate of the TCA cycle is utilized in gluconeogenesis?
Malate.
What intermediate of the TCA cycle is utilized for fatty acid synthesis?
Citrate.
If all these intermediates are utilized for other pathways - how are they restored to prevent depletion of oxaloacetate?
Aneplerotic reactions
What are the two main aneplerotic reactions?
Pyruvate carboxylase converting pyruvate to OAA.

Synthesis of Succinyl CoA from Ile and Val.
Leigh's disease can be caused by what to type of deficiencies?
Pyruvate carboxylase deficiency and pyruvate dehydrogenase complex deficiency.
Pyruvate carboxylase is regulated by...
Acetyl CoA (once enough Acetyl CoA is formed, then it will promote the transformation of pyruvate to OAA)
List the order of the electron transport chain...
Complex I (NADH DH)
Coenzyme Q
Complex III (b-c)
Cytochrome C
Complex IV (cytochrome c oxidase)
What gradient is responsible for the functionality of ATP-synthase?
H+ gradient.....high in the intermembrane space and low in the matrix.
One turn of the rotor requires ______ protons and synthesizes _____ ATPs.
11-12; 3.
Since Coenzyme Q contains a quinone....this can lead to the formation of....
Semiquinones (free radicals)
Each cytochrome contains a bound ______ and therefore requires _____.
heme; iron
In anemic patients - not only is Hb and Mb function affected, but also....
Electron transport chain (because of the dependence of each complex on iron).
What is Hemoglobin M?
Stabilization of methoglobin (oxidized form of hemoglobin) which can be fatal if homozygous (leads to cyanosis)
Only 30-40% of energy gained by NADH and FADH2 are utilized for ATP synthesis....where does teh rest go..
Transportation of anions and formation of heat.
Anoxia results in the stopping of the _____ ______ _____.
Electron transport chain.
Carbon monoxide, cyanide, and azide poisoning bind to ________ and stop ____ ____ _____ thus mimicking the effects of _______.
cytochrome c oxidase, electron transport chain, anoxia
What is a concern when administering sodium nitroprusside for hypertension?
Liver can metabolize this drug into cyanide and therefore the thiocyanate levels in the blood must be monitored.
OxPhos diseases are typically linked to problems in _____.
Mitochondrial DNA.
When oxidation is not coupled to phosphorylation what happens?
ATP is not synthesized - so the body keeps signaling higher activity of ETC (thus you are increasing the H+ gradient) - so there is increased oxygen consumption and heat production.
Dinitrophenol is an example of a....
chemical uncoupler
Salycilate poisoning (a metabolite of aspirin) can cause....
OxPhos uncoupling and therefore overstimulation of glycolytic pathways without ATP production (thus metabolic acidosis)
Uncoupling proteins (UCP) can reduce the likelihood of radical formation from Coenzyme Q because....
Uncoupling stimulates ETC, thus CoQ is more occupied.
How does Pyruvate and ADP enter the matrix of the mitochondria?
Through voltage-gated channels
How does pyruvate and inorganic phosphates enter the matrix?
Via symport with hydrogen ions.
How does calcium enter the matrix?
Through a uniport (driven by the water potential)
How does ATP leave the matrix once produced by ATP-synthase?
First by an antiporter (ATP-translocase) with ADP into the intermembrane space, and then via a voltage-dependent anion channels in the outer membrane.
What is the key component (intermediate) required to bring metabolites into the matrix of the mitochondria?
Malate.
The Mitochondrial Permeability Transition Pore (MPTP) is a complex of....
Antiporter and Voltage-dependent Anion channel.
The proton gradient helps keep the ______ (open/closed) in the MPTP.
ANT (antiporter) closed.
High amounts of ______ and _____ keep the VDAC in the MPTP (open/closed)
hydrogen ions, ATP - keep it closed.
Why is it criticial to know that Bax and Bcl-2 connect/control the VDAC?
Bax is pro-apoptotic, and Bcl-2 is anti-apoptotic - so cellular signaling may stimulate Bax to open VDAC and cause mitochondrial/cell lysis.
Calcium and reactive oxygen species both promote the _______ of the antiporter.
Opening.
What is ischemia reperfusion injury?
If the patient has been oxygen-deprived too long, and you "shock" the patient - the reversal of the ATP synthase at this point will cause the addition of oxygen to induce more problems.
Glycolysis is the main energy-producing pathway in the _____ and ______.
brain and red blood cells.
Why is AMP instead of ADP used as an allosteric activator of PFK-1?
Concentrations are usually kept low by adenylate kinase - therefore any alterations/changes will have a greater effect/sensitivity.
The transport of NADH into the mitochondria occurs by two pathways, which are....
Malate-aspartate shuttle
Glycerol-3-phosphate - DHAP shuttle.
Which NADH shuttle pathway reduces the amount of energy available afterwards?
G3P - DHAP shuttle
If we assume 3 ATP/NADH and 2 ATP/FADH2, what is the total energy production from 1 mol of glucose?
38 ATP.
If there is an issue with fatty acid metabolism, the clinical symptom that will almost always appear is...
Episodes of hypoglycemia during fasting/sleeping.
Fatty acids bind to ______ in the blood.
Albumin.
How do you activate a fatty acid for metabolism?
Attach fatty acid to Acyl CoA via a thiokinase.
How is the activated fatty acyl CoA transported into the mitochondria?
Carnitine.
What is the destination of Acetyl CoA in the body?
Oxidation in the TCA or ketone body synthesis.
Why is CPT I and CPT II required in the cell?
Fatty acyl CoA cannot diffuse into the mitochondrial matrix without the attachment to carnitine which is facilitated by CPT I and CPT II.
CPT II deficiency leads to ______.
hypoglycemia and myoglobinuria.
After each cycle of Beta-oxidation how many carbons are lost?
Two.
Each cycle of Beta-oxidation produces ....
1 FADH2 and 1 NADH
If you have a 20C fatty acid, what is the product?
9 FADH2, 9 NADH, and 10 Acetyl CoA.
What are the four steps of Beta-oxidation?
1. oxidation
2. hydrolysis
3. oxidation
4. addition of Co-A
In Medium-chain acyl coA (MCAD) deficiency what is the clinical manifestation?
Acylcarnitines in the blood/ruine, and hypoglecemia and hypoketosis.
What is the main difference (in clinical signaling) between MCAD and LCAD deficiency?
Acylcarnitines are only in blood and does not show up in urine
What happens in an odd-chain fatty acid?
Last two products are propionyl CoA and acetyl CoA.
What is the product of propionyl CoA?
Converted to succinyl CoA with the help of cobalamin (B12) which then goes into glycolysis or heme production.
High amounts of FADH2 and NADH (inhibit/stimulate) Beta-oxidation.
Inhibit.
How does Acetyl CoA indirectly regulate the Beta oxidation?
Acetyl CoA is converted to Malonyl CoA thus inhibiting Beta oxidation to prevent further production of Acetyl CoA.
How is Acetyl CoA carboxylase regulated and what is its function.
Converts Acetyl CoA to malonyl CoA - inhibited by high AMP levels and promoted by insulin.
Name two alternate pathways of fatty acid metabolism.
Peroxisomal Beta-oxidation and microsomal omega-oxidation.
Why doesn't peroxisomal oxidation result in energy production?
Because the FADH2 is utilized for reduction of hydrogen peroxide and the remaining 4-6 carbon fatty acid is shifted to the mitochondria.
What enzyme converts hydrogen peroxide to water?
Catalase.
What is the purpose of alpha-oxidation?
Due to the additional methyl groups - it only reduces it by one carbon so further Beta oxidation can avoid the methyl groups.
What is the purpose of omega oxidation?
Converts terminal carbon in fatty acid to carboxylic acid (thus becomes a dicarboxylic acid)
What is the regulatory mechanisms of fatty acid oxidation?
Substrate levels and THAT'S IT!!
If there is any deficiency in beta-oxidation or carnitine what will this manifest as in a clinical test?
Presence of dicarboxylic acids in the urine.
What is the product of ketone body synthesis?
Acetone and Beta-hydroxybutarate.
What is an important intermediate of ketone body synthesis and what is an alternate synthetic pathway?
HMG CoA....can go into cholesterol production.
Ketone bodies can be fuel for the ________ and _______ , but NOT the ______.
Skeletal muscle, brain, liver
What is the energy production of acetoacetate (main ketone body functioning).
Two acetyl CoAs (so 20 ATP total) minus 1 ATP for conversion of acetoacetate to acetyl CoA.
What is a possible treatment for pyruvate dehydrogenase deficiency?
Ketogenic diet (avoids the need for pyruvate to convert into Acetyl CoA)
Why do you see a larger rise in ketone bodies once the body has compensated via fatty acid metabolism?
Because the skeletal muscle has lower energy demands - thus not utilizing glucose nor ketone bodies.
What is the Jamaican vomiting sickness?
ingestion of hypoglycin inhibits acyl CoA dehydrogenase - thus beta oxidation is inhibited and sever hypoglycemia occurs.
Which are the only two TRUE ketogenic amino acids?
Lysine and leucine.
What is propionic acidemia and aciduria?
Defect in propionyl-CoA carboxylase leading to an accumulation of propionyl CoA ... as a false substrate to isocitrate....will cause depletion of oxaloacetate and increase in methylcitrate in the urine.
What enzyme allows the recycling of biotin in the body?
Biotinidase
What is methylmalonic aciduria?
Defect in methylmalonyl CoA mutase - thus excretion of methylmalonate in the urine is very high.
Defects in branched chain alpha-keto acid dehydrogenase leads to....
Maple-syrup disease (due to increase in ketoacids in the urine leading to syrup odor"
If there is a defect in phenylalanine hydroxylase this causes...
Phenylketonuria (phenylalanine is converted in the body to phenylketones)
What is the disorder that leads to urine that immediately stains upon exposure to air?
Defect in homogentisate oxidase called alkaptonuria.
What is tyrosinemia I?
Defect in fumarylacetoacetate hydrolase or maleylacetoacetate isomerase which leads to a cabbage-like odor and liver failure.
How do you detect Tyrosinemia I?
Succinylacetone accumulates in the body.
Is Tyrosinemia I more or less severe than Tyrosinemia II?
More severe, Tyr. II has a defect earlier in the pathway at tyrosine aminotransferase.
What are all the essential amino acids?
FILM TV WK HR
phenylalanine, isoleucine, leucine, methionine, threonine, valine, tryptophan, lysine, histidine, arginine
Are amino acids a major source of energy?
No - fatty acids and glucose are main sources of energy.
What are all the ketogenic amino acids?
Why Fuck Your King In London?

Tryptophan, phenylalanine, tyrosine, lysine, isoleucine, leucine.
Which is more likely to diffuse into the cell - large or small molecules?
Small molecules.
Which is more likely to diffuse into the cell - polar or non-polar molecules?
Non-polar.
Which is more likely to diffuse into the cell - ionic or neutral molecules?
Neutral.
The concentration of all ions are greater outside the cell EXCEPT ______.
K+ and proteins.
What are the two types of passive diffusion?
Simple diffusion (osmosis) or facilitated diffusion.
There are two types of facilitated diffusion...
Channel and carrier proteins.
Which type of facilitated diffusion is non-specific...channel or carrier?
Channel.
What are the different types of ways channel proteins are selective (without binding to a specific molecule)?
Voltage-gated
Ligand-gated
Stress-induced
What are the three types of active transport?
Symport, antiport, and uniport
Does all active transport require energy?
YES!
The cytoplasmic side of the membrane is usually ______ and the extracellular side is usually ______.
Negative; positive
What is the significance of the Na-K pump?
Maintains the potassium levels high inside and the sodium levels low within the cell.
Active transport that utilizes ATP is called...
Primary active transport.
Active transport that utilizes an electrochemical gradient is called...
Secondary active transport.
What is the advantage of having very low Calcium concentrations within the cell?
Allows sensitivity for signaling (much like how AMP is more sensitive than ADP as a signaling molecule)
If the extracellular area is hypertonic then what happens....
Cellular water will leave the cell and cell will shrink.
If the extracellular area is hypotonic then what happens....
Water will flow into the cell and cell will lyse.
How is water permeable in the cell membrane?
Aquaporins!
What percent of the total body weight is water?
60%
What is the extracellular fluid percentage in the body? Intracellular?
Intracellular is 40%....extracellular is 20%
What two forces work for flow of fluids in and out of capillaries?
Hydrostatic and oncotic pressures.
What is the main change in arterioles and venuoles that allow fluid to leave arterioles and fluid to re-enter capillaries?
Hydrostatic pressure changes.
Vasodilation will ______ hydrostatic pressure while vasoconstriction will ______ hydrostatic pressure and typically acts on the _____.
decrease; increase, venous return.
What is the purpose of lymphatic system?
Not all fluid perfectly returns to the blood circulation - this extra fluid goes into the lymphatic system.
What is edema?
Excess accumulation of fluids in the interstitial compartment.
What is the difference between paracrine and endocrine signaling?
Paracrine is nearby, endocrine travels through the blood.
How do G-protein linked receptors work?
GTP binds, alpha unit splits from beta-gamma unit, cascade of proteins, when action done - GTP hydrolyzed and G-protein reassociates.
What is an example of contact signaling?
Immune response and antibodies!
What is a critical characteristic of those molecules that are intracellular receptors?
Lipid-soluble! They must transgress through the membrane.
What does nitric oxide stimulate?
Guanylyl cyclase....increases cGMP.
What does sildenafil do?
Inhibits phosphodiesterase, prolongs action of cGMP.
What are enzyme-linked receptors?
Receptors that have enzymatic activity on their own (growth factors).
What mechanism do cGMP and guanylyl cyclase follow?
Intracellular receptors.
What are the two types of secondary messengers in G-protein coupled receptors?
cAMP and DAG/IP3 (protein kinase)
What is the enzyme-linked receptor pathway mechanism?
Receptor dimerizes, cross-phosphorylation, SH2 then Ras binds - Ras signals a series of protein cascades.
30% of cancers are involved with mutations of _____.
Ras.
What are the two other signaling pathways that resemble enzyme-linked receptors?
JAK/STAT - fewer intermediates than enzyme-linked.

TGF-Beta - kinases activate SMADs directly affect gene transcription
What are the pro-apoptotic proteins?
Bax, Bak
BH3 (separate family of proteins)
What are the anti-apoptotic proteins?
Bcl-2
Once the apoptotic enzymes are activated...what happens...
Binds to Apaf to create the apoptosome which then initiates the caspases which lyse specific parts of the cell.
Apaf is an ______ protein while the caspases are the ______ proteins.
adaptor; effectors
How are the pro-apoptotoic proteins activated?
Stress-induced or loss of cytokine signals induces the BH3 to throw off the equilibrium of Bcl/Bax (less Bcl) - thus beginning the apoptotic pathway.
Aside from stress-induced signals, what is another intracellular apoptotic signal?
p53 can induce transcription of Bax or BH3....it can ALSO stimulate death receptors for the EXTRINSIC pathway.
What is the extrinsic apoptotic pathway?
Death receptors induce tumor necrosis factor (TNF) and induces cytoplasmic proteins which activate caspase 8.
What role does cytotoxic T lymphocytes play in apoptosis?
Granzyme B from T-cell or NK cell enters the cell via the perforin pore which then activates caspases.
How does hypoxia induce cell injury?
Lack of oxygen, ATP not produced sufficiently, ion equilibrium lost, ions swell into the cell and lysis can occur unless reversed.
What is ischemia?
Loss of blood flow to an area.
What is anoxia?
Extremely low to no oxygen in the system.
What is hypoxia?
Lack of oxygen (either induced by ischemia or lack of oxygen inspiration)
Necrosis is cell ______ while apoptosis leads to cell ______.
swelling; shrinking
In time of cell injury all activity in the cell decreases except for.....
Proteases.
High levels of calcium stimulate...
degradative enzymes...phospholipases, proteases, and the MPTP is opened.
WHat is the Fenton reaction?
Utilizes iron as a co-reactant to oxidize peroxide into a hydroxyl and a reactive hydroxyl molecule.
What type of cellular damage can ROS cause?
Fragmentation of membrane proteins and oxidative damage to nuclear DNA (thymidine and quinine)
What are the two reactions by which hydrogen peroxide is reduced to water?
1. Catalase (peroxisomes)
2. Glutathione reductase then peroxidase (uses NADPH)
What is atrophy?
Cell shrinkage.
What is hypertrophy?
Increased cell size (typically due to increased functional demand).
What is hyperplasia?
Greater NUMBER of cells.
What is metaplasia?
Differentiation into a different type of cell than what is normal.
Hypertrophy can occur in ______, ______, and ______ muscles BUT hyperplasia can only occur in ______ muscles.
Smooth, cardiac, skeletal; smooth
What is psoriasis?
A type of hyperplasia of the skin.
What are keloids?
Hypertrophic scars.
Metaplasia in the conjunctiva (goblet cells isntead of squamous epithelial) is caused by....
Vitamin A deficiency.
What is a hydropic change?
Accumulation of water due to ionic changes (typically induced by lack of ATP)
What is steatosis?
Fat accumulation in the liver.
What are xanthomas?
Intracellular accumulation of cholesterol (hyperlipidemias, atherosclerosis, inflammation)
What is anthracosis?
Accumulation of carbon in the lungs (not necessarily harmful).
What are mallory bodies?
Accumulation of proteins typically due to liver damage.
What is pneumoconiosis?
inflammatory response to carbon/silica dust particles.
What is hemosiderin and how can you detect it in cells?
Hemosiderin is present in high-iron cells (can be caused of hemolytic anemias) - stains Prussian blue.
What is hemochromatosis?
Excessive iron in the liver due to cirrhosis and heart failure.
What is bilirubin?
End product of hemoglobin metabolism.
What can cause the accumulation of bilirubin in the hepatocytes? (brown-pigment staining)
Excess bilirubin production, impaired bile flow, impaired excretions, reduced hepatic uptake.
What is lipofuscin?
Can occur in aging people - completely normal "wear and tear" pigment.
What are the four types of necrosis?
Coagulative, liquifying, caseating, and fat.
True/false: Necrosis releases proteolytic enzymes that can damage adjacent cells.
True.
What usually causes coagulative necrosis?
Ischemia/hypoxia injuries leads to dead myocytes.
What other type of response will you see around sites of necrosis?
Acute inflammatory response - apoptosis comes from macrophages.
Bacterial and fungal infections along with brain ischemia leads to what type of necrosis?
Liquefactive necrosis.
Caseating necrosis normally associated with....
tuberculosis.
Pancreatitis and direct trauma to the breast typically lead to what type of necrosis?
Fat.
How do -PRIL drugs work (like captopril)?
ACE inhibitors thus preventing vasconstriction - treats hypertension.
What does Atorvastatin inhibit?
HMG CoA Reductase (reduces cholesterol synthesis...treats high cholesterol).
How does digoxin function?
Inhibits the Na-K ATPase pump - slows down stimulation of action potentials (for cardiac arrythmias).
What drugs inhibit cyclooxygenase?
NSAIDs - treats inflammatory responses and headaches, etc.
How does nitroglycerine function?
Promotes guanylyl cyclase and increases production of cGMP.
Why is nitroglycerine and sildenafil (viagra) a dangerous combination?
Both increase the levels of cGMP - this excess prolongation of sympathetic stimulation can reduce the blood pressure to dangerous levels.
How does heparin function?
Inhibits prothrombin III to prevent clotting (anticoagulant).
How does minoxidil work?
ACTIVATES potassium-channels to promote relaxation of smooth muscle (was for hypertension....now for hair loss!)
What does dilitazem, nifedipine, and lidocaine function as?
Ion-channel blockers.....typically reduces the signaling/excitation - can treat arrythmias.
How does omeprazole work?
Inhibits hydrogen pump that releases acid into the gut - treated for acid reflux and ulcers.
How does fluoxetine work?
Inhibits uptake of serotonin - for antidepression.
What is amytryptaline?
Inhibits norepinephrine uptake - for anti-depression.
How do diuretics function?
Ethacrynic acid and hydrochlorothiazide both block ion reabsorption in the renal tubules (promotes excretion).
How does epinephrine work?
Agonist to Beta receptors - induces cAMP - stimulates heart rate.
How does isoproterinol work?
Beta-2 receptors for the lungs - bronchodilation treated for ashtma.
What are beta-blockers used for cardiology?
Propanolol (other -OLOL drugs) block these receptors to reduce the stimulation for hypertension.
What does naloxone do?
Morphine receptor blocker - treats morphine overdose.
What is losartan?
Angiotensin II receptor blocker - prevents vasconstriction for hypertension.
How do allergy medications work?
They are histamine receptor blockers to stop inflammation response!
Give two examples of drugs that function as enzyme-linked receptors?
Growth factors and insulin.
How do steroids work?
They are intracellular receptors and alter transcription.
What is omalizumab for?
Binds to IgE to control asthmatic response.
What is cisplatin?
Alkylating agent to treat cancer.
What is vincristine?
Binds to microtubules to control cancer growth.
If the reflection coefficient is 1 what does that mean about permeability?
The membrane is IMPERMEABLE.