• 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

1/366

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;

366 Cards in this Set

  • Front
  • Back
proteins/amino acids are linked by ______ bonds
peptide
Which level of protein structure participates in backbone H-bonding?
Secondary
Why is histidine very special?
It's a good buffer
1) What amino acid can form disulfide bonds?

2) And what happens if there is too much of this AA?
1) Cystine

2) can form stones
In an alpha helix, H bonds occur between backbones of SAME or DIFFERENT strands?
different
In a beta sheet, H bonds occur between backbones of SAME or DIFFERENT strands?
different
Name the FOUR types of bond/interactions that can drive tertiary and quaternary structure formation
H-bonds
ionic bonds
disulfide bonds
hydrophobic interactions
Name of a protein composed of 3 different subunits
heterotrimer
What is the main function of glutathione (2 GSH)?
Reduce disulfide bonds
What molecule does glutathione (2 GSH) become during the process of reducing disulfide bonds?
GSSG
What kind of structure is insulin secreted as?

What kind of structure does insulin have to be in order to get absorbed?
hexamer


monomer
How does Lispro increase activity of insulin?
it destabilizes the hexamer formation, increasing insulin break down into monomer
Describe the overall structure of immunoglobulins?

(types of chains, bonds and regions)
-2 light chains & 2 heavy chains

- inter and intra chain disulfide bonds

- variable & constant regions
What is a holoprotein?
a protein with ALL of its groups
What is an apoprotein?
a protein WITHOUT all of its group
In what state does the iron have to be in in hemoglobin for it to bind oxygen? (give name and oxidation state)
Ferrous

2+ oxidation state
When no oxygens are attached to hemoglobin, what form is it in?
taut form
What is cooperative binding of oxygen to hemoglobin?
binding of 1st oxygen to Hb increases the affinity of the other oxygens to bind

unloading of oxygen from hemoglobin occurs in the same way
Oxygen cooperative binding is also called __________ binding
allosteric (binding is affected by changes at a different site)
2,3-BPG is an example of ______ _____________
self regulation
Describe where 2,3-BPG is found in the body and its main function
found in RBC

stabilizes the taut Hb form to release oxygen into tissues that need it more readily
What is MetHb?
-methomeglobin
-ferric state (Hb-Fe^3+) of hemoglobin that cannot bind oxygen
What is the role of NADH methemoglobin reductase?
it's an enzyme which converts methemoglobin (Hb-Fe^3+) to hemoglobin (Hb-Fe^2+)
Name 3 potential cases of methemoglobinemia
1) drugs/toxins can oxidize Fe^2+

2) deficiencies in NADH methemoglobin

3) MetHb mutations can make it resistant to reduction by NADH methemoglobin reductase
An increase in cross-links in the serum suggests what?
increased bone degredation
An increase in procollagen levels in the serum suggests what?
increased bone synthesis
High carboxyhemoglobin levels in the body indicate what?
CO poisoning
Where does synthesis of collegen take place?

By what means does synthesis of collegen occur?
in the RER


hydroxylation and glycosylation
Where does cleavage of procollagen occur?
outside of the cell
If a protein is found that cleaves 2,3-BPG is expressed in the body, would this increase, decrease or have no effect on the oxygen content of Hb?
increase (2,3-BPG increases oxygen unloading from Hb, so if 2,3-BPG is broken down (cleaved), there would be more oxygen still bound to Hb)
What is a polar molecule?
molecule in which electrons are shared UNEQUALLY between other molecules
What type of molecules can dissolve readily in water?
polar, hydrophilic molecules (like dissolves like)
What would happen to a cell if placed in a hypertonic environment?
Hypotonic?
Isotonic?
Shrink

Swell (can cause the cell to rupture)

No change in cell shape
Will bacteria rupture if placed in a hypotonic solution?
No (the have a cell wall)
To dissolve an acid, you must place it in (stronger/weaker) (acid/base). Why?

choose one from each parenthesis group
stronger acid

(a stronger acid solution will have more protons to donate)
To dissolve a base, you must place it in (stronger/weaker) (acid/base). Why?

choose one from each parenthesis group
stronger base

(a stonger base solution will be looking to gain a proton)
Is Hendersen-Hasselbalch equation log (unprotonated/protonated) or log (protonated/unprotonated)?
log (unprotonated/protonated)
Which extracellular biological buffer system (that we've learned thus far) is used to maintain acid-base balance?
bicarbonate buffer system

Effective pKa = 6.1

(CO2 adjusted by breathing)
Which intracellular biological buffer system (that we've learned thus far) is used to maintain acid-base balance?
phosphate buffer system

Effective pKa = 7.2
Something that's oxidized is also called the _______ agent
reducing
Something that's reduced is also called the _______ agent
oxidizing
Name of carbohydrates that differ only at ONE asymmetric carbon
epimers
molecules that have the same molecular formula and sequence of bonded atoms, but differ ONLY in the three-dimensional orientation of their atoms in space
stereoisomers
The structure of monosaccharides in SOLUTION
cyclic

(their stereochemistry at the anomeric carbon can also change)
What compounds can form glycosidic bonds?

bonds are formed between what?
sugars

bonds between anomeric carbon and the O or N group of another molecule
N-glycosidic bonds are found where?
nucleosides & glycoproteins
O-glycosidic bonds are found where?
disaccharides, oligosaccharides, polysaccharides & glycoproteins
Name the 3 monosaccharides
glucose
galactose
fructose
Name the 3 disaccharides
sucrose
lactose
maltose
What is sucrose composed of?
glucose-fructose
What is lactose composed of?
glucose-galactose
What is maltose composed of?
glucose-glucose
What is the only sugar that will give a negative reducing sugar test?
sucrose
How many double bonds do saturated fatty acids have?

Is the melting point higher or lower compared to unsaturated fatty acids?
none (each carbon has a maximum amount of single bonds)

melting point is HIGHER than unsaturated. (introducing impurities such as double bonds will lower the melting point)
Name the 2 types of unsaturated fatty acids

How many double bonds do they have?
monounsaturated (1 double bond)

polyunsaturated (>1 double bonds)
Can cis-structured fatty acids stack?

Is their melting point higher or lower than trans fatty acids?
No.

Less stacking mean lower melting point. (that's why trans fatty acids are so bad for you --> higher melting point)
Instead of glycerol, sphingolipids have what common core component?
ceramide
What common core component do steroids have?
cholesterol
Proteins fold into correct stuctures due to their __________ sequence of AA
primary
Heat shock proteins use energy from ATP hydrolysis to overcome _________
kinetic barriers
The role of isomerases in protein folding
helper proteins that help overcome kinetic barriers
What are chaperons/chaperonins?
heat shock proteins that assist in protein folding
Proteases are enzymes that break the ______ ________
amide backbone (& separates the protein into its individual AA)
Proteases work best in basic or acidic environments?
acidic
What is ubiquination?
proteins being tagged with ubiquitin for degradation
What is the role of ubiquitin after it has been tagged to a protein?
brings it to a proteasome, where the protein will be degraded into individual AA's
What is a proteasome?
protein chopping factory within a cell

(protein must have ubiquitin added to be degraded by proteasome)
What attaches the ubiquitin to a protein?
ligases
PEST sequences are associated with proteins that have a ______ ____________ _________
short intracellular half-life
PEST sequences are found in proteins with high or low turnover rates?
high (thus acting as signal peptides for protein degradation)
Explain what happens when there is too much glucose in the body. (in terms of protein modification and glycosylation/glycation)
increase glucose = increase in glycosylation/glycation (bad) = increase in AGEs (aggregates) = increase in complications
Are proteins with high or low turnover rate affected more by Glycosylation/glycation
low turnover rate
Why is hemoglobin more affected by sugar levels than other proteins?
once sugar binds to Hb, oxygen cannot bind as readily

(so as glucose increases, glycosylation increases, and the amount of oxygen bound to Hb DECREASES)
What does the term AGE stand for?

When are AGE levels high?
advanced glycation end products

AGE levels are high when blood glucose is high (diabetics)
What does HbA1c test measure?
hemoglobin glycation in RBC's, who's life span is 120 days.

can be used to ***** what glucose levels have been over a long period of time
If you want to assess how well a pt has controlled their sugar levels over time, what test do you use?
HbA1c
Two proteins that are the same, but encoded by different genes and that function in different tissues
isoforms
When a variant occurs in a population; NOT a mutation, just a difference
polymorphism
Creatine kinase is an example of _______-_______ variation
tissue-specific
Creatine kinase is important in regenerating _____
ATP
If CK-MM is detected, where is the problem?
CK-BB?

CK-MB?
CK-MM = skeletal muscle

CK-BB = brain

CK-MB - cardiac
creatine kinase M and B are examples of what? (think structure)
isoforms (2 proteins that have same structure but encoded by different genes to work in different tissues)
A2γ2 is a structure for what type of hemoglobin

Does it have increased or reduced 2,3-BPG binding?
HbF

Reduced, since oxygen must be transferred from maternal to fetal Hb
α2δ2 is a structure for what type of hemoglobin?
HbA - a normal Hb variant
HbC is abnormal hemoglobin where the normal glutamic acid residue at the 6th position of the β-globin chain is subsituted by _________
lysine residue
HbS is abnormal hemoglobin (sickle cell) where the normal glutamic acid residue at the 6th position of the β-globin chain is subsituted by _________
valine residue
Polymerization occurs only when Hb is in the ______ form.

Thus, it is more common in the ________ tissues.
taut (or deoxyginated)


peripheral
What are proteinaceous infectious particles? (AKA infectious proteins)
PRIONS = non-living structures (proteins) that are involved in protein misfolding (ex. turning normal PrPc into PrPsc, which form aggregates/clumps in the brain)
What is mutant CJD?
when your own proteins mutate, forming prions
What is variant CJD?
when you ingest a prion (ex. PrPsc)
inheritance of alleles at one locus does not influence inheritance at other loci
independent assortment
test to see if two mutants with the same phenotype are produced by mutations of the same gene, or by mutations of separate genes
complementation
In complementation, mutations in different genes are _______________

mutations in same genes are _______________
complementary

non-complementary
genetic interactions other than the autosomal recessive pattern
non-mendilian genetics
when different alleles have equal effects and heterozygotes have intermediate phenotypes
semidominance
different alleles have equal effects, but there is no blending of phenotypes
codominance
multiple alleles for the same gene/locus
polymorphism
multiple alleles for the same trait
polygenic inheritance
Explain epistasis
when one separate GENE masks the phenotypes of a totally separate gene
Define autosomes
all CHROMOSOMES besides X and Y
define autosomal
a GENE not located on X or Y
What is sex influenced inheritance?
AUTOSOMAL GENES which are expressed differently in males and females
Do exergonic reactions have a positive or negative ΔG?
negative
What do cells do to move unfavorable reactions forward?
couple them with more favorable reactions, so the overall ΔG is negative
What is an important carrier of chemical energy in muscle cells?
creatine phosphate
If [substrate] is raised, would ΔG become more favorable or unfavorable?
favorable
ATP/ADP ratios are maintained through _______ _________
aerobic respirations
Name the 3 steps in aerobic respiration
1) oxidize fuels (turn everything into acetyl CoA, oxidize acetyl CoA in the TCA cycle to make CO2m FADH2 and NADH)
2) Make ATP (oxidative phosphorylation)
3) Use ATP (will make ADP)
Do enzymes change the ΔG of a reaction?
NO (they decrease activation energy AND increase reaction rate)
What is a coenzyme, and what happens to it during a reaction?
Coenzyme/cofactors are derived from VITAMINS

They get MODIFIED during a rxn
inactive or less active enzymes that do not have its cofactors
apoenzyme
active enzyme (apoenzyme + cofactor)
holoenzyme
What does the mnemonic NVYC (envy c) stand for?
non-competative
vmax involved
y-intercept changed
catalytic rate affected
what is Vmax in a reaction?
maximum velocity at which enzyme can catalyze the reaction

(adding more substrate will no increase the reaction since all enzymes are saturated)
what is Km in a reaction?
substrate affinity

high affinity = low kM
How is Km different from Kd?
Km is related to substrate AFFINITY

Kd is related to substrate BINDING
A ligand or drug that does NOT activate signaling upon binding
receptor antagonist
A ligand or drug that activate signaling upon binding
receptor agonist

(a competative binder with the natural ligand at the same site)
Do competative inhibitors affect the Km or Vmax?
Km
Do noncompetative inhibitors affect Km or Vmax?
Vmax
Define product inhibition
where the product of the reaction goes ack to the enzyme and binds to the allosteric site and inhibits the reaction from producing more product
name for inhibitors that bind to enzymes with non-covalent bonds such as H-bonds, hydrophobic bonds or ionic bonds
reversible inhibitors

add more substrate so inhibitor can be overwhelmed
name for inhibitors that bind to enzymes with covalent bonds
irreversible inhibitors. (can't remove them).

Only way to make rxn proceed is to add more enzyme
Name for an inactive enzyme precursor
zymogen
What do zymogens require in order to become active?
require biological change, such as CLEAVAGE
How does the activation of chymotrypsin differ to that of pepsin?
chymotrypsin requires outside source to be cleaved (trypsin)

pepsin cleaves itself (when in the stomach)
What are futile cycles?
where 2 pathways run simultaneously in opposite directions and have no overall positive effect
metabolic products of one enzyme are passed directly to another enzyme w/o having to diffuse through the solution
substrate channeling
What forms the primary clot plug?
platelets
What things activate platelets?
Collagen
Thrombin
ADP
Epinephrin
Immune complexes & stress
The 3 roles of platelets
activation (Change shape)
aggregation
adhesion to vessel wall
Platelets are derived from
megakaryocytes
life span of a platelet
10 days
Do platelets contain a nucleus?
No
vWF is responsible for anchoring
Platelets to the subendothelial collagen
Phospholipase A2
is responsible for cleaving a phospholipid and releasing arachidonic acid in platelets
Arachidonic acid
is used to make thrombaxane (TXA2)
Is TXA2 a vasoconstrictor or vasodialator?
vasoconstrictor
What are the six sites of inhibiting platelet activation?
1. Cyclooxygenase activity (aspirin)
2. Thromboxane synthetase
3. Thromboxane receptor
4. GPII antagonists
5. ADP inhibitors
6. ADP receptor inhibitor
What's the cause of von Willebrand disease?
A deficiency of vWF (can't fold a scaffold to which platelets can bind)
Cause of Bernard Soulier syndrome?
A deficiency of GP1b-IX
What is thrombocytopenia?
Low Platelet count
Activation of factor VII requires
factor III (Tissue factor)
Activation of factor XI requires
factor XII
Activation of factor IX requires
factor XI
Activation of factor II requires
factor X and factor V
What's so special about Factor XIII?
Factor XIII is the only non-serine protease enzyme. This enzyme is a transglutaminase that forms cross-links in fibrin by removing NH3
Breakdown of fibrin requires which protien?
plasmin
Protein C (APC)
inhibits coagulation through the protein degradation of coagulation factor VIII and factor V
Cause of Hemophilia A
Deficiency in factor VIII
Cause of Hemophilia B
Deficiency in factor IX
The most common bleeding disorder
vWF disease
What are PGI2 and NO made by, what are they activated by and what are their functions?
Produced by the endothelial cells. Activated by APC (activated protein C) and is used to vasodialate.
Vitamin K Requiring Coagulation factors
Factor II, X, IX, and VII as well as protien C/S.
Warfarin
Inhibits Vitamin K Epoxide Reductase thus reducing the activation of coagulation factor II, X, IX, and VII.
Antithrombin III
Inhibits the activity of factor II or thrombin
Heparin
Induces antithrombin III's activity which inhibits thrombin's activity.
Prothrombin time (PT) measures
the extrinsic pathway & the common pathway
Activated partial thromboplastin time (PTT) measures
the intrinsic pathway and the common pathway
Endothelial cells inhibit coagulation by
producing PGI2, nitric oxide and inducing APC
Endothelial cells block platelet binding by being
negatively charged
The D-dimer test will measure the
breakdown of fibrin
Glanzmann's thromboasthenia affects what?
GPIIb
How does fibrinogen deficiency affects platelets?
fibrinogen is used to aggregate platelets TOGETHER, so a deficiency would decrease aggregation
What factors does Thrombin activate?
Factors V, VIII, XI, XIII(when calcium is present)
What makes a soft clot?
Fibrin (Factor I)
How is the secondary clot plug made?
when fibrin (factor I) hooks up with factor XIII
How does thrombin indirectly inhibit coagulation?
thrombin binds to the thrombomodulin receptor in the endothelial cells to activate protein C
Heme is a component of what 3 things?
hemoglobin
myoglobin
cytochromes
Glutathione (reduces/oxidizes) oxidizing agents

-pick the correct choice
reduces
In the process of reducing oxidizing agents, glutathione becomes (reduced/oxidized)

-pick the correct one
oxidized to become GSSG
What re-reduces oxidized GSSG back to glutathione?
NADPH
What happens to NADPH when it re-reduces GSSG to glutathione?
it becomes oxidized to NADP+
What pathway re-reduces NADP+ back to NADPH?
pentose phosphate pathway
When methoglobin reductase reduces Fe^3+ to Fe^2+, what molecule is oxidized in the process?
NADH
What pathway re-reduces oxidized NAD+ back to NADH?
glycolysis pathway
Is the porphyrin ring recycled?
No, it's degraded into bilirubin
Where does heme synthesis take place?
reticulocytes, hepatocytes and other heme synthesizing proteins
What 3 things are needed for heme to be synthesized?
Succinyl CoA
Glycine
Fe^2+
What is the rate-limiting step in heme synthesis?
Aminolevulinic Acid (ALA)
What is the cofactor of Aminolevulinic Acid (ALA)?
Vitamin B6
Question: The pKa of hydroxybutyric acid is about 4.7. At what pH (2 or 12) would almost all of the hydroxybutyric acid be protonated?
at pH of 2. (weaks acids need to be in stronger acids to get protonated)
Which amino acid enables intramolecular covalent bonding?
Cysteine. Disulfide bonds are covalent bonds
Purpuric spots suggest what?
thrombocytopenia
Name some things that cause or can cause defects in heme synthesis.
Lead poisoning
iron deficiency
porphorias
B6 deficiency
Thalassemia
With what method does Antithrombin III bind to thrombin?
irreversibly
Warfarin (increases/decreases) (PT, PTT)?

chose one answer from each parenthesis
increases PT
If a patient has insufficent NAD in their body, what do you give them to treat the problem?
Niacin (B3) supplements
Long term iron storage is made by possible by binding iron to ________
hemosiderin
ferritin is responsible for _______ _______ _______ of iron and assists in gut absorption
short term storage
Name for genetic defects in ENZYMES catalyzing heme synthesis, which can lead to neurological disorders
porphyria's
Iron is transported in the blood by being bound to _________
transferrin
Where is most of the iron stored in the body?
liver
What is responsible for the reddish color of stool?
bilirubin
Where is bilirubin produced in the body?
reticulo-epithelial cells and spleen
What is produced by hepatic conjugation of bilirubin?
bilirubin diglucuronide
What 2 things are needed for bilirubin to become conjugated?
1) glucuronyl transferase
2) UDP-substrate
What converts bilirubin to urobilinogen?
bacteria in the gut
Is presence of some indirect/unconjugated serum bilirubin normal?
yes
If indirect bilirubin is abnormally high in the serum, what 2 places could the pathology be?
pre-hapatic or hepatic pre-conjugated
If direct bilirubin is detected in the serum, what 2 places could the pathology be?
hepatic post-conjugated or
post-hepatic
Is presence of some direct/conjugated serum bilirubin normal?
No
What is the intermediate in the process of conversion of heme to bilirubin?
biliverdin

(heme-->biliverdin-->bilirubin)
Why does bilirubin need to be converted to bilirubin diglucuronide first before excretion?
bilirubin diglucuronide is more water soluable
How does heme synthesis play a role in globin synthesis?
heme stimulates synthesis of protein globulin by maintaining the ribosomal initiation complex for globin synthesis in an active state
How are glycolysis and the methemoglobin reductase reaction related?
they both reduce oxidizing agents
Are introns in both prokaryotes and eukaryotes?
no, only eukaryotes
The central dogma of biology
DNA -> RNA -> protein
HDAC
stands for histone deacetylase and is responsible for the deacetylation of histones (lysine residues), to make DNA INACTIVE
HAT
Histone acetyltransferase and is responsible for acetylating histones (the lysine residues), to make DNA ACTIVE
Define polycistronic DNA and what organisms they are found in
one mRNA transcribed MANY GENES
found in prokaryotes
What do prokaryotic operons contain?
promoter + structural genes
Regulation of chromatin structures achieved by regulating what?
enzymatic activity of HDAC and HAT.
Transcription Factors do what?
control gene expression by regulating or inhibiting gene transcription.
RNA processing involves what 4 steps?
7mG guanicine cap, splicing, polyadenylation, and RNA editing.
What are the four elements found within an intron?
a 5' & 3' splice site, branch point, and a polypyrimidine tract.
Can splicing be regulated?
ya
How is splicing regulated?
Through the activity of SR proteins and hnRNP proteins.
T/F - Alternative splicing is a way to increase protein diversity.
True
T/F - When iron levels are low, ferritin mRNA is less stable.
True
T/F - When iron levels are high, Ferritin mRNA is more stable
True
T/F - When iron levels are low, TfR (transferrin receptor) mRNA is less stable.
False
T/F - When iron levels are high, TfR mRNA is less stable.
True
T/F - Nonsense mediated decay is a pathway for regulating RNA degradation.
True
Prader-Willi Syndrome is a result of what
paternal deletion of chromosome 15 q11-q13.
Angelman syndrome is a result of what
maternal deletion of chromosome 15 q11-q13.
Where does DNA methylation commonly occur?
GC rich regions of the promoter.
T/F - DNA methylation is a method of turning off gene expression.
True
T/F - Transcription and Translation in Prokaryotic organisms are coupled?
True
T/F - In prokaryotes, a repressor is responsible for inhibiting gene expression.
True
HDAC will (inactivate or activate) gene expression.

-pick one answer
inactivate
HAT will (inactivate or activate) gene expression.

-pick one answer
activate
Tautomeric shifts produce what type of mutations?
transitions (electron exchange exchanges purines to purines, and pyrimidines to pyrimidines)
If an exon has an ESE sequence attached to it, will it be included or excluded in the final mRNA product?
included
Is phosphorylation a type of post-transcriptional or post-translational modification?
post-translational
T/F - Use PCR to amplify RNA?
False. PCR is used to amplify DNA only
F and V class ATPases are examples of what?
proton pumps
Liver damage is likely to cause an decrease of what in the body?
albumin
Which part of snRPS actually does the splicing?
RNA portion

(snRPS has an RNA and protein portions)
When there is low iron, IRE-BP are (bound/unbound) to transferrin IRE's.

-pick one
bound (binding prevents degredating of transferrin).
When there is high iron, IRE-BP are (bound/unbound) to transferrin IRE's.

-pick one
unbound (unbound IRE's lead to degredation of transferrin)
Schmid metaphyseal chondrodysplasia occurs when
AA codon is made into a stop codon (nonsense mutation)
Prader-Willi and Angelman syndromes are examples of what?
DNA Imprinting - male & female methylate DNA differently
Three characteristics of facilitated diffusion.
1) specific
2) increases rate of diffusion
3) diffusion rates are saturable
Name two differences between transport proteins and gap junctions
1) Gap junctions are cell-cell whereas transport proteins are cell-extracellular
2) Gap junctions are non-specific, transport proteins are
Name the 3 types of transport proteins
1) Channels
2) Transporters
3) ATPase pumps
T/F - Transporters require a conformational change in the protein to let things pass
true
Name the subunits used in P Type ATPase pumps and their function
has 2 α subunits that are used for transport & 2 β subunits used for regulation
What is the largest and most diverse class of ATPase pumps?
ABC class pumps (ATP Binding Cassette)
Name the ABC class pump in the liver that pumps out hydrophobic toxins out
MDR-1
In CFTR lungs, why does mucus become viscous?
Because NaCl can't go into the mucous and attract water.
What is the biggest reason people with CFTR die?
infection due to Pseudomonas aeruginosa (which is usually harmless in normal lungs)
Which confirmational state, E1 or E2 is the resting state?

At this state, which binding site is open (high or low affinity)?
E1

high affinity
In P Type ATPases, what chages the conformation from E1 to E2?
phosphorylation
In Na/K pump, how many of what are pumped into the cell, and what and how much is pumped out?
3 Na are pumped OUT
2 K are pumped IN
(for every ATP hydrolyzed)
In Na/K pump, what prevents K from building up inside the cell?
K channels
What is a receptor?
something that recognizes, binds a SPECIFIC ENZYME, and INDUCES A BIOLOGICAL RESPONSE
What are isoreceptors?
different receptors are bound and activated by the SAME ligand (produces different response)
Do carbohydrates act as ligands?
NO
T/F - Nuclear receptors are transmembrane receptors.
False
In nuclear receptors, hydrophobic ligand binding causes what to happen to the receptors?
dimmerization
What are the three parts to a transmembrane receptor?
1) ligand binding domain (hydrophilic)
2) transmembrane domain (hydrophobic)
3) signal transduction domain (hydrophilic)
Name 4 second messangers we studied
cAMP, DAG, IP3, Ca
Definition of second messanger
effectors that can change concentration
In TGFβ signaling pathway, what are the main receptor used?
Serine/Threonin Kinase receptor
In TGFβ signaling pathway, describe what is upstream and what is downstream of Type II receptors
Upstream - TGFβ ligand
downstream - Type I receptor
What is the TGFβ signaling pathway used to control?
Development and Cellular Differentiation (NO physiological role!)
In TGFβ signaling pathway, what needs to happen in order for Smads to associate with coSmads?
phosphorylation of Smads (AKA receptor regulated Smads) by Type I receptors
In the Ras pathway, what receptors are used?
tyrosine kinase
What does the Ras pathway control?
Physiologic things such as blood glucose levels. Also does development and cellular differentiation
In the Ras pathway, what happens to the TK receptors after the ligand binds?
The TK receptors form dimers
Transautophosphorylation occurs in what signaling pathway?
Ras pathway
In the Ras pathway, what happens after the TK receptors are phosphorylated?
they bind adaptor proteins
In the Ras pathway, adapter proteins recruit what?
GEF
In the Ras pathway, what happens when GEF is activated?
GEF recruits Ras(GDP) and plucks of the GDP
In the Ras pathway, what does RAS(GTP) phosphorylate?
RAF
In the Ras pathway, what does RAF do?
phosphorylates MEK
In the Ras pathway, what does MEK do?
phosphorylates MAP
In the Ras pathway, what does GAP do?
causes dephosphorylation of Ras(GTP) to Ras(GDP)
In the Ras pathway, what do phosphates do?
dephosphorylate downstream effectors such as Raf, MEK & MAP)
In the cAMP pathway, what receptor is used?
7-transmembrane receptor & G-proteins
What is the cAMP pathway used for?
physiological (glucose and metabolism), inflamation and menstrual cycle
In the cAMP pathway, which sections of the 7-transmembrane receptor interact with the G-protein?
C-terminus; loop between helix 5 & 6
In the cAMP pathway, which structures are attached in the inactive state?
GαGDP is attached to the 7-transmembrane protein in the inactive state
In the cAMP pathway, what happens when the ligand first binds?
GαGDP becomes GαGTP
In the cAMP pathway, what does GαGTP do?
activates adenylate cyclase
In the cAMP pathway, what is the role of adenylate cyclase?
convert ATP to cAMP
In the cAMP pathway, what is the role of cAMP?
acts as a second messenger to activate Protein Kinase A (PKA)
In the cAMP pathway, what is the structure of Protein Kinase A?
Has 2 R (regulatory) subunits and two C (catalytic) subunits
In the cAMP pathway, when Protein kinase is activated, what goes on to phosphorylate other things?

What gets phosphorylated?
the C subunit goes on to phosphorylate serine & threonine residues of specific proteins
In the cAMP pathway, what role does phosphodiesterase play?
Deactivates the cAMP pathway by converting cAMP to AMP
In the cAMP pathway, what role do GAP domains play?
GAP always changes GTP to GDP
In the cAMP pathway, what inhibits GAP domains?
cholera toxins (therefore, can't stop pathway)
In phosphoinositide pathway, what recetor/s are used?
USUALLY the 7-transmembrane receptor w the G-proteins.

BUT one class uses tyrosine kinase receptors
What is the phosphoinositide signaling pathway used for?
physiological (hormones, metabolism), transport protein activity, cardiac & neuronal electric signal propagation & mental health
In phosphoinositide pathway, if a ligand binds a tyrosine kinase receptor, what happens next?
TK receptor phosphorylates PLC
In phosphoinositide pathway, if a ligand binds a the 7-transmembrane receptor, what happens next?
the 7-transmembrane receptor phosphorylates GαGDP to GαGTP, which then goes on to phosphorylate PLC
In phosphoinositide pathway, once PLC is activated, what does it do?
catalyzes cleavage of PIP2 into IP3 & DAG
In phosphoinositide pathway, which of the following factors are integral membrane and which are released into the cytosol? (PIP2, IP3, DAG)
-PIP2 and DAG are integral membrane factors

-IP3 is released into the cytosol
In phosphoinositide pathway, what does IP3 do?
released into the cytosol to activate Ca channels which then go on to activate Protein Kinase C (PKC)
In phosphoinositide pathway, what does DAG do?
Activates Protein Kinase C (PKC) when it reaches DAG (allows it go go and activate effectors downstream)
In phosphoinositide pathway, what is the purpose of Protein Kinase C (PKC)?
it phosporylates various proteins such as transcription factors and enzymes to induce various cellular responses
In phosphoinositide pathway, how is the signal inactivated?
when IP3 & DAG rejoin to regenerate PIP2
In phosphoinositide pathway, what role does pertussis toxin play?
It prevents the release of the GDP from Gα, which blocks PLC activation and inhibits the pathway
In phosphoinositide pathway, what role does lithium play?
lithium blocks the enzyme that rejoins IP3 & DAG to reform PIP2
How is Gα similar to Ras & GAP?
Gα has domains that act like Ras (which binds GTP) and also has GAP-type domains, which induces hydrolysis of GTP to GDP
What is the abbreviation for phosphatidylinositol biphosphate?
PIP2
What is the abbreviation for diacylglycerol?
DAG
What is the abbreviation for inositol triphosphate?
IP3
How is proto-oncogene different than oncogene?
Proto-oncogenes are normal functioning genes that that have the ability to proliferate (can have a gain-of-function mutation)

Oncogenes are proto-oncogenes that have the gain-of-function mutation and can increase the chance of tumors
In Gel electrophoresis, does DNA travel from - side to + side, or + side to - side?
- side to + side. DNA itself is - , so it will travel to the + side
What is Southern Blot used to identify?
DNA
What is Northern Blot used to identify?
RNA
What is Western Blot used to identify?
Proteins (labeled with antibodies)
What are palindromes?

And could they be a problem?
A strand of DNA that reads the same as its complementary sequence when read backwards
CATATG
GTATAC (same as above, but backwards)

These are potential restriction sites!
What is DNA sequencing used for?
Determining the sequence of nucleotides in a DNA strand

Can be used in finding genetic disorders
What type of sequence/structure blocks the the translation of VEGF mRNA and also induces its degradation??
RNAi
What is the abbreviation for phospholipace c?

What signaling pathway is it used in?
PLC

Used in the phospoinositide pathway
What are quantitative multifactoral traits?
can be assigned a numerical value (IQ, height, etc)
What are qualitative multifactoral traits?
all or not phenomenon. Either occur or not. (heart attacks, neural tube defects)
If a genetic problem tells you the incidence of something, is it telling you the q^2, or q?
q^2
What is the primary assumption of the Hardy Weinberg model?
population is in EQUILIBRIUM
What are the secondary assumptions of Hardy Weinberg model?
These are mechanisms for changing gene frequency:

-Mutation
-Genetic Drift (plays a big role in small population)
-Migration
-Selection (fitness..must pass genes to be "fit")
What is a proband?
its the first family member identified with the inherited trait of interest
What are consanguineous individuals?
Any two individuals who are descended from a common ancestor
Vertical transmission in pedigrees is an characteristic of what inheritance pattern?
autosomal dominant
Horizontal transmission in pedigrees is an characteristic of what inheritance pattern?
autosomal recessive
A proto-oncogene
typically induces cell transformation
What is oncogenesis?
Oncogenesis is the development of cancer cells. Cancer cells commonly inactivate inhibitor growth signals, evade apoptosis, have a limitless replicative potential, invade tissues, a sustained mutagenesis and undergo angiogenesis.
List examples of oncogenes
Growth factors/receptors
Signal Transduction pathways
Transcription factors,
Cell cycle control proteins, DNA repair enzymes and apoptosis proteins.
By what mechanisms do proto-oncogenes become oncogenes?
1. A point mutation
2. Chromosomal translocation
3. Multiple copies of DNA
4. A frame shift mutation
Cyclin D does what?
regulates the G1 phase of the cell cycle.
Cyclin D binds to what in the cell cycle?
CDK4/6
CDI (also known as CDK-I..)
is inhibitors of the cell cycle. Cyclin-dependent inhibitors help to control the progression through the cell cycle.
p16 inhibits
the cylin D/Cdk 4/6 activation of RB.
p21 inhbits
most cylin/cdk complexes, thus it inhibits the cell cycle.
p53
monitors DNA damage and activates p21 to inhibit cell division, and initiates DNA repair.
Describe the make-up of Ras
is a monomeric G protein and a proto-oncogene.
pRB
stands for retinoblastoma and is an example of a tumor suppressor.
DNA repair is initiated by:
p53
Tumor suppressor genes become oncogenic:
by deletions, point mutations, or DNA methylation.
A loss-of-function mutation is an example of
tumor suppressors
A gain-of-function mutation is an example of
proto-oncogenes
MAPK activates
Fos (a transcription factor)
T/F - Ras induces cell proliferation.
True
T/F - Hedgehog is a protoncogene.
True
T/F - The EGF receptor is an example of a protooncogene.
True
What are the two types of mechanisms that induce apoptosis?
1. The mitochondrial pathway
2. The membrane receptor pathway
T/F - Activation of caspase-8 induces apoptosis
True
T/F - Activation of the death receptor activates apoptosis through the activation of caspase-8.
True
T/F - The activation of the mitochondrial pathway is elicited through the activation of Apaf-1, Cyt c, ATP, and pro-caspase-9.
True
Bcl-2 is what type of protein?
anti-apoptotic
Bax is what type of protein?
pro-apopotic
What is the multiple hit hypothesis?
The multiple hit hypothesis is an idea that more than one mutation is required for a normal cell to become a tumor.
List the chemicals that can cause DNA damage:
Benzopyrene(Cigarettes)
Aflatoxin B
reactive oxygen species
DNA crosslinking agents
Alkylating agents
and various other chemical mutagens
Xeroderma pigmentosum is caused by
the inability to repair thymine dimers which are caused by UV irradiation.
Telomeres
are multiple TTAGGG repeats found at the ends of chromosomes.
What is telomerase
is the enzyme responsible for creating telomeres and is often inactivated in adult cells, but activated during oncogenesis.
Define senescence
a limited capacity to divide before cells enter a non-proliferate state.
Burkitt Lymphoma is caused by what?
a translocation of the myc gene from chromosome 8 to 14.
Chronic Myelogenous Leukemia is caused by what?
by a translocation between chromosomes 9 to 22.
Cofillin and N-WASPS are involved in what process?
Invadopodia (tumors degrading the basement membrane of normal cells)
In the Hedgehog pathway, what is the proto-oncogene and what's the tumor supressor? (hedgehog or patched?)
Patched inhibits smoothed (the thing that starts the pathway), so it's a tumor suppressor.

Hedgehog inhibits patched, so the pathway can continue, so its a proto-oncogene.
What are Beta-catenins and what's their role in cancer?
are subunits of cadherins, and are found to be increased in cancer
Whats the roll of topoisomerases?
maintain a certain level of DNA supercoiling
What are Geimsa stains?
Stains the chromosome banding patterns