• 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

1/263

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;

263 Cards in this Set

  • Front
  • Back
what makes RNA more susceptible to degradation than DNA?
The 2' OH group.
what happens to uracil in DNA?
Deamination of cytosine to uracil is potentially mutagenic so Uracil bases in DNA are repaired
nucleoside
a base bonded to a sugar (minus the phosphate)
nucleotide
a nucleoside bonded to phosphate groups (by ester linkages)
secondary structures of DNA
hairpins: short turn with 6-8 nucleotides

stem loops: single stranded loop with hundreds of nucleotides
tertiary structures of DNA
pseudoknot - (like a figure eight)
how are three dimensional DNA structures formed
by hydrogen bonds
ribozymes
RNA molecules that catalyze reactions (the proof of the RNA world hypothesis)
ribopolymerase
a ribozyme that is an RNA-directed RNA polymerase
retrovirus
a virus containing RNA and reverse transcriptase. the RNA gets converted to cDNA in the host cell and integrated into the genome for stability.
three basic RNA types in bacteria
rRNA (80%)

tRNA (15%)

mRNA (5%)
shine delgarno sequence
a sequence of RNA that base-pairs with rRNA.

known to be purine-rich (A/G)

it is located upstream of the AUG initiation codon that codes for the fMet residue.
polycistronic
characteristic of most bacterial mRNAs.

it means that one mRNA can code for multiple proteins.

Intervening sequences between coding sequences serve as docking sites for ribosomes (shine delgarno sequences
what intitiates translation in eukaryotic mRNA?
the presence of the first AUG. it is recognized by the tRNA and then the ribosomes are brought over.
stop codons
UAA, UAG, UGA

these are read by specific release factors which causes the ribosomes to release the protein
release factors
proteins that recognize stop codons and cause the release of the protein from the ribosome.
how to detect introns by electron microscopy
hybridize an mRNA to genomic DNA. if there is one loop, then the DNA sequence is continuous.

if there are two loops (or more) then the gene contains an intron (or more)
prokaryotic ribosome structure
70S --> 50S + 30S

50S --> 23S + 5S + 31 proteins

30S --> 16S + 21 proteins
eukaryotic ribosome structure
80S --> 60S + 40S

60S --> 28S + 5.8S + 5S + 50 proteins

40S --> 18S + 33 proteins
tRNA structure
phosphorylated 5' terminus

amino-acid attachment site (-OH on an CCA) at the 3' terminus

cloverleaf pattern

anti-codon loop near the center

half of the molecule is base-pairing

4 dS regions stack to form an L 3D shape.
aminoacyl tRNA
a tRNA that is bonded with a residue.

the residue is esterified to the 2' or 3' OH of the terminal A of the tRNA
main difference between transcription/translation in prokaryotes and eukaryotes
prokaryotes: can take place simulatenously (no compartmentalization

eukaryotes: take place in different places so are spatially and temporally separated
other RNA that is important in protein synthesis in eukaryotes
snRNA: participate in splicing

microRNA: small mRNA that hybridize and inhibit the translation of complementary mRNA

siRNA: (small interfering RNA) can silence mRNA through similar mechanisms as miRNA

snRNP: ribonucleoproteins (i.e. ribozymes)
how to label the 2 strands of DNA
template strand (the strand that is complentary to the RNA)

coding strand (the strand that is identical to the RNA...except for the T to U)
what subunit of RNA polymerase (in bacteria) recognizes the promoter?
the sigma site (σ)
the subunits of RNA polymerase in bacteria
alpha (2 identical subunits)
beta (2 dissimilar subunits)
sigma
highly conserved promoter sequence in prokaryotes:
at -35: TTGACA

at -10: TATAAT (Pribnow box)
highly conserved promoter sequence in eukaryotes:
at -25: TATAA (TATA box)

sometimes there is a CAAT box at -75.

there are also enhancers that can enhance the binding ability of the polymerase
different sigma subunits in RNA polymerase
sigma 70: the default subunit

sigma 32: synthesized when the cell is starving, it specifically binds to genes that synthesize heat shock proteins (different promoter sequences)
the active site of RNA Polymerase
similar to DNA polymerase:
what types of modifications are done to mRNA in prokaryotes?
NONE!!
what types of modifications are done to mRNA in eukaryotes
5' cap

3' polyA tail

splicing

editing (deamination of cytidine)
how does RNA polymerase attach to the DNA?
the sigma subunit runs along the DNA until it reaches the -35, -10 promoter sequences.

then it recruits the other subunits to start transcription.
rna polymerase active site (prokaryotes)
there are 2 metal ions (Mn or Mg). One attaches to the enzyme and the other comes attached to the nucleoside

the main amino acid is aspartate.

The 3’ hydroxyl group of the growing chain attacks the alpha phosphoryl group of the incoming nucleoside triphosphate.
de Novo
means without primers.

in transcription, the RNA polymerase works without primers.
what is found at the 5' end of RNA (before modifications)
a triphosphate group

the first RNA base is usually a purine
what is the reaction that RNA polymerase catalyzes:
(RNA)n + NTP --> (RNA)n+1 + PPi

requires NTP and metal ions
what can RNA polymerase do other then catalyze the addition of nucleotides?
it unwinds DNA (about 17bp) and rewinds it in the rear.
transcriptional stop signal
the simplest version is a palindromic GC rich region followed by an AT rich region containing 4 Uracil residues

Polymerase pauses after synthesizing a hairpin – the U-A base pairs are weak – causing the nascent RNA to dissociate

the Rho signal (without it, the RNA would be much longer)
mechanism of Rho termination
Rho binds to C rich region of ssRNA and hydrolyzes ATP to dissociate the RNA
rifampicin
antibiotic that inhibits transcription by binding with the polymerase
actinomycin
antibiotic that inhibits transcription by binding tightly and specifically to double helical DNA preventing it from being an effective template for RNA synthesis.

can inhibit RNA transcription while retaining DNA synthesis!
how are rRNA and tRNA processed in prokaryotes
they are transcribed as one long transcript and then cleaved and modified.

rRNA: RNAase III cleaves the 5S, 16S and 23S

tRNA: RNAse P generates the correct 5' end

other enzymes add CCA to the 3' end and modify bases
how can nucleotides of RNA be modified?
can be methylated or isomerized
how many eukaryotic RNA polymerases?
3. Pol I, II, and III
RNA Pol I

what does it encode?
TF's?
Location?
Regulated?
synthesizes rRNA (except 5S)

located in nucleolus

has 2 TF's

not very regulated

rapid continous transcription
Pol II

what does it encode?
TF's?
Location?
Regulated?
synthesizes mRNA and snRNA's

located in nucleus

many TF's inhibitors and enhancers

highly regulated

transcription only occurs when proper proteins are bound to the promoter
Pol III

what does it encode?
TF's?
Location?
Regulated?
encodes tRNA and 5S rRNA

located in nucleus

has 3 TF's: TFIIIA, TFIIIB, TFIIIC

Regulated by DNA sequences within the transcribed region

no promoter region needed
what is common about all RNA polymerases in eukaryotes
TF's are needed in addition to RNA Pol for transcription to take place
alpha amanitin
poisonous mushroom has effects on RNA Pol:

I: nothing
II: inhibited by low concentrations
III: inhibited by high concentrations
CTD
carboxy terminal domain.

found at the 5' end of RNA Pol II

is inactive when not phosphorylated (i.e. during the preinitiation complex) can be phosphorylated by TFIIH which starts elongation

also plays a major role in post-transcriptional modifications (recruiting the necessary proteins)
similarities between eukaryotic and prokaryotic RNA Polymerases
eukaryotic has Beta like subunits and Alpha like subunits. but they then have many additional subunits
omega subunit
it is an essential subunit in eukaryotic RNA polymerase activity. however in prokaryotes, it is now considered part of the core for the polymerase but it is not essential for transcription. Rather it stabilizes enzyme and assists in assembly of subunits
Pol I transcription factors
upstream activator element (UAF) binds to UCE (upstream control element) of DNA. it is located at -100. it also binds to the core element directly upstream from the transcriptional start site

then it recruits selectivity factor I complex (SFI and TIF-IB) which then recruits the polymerase for transcription
Pol III Transcription factors
TFIIIC binds to the two control elements upstream of the transcriptional start site

it then recruits TFIIIB to bind to sequence 30 bp upstream from TSS

TFIIIB has TBP (interact w/ DNA) and BRF (interacts with Pol III)

TFIIIB then recruits Pol III for transcription

TFIIIA is only used for 5S transcription. it binds to DNA using zinc-fingers. it then recruits TFIIIC and then follows the normal transcription route
Pol II promoter/enhancer sites
TATA box and Inr.

housekeeping genes have neither and are transcribed at very low constant levels.

to get the highest level of transcription, should have both TATA and Inr

Dpe: downstream enhancer
Pol II Transcription Factors
TFIID (contains TBP and TAF's) bind to DNA at the TATA, Inr and Dpe sequences

then TFIIB interacts with C-terminus of TBP and DNA.

it then recruits TFIIF which is associated with Pol II.

Pol II comes with TFIIF, TFIIE, TFIIH, and SRB

TFIIE activates TFIIH

TFIIH has helicase activity, kinase (for CTD), and DNA repair and cyclin dependent activity.

SRB remodels chromatin, activates splicing, is connected to RNA Pol II through the CTD
TBP
TATA binding protein

binds TATA with high affinity and interacts with the minor groove and bends the DNA.

it interacts with TBP associated factors (TAF's) for modulation

C-terminus is highly conserved and N-terminus is not but both are neccessary
how does eukaryotic transcription occur after the preinitiation complex has completed
TFIIH hydrolyzes ATP and phosphodiester bond is formed.

then TFIIH phosphorylates CTD then ELONGATION.

then TFIIB and TFIIE leave

to dissociated RNA Pol II, CTD needs to be dephosphorylated.
6 steps in protein regulation
1) transcriptional control (***KEY***)

2) RNA processing control

3) RNA transport/localization control

4) translational control

5) mRNA degredation control

6) protein activity control
trans activators and cis regulators
proteins that bind to DNA sequences far away from the gene (or very close (cis)) but can help stablize the preinitiation complex and activate transcription
cis-acting elements regulatory role in transcription
- some are necessary for basal activity of gene (TATA box)

- some are designed for gene activation in response to changes in environment

- some are meant to be activated in specific tissues (tissue specific elements can bind and increase transcriptional rate)
how do DNA binding proteins interact with DNA?
most use the major groove because it has more connections available

- zinc finger (c2h2, c4, or c6)

- leucine zipper

- helix-loop-helix

- homeobox
transcriptional co-activators
proteins that bind to sequences that are very far away from the transcriptional site can still influence transcription by DNA looping and can interact with TF's and influence their activity and affinity
how to determine if a protein binds to a region of DNA
DNA footprinting
zinc finger
c2h2: 3 fingers
c4, or c6

each finger can contact three nucleotides in the major groove through alpha helix

can form heterodimers and inhibit
homeobox
popular by the fly having a leg in its face!

one helix (60 aa) fits in major groove of DNA. highly conserved
leucine zipper
2 domains dimerized. basic amino acids bind to the DNA.

The dimerization domain contains leucine or other hydrophobic amino acids every seventh amino acid which form hydrophobic bonds between monomer helices - coiled coil.
helix-loop-helix
Helix-loop-helix motif has an N-terminal basic helix, a middle loop region, and a C-terminal region with hydrophobic amino acids spaced at intervals characteristic of an amphipathic alpha helix.

kind of like leucine zipper

can form heterodimers and inhibit!
use of heterodimers
expands the number of potential dna binding sequences.

they can only be formed between members of the same class!!
what is unusual about the 5' cap
usually, the 5' P attacks the 3' OH on the ribose.

here the 5' P (diphosphate) attaches to the alpha 5' P of the GTP and so there is a 5'-5' linkage as opposed to 5'-3' linkage

N-7 of G is methylated in all caps
difference between cap 0, 1, 2
cap 0: only N-7 is methylated
cap 1: N-7 and the first ribose (past GTP)

cap 2: N-7 and the first two riboses (past GTP)
tRNA modifications
- chemically modified bases

- 3' end cleaved and CCA added

- splicing (sometimes in euk)

- 5' end cleaved
rRNA modifications
- cleavage

- nucleotide modifications

- 5' splicing
5' cap's role
stabilizes mRNA

enhances translation
role of polyA tail
NOT needed for transport

enhances translation **KEY

stabilizes mRNA
how is polyA tail added
the termination sequence of RNA is A rich and recognized by an endonuclease which cleaves it and another enzyme adds 250 or so A using ATP!
how can you have two identical pre-mRNA's encoding proteins of different lengths?
alternative splicing...

OR cytidine deamination (CAA-gln UAA-STOP!)
typical intron sequence:
upstream exon AG GUAAGU..........Branch site/A...........(Pyrimidine)10 NCAG G downstream exon
how does intron begin and end
begins with GU and ends with AG
what is upstream exon sequence, branching sequence, and downstream exon sequence in splicing
usptream: AG

branch: A

downstream: G
spliceosome
A large complex of proteins and small RNA responsible for splicing.
spliceosome reaction
1. Cleavage of phosphodiester bond between exon 1 and
the 5’ end of the intron

The attacking group is the 2’-OH
group of an adenylate residue in the branch site.

2. A 2’ - 5’ phosphodiester bond is formed between A and
5’ terminal phosphate of the intron (transesterification). this results in the release of exon 1

3. The 3’-OH terminus of exon 1 attacks the
phosphodiester bond between the intron and exon 2, to join
exon 1 and 2 and the intron is released.
snRNP
small nuclear ribonucleoproteins

they are RNA's in complex with proteins
alternate splicing
in a gene with X introns (or X+1 exons) the number of possible mRNAs due to alternate splicing are

2 ^ X
what is the 3' RNA sequence that signals the endonuclease to cleave and then attach a polyA tail?
AAUAAA
rRNA splicing
can be self-spliced. the introns are called group I introns.
rRNA processing
Pol I --> 45S

45S --> 28S, 18S, 5.8S

18S + proteins --> 40S

28S + 5.8S + proteins --> 60S
what enzymes process rRNA
sno RNPs modify ribose and base components in the rRNA (e.g. aiding methyl groups and changing uridine to
pseudouridine)

nucleases cleave the large precursor to the three mature RNA's.
tRNA processing
1. The 5’ leader is cleaved by RNAse P

2. The 3’ trailer is removed and CCA is added by CCA adding
enzyme

3. Ribose and bases are modified (as the rRNA)

4. Many eukaryotic pre-tRNA are spliced by an endonuclease and a ligase to remove introns

1 in every 10 bases are modified
β-galactosidase:
enzyme that converts lactose into glucose and galactose.

there are small amounts of allolactose (glucose+galactose in a 1,6 bond)

encoded by Lac Z
Galactoside permease:
protein that transports lactose across the bacterial membrane

encoded by Lac Y
Thiogalactoside transacetylase:
detoxifies compounds that are brought in by permease

encoded by Lac A
operon
A transcription unit in prokaryotes that consists of regulatory regions and several genes in a cluster.
basic fundamental functions of the lac operon in the presence/absence of lactose
in the presence of lactose, the
genes Z, Y, A are activated.

In medium containing glucose
the activity is turned off within minutes.
how can one follow beta-galactosidase activity?
IPTG: is a small molecule that resembles lactose and can greatly induce the expression of beta-galactosidase

also, X-GAL is transported by lactose permease and hydrolyzed
by β-galactosidase, producing a blue product which gives the colony a blue color.
2 types of regulatory proteins in bacteria
- positive acting proteins

- negative acting proteins:

Negative-acting proteins are called Repressors, and the DNA site to which they bind is call Operator. The operator sequence is always close to the promoter sequence.
repressors
they are negative acting proteins that bind to an operator which is always close to the promoter sequence.

they bind to effectors

Effectors can be inducers

Effectors can also be Corepressors to activate the repressor
effector
compound that can bind to a repressor and either activate it or deactivate it.
in absence of lactose, how does the lac operon get affected?
the repressor mRNA is transcribed using the first promoter.

The repressor protein binds to the operator and prevents transcription of the downstream promoter
what happens to lac operon when lactose is present
lactose acts as an effector (inducer) to repress the repressing activity of the repressor.

it is a natural inducer. thus the Lac A, Y, Z can be expressed
cAMP and the lac operon
in the absence of glucose, cAMP levels INCREASE

high cAMP levels are a signal for starvation and induce beta galactosidase expression but cAMP alone is NOT enough.

need CRP (cAMP Receptor Protein aka CAP)

cAMP-CAP and RNA polymerase bind to each other weakly in the absence of DNA.

By itself, RNA polymerase binds fairly weakly to the lac promoter.

However, when cAMP-CAP and RNA polymerase bind to the lac operon together, they stimulate each other’s binding
dibutyryl cAMP
acts as a transcriptional activator for the lac operon.
cooperative binding
situation where two proteins bind with a greater affinity to a substrate when they are both present in solution (i.e. cAMP-CAP and RNA Pol)
two lines of defense of the human body
innate immune system: responds rapidly to features present in many pathogens

adaptive immune system: responds to specific features in a given pathogen
how does the immune system work?
identifies the features on disease-causing organisms and then works to eliminate them.
specificity and self tolerance (immune system)
specificity: recognizing foreign antigens

self-tolerance: destroying them without harming the host
examples of the innate immune response
monocytes, macrophages, dendritic cells, natural killer cells, granulocytes
examples of the adaptive immune response
B lymphocytes

T lymphocytes
two lines of defense of the human body
innate immune system: responds rapidly to features present in many pathogens

adaptive immune system: responds to specific features in a given pathogen
how does the immune system work?
identifies the features on disease-causing organisms and then works to eliminate them.
specificity and self tolerance (immune system)
specificity: recognizing foreign antigens

self-tolerance: destroying them without harming the host
examples of the innate immune response
monocytes, macrophages, dendritic cells, natural killer cells, granulocytes
examples of the adaptive immune response
B lymphocytes

T lymphocytes
immunodeficiency
the breakdown of the immune systems ability for specificity
autoimmune
break down of self-tolerance
what is the rate limiting step in protein folding?
from secondary to tertiary structure
domains of hsp 70
ATPase domain, peptide binding domain, and a cap
atp binding region of hsp 70
highly conserved region.

requires Mg and K
peptide binding domain of hsp 70
highly variable

binds short hydrophobic regions
dnaK
another name for hsp70 in bacteria
why are the experiments done on protein refolding different than the real deal?
in the real deal, the protein is coming out slowly from the ribosomes, and you want to make sure that no unwanted folding takes place until the entire protein is out.
what happens if the protein does not fold correctly
hsp 70 recognizes this by the hydrophobic regions still sticking out, so hsp70 rebinds and then gives the protein another chance!
hsp70 bound to adp vs atp
when bound to adp: the latch is closed and the polypeptide cannot break free

when bound to atp, the latch is open and the polypeptide can break free
DNA J
peptide that works with hsp 70. it binds to the DNA and recruits the hsp70 and promotes atp hydrolysis to latch onto the protein
peptidyl-prolyl isomerase
enzyme that sits at the end of the ribosome to isomerize certain residues before hsp70 latches to it.
what does hsp70 stand for? why?
heat shock protein.

because it protects proteins during heat shock. it binds to the denatured proteins to prevent unwanted interactions between denatured proteins.
hsp 10
the cap for the hsp 60 chaperone
how does hsp60 work?
it has 7 subunits. inside there are hydrophobic regions
how is hsp60 different from hsp 70
it doesn’t bind to COMPLETELY unfolded protein. It likes preformed secondary structures unlike hsp70.

also it completely encases the protein unlike the hsp 70
aggregates
form when misfolded proteins accumulate.

small aggregates are far worse then large ones. they form plaques.

many cells have machinery to prevent the aggregates (except animals)
how to get rid of aggregates?
use hsp 104 (looks like hsp60)

has slicers on its side to pick up and carry away the small misfolded proteins

binds atp. when it binds, the two rings grind against one another

in animals the aggregates are brought using microtubule/dynein to the centrosomal area and can harmlessly accumulate there
what is the difference between cytosolic and non-cytosolic environment
cytosolic: oxidizing environment (i.e. bad for cys and met)

non-cytosolic: reducing environment
how to measure the rate of degradation of a cell?
flood a cell with radioactive S and then measure the rate of excretion (this is the rate of degradation)
what is the probable function of short-lived proteins
rapid response
what is the probable function of long-lived proteins
a protein that should remain at constant levels.
Jun protein function
a transcription factor for stress response
major differences between cellular digestive and organism digestion?
specificty, processivity, and energy consumption
specificty (digestion)
stomach: none! want to chew everything up!

cell: YES!! must be very specific so that the cell isnt eaten!
processivity (digestion)
stomach: doesnt have to be completely processed, there could be fragments.

cell: must be completely broken down into aa and other molecules
energy consumption: cell v gut
stomach: no energy used

cell: uses energy
clpP
degradation protein in bacteria

has a chamber (like hsp proteins) but has scissors inside to cut the protein. this is all energy independent.

the energy is needed to selectively bring the protein in!
clpA
part of protein degradation in bacteria.

sits on top of clpP. responsible for bringing in the proteins that need to be degraded.

it is like a chaperone that recognizes the protein, and unfolds it and sends it into clpP.

there are a variety of homologs of clpA that recognize different signals for degradation
Lon protease
very large protease.

pretty much a combination of clpA and P.

has both chambers for bringing proteins in and digesting them.

could be an evolutionary precursor to regular chaperones since if a mutation occurs in the protealytic site, than it would behave much like hsp60!
2 sites of degradation in eukaryotes
proteasome and lysosome
which proteins are sent to proteasome
-short lived regulatory proteins

-small amount of long-lived proteins

-abnormal proteins
which proteins are sent to lysosomes
-most long-lived proteins

-membrane proteins

-aggregated proteins
ubiquitin
a polypeptide shaped like a noose

it marks substrate proteins for degradation. To be delivered to proteasome, it needs to be covalently attached to multiple ubiquitins
most important residues on ubiquitin
glycine residue: to attach to cys of E1(c terminus)

lysine residue: to attach to the next ubiquitin
sequence of events to ubiquitination
E1 binds to U.

then it transfers U to E2.

E3 meanwhile, binds to the protein and then E2 finds E3 and transfers the U to the protein--attaches to lysine on the protein
how many E1, E2 and E3's are there and why?
E1: 1
E2: a few
E3: thousands

the reason is because you only need one E1 and a few E2's to recognize the thousands of E3's which need to vary so that it can bind to different domains on degradable proteins
structure of proteasome
4 subunits. in bacteria they are all the same but in eukaryotes they can be different.

the beta chambers in the middle are for degradation. but the protein must be denatured first.

another peptide (analogous to clpA) recognizes the ubiquitin markers and binds the protein and denatures it. it then sends it to the 20S proteasome for digesiton.
functions of the lid (19S structure that assists the proteasome)
recognize the 4 ubiquitin chain.

recognize substrate and transfer to the base

isopeptidase (destroy the peptide bond between the ubiquitin branches)

release the free ubiquitin.
how does proteasome chew a protein?
like a dog. first cuts into large pieces then smaller.

the first bite is with trypsin (arg, lys)
HDAC 6
histone deacetylation complex

doesnt actually do that since it is in the cytosol.

binds to ubiquitin and sends it to dynein to carry off to the aggresome.
how can ligands affect expression?
Protein-protein interactions occur between DNA binding proteins and co-activators (i.e. the ligands) to regulate transcription.

sometimes the ligand can cause the release of a repressor or activation of an enhancer,

or the opposite.
glucocorticoid receptor and how it works with nuclear localization
wihtout the ligand, there is a repressor bound to the protein.

when the ligand binds, the repressor leaves and exposes the NLS. so the complex goes to the nucleus and regulates transcription
what happens after chromatin is activated
transcription factors can bind, and cis-acting proteins, mediators, co-activators...

transcription begins!
histones
proteins that help bundle up DNA into inactive chromatin. they interact with DNA through the minor groove

4 different kinds. H1-H4

H1 is the most unique, it forms as a linker between nucleosomes
dna interaction with histones
histones bind at the minor groove.

DNA surrounds the 4 core histones with an amino tail protruding out.

this tail is important in docking proteins
HAT
histone acetyl transferase.

acetylates histones which activates the chormatin to allow for transcription.

does so by transferring acetyl from acetyl CoA to the lysine of the amino tail of the histones which decreases affinity for DNA and increases affinity for bromodomains which are characteristic of many TF's and other DNA binding proteins
TFIID and histones
TFIID contains a bromodomain and an acetyl transferase.

so it is capable of activating chromatin and recruiting Pol II
2 ways of activating chromatin
- acetylation

- chromatin remodeling (must be acetylated first before recruitng the remodeling engine)
2 ways of deactivating chromatin
- deacetylation

- remodeling
epitope
region of the antigen that binds to the antibody
how does epitope interact with antibody?
non covalent interactions
where are t and b cells synthesized?
in the central lymphoid organs: bone marrow
where are t cells maturized
in the thymus
where do t and b cells go after syntehsis
to the spleen and other peripheral lymphoid organs and filter the blood for antigens
constant regions of the heavy chain?
responsible for antibody-antibody interactions such as dimerization or pentamerization...

hold the chains together

also, carbohydrates are present to stimulate dimerization and phage recognition
variable regions of the chains
responsible for binding the antigen
secondary structure motif of b cells
beta sandwich. many similar antiparellel beta sheets with loops that are variable.

these loops are where the epitope binding occurs
CDR
the loops of the secondary structure of the b cell.

these are responsible for the variety in b cells
avidity vs affinity
avidity: the strength of an antigen binding to the antibody on both of the variable chains.

affinity: the strength of an antigen binding to the antiboy on one chain

avidity is hundreds times stronger than affinity
Fab
the two slants of the Y of the b cell.

THese are the parts of the b cell that have the binding to the antigen
Fc
the stem of the Y of the b cell

mostly repsoible for dimerization and such
IgA

-secreted form
-subclass
-H chain
-L chain
dimerized with J

has IgA1 and IgA2

H chain has alpha1 or alpha2
L chain has kappa or lambda
IgM

-secreted form
-subclass
-H chain
-L chain
penatmeric with J

none
H chain has Mu

L chain is kappa or lamnda
IgD

-secreted form
-subclass
-H chain
-L chain
monomeric

none

H chain has delta

L chain has kappa or lamda
IgG

-secreted form
-subclass
-H chain
-L chain
monomeric

none

IgG1-4

H chain has gamma1-4

L chain has kappa or lamda
IgE

-secreted form
-subclass
-H chain
-L chain
monomeric

none

H chain is epsilon

L chain is kappa or lamda
different Ig isotypes

what determines them?
IgA, IgD, IgE, IgG, IgM

the amino acid sequence of the CH chain determines which!
B cell differentiation to antibody secretion
as B cells proliferate, they become B blasts with less memrbane bound antibodies

and then eventually become plasma cells which secrete IgM class first!
somatic hypermutation
when a B cell gets activated by an antigen, it proliferates and an enzyme AID (activation induced deaminase) helps the activated B cell hypermutate in the hopes that one mutation will develop a greater avidity/affinity for the antigen and will become the new primary antibody
AID
activation induced deaminase

an enzyme that helps b cells hypermutate by deaminating cytidine to cause transition mutations
how many classes of antibodies can a single b cell release
1
how many antibodies are present in an immune response
many
which class of antibodies are responsible for the primary response?
IgM
which antibodies are responsible for the secondary response
IgG, IgA, IgE. and they have greater affinity so this makes the secondary response greater.

example of memory
t cell receptor

2 different types
2 chains with globular domains.

V and C regions

contain CDR's

1) alpha-beta
2)gamma-delta
t cells
they do not secrete anything, rather they bind to the antigen and either kill it themselves or recruit other molecules to kill it
how do t cells work?
each TCR is capable of recognizing foreign proteins that are held in the MHC (major histocompatibility complex).

they then bind to it for an immune response
MHC
major histocompatibility complex, brings samples of intracellular proteins out of the cell and holds them in place outside the cell for recognition
which receptors have a VJ chain

which have a VDJ chain
VJ: B--> L chains (kappa and lamda)
T: alpha and gamma

VDJ: B-->H chains
T: beta and delta
how does the C region go?
mu-delta-gamma3-gamma1-alpha1-gamma2-gamma4-epsilon-alpha2
how does rearrangement go at the site of the DNA locus
All the genes upstream and downstream of the rearranging genes remain in the locus
sources of diversity in T and B cells
(9 total)
combinatorial: alignment, cleavage, and religation

inexact joining: deletion/insertion, extra D or no D region (T only) or reading of D in all three regions (T only)

random chain association in TCR's
(1000 alpha x 1000 beta = 1,000,000 possible TCR)

class switching (B only)
somatic hypermutation (B only)
where does somatic hypermutation and class switching occur?
peripheral lymph organs
peripheral lymph organs
spleen, lymph, tonsils, peyers patches
where are switch regions located in b cell blasts?
after every constant region except in between mu and delta
DNA locus of rearrangments
contains all the V regions up to the V of choice.

contains only the D region of choice (if it has a D region)

contains the J region of choice and all J's after (they eventually get spliced)

contains the C region of choice and all C's after that
Primary RNA transcript of recombination vs mature RNA
primary: only contains the V of choice. only contains the D of choice, contains the J of choice and all J's after, contains the C of choice and all C's after

mature: only contains the V of choice, only contains the D of choice, only contains the J of choice, and only contains the C of choice.
3 stop codons
UAA, UAG, and UGA
wobble
explains how one tRNA can recognize multiple codons.

the third codon doesn't bind well with the tRNA because it is very far away and the two compounds react in a strict orientation, so it is mostly irrelevant
how can nucleotides interact in a wobble?
in the anti-codons:

G--> C or U
C--> G
A--> U
U--> A or G
I--> A, U or C
reaction of aminoacylation:
ATP + amino acid --> AMP-AA + PPi

AMP-AA + tRNA --> AA-tRNA + AMP

tRNA binds to AA at the 3'OH
mechanism of amino-acylation reaction
one enzyme does everything. the 2 step process increases the fidelity of the reaction

the enzyme binds an aa and makes aminoacyl adenate. there is specificity in recognition

After that, the enzyme binds tRNA and this leads to conformational changes in enzyme. Enzyme pauses for a little and decides whether this particular aa corresponds to this particular tRNA.

If it understands that it’s a match, it makes the finished product.
why would aggregate form in purkinje cells more often then other cells?
bc purkinje cells do not divide and so the aggregates are not dilluted.
prokaryotic ribosomes
70S --> 50S and 30S

50S --> 23S+5S+34 proteins

30S --> 16S + 21 proteins

the RNA are the catalytic parts. the proteins are there for structure.

in fact, if there were no proteins, the ribosome would still assemble
where does the mRNA attach to the ribosome? (pro)
30S subunit
3 steps of translation in pro karyotes?
initiation, elongation, termination
initiation of translation in prokaryotes
30S subunits slides upstream until it reaches AUG (met) next to the shine delgarno sequence that binds with the 16S rRNA.

special initiator tRNA is used for the first aa.

IF2 binds GTP+fMet+tRNAf (cannot recognize any other tRNA)

IF1,3 bring in mRNA and 30S complex

then 50S joins and IF1,2,3 leave and GTP is hydrolyzed

the formation of 70S marks the end of initiation
3 spots in the 50S subunit of ribosome (prokaryotes)
A(aminoacyl), P(peptidyl), E(exit)
elongation in prokaryotes translation
Ef-Tu binds with aa-tRNA and GTP and brings it to the A spot
(Tu interacts with any tRNA except tRNAf)

if the tRNA matches with the codon of the mRNA in the A spot, then GTP will hydrolyze and "click"
if not, the aa-tRNA will dissociate.

the GTP hydrolysis causes GDP+EF-Tu to leave and tRNA-aa stays in the A spot.

then the peptide in P spot attacks the aa in A spot. (transpeptidation)

Now there is an empty tRNA in P spot and peptidyl-tRNA in A spot.

Ef-G with GTP binds to the A spot of the ribsome, hydrolyzes GTP and makes the mRNA (with attached tRNA's) move one spot over in the 5' direction. (translocation)

tRNA in E spot leaves, and there is an A spot open and P spot has peptidyl-tRNA
what reactivates EF-Tu+GDP?
Ef-Ts
translocation (prokaryotes)
Ef-G binds and hydrolyzes GTP to make mRNA shift one spot over and cause all the tRNAs to shift one spot over.
termination (prokaryotes)
Rf1: UAA, UAG
Rf2: UAA, UGA

when stop codon appears, the Rf attaches instead of tRNA

recruits Rf3 which causes ribosomes to detach.
Streptomyocin
antibiotic: in low concentrations causes ribosomes to make a lot of mistakes.

it interferes with binding of aa-tRNA into the A site.

If you increase the concentration, it will stop the binding of tRNAf and block translation
Tetracyclin
antibiotic: inhibits binding to A site
Erythromycoin
antibiotic: binds to 23S rRNA and specifically inhibits translocation step.
Chloramphenicol
antibiotic: inhibits peptidyl transferase on 50S subunit.
Puromycin
antibiotic: mimics aa-tRNA and can get transiently into A site instead of aa-tRNA. That ribosome will put the growing peptide on puromycin instead of aa-tRNA. But association of puromycin is transient and weak and readily dissociates with the bound nascent peptide!! If you add puromycin, you will have formation of a lot of premature terminations
important differences between prokaryotic and eukaryotic translation
1) euk takes place in different compartment so there is more added levels of regulation. while in pro, translation takes place during transcription so there is no regulation.

2) euk mRNAs have longer half-lives and are more stable

3)euk are always monocistronic and pro are always polycistronic

4) euk mRNA is modified, pro does have a polyA tail but it specifically signals for degradation!

3)
mature mRNA in eukaryotes have:
5' cap
3' polyA tail
5' UTR with the kozak sequence
3' UTR, ORF (starting with AUG)
kozak sequence, how is it different from shine delgarno?
sequence in 5' utr that signals ribosomes in eukaryotes.
A/GCCAC

different because it has no homology to the rRNA (unlike SD and 16S rRNA)
rather it increases affinity for transcription factors
eukaryotic ribosomes
80S --> 60S and 40S

60S --> 28S, 5.8S, and 5S + 50 proteins

40S --> 18S and 30 proteins
eiF-6
maintains equilibiurm between 80S and 60,40S by inhibiting the interactions between 60,40S subunits
translation pre-initiation of mRNA in eukaryotes
The 5' cap structure is recognized by eIF-4E.

4G (scaffold protien) then binds to 4E and mRNA.

4G then recruits 4A which has RNA helicase activity--along with cofactor 4B
translation pre-initiation of tRNA in eukaryotes
eIF2-GTP-Met-tRNAi bind to 40S with assistance from eiF-1,3,5.

this forms the 43S preinitiation complex
translation initiation in eukaryotes

mechanism?
take the 43S preinitiation complex and the activated mRNA complex together.

this makes the 48S initiation complex

the mechanism of interaction is between 4G, 3, and the polyA tail

48S complex scans mRNA for AUG. when AUG is found, there is a "click" and eiF-1 dissociates causing GTP hydrolysis.

this causes the release of all factors from the complex. eiF-2 has the GDP

then 60S subunit joins.
why is the 3'UTR so important for translation?
because of the polyA tail and its importance in combining the two pre-initiation complexes in the right spot. this brings the 3'UTR right at the beginning where its bound translation factors can work.
why is eiF-1 so important?
prevents the premature hydrolysis of GTP in the initiation complex

also releases mRNA from 40S subunit in termination
how is elongation and termination different in eukaryotes vs prokaryotes?
euk has eF1 (like Tu and Ts of pro) and eF2 (like G of pro)

Erf1 (binds to all stop codons unlike Rf1 and Rf2 in pro)

Erf3 (releases polypeptide in GTP dependent manner)

in euk: the ribosomes are not dissociated by factors, rather the polypeptide is dissocated from the ribosome.
why is eiF-3 important?
causes dissociation of 60 and 40S subunits in termination

also helps bind the tRNA complex to the mRNA complex in the right spot during initiation
reinitiation in eukaryotes
takes place in the few polycistronic mRNA's

occurs after eiF-3 causes dissociation but before eiF-1 breaks apart the mRNA from the 40S.
how do viruses affect translation in eukaryotes while still allowing translation of their own proteins?
first of all they inhibit eif-4G to stop initiation.

but they work because they have:
1) IRES site which binds with eiF-3 and bypasses the 4G requirement

or

2) they have polyA tail at the 5' end which bypasses the need for 4G or 3 altogether!
regulatory role of eiF-2 phosphorylation
three subunits:
alpha - gets phosphorylated
beta - holds together
gamma - interacts with tRNA, mRNA

if phosphrylated, cannot initiate translation and all translation stops.
if only 30% of eiF-2 is phosphorylated, then why is 100% of translation inhibited?
Because eIF-2B is needed to get rid of GDP and make GTP. and if the eIF-2 is phosphorylated, it will bind eif-2B and NOT LET GO.

there is a very small amount of eif-2B and once they are all bound up, 100% of translation stops
4 ways to phosphorylate eif-2
1) hemoglobin pathway: excess heme activate kinase to phosphrylate eif-2

2)GAAC pathway: if there is not enough aa's, then there will be many empty tRNA molecules. the empty tRNA molecules active GCN2 which is kinase that phosphorylates eIF-2 until enough aa's are available to inactivate GCN2 (GCN4 still translates at a higher rate to produce more amino acids)

3) dsRNA activates PKR which phosphrylates eif-2

4) PERK (activated during unfolded protein response) can phosphrylate eif-2
cells defense against dsRNA (3)
1) dsRNA activates PKR which phosphrylates eif-2 to stop translation

2) activates oligosynthetase which activates a nonspecific RNAase to kill all RNAs

3) activates RNAi
how does dicer work?
PAZ domain, 1 and 2 RNAase domains.

functions as a molecule ruler

cuts down dsRNA into two short ds fragments with overhanging ends.

dicer then carries the short ds fragments to RISC
what does RISC do?
RISC gets short dsRNA from dicer and unwinds into guide/passenger strand.

the RNA with the less stable 3'OH overhang becomes the passenger.

RISC+guide strand find complementary ssRNA and Ago2 subunit cleaves or blocks translation by binding to 3'UTR
what proteins are synthesized in the ER?
secreted and membrane bound proteins
why are stability requirements for proteins in ER so much higher then free ribosomal proteins
because these proteins can leave the cell, so need to be more careful.
how can secreted proteins be more stable
disulfide bonds (reducing env)
glycosylated
how does cell know what protein needs to be secreted?
signal sequence
what is a signal sequence
not a conserved sequence, rather a moderate hydrophobic region in the N terminus that signals the SRP to take the ribosome to attach to the ER.

it gets cleaved by signal peptidase once mature protein emerges in the lumen of RER.
SRP and how it stops translation
signal recognition peptide

recognizes the signal sequence and carries the unfinished protein to the ER for release into the lumen

consists of 7S RNA and p54 protein which has gtpase activity.

p54 binds to the hydrophobic region and another subunit of SRP inhibits eF2 (translocation) and stops translation
how does SRP interact with ER?
SRP interacts with the alpha receptor and hydrolyzes gtp.

this brings Sec61 translocon close to it and interacting with the ribosome so that it sits on top of it.

lastly, the now hydrolyzed srp loses all interaction affinity with the ribosome and the srp receptor on the er
bip and grp94
chaperones in the RER that are analogous to hsp70 and 90 respectively
modification enzymes in RER
chaperones (no hsp60 but yes hsp70-bip)

disulfide isomerase - to give disulfide bonds

N-glycosylation
how does protein become transmembrane?
during translation, a large hydrophobic sequence is translated and gets inserted into the membrane...no known mechanism...

can loop more then once.
type 1 vs type 2 transmembrane proteins
type 1: N terminus is in the lumen
type 2: C terminus is in the lumen
N-glycosylation vs O-glycosylation
N - done in the ER and on asn residue

dolichol phosphate attaches: 3 glu, 9 mannose, 3 n-acetylglucosamine to residue as it is translated

O - done in golgi and on thr or ser residues
calnexin and calreticulum
binds to sugar molecules on proteins in ER lumen
what happens if protein folds incorrectly in ER?
sugar molecules are cleaved off 1 at a time (3 glu and 1 mannose) and it is sent back out to the cytosol (ERAD)

after 4 times, it is sent to degradation (ubiquitin)
unfolded protein response
1) ATF 6 sensor binds with BiP. when Bip levels go down (meaning lots of unfolded proteins), ATF 6 gets sent to golgi and TF region is freed to go to nucleus and activate BiP transcription.

2) IRE1 has sensor region that binds Bip. when bip decreases, IRE1 can dimerize and cross phosphorylate which activates the RNAase regions that splices mRNA for HAC1 (unusual since splicing occurs in nucleues only)

the two exons are ligated by tRNA ligase.

and HAC1 is now activated and is a TF for expression of Bip and grp94.

also potentiates the erad pathway

lastly, PERK is activated to stop translation
cop II
proteins that coat the vesicles that transport between ER and golgi
how do you know cells belong in ER
they have KDEL sequence. if they get transported to golgi, then they get sent back to ER
cop I
proteins that coat vesicles that send between golgi and ER
tag that sends proteins to lysosomes
mannose-6-phosphate
MSF
mitochondrial import stijmulating factor.

recognizes the mitochnodrial import sequence: amphiphilic alpha helix with positive charges on one side and hydrophobic residues on the other side.

brings the protein over to the mitochondria
what is the role of SRB?
chromatin remodeler

co-activator

helps with splicing