Cumulative exam Flashcards

1
Q

direction of DNA sequence

A

written/read 5’ to 3’

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

nucleotide linkages

A

3’, 5’-phosphodiester bridges

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

RNA vs DNA

A
  • uracil instead of thymine in RNA
  • 2’ OH missing in DNA: this can normally facilitate hydrolysis of phosphate backbone, therefore DNA is more stable
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

division of common DNA/RNA bases

A

1) purines: two rings, includes adenine and guanine
2) pyrimidines: one ring, includes cytosine, uracil and thymine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

pyrimidine numbering system

A

1) assign priority to nitrogen adjacent to C=C
2) number in direction of second nitrogen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

purine numbering system

A

1) assign priority to the nitrogen in the larger ring, farthest away from the ring junction (C=c)
2) number in direction of second nitrogen in larger ring
3) move onto the smaller ring, number the double bonded nitrogen.
4) go clockwise for the rest of the atoms in the smaller ring

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

ribose sugar numbering

A
  • 1’ is anomeric carbon, continue clockwise
  • ’ is associated with sugars
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

nucleoside

A
  • base attached to sugar with beta-glycosidic linkage (anomeric carbon to N, aka N-glycosidic linkage)
  • purine: N9
  • pyrimidine: N1
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

nucleotide

A
  • base + sugar + one or more phosphate
  • phosphate attached to 5’ carbon through phosphoester link
  • phosphates attached to each other through phosphoanhydride link
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

phosphates in ATP

A
  • closest to ribose sugar is alpha, then beta, gamma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

why does DNA contain thymine?

A
  • cytosine can spontaneously convert to uracil (hydrolytic deamination)
  • DNA repair enzymes recognize these “mutations” to replace Us with Cs
  • thymine used in place of uracil in DNA to signal that T belongs in DNA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

purine derivatives

A

1) hypoxanthine: deaminate A
2) xanthine: deaminate G
3) inosine: hypoxanthine attached to ribose sugar

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

what is special about inosine?

A
  • wobble pairing: can base pair with C, A or U
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

pyrimidine derivatives

A

1) thiouridine: uracil carbonyl replaced with thiol
2) ribothymidine: thymine + ribose sugar
3) pseudouridine: uracil linked to ribose sugar at C5 instead of N1
4) dihydrouridine: hydrogenation of uracil double bond

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

where are pseudouridine and dihydrouridine found?

A

tRNAs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

timeline of DNA double helix discovery

A

1) nuclein
2) 4 distinct nuclein species identified (pentose + nitrogenous base)
3) carbohydrate or phosphate polymer
4) nucleosides crystallized, sugar-base connectivity defined
5) 1:1:1:1 base ratio incorrectly reported, led to tetrameric structure and cyclic tetramer theories
6) MW ~10^6 inconsistent with tetramer (heavier than expected), X-ray diffraction of extended structure found 3.4 angstrom spacing between nucleotides = penny stack
7) H-bonding discovered in enolic tautomers
8) 3-stranded structure, Chargaff reports
9) DNA double helix

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

phosphate-based polymer (Steudel)

A
  • linkages through phosphate
  • problems: no negative charge, base bound to sugar attached to phosphates
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

carbohydrate-based polymer (Levene/Jacobs)

A
  • linkages through carbohydrates (ribose sugars)
  • base/phosphate hang off that backbone
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

cyclic tetramer (Takahashi’s tetranucleotide hypothesis)

A
  • correct base-sugar-phosphate connectivity, but overall structure wrong
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

tetranucleotide hypothesis (Levene/Tipson)

A
  • linear, single-stranded tetramer units
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

single-stranded DNA structure (Astbury/Bell)

A
  • aka penny stacking model
  • problems: single stranded DNA, planar sugars and bases stacked on top of each other
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Gullard’s discovery of H-bonding

A
  • discovered in enolic tautomers
  • H-bonds were in the wrong locations
  • suggests possibility of multiple strands
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

why do tautomers have different H-bonding paterns?

A
  • enol form has OH (H-bond donor) instead of C=O (H-bond acceptor)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Pauling’s inside-out DNA structure

A
  • three strands
  • backbone on inside, but this would have too many negative charges and would be unstable
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Chargaff reports
- observed 1:1 ratio of A:T, G:C, purines:pyrimidines - facilitated understanding of H-bond pattern (Watson-Crick base pairing)
26
Watson-Crick base pairing
- 3 bonds between G-C - 2 bonds between A-T - purine uses larger ring (1, 2 for guanine only, 6) - pyrimidine uses face opposite to beta-glycosidic bond (4, 3, 2 for cytosine only)
27
DNA double helix
- Watson and Crick, using x-ray crystallography by Franklin - also paved the way for understanding DNA replication
28
forces that stabilize DNA structure
1) hydrogen bonding 2) dipole interactions 3) hydrophobic effect
29
hydrogen bonding in DNA
- between base pairs - three types 1) canonical Watson-Crick base pairs: large distance between sugar C1s 2) Hoogsten base pair: any other variation, smaller C1 distance 3) wobble base pairs: important in anticodon/codon pairing and degeneracy of codons
30
guanine H bonding in telomeres
- guanine rich strands can form quadruplexes using Hoogsteen hydrogen bonding - these can be found in telomeres
31
measuring DNA melting temperature
- denaturation causes A260 to increase (exposes inner bases that have rings)
32
DNA melting curve
- sigmoidal shape = cooperativity - melting temperature (Tm) is middle of curve, where there is 50% ds and 50% ss DNA - Tm is reflective of DNA stability
33
G-C vs A-T melting temperature
- G-C has more H-bonds so higher Tm
34
dipole interactions in DNA
- weak dipole-dipole interactions between adjacent bases (individually has minimal effect, but cumulative is stronger) - aka base-stacking - changes to atoms present can also affect Tm, ex. less polar = less dip-dip = lower Tm
35
non-polar solvents and DNA
- destabilize duplex DNA - indicates that H-bonds are not the primary force in DNA stability
36
hydrophobic effect in DNA
- most important driving force - aromatic hydrophobic base rings bury themselves inside DNA away from water, then base-stacking occurs - entropy driven: H-bond formation releases water molecules, also disruption of "iceberg" structure - more hydrophobic = more stable
37
DNA intercalators
- hydrophobic, planar agents that use the hydrophobic effect to insert between base pairs - can fluoresce (visualization), but destabilize DNA so that it can't be effectively used for replication or transcription (carcinogenic)
38
factors controlling DNA/RNA structure
1) base-sugar orientation 2) sugar pucker
39
base-sugar orientation
- rotation around glycosidic bond can lead to syn (same side) or anti conformation
40
syn vs anti base-sugar conformation
- syn less stable because of steric effects - syn more unfavourable for pyrimidines (basically does not exist) because of C=O
41
sugar pucker
- C2 or C3 points out (C-2' or C-3' endo) - C-2' favoured for deoxynucleotides (DNA), leads to larger interphosphate distance (7.0 angstroms) - C-3' in ribonucleotides (RNA helices), smaller interphosphate distance (5.9A)
42
types of helices
1) A-form (RNA) 2) B-form (DNA) 3) Z-form
43
A-form helix
- short and broad - 2.3A per base pair - right-handed helix - narrow and deep major groove - broad and shallow minor groove (protein-binding here) - anti base-sugar orientation - C-3' endo
44
B-form helix
- longer and thinner - 3.32 plus/minus 0.19A per base pair - right-handed helix - wide and intermediately deep major groove (protein-binding here) - narrow and intermediately deep minor groove - anti base-sugar conformation - C-2' endo
45
Z-form helix
- elongated and slim - 3.8A per base pair - left-handed - flattened out major groove on helix surface - extremely narrow and deep minor groove - anti base-sugar at C, syn at G - C-2' endo
46
protein-DNA interactions
- binding to major-groove with certain motifs (basic and polar AAs) - ex. Leucine zipper, helix-turn-helix (ex. Lac operon), zinc finger
47
eukaryotic chromosome structure
- very compressed (2m to 5micrometer diameter nucleus) - DNA wrapped around nucleosomes, packed in helical filaments, arranged in loops associated with nuclear matrix
48
nucleosomes
- fundamental unit of chromatin - 2x H2A, H2B, H3 and H4 create octameric core structures - (H1 is another histone that also exists, histones are positively charged) - DNA wound ~2 turns around core to create nucleosomes
49
structures in increasing compression in DNA
1) DNA double helix 2) beads on a string chromatin 3) solenoid (6 nucleosomes per turn) 4) loops (50 turns per loop) 5) miniband (18 loops) 6) chromosome (~10^6 stacked minibands)
50
RNA structures
- unlike DNA, complex secondary and tertiary structures are important for function - dsRNA structures: 1) transfer RNA (tRNA) 2) ribosomal RNA (rRNA) 3) catalytic RNAs (ribozymes) 4) riboswitches - ssRNA structures: 1) messenger RNA (mRNA) 2) microRNA (miRNA)
51
how do RNA structures form?
- complementary sequences can join via intrastrand base pairing - when base pairing is incomplete, bulges and loops can form (ex. hairpin stem-loop structures) - bases that stick out in bulges or loops can facilitate noncovalent interactions in tertiary structures
52
tRNA characteristics
- carry amino acids at 3' end to ribosomes for protein synthesis - each amino acid has at least one unique tRNA, sometimes more (because codon degeneracy) - small polynucleotide chains (73-94 residues) - contain modified bases and multiple interactions that create L-shaped molecule
53
tRNA primary structure
- reflect different amino acid specificities ex. anticodon region - some highly conserved regions (sequences and post translational modifications) that are important for overall structure and general function
54
tRNA secondary structure
- cloverleaf structure with four distinct base-paired regions + one variable loop: 1) acceptor stem: 5' and 3' end, tRNA nucleotidyltransferase attaches CCA to 3' end, terminal A attaches to AA 2) TψC loop: conserved T, pseudouridine, C 3) variable loop: lacks conserve sequence, varying length 4) anticodon loop: contains anticodon, which pairs with mRNA 5) D loop: conserved dihydrouridines
55
tRNA tertiary structure
- TψC loop and D loop brought into close proximity - stabilized by internal noncovalent interactions: 1) Watson-Crick base pairing 2) Hoogsten pairing 3) triple base pairing - also stabilized by: 4) divalent metal ion binding 5) polyamine binding
56
modified bases in tRNA
- function to facilitate specific interactions and prevent unwanted interactions - ex. methylation: 7-methylguanosine, 5-methylcytidine, 1-methylguanosine - also inosine and other derivatives
57
Hoogsten basepairs in tRNA
- G18:ψ55 - two H-bonds (NH2 on C2, N1) from guanine to one C=O (C4) on pseudouridine
58
how would pseudouridine synthase mutation affect tRNA structure or function?
- structure: different H-bonding pattern but minimal effect because only 2 H-bonds affected - function may be altered because ψ is conserved: could move H-bonding with ribosome (misaligned)
59
triple base pairs in tRNA
- U12:A23-A9 - U12:A23 = normal WC base pair - A9 binds entering through major groove with 1 H-bond to phosphate backbone, 2-H bonds to A base
60
divalent metal ions in tRNA
- Mg2+ interactions with RNA: 1) Mg2+/phosphate --> direct interaction 2) Mg2+/H2O/base --> indirect 3) Mg2+/H2O/2'-OH in sugar --> indirect
61
polyamines in tRNA
- have positive charge at physiological pH because amines protonated (pH < pKa) - positive charges interact with negatively charged phosphate backbone
62
rRNA characteristics
- about 2/3 of ribosome (other 1/3 is protein) - structural foundation for ribosomal proteins - contain certain modified nucleotides (ribothymidine, pseudouridine) - different species referred to by sedimentation coefficients
63
sedimentation coefficient
- aka Svedberg units - larger, more dense settle faster - dependent on surface area, so not additive, ex. 16S + 16S does not make 32S (would be less)
64
rRNA secondary and tertiary structures
- complex, vary between species - however, common function = homologous, similar structures
65
ribozyme characteristics
- RNA folding creates active sites - play a role in catalysis via hydrolysis or transesterification reactions in the absence of proteins (but metal ions may be important) - ex. self-splicing (removal of introns, alternatively done by spliceosome) - ex. RNAse P is a type of ribonuclease that processes tRNA 5'-ends (phosphate backbone cleavage)
66
riboswitch characteristics
- found on mRNA transcripts - binding of small molecules affects structure and can turn RNAs on/off
67
riboswitch transcription regulation
1) ligand binding to aptamer region 2) terminator stem loop region formed, signals stop to RNA polymerase 3) no transcription
68
riboswitch translation regulation
1) ligand binds to aptamer 2) ribosome-binding sequence hidden 3) ribosome cannot bind, no translation
69
aptamers
- ssRNA protein-binding sequences - can be synthesized, ex. as complementary to a biomarker (analogous to and cheaper than antibodies)
70
techniques for studying nucleic acids
- reading or locating sequences: 1) restriction analysis 2) synthetic DNA 3) blotting and hybridization 4) DNA sequencing - manipulating sequences (i.e. to study gene function) 1) PCR 2) site-directed mutagenesis 3) recombinant DNA technology 4) cloning
71
restriction nucleases
- discovered in bacteria - enzymes that recognize 4, 6, or more base sequences with 2-fold axis of symmetry - cleave to create blunt or sticky (overhangs) ends - endonuclease: cleave phosphodiester bond within sequence - exonuclease: remove from ends
72
restriction enzyme nomenclature
- 1st letter = genus - 2nd/3rd letters = species - 4th letter = strain (capital) - roman numeral for order of discovery
73
restriction enzyme types
- type I cleaves randomly - type II/II cleave at selected sites - type II have no ATP requirement
74
isoschizomers
restriction enzymes that recognize the same site
75
restriction enzyme binding
- bind as dimers (noncovalent binding)
76
gel electrophoresis
- samples loaded on to gel - negatively charged DNA migrates towards positive anode - larger fragments migrate slower - stain with intercalator to visualize - molecular weight ladder of known fragment sizes
77
restriction enzyme digest analysis
- using restriction enzymes to analyze DNA sequences
78
restriction enzyme analysis for disease dignostics
- can detect single base pair mutations that cause restriction cleavage sites to appear/disappear - can identify healthy, carrier or diseased individuals - ex. CTFR gene in cystic fibrosis
79
why is synthetic DNA needed?
- probes and primers for blots or PCR
80
solid phase synthesis nucleotide modifications
1) di-isopropyl group bound to the 3’ phosphate: good leaving group 2) β-cyanoethyl (βCE) group bound to the 3’ phosphate: protects backbone 3) dimethyoxytrityl (DMT) group bound to the 5’ end: protects 5'OH so that 3' phosphate reacts with another nucleotide (not itself) 4) base is also protected: preserve functional groups
81
solid phase synthesis steps
1) coupling: growing chain (first base bound to resin) 5'OH attacks activated monomer 3' phosphate = phosphite triester intermediate 2) oxidation by I2 = phosphotriester intermediate (more stable) 3) deprotection with dichloroacetic acid: removes DMT to facilitate addition of next nucleotide = elongated chain 4) after all bases added, other protecting groups are removed
82
solid phase vs natural DNA synthesis
- solid phase is reverse direction - leaving group in natural = pyrophosphate (PPi hydrolyzed to 2Pi drives reaction) - LG in solid phase is di-isopropyl group
83
southern blotting steps
1) cleave DNA (ex. with restriction enzymes) 2) separate fragments (gel electrophoresis) 3) transfer to nitrocellulose in alkaline solution (capillary action), this also denatures 4) incubate with radiolabeled probe 5) visualize with autoradiogram
84
southern blot probes
- generated from solid phase-synthesis - ideally want to bind with high specificity (good probe) - non-specific or imperfect pairing = bad-probe
85
southern blot application example
forensics --> DNA match of suspects to crime scene evidence
86
fluorescent in situ hybridization
- fluorescently tagged probes hybridize to chromosomes - visualized using fluorescence microscope - ex. used diagnostically to identify multiple copies of HER2 gene
87
DNA sequencing methods
1) Maxam-Gilbert 2) Sanger chain-terminator 3) automated 4) single-molecule 5) proton (electronic) 6) pyrosequencing
88
Maxam-Gilbert sequencing
1) radiolabel DNA sample for identification (5' P32 label) 2) react with different reagents - dimethyl sulfate (neutral) cleaves G - dimethyl sulfate (acidic) cleaves G and A - hydrazine + 1.5M NaCl cleaves C - hydrazine alone cleaves C+ T 3) each sample treated with piperidine: cleaves phosphate backbone 4) run samples on gel and visualize: 5' at bottom of gel
89
Sanger chain-terminator sequencing
- use DNA polymerase - add low concentrations of dideoxyribonucleotides that stop polymerization (also missing 3'OH)
90
Sanger sequencing steps
1) radiolabel primer 2) add ddNTPs, dNTPs and DNA polymerase: one tube for each; since ddNTPs are at low concentrations, they are randomly incorporated and can terminate synthesis at various points 3) electrophoresis and visualization: 5' at bottom, complementary to original template strand
91
automated DNA sequencing
- also uses ddNTPs, but each has a different colour fluorescent tag - gel electrophoresis with capillary gel in sequencing machine = high resolution reading of sequence - emitted photons upon excitation identifies base
92
DNA sequencing efficiency
- becoming more efficient and less costly - especially with next-generation sequencing techniques: single molecule, proton flow and pyrosequencing
93
single molecule sequencing
- fluorescently labelled dNTPs used to determine sequence as it is being synthesized - bases flowed in through nanopore one at a time - incorporation of base leads to pulse of fluorescence
94
proton (electronic) sequencing
- measures DNA sequencing byproducts (hydrogen and pyrophosphate) - flow bases one at a time, ion sensor detects byproduct release if base is incorporated (several in a row = multiple H+ released) - flow of H generates current
95
pyrosequencing
- measures PPi release - after released, sulfurylase incorporates APS (adenosine 5' phosphosulfate) to make ATP - ATP and luciferin --> oxyluciferin via luciferase, this emits light that is detected (proportional to initial amount of PPi released)
96
PCR reagents
1) dsDNA template 2) DNA polymerase, usually Taq because temperature resistant 3) Mg2+ 4) dNTPs 5) primers
97
PCR steps
1) denaturation (heat) 2) annealing of primers 3) extension by polymerase using primers, dNTPs and Mg2+ (cofactor) 4) repeat cycle to amplify (exponentially)
98
PCR application as a diagnostic tools
1) amplify regions of DNA that vary between individuals (ex. short tandem repeats or STRs vary in length aka variable number of tandem repeats/VNTR) 2) clone pieces of DNA using primers 3) introduce mutations (site-directed mutagenesis) 4) real-time PCR to detect and quantify presence of specific nucleotide sequences
99
site-directed mutagenesis
- mismatched nucleotide in primer added to sequence - PCR to make many copies of plasmid with new mutation
100
real-time PCR (qPCR)
- primer with reporter and quencher on different poles - quencher blocks reporter fluorescence because in close proximity - once synthesis done and primers are cleaved/released, reporter detected - measurements made during each cycle of polymerization: exponential increase in product can quantify the number of copies
101
diagnosis of CML using qPCR example
- primers designed to flank bcr-abl sequence - positive = fluorescence - negative = no fluorescence
102
recombinant DNA technology
- cleave DNA fragment and vector with restriction enzymes (flank sequence, best to have two different ones) - anneal DNA fragments and rejoin with DNA ligase - check for bacteria with vector (antibiotic resistance) - bacteria replicate DNA - fusion proteins can be expressed and purified
103
green fluorescent protein
- acts as reporter protein, identifying location of fusion proteins
104
RNA base nomenclature
- ribonucleoside: adenosine, guanosine, uridine, cytidine - ribonucleotide (nMP): adenlyate, guanylate, uridylate, cytidylate
105
DNA base nomenclature
- deoxyribonucleoside: deoxyadenosine, deoxyguanosine, thymidine, deoxycytidine - deoxyribonucleotide: deoxyadenylate, deoxyguanylate, thymidylate, deoxycytidylate
106
roles of nucleotides
1) precursors of DNA and RNA 2) universal energy (ATP, GTP) 3) components of cofactors (NAD+, FAD, CoA) 4) molecule activation (UDP-glucose, PRPP) 5) signal transduction (cAMP, cGMP)
107
nucleotide metabolic pathways
1) de novo synthesis: base/sugar from simple precursors 2) salvage pathway: recycle free bases and nucleosides from polynucleotide breakdown 3) nucleotide degradation: modification of bases to generate products for excretion or reuse
108
nucleotide metabolic pathway defects
1) de novo synthesis: orotic aciduria, causes anemia and growth retardation 2) salvage pathway: Lesch-Nyhan syndrome, causes neurological impairments and self-mutilation 3) nucleotide degradation: gout, causing uric acid crystal build up in joints and acute inflammatory arthritis
109
nucleotide metabolism overview
1) 70% tissue de novo nucleotide synthesis (in liver) and 30% dietary purines and pyrimidines 2) contribute to cellular nucleotide use, leads to... 3) complete pyrimidine breakdown or purine breakdown to uric acid 4) salvage pathways from 3) recycle for more cellular use 5) uric acid: 70% excreted through kidneys, 30% excreted through large intestine
110
common nucleotide metabolism intermediates
- pyrimidine: uridylate (UMP) - purine: inosinate (IMP) - xanthine, hypoxanthine
111
pyrimidine de novo nucleotide synthesis
1) bicarbonate + NH4 --> carbamoyl phosphate using 2 ATP 2) carbomyl phosphate + aspartate = pyrimidine ring 3) add sugar (PRPP) to make UMP 4) UTP --> CTP (RNA) 5) CTP --> dCTP, TMP (DNA)
112
purine de novo nucleotide synthesis
1) rings built off 5-phosphoriribosyl-1-amine (amine becomes N9, add glutamine, glycine, aspartate, N10-formyl-tetrahydrofolate) 2) IMP 3) ATP or GTP (RNA) 4) dATP or dGTP (DNA)
113
substrate activation by phosphorylation
1) ATP molecule to phosphorylate C=O, creating a good leaving group 2) nucleophilic attach from amine group to form C-N bond
114
displacement reactions and PRPP
1) ribose 5-phosphate --> phosphoriboyl diphosphate (PRPP) via PRPP synthetase (two Pi added) = pyrimidine pathway 2) PRPP --> 5-phosphoribosyl-1-amine: glutamine hydrolysis makes glutamic acid with amine group, amine attacks PRPP (PPi is good LG) = pyrimidine pathway, catalyzed by PRPP amidotransferase
115
AMP regeneration to ATP
AMP + ATP --> 2 ADP --> ATP
116
pyrimidine synthesis: production of carbamoyl phosphate
1) bicarbonate phosphorylated with ATP to make carboxyphosphate 2) NH3 from Gln displaces Pi from carboxyphosphate to make carbamic acid 3) carbamic acid phosphorylated with ATP to make carbamoyl phosphate
117
carbamoyl phosphate synthetase
- 3 active sites: 1) glutamine hydrolysis: hydrolyze NH3 from glutamine, add to carboxyphosphate 2) bicarbonate phosphorylation 3) carbamic acid phosphorylation - substrates travel through with noncovalent interactions
118
pyrimidine synthesis: ring formation
- rate-limiting step (regulated) 1) carbamoyl phosphate + Asp --> carbamoylaspartate + Pi via aspartate transcarbamolyase (ACTase) 2) ring formation: dehydration to dihydroorotate and redox reaction (CoQ or NAD+ reduced) to orotate (oxidized to make double bond)
119
ACTase structure
tetramer with 2 regulatory and 2 active sites
120
pyrimidine synthesis: UMP formation
1) orotate + PRPP --> orotidylate via orotate phosphoribosyltransferase 2) orotidylate decarboxylase: to UMP
121
nucleoside monophosphates to triphosphates
1) UMP + ATP --> UDP + ADP (uridylate kinase) 2) UDP + NTP --> UTP + NDP (nonspecific kinase) *all with delta G = 0 (break and reform phosphate bond)
122
pyrimidine synthesis: UTP to CTP
- cytidylate synthetase - substrate activation by phosphorylation (ATP) + ammonia from glutamine hydrolysis adds NH2 group
123
feedback regulation of pyrimidine synthesis
- CTP negative feedback (inhibits) to ATCase - ATP activates ATCase
124
pyrimidine de novo synthesis ATP cost
- 4 ATP: 2 for carbomoyl phosphate synthesis, 2 to make PRPP (ATP --> AMP functionally uses 2 ATP)
125
deoxyribonucleotide synthesis
- enzyme = ribonucleotide reductase (RR), which is diphosphate specific - ribonucleoside diphosphate + NADPH + H+ --> deoxyribonucleoside diphosphate + NADP+ + H2O - diphosphokinases add 3rd phosphate
126
ribonucleotide reductase structure
- R1 dimer: two alpha subunits, contains active site - R2 dimer: two beta subunits, contains tyrosyl-radical site (iron cluster to facilitate radical transfer to Tyr122)
127
ribonucleotide reductase mechanism
1) radical transferred from R2 to Cys439 in R1 2) radical transferred to nDP sugar 3' carbon (H donated to Cys) 3) radical promotes release of water (2'OH + H from SH on Cys462) 4) Cys225 donates H to 2' carbon, disulfide bond formed between Cys225-Cys462, radical transferred back to Cys439 5) NADPH reduces disulfide bond (thioredoxin reductase)
128
ribonucleotide reductase regulation
- R1 dimer contain two allosteric sites per subunit 1) specificity-determining site: - dATP/ATP: enhance pyrimidine (CTP, TTP) reduction - dTTP: inhibits pyrimidine reduction, promotes GDP reduction - dGTP: stimulates ADP reduction 2) activity-determining site: - ATP binding stimulates - dATP binding inhibits
129
pyrimidine synthesis: thymine problem
- DNA polymerase cannot distinguish between TTP and dUTP - dUTP converted to dUMP by dUTP pyrophosphatase (hydrolyses PPi) - methyl group added using tetrahydrofolate derivative by thymidylate synthase to make (d)TMP - TMP phosphorylated to TTP
130
TMP synthesis
1) enzyme Cys residue attacks C6 of dUMP 2) proton abstracted to enzyme Cys, hydride transferred to add methyl group, enzyme released
131
drugs targeting TMP synthesis
- to inhibit DNA synthesis, since thymidylate synthase is the rate-limiting step in DNA synthesis 1) fluorouracil inhibits thymidylate synthase 2) analogs of dihydrofolate: aminopterin, methotrexate (amethopetrin) --> inhibit dihydrofolate reductase (which makes tetrahydrofolate --> N5,N10-methylene-THF with Ser to Gly) - many off target effects: healthy cells, also tetrahydrofolate needed in other pathways
132
suicide inhibition of thymidylate synthase using fluorouracil
- fluorodeoxyuridylate has F instead of H: cannot be abducted to regenerate the enzyme - used for treatment of skin, colon, breast and stomach cancer
133
purine synthesis: inosinate formation parts
1) 5-membered heterocycle installed on ribose 2) additional 6-membered heterocycle formed to complete purine
134
inosinate formation part 1: 5-membered ring
1) glycine (N7, C4/5) attached to sugar using ATP 2) carbonyl (C8) added from 10-formyltetrahydrofolate 3) substrate activation by phosphorylation and glutamine hydrolysis to form C4=N 4) ring closure using ATP, C=N becomes amine group (N3) 5) bicarbonate (C6) activated by phosphorylation, attacked by C5) to start 2nd ring
135
inosinate formation part 2: 6-membered ring
6) bicarbonate C=O activated by phosphorylation, attached by Asp amino group 7) fumarate removed, only nitrogen (N1) remains 8) 10-formyltetrahydrofolate donates C=O by forming C-N bond (C2) 9) dehydration reaction closes ring
136
purine synthesis overall requirements
- 5 ATP - 1 Gly - 1 Gln - 1 Asp - 2 THF - 1 bicarbonate - produces 1 H2O
137
functionalization of purines: IMP to AMP
1) inositate carbonyl activated by phosphorylation (using GTP!), attacked by Asp amino group 2) fumarate released to make AMP
138
functionalization of purines: IMP to GMP
1) redox reaction (NAD+ reduced), inositate oxidized to form xanthylate (C2 carbonyl) 2) C2 carbonyl phosphorylated by ATP, glutamine hydrolysis adds amine to make GMP
139
purine synthesis regulation
1) excess IMP, AMP, GMP: inhibit PRPP --> phosphoribosyl-amine 2) AMP: inhibits IMP --> adenylosuccinate 3) GMP: inhibits IMP --> xanthylate
140
the DNA replication task (humans)
- 3x10^9 base pairs (haploid) - 10bp = 1 turn = 3.4nm/turn = 1.02m DNA per genome = 2.04m DNA per cell (diploid) - has to be replicated in S phase: 6-8 hrs
141
the cell cycle
G0: resting G1: growth and metabolism (6-12hrs, DNA = 2n) S: DNA replication, (6-8hrs, DNA = 2n to 4n) G2: growth of structural elements (3-4hrs, DNA = 4n) M: mitosis (1hr, DNA = 4n to 2n)
142
Lehman DNA polymerase assay components
1) E. coli: rapid growth cycle = constant DNA replication 2) ATP: energy for anabolic process 3) Mg2+: commonly associated with ATP 4) C14 labeled deoxythymidine: unique to DNA
143
Lehman DNA polymerase assay steps
1) combine radiolabeled T and E.coli with other components 2) add acid and centrifuge: kills E.coli, separates by solubility --> acid soluble nucleosides in supernatant, acid insoluble polynucleotides and proteins in pellet 3) radioactivity detected in pellet (0.01%), but T could be bound to proteins, not necessarily DNA 4) resuspend pellet 5) add DNase 6) add acid and centrifuge --> acid soluble nucleosides in supernatant (denatured DNA), acid insoluble proteins only in pellet 7) radioactivity (0.01%) detected in supernatant!
144
what was the issue with Lehman's initial assay?
- not enough DNA polymerization activity - needed to optimize
145
Lehman DNA polymerase assay optimization
1) better substrates: - thymidine kinase: deoxythymidine + ATP --> dTMP + ADP - nucleotide kinase: ATP + dTMP --> ADP + dTDP - nucleoside diphosphate kinase: ATP + dTDP --> ADP + dTTP = strongest signal = best substrate 2) P32 as more sensitive radiolabel than C14: - label gamma phosphate of ATP: added as alpha phosphate on dTTP, which remains when incorporated into DNA - also a better substrate
146
purifying DNA polymerase by size
1) fractionation column separated into simpler fractions 2) add radioactively labelled dTTP, Mg2+ and ATP 3) acid precipitation 4) check for which pellet has radioactivity: contains DNA polymerase and other necessary components for DNA polymerization
147
purifying DNA polymerase by solubility
1) add streptomycin (+) to precipitate (-) proteins 2) Lehman DNA polymerization assay on supernatant and resuspended pellet = no activity in individual, but activity in control *conclusion: need components from both for DNA polymerization
148
additional DNA polymerization components purified with fractionation
1) supernatant: - kinases: to convert dNMPs to dNTPs - dNTPs: all four needed 2) pellet: - DNA polymerase --> purified finally by ion exchange chromatography - DNA template
149
revising the hypothesis of DNA polymerization
- original thought: existing DNA chains elongated by DNA polymerase - however... 1) other dNTPs needed to allow dTTP to be incorporated 2) new DNA had same AT:GC 3) composition of new DNA not affected by dNTP concentration - revised: DNA polymerization is directed by a DNA template
150
other findings about DNA polymerase I
- can't start DNA chains de novo; needs a primer - synthesis in 5' to 3' direction only - not highly processive or rapid - contains both polymerase and exonuclease activity
151
DNA polymerization reaction
dNTP + (dNMP)n <-> (dNPM)n+1 + PPi --> 2 Pi 1) base-pairing between new substrate and parental (template) strand: ensures correct fit and brings two into close proximity 2) nucleophilic attack: 3'OH attacks alpha phosphate, leading to release of PPi (hydrolysis into 2 Pi drives reaction forward through equilibrium)
152
DNA polymerase fidelity factors
~1 error in 10^10 bases added 1) base-pairing: incorrect shape does not fit well (10^-2) 2) active site: shape = fit and amino acid interactions with P backbone (10^-3) 3) proofreading: exonuclease activity (10^-2) 4) mismatch repair: other enzymes recognize altered shape (10^-3)
153
DNA proofreading vs synthesis
*not reverse reactions - polymerization active site: (dNMP)n + dNTP --> (dNMP)n+1 + PPi (normally faster rate of reaction) - exonuclease active site (dNMP)n --> (dNMP)n-1 + dNMP (faster if incorrect base is incorporated)
154
DNA polymerase polymerization vs editing mode
1) polymerization: DNA 3'OH in active site (between fingers and thumb) 2) editing: distortion of DNApol due to incorrect nucleotide incorporation = 3'OH in active site of exonuclease domain (below palm)
155
is DNA polymerase I enough?
- too slow for E. coli replication rate (every 20 mins) - therefore there must be faster polymerases
156
searching for DNA polymerase mutants
1) random mutagenesis of E. coli strains 2) Lehman assay 3) detection of mutant with no DNA pol I activity --> proved DNA pol I is not essential for life, and therefore does not replicate the genome! 4) search mutant fraction for other polymerases: discovered DNA pol II/III
157
DNA polymerase type functions
I) prototype enzymatically and structurally, involved in DNA repair, removing RNA primer, filling primer gaps II) DNA repair III) replication and synthesis of DNA
158
types of DNA polymerases in eukaryotes
1) DNA-dependent DNA polymerases: DNA polymerases 2) RNA-dependent DNA polymerases: reverse transcriptase, telomerase 3) DNA-dependent RNA polymerase: RNA polymerases
159
DNA polymerase alpha
- replication (complex with primase, aids in starting primer) - DNA repair - no exonuclease activity
160
DNA polymerase beta
- DNA repair only - no exonuclease activity
161
DNA polymerase gamma
- DNA replication in mitochondria - 3' to 5' exonuclease
162
DNA polymerase delta
- replication (processive synthesis on lagging strand) - DNA repair - 3' to 5' exonuclease
163
DNA polymerase epsilon
- replication (leading strand) - DNA repair - 3' to 5' exonuclease
164
DNA polymerase kappa, eta, xi, iota
- DNA repair (bypass polymerase) - no exonuclease activity
165
biotechnologies based of DNA polymerase understanding
PCR, DNA sequencing, CRISPR
166
retrovirus replication
1) entry into cell 2) uncoating of capsid, release of RNA 3) reverse transcription 4) duplex DNA integrated into host chromosome 5) viral RNA synthesis 6) viral protein translation 7) virus assembly 8) release of new viruses
167
AZT and HIV
- AZT has 3' N3 instead of OH - prevents next base from being added onto growing DNA chain, so prevents HIV replication - less impact in eukaryotes because exonuclease activity can remove altered base
168
Meselson and Stahl experimental design
1) E.coli undergoing replication 2) grow in 15N for a few generations 3) switch to 14N 4) separate by CsCl gradient centrifuge: heavier closer to bottom of tube 5) visualize with intercalators (280nm UV absorption)
169
Meselson and Stahl expected results
1) conservative: one heavy, one light --> heavy, more light 2) semiconservative: HL mix --> L, HL mix 3) random dispersive: HL mix --> lighter HL mix
170
Meselson and Stahl actual results
- semiconservative
171
possible ways for semi-conservative replication
1) continuous on both strands 2) semi-discontinuous 3) discontinuous on both strands 4) pieces
172
Okazaki et al. experimental design
1) grow E. coli with 3H-thymidine to label newly synthesized DNA --> stop reactions with cyanide and ice (kills cells) before replication is completed 2) separate DNA by size: ultracentrifugation in alkaline (denatures) sucrose gradient = small at top 3) divide centrifuge tube into fractions, measure radioactivity
173
why does alkaline solution denature DNA?
amino groups deprotonated on bases = inhibits H-bonding
174
Okazaki et al. expected results
1) continuous = long fragments only, growing longer over time 2) discontinuous (both strands) = short fragments growing, long appears later 3) pieces = only short fragments 4) semi-discontinuous = short fragments roughly same size decrease by the end (ligation), long fragments getting longer and longer
175
Okazaki et al. results
1) 2 peaks: short and long 2) both increase in number over time 3) long gets longer, short stays the same = semi-discontinuous model
176
Huberman and Riggs experimental design
1) cells (Chinese hamster ovaries) grown with 5fdU = stops thymine synthesis, all cells primed and ready to go in G1 2) pulse: add 3H thymidine (hot) for a short time 3) chase: change to excess 1H-thymidine (cold) 4) lyse cells, spread DNA on slide 5) coat with photographic emulsion (silver halide crystals) 6) fibre autoradiography 7) develop and view under microscope
177
Huberman and Riggs expected results
1) bidirectional: chase, pulse, chase 2) unidirection: chase, pulse --> only decreases in one direction
178
Huberman and Riggs actual results
1) replication is bidirectional 2) mammalian cells have multiple origins of replication
179
problems DNA polymerase cannot address on its own
1) DNA problem: can't unwind dsDNA, ssDNA template is unstable (reforms dsDNA too quickly or forms hairpins) 2) starting problem: can't start DNA chains de novo (need 3'OH to build off) 3) speed: too slow and distributive 4) lagging strand: no 3' to 5' polymerase activity
180
solving the DNA problem
1) DNA helicase unwinds DNA 2) ssDNA binding proteins (SSBs) prevent re-annealing and protect DNA from damage 3) topoisomerase relieves supercoiling
181
DNA helicase characteristics
- toroid hexamer - homo in prokaryotes, hetero in eukaryotes - binds dsDNA and ssDNA - ATPase - positively charged center
182
DNA helicase mechanism
*not fully understood 1) two helicase bind to DNA, DNA fed 3' into the channel (unwinds 3' to 5') 2) DDK (kinase) phosphorylates both to start 3) ATP cycling causes rotation along the strands in opposite directions 4) leads to unwinding
183
DNA helicase assay
- radiolabelled DNA under various conditions with ATP: 1) boil = positive control = ssDNA only 2) no boil = negative control = dsDNA only 3) increasing amounts of DNA helicase = increasing ssDNA, decreasing dsDNA
184
ssDNA binding protein characteristics
- replication protein A (RPA) - heterotrimer: RPA70, 32, 14 (largest to smallest) - multiple DNA binding domains (3) - bend DNA around protein - prevent reannealing, protect from damage
185
topoisomerase characteristics
- break phosphodiester bond downstream of replisome, relieving supercoiling (torsional strain) - type 1 = single-strand break - type 2 = double-strand break - reanneals later (has nuclease and ligase active sites
186
replication termination
- helicases meet, replication near complete but DNA is tangled - Topo II (type 2 topoisomerase) cleaves to untangle
187
resolving the starting problem
- RNA polymerases don't need 3'OH group (can hold two bases in active site) - two polymerases, four subunit complex 1) DNA-dependent RNA polymerase (primase): slow, adds 11-14 RNA bases 2) DNA-dependent DNA polymerase (Pol alpha): faster, extends by 10-20 bases, NO proofreading - conformational change to switch between the two
188
why does the primase/pol alpha complex not need high-fidelity?
- primer is removed from final DNA product - prokaryotes: DNA pol I - eukaryotes: RNase H and Fen endonuclease
189
solving the speed problem
- sliding clamp keeps DNA pol from sliding off DNA - called proliferating cell nuclear antigen (PCNA) in eukaryotes
190
PCNA characteristics
- homodimer - toroid - no ATPase activity - binds DNA pol epsilon and delta
191
PCNA prokaryote equivalent
- beta clamp
192
PCNA affect on DNA polymerase processivity
- dramatic increase because can stay on DNA for longer and create longer strands 1) high conc. DNA pol with no pol delta auxillary protein or PCNA = small fragments only 2) low conc. DNA polymerase with poly delta aux or PCNA = large fragments
193
loading sliding clamp onto DNA
1) RFC1/RFC5 (replication factor C) N-termini bind PCNA 2) ATP binds to open up PCNA 3) DNA attaches 4) ATP hydrolyzed, ADP released = PCNA separated and loaded onto primer-template junction (ss-dsDNA junction)
194
solving the lagging strand problem
- replisome coordinates replication: 1) topoisomerase 2) primosome: DNA helicase + DNA primase 3) sliding clamps on both strands 4) ssDNA binding proteins - lagging strand wraps around to keep complex close together
195
lagging strand synthesis
1) first round of synthesis 2) unwinding of parental strands, second round starts with new Okazaki fragment at replication fork 3) RNA primers removed 4) gaps filled with repair DNA polymerase 5) Okazaki fragments joined by DNA ligase
196
DNA ligase
- forms phosphodiester bond to fill gaps
197
DNA replication vs PCR
similarities: 1) function: copy DNA template 2) substrates: dNTPs, DNA template differences: 1) polymerase: epsilon/delta vs Taq 2) cofactors: Mg vs Mg + buffer 3) DNA problem: solved by helicase, SSBs, topoisomerase vs heat 4) starting problem: primase/DNA pol alpha vs primers 5) speed: PCNA vs E507K mutation 6) lagging strand: Okazaki fragments vs no lagging strand
198
end replication problem
- no place for the final bases once the primer is removed on 5' end - leaves exposed ssDNA that is susceptible to nucleases
199
solving the end replication problem
- telomerase (RNA-dependent DNA polymerase) adds repeat sequence (contains its own complementary template) - provides space for primase to add another primer
200
norfloxacin mechanism
- quinolone antibiotic that inhibits ligation activity of bacterial topoisomerase (gyrase) - prevents DNA replication - nuclease activity remains = DNA damage = cell death - specific to bacteria (differences in AAs)
201
is DNA very stable?
- wrong assumption - DNA is susceptible to damage: nicks, breaks, base alterations, chemical reactions - repair mechanisms prevent damage from becoming mutations
202
endogenous vs exogenous DNA damage
- endogenous = spontaneous - exogenous = environmental
203
DNA damage vs mutation
- damage = physical alteration to structure of DNA - mutation = change in sequence after replication = inherited by daughter cell, cannot be repaired
204
types of DNA damage
endogenous: 1) depurination / depyrimidination 2) deamination 3) oxidative damage exogenous: 1) ionizing radiation 2) UV radiation 3) alkylation
205
depurination / depyrimidination
- loss of nitrogenous base through hydrolysis of N-glycosidic bond - results in apurinic (more common) / apyrimidinic / abasic (AP) site = loss of coding info for DNA replication or transcription - also risk of DSB
206
DSB from depurination / depyrimidination
1) free OH = potential for linearization of sugar 2) beta-elimination: base attack leads to loss of 3' connection to phosphate 3) no 3'OH to help with repair = DSB
207
translesion (bypass) synthesis polymerase
1) replication or transcription stalls at site of damage (ex. AP site) 2) PCNA ubiquinated 3) recruitment and switch to bypass polymerase (eta), fills gap with random nucleotide 4) PCNA deubiquinated, switch back to replisome to continue process
208
deamination
- exocyclic amine group replaced with C=O - can occur on A, G, C - loss of two H-bond donors, replaced with two H-bond acceptors - ex. C --> U changes base pairing (G-C to A-U)
209
oxidative damage
- from ROS generated in cellular processes: ETC, drug metabolism, inflammation
210
ROS reactions
O2 + electron = O2- (superoxide) O2- + e, 2H+ = H2O2 (hydrogen peroxide) H2O2 + e, H+ = H2O + OH radical OH radical + e, H+ = H2O
211
protective mechanisms against ROS
- enzymes and anti-oxidants protect DNA, protein and lipids from ROS - ex. SOD, catalase
212
oxidative damage example: guanine
- guanine + OH radical = 8-oxo-G - steric clash introduced = syn conformation preferred - base pairs with adenine instead of cytosine - 2nd round of replication = mutation
213
ionizing radiation
- produces ROS (splitting water) - also directly damages DNA bonds
214
UV radiation
- produces pyrimidine dimers - pi bond electrons excited by UV light, can cause stacked pyrimidines to bond
215
pyrimidine stacking effects
1) pulls bases closer together (3.4 to 1.5A) 2) distortion (kink/bend) because of disrupted H-bonding 3) interferes with polymerase activity (binding)
216
types of pyrimidine dimers
1) two bonds = cyclobutane pyrimidine dimer (CPD) 2) one bond = 6-4 photoproduct
217
TLS in pyrimidine stacking
- fills gap to allow replication or transcription to continue - preference for adding A:A dimer = potential mutation
218
alkylation
- transfer of methyl to base - ex. from SAM - ex. methylation of O6 on guanine alters H-bonding, can now base pair with T
219
relative frequencies of DNA damage
1) UV radiation: 100 000 lesions/cell/day 2) depurination: 10 000 3) alkylation: 5 000 4) depyrimidination: 500 5) deamination: 100
220
how does sunscreen protect skin from DNA damage?
- delocalized electrons in organic compounds absorb some energy from UV radiation - released photons are at lower energy
221
DNA repair mechanisms
1) direct repair 2) base excision repair (BER) 3) nucleotide excision repair (NER) 4) mismatch repair 5) post-replication repair 6) DSB repair
222
direct repair
- fixes damaged base with specific enzymes that reverse the damage
223
O6-methylguanine-DNA methyltransferase (MGMT)
1) recognizes kink caused by G methylation 2) interdigitation: flips damaged base out into active site and inserts Arg into DNA helix to stabilize 3) Cys145 attacks methyl = suicide enzyme (degraded)
224
general steps to replacing DNA damage
1) damaged DNA recognized by protein 2) enzymes recruited to break phosphodiester bond to remove damaged area 3) DNA polymerase rebuilds removed area 4) DNA ligase re-seals phosphodiester bonds
225
base excision repair
remove and replace damaged nitrogenous base: 1) proteins recognize changes in DNA bases 2) enzymes recruited: glycosylase breaks N-glycosidic linkage to generate an AP site (specific for each type of DNA damage) 3) endonuclease nicks backbone 4) dRP lyase removes deoxyribose 4) DNA polymerase replaces nucleotide 5) DNA ligase seals phosphodiester bond
226
nucleotide excision repair
remove and replace damaged nucleotide 1) XPC-HR23B recognizes changes in helix structure 2) TFIIH (XPB/XPD) is recruited to unwind DNA: XPC-HR23B interdigitation enhances kink severity to attract 3) RPA protects ssDNA (TFIIH bound to damaged strand only) 4) endonucleases (3' XPG/XPA and 5' XPF) cleave phosphodiester bond, removing larger stretch of nucleotides 5) replisome fills gap 6) DNA ligase seals phosphodiester backbone
227
DNA damage and associated repair mechanism
1) direct repair = alkylation 2) BER = depurination / depyrimidination, deamination, oxidative damage 3) NER = UV radiation
228
mismatch repair
- fixes DNA polymerases mistakes (mismatches and small insertions/deletions) - recognizes newly synthesized strand because of lack of methylation (epigenetic marks copied after complete replication) --> demonstrated in prokaryotic cells, only PREDICTED for eukaryotic cells
229
mismatch repair steps
1) unwind DNA 2) endonucleases cleave phosphodiester bond 3) replisome replaces nucleotides 4) DNA ligase seals phosphodiester backbone
230
xeroderma pigmentosum (XP)
- inherited condition (mutations in XPC and POLH (DNA pol eta)) - extreme sensitivity to UV radiation and risk of UV-induced cancers - XPC mutations = NER inhibited - POLH mutations = TLS inhibited = more damage, apoptosis
231
single-strand breaks
- phosphodiester bond breaks on one side of the DNA - leads to DNA damage response
232
causes of SSBs
1) ionizing radiation: ROS (sugar oxidation) and direct DNA damage 2) inhibition of DNA ligase = unligated Okazaki fragments 3) inhibition of topoisomerase 1 ligation activity 4) stochastic base damage, cytosine demethylation/alkylation, guanine deamination = BER (if fails, leads to SSB) 5) depurination = AP site = beta-elimination (or BER)
233
DNA damage response for SSBs proteins involved
1) lesion identification: PARP1, XRCC1 2) repair of DNA ends: PNKP 3) replace DNA bases: a) short-patch = pol beta b) long-patch = pol beta, then epsilon/delta add more, then FEN1 endonuclease removes primers 4) ligation: DNA ligase
234
indirect SSB (BER) DNA damage response
1) detection: APE1/lyase 2) end processing: XRCC1 scaffolds for PNKP and other proteins 3) short-patch filling 4) ligation
235
direct SSB (ex. sugar damage) DNA damage response
1) detection: PARP1/PARG 2) end processing: XRCC1 scaffolds for PNKP and other proteins 3) long-patch filling with PCNA 4) ligation
236
TOP1-SSB DNA damage response
1) detection: RNAP and other lesions 2) end processing: XRCC1 scaffolds for PNKP and other proteins 3) long-patch filling with PCNA 4) ligation
237
double-stranded breaks
- break of phosphodiester bond on both sides of DNA (can be blunt or sticky end) - more serious than SSB, requiring immediate action - if not repaired, apoptosis initiated
238
causes of DSBs
1) ionizing radiation 2) ROS 3) type II topoisomerases 4) meiosis: recombination events 5) SSBs during replication: if replication fork encounters SSB, lack of non-covalent interactions can cause replication arm to separate from the fork
239
DSB repair pathways
1) if resection = homology-directed repair (HDR) 2) if no resection = non-homologous end-joining (NHEJ)
240
which DSB repair pathway dominates?
- competitive binding of protein complexes - if MRN binds first, resection occurs = HDR - if Ku binds first, resection blocked = NHEJ
241
NHEJ characteristics
- quick and messy - error-prone - all phases of cell cycle
242
HDR characteristics
- finding match - fewer errors - S and G2 phases
243
HDR types
1) homology between tails = single strand annealing 2) no homology between tails = homologous recombination
244
resection
- removal of some bases around the break to create 3' overhang
245
NHEJ vs HDR protein complexes
- NHEJ = Ku70/80 heterodimer - HDR = MRN complex
246
NHEJ steps
1) Ku recognizes DSB, acts as scaffold 2) repair proteins recruited 3) ends of chromosome brought together 4) any homology used to reconnect DNA 5) IF NEEDED: gaps filled or excess DNA is removed (pol beta, gamma, mu) 6) backbone ligated together
247
possible outcomes of NHEJ
1) perfect synapsis = no sequence change 2) incorrect microhomology = addition or deletion (indels)
248
HDR (homologous recombination) steps
1) MRN (scaffold) recognizes DSB 2) CtIP recruited, activated by phosphorylation (ATM) and ubiquitylation (BRCA1) 3) MRN/CtIP begins resection: CtIP 5' to 3' endonuclease removes ~100nt 4) Exo1 continues: more efficient (~4kb/h), produces large ssDNA overhangs 5) RPA binds overhangs 6) BRCA2 replaces RPA with RAD51 and paralogs 7) strand invasion: RAD51 coated ssDNA invades dsDNA, searching for homologous region 8) D-loop formed 9) RAD54 removes RAD51, allows DNA polymerase to bind and replicate DNA from template 10) second strand invasion = Double Holiday Junction 11) nucleases resolve junction
249
HDR template options
1) sister chromatid: ideal, because exact same sequence 2) homologous chromosome: could be different allele 3) retrotransposon (homologous sequences): detrimental effects ex. chromosomal translocation and rearrangement 4) artificially introduced repair templates
250
BRCA2 evidence study components
1) long dsDNA with 3' overhang coated with RPA 2) radiolabeled short dsDNA repair template 3) RAD51: same conc. in all samples 4) BRCA2: various conc.
251
BRCA2 evidence study methods
gel electrophoresis: 1) long fragment visible = transfer or repair template = strand invasion occurred 2) short fragment visible = no reaction
252
BRCA2 evidence study results
1) negative control: RPA only = only template 2) positive control: RAD51 only = template and product 3) RPA + RAD51 +increasing BRCA2 conc. = increasing product
253
why are BRCA1/2 mutation associated with increased cancer risk?
- HDR cannot be initiated - DSBs repaired using NHEJ, which is more error-prone
254
sickle cell anemia
- single amino acid change (Glu6Val) in beta globin gene - hemoglobin cannot form tetramers, instead form rod-like aggregates that deform RBCs and occlude blood flow - can be treated with gene therapies
255
genome editing
- alteration of genetic material of living organism by insertion, replacement or deletion of DNA sequence - typical aim: improve some characteristic of crop/farm animal or correct genetic disorder
256
genome editing exampels
1) site-directed mutagenesis 2) restriction cloning 3) random mutagenesis 4) recombinant DNA cloning
257
recombinant DNA cloning mechanism
1) artificially introduced recombinant DNA molecule triggers DSB repair pathway 2) molecule is homologous to gene of interest, leading to gene replacement
258
recombinant DNA cloning efficiency
- low efficiency - worked best in organisms that prefer HDR over NHEJ, ex. yeast
259
how to improve genome engineering efficiency
- introduce DSB in genome as well (in target gene) - end resection occurs on both genome and artificial DNA, allowing for single-strand anealing
260
single-strand annealing
- short-cut for HDR - homologous tails base pair, flaps are cleaved, DNA ligated
261
which proteins can be used to create DSBs?
1) topoisomerase II 2) restriction enzymes 3) nucleases 4) CRISPR-Cas
262
limitations of topoisomerase, restriction enzymes and engineered nucleases in genome editing
- topoisomerases: cannot target specific sequences - REs: limited by specific target sequence - engineered nucleases: require extensive protein engineering
263
types of engineered nucleases
1) zinc-finger nucleases 2) transcription activator-like effector nuclease (TALENs)
264
ZFN vs TALEN
- both have nuclease attached to engineered proteins that recognize DNA sequences - ZFN: each domain recognizes 3nt - TALEN: each domain recognizes 1nt
265
what does CRISPR do in bacteria?
1) invasive viral nucleic acids cleaved and incorporated into CRISPR site of bacterial genome 2) expressed as RNA 3) forms complex with endonuclease 4) complex searches for matching viral sequences 5) breaks them = protects against viral infection
266
spCas9
Cas9 from streptococcus pyogenes
267
spCas9 domains
- 2 nuclease domains: RuvC and HNH - C-terminal domain = PAM-recognition site - recognition (REC) lobe = sgRNA binding site - Arg-rich helix = DNA binding
268
spCas9 conformational changes
- apo state: PAM site disordered, nuclease domains collapsed on each other - sgRNA bound: central channel opens, accommodating dsDNA
269
Cas9 searching
- loaded with guide RNA - searches dsDNA for PAM site: protein-DNA interaction - if no site, Cas falls off
270
spCas9 PAM site
5'-NGG-3'
271
Cas9 mechanism
1) dsDNA unwound, stabilized by Arg-rich helix 2) interaction allowed with sgRNA: if enough complementary sequence is found, another conformational change occurs 3) nuclease domains brought into position 4) cleave on both sides to make blunt end: HNH on target strand, RuvC on non-target
272
Cas9 off-target effects
- some mismatch permitted
273
what part of sgRNA has greatest impact on binding?
sgRNA-DNA base-pairing nearest to PAM site
274
what happens after CRISPR-Cas9 cleavage?
- DSB repair or cell death - NHEJ: can knock-out gene, silent mutation or no mutation - HDR: can provide repair template for targeted mutation SSA
275
advantages of CRISPR/Cas9
1) targeted 2) sgRNA is cheap and highly efficient (compared to proteins) 3) Cas9 can be modified and/or be a transport protein (dCas9 = no nuclease activity)
276
what proteins can Cas9 transport?
1) nickase: causes SSB (one Cas9 nuclease site mutated) 2) transcriptional regulators 3) chromatin modification: epigenetic modulators 4) tags: ex. GFP 5) base-modifiers
277
applications of CRISPR
1) research: KO models, add tags/domains/modify proteins, change gene expression 2) biotechnology: agriculture and manufacturing 3) healthcare: correct gene mutations, treat disease
278
gene-editing to treat sickle cell anemia
1) screen 2) collect patient stem cell (low risk of rejection) 3) gene-edit, test for function - turns off expression of BCL11A to allow gamma-globin expression to increase = fetal hemoglobin produced 4) infuse cells
279
ethical concerns around genome editing
1) somatic (adult cells) vs germline (embryos) editing 2) preventing/treating disease vs enhancement 3) risk/benefit profile: availability of reasonable alternatives 4) supporting clinical or pre-clinical data 5) independent ethics reviews
280
CCR5
receptor that HIV binds to get into T cells, may have other important functions that are not completely understood
281
CCR5delta32
- leads to frameshift + premature stop codon - homozygous individuals for this mutation are resistant to some strains of HIV - HIV can still infect by interacting with other receptors
282
He experiment (CRISPR babies)
1) CRISPR engineered mutations in CCR5 gene in embryos (mother HIV negative, father HIV positive) 2) no repair template = NHEJ 3) Nana: homozygote with different mutations (1bp insertion, 4bp deletion) 4) Lulu: WT + 15bp deletion (heterozygote) 5) both displayed mosaicism after birth because editing only occurred in some cells of embryo 6) no apparent off-target mutations
283
ethical concerns of He experiment
1) other more effective ways to prevent HIV infection, ex. IVF: remove HIV from sperm first 2) no repair template = cannot guarantee mutation provides resistance 3) debatable informed consent
284
central dogma of molecular biology
DNA --> RNA --> protein
285
prokaryotic vs eukaryotic RNA polymerase similarieties
- overall structure quite similar - 3 stage synthesis: initiation, elongation, termination
286
RNA polymerase functions
1) recognize initiation sites and promoters 2) helicase activity: unwinds dsDNA 3) correct ribonucleotide triphosphate selection: unidirectional/processive 4) termination 5) activation/repression: transcription factors 6) fundamental reaction: 3' OH attacks alpha P to create phosphodiester bond
287
cis-acting elements
cis = on template strand being transcribed
288
RNA synthesis direction
5' to 3', DNA is read 3' to 5'
289
RNA polymerase vs DNA polymerase
1) no primer required 2) no proofreading capability (1 in 10^5 error rate vs 10^10): acceptable because of codon degeneracy 3) slower (50nt/s vs 800)
290
E. Coli RNAP structure
- 400kDA - 4 subunits - holoenzyme is two alpha, one beta, one beta prime, one sigma (pentamer) - alpha2betabeta = core enzyme, contains catalytic site
291
RNAP sigma subunit function
- decreases DNA binding affinity - released after 10nts synthesized, goes to bind another core enzyme
292
RNAP active site
- similar to DNAP - Asp residues interact with DNA and Mg2+ - another Mg2+ cofactor interacts with ribonucleoside triphosphate gamma and beta phosphate = good LG
293
footprinting
1) PCR amplification with radiolabeled DNA 2) add DNase I: cleaves randomly 3) denature dsDNA 4) gel electrophoresis 5) compare DNA/protein complex to naked DNA: gaps show where protein binds (DNase cannot access) 6) can use single base analysis to find sequence
294
identification of prokaryotic promoter sites
- footprinting - comparisons between different genes reveals similar initiation sites
295
prokaryotic promoter sites
core promoter = 40nts, contains: 1) -35 region 2) -10 TATA (pribnow) box
296
consensus sequence
- average of different sequences of genes - number below shows percent frequency
297
gene nucleotide numbering
+1 = initiation site negative = upstream positive = downstream
298
RNAP interaction with promoter
- recognition helix in sigma subunit makes transient bonds with T and A - residues: Tyr, Trp, Thr, Gln, Arg
299
non-template strand
coding or sense strand
300
template strand
non-coding or antisense strend
301
RNA transcript resembles...
non-template strand with U instead of T
302
strong vs weak promoters
- strong = frequently transcribed (housekeeping genes), 17nt between -10/-35 is ideal - weak = multiple substitutions in -10/-35 regions, less frequently transcribed
303
factors that impact promoter strength
1) regulatory proteins: bind near or to promoter regions 2) UP elements
304
UP (upstream) elements
- bound by alpha subunit C-terminus - increases transcription efficiency - not highly conserved - typically A/T rich - 40-60 nts upstream
305
alternative promoter sequences in E. coli
- bound by different variants of sigma subunit, ex: 1) standard = 70 2) heat-shock = 32 3) nitrogen-starvation = 54 - bacteria upregulates synthesis of different subunits in response to environmental changes
306
prokaryotic transcription initiation mechanism
1) holoenzyme (sigma recognition helix) forms transient H-bonds with base pairs: rapid (10^10 M^-1 s^-1) and random search 2) recognition of promoter = closed promoter complex (STILL REVERSIBLE) 3) unwinding of DNA to form open promoter complex (IRREVERSIBLE) 4) RNA polymerase starts transcription, usually by adding a purine 5) sigma subunit falls off after 2-10 nts synthesized 6) core enzyme remains to continue to elongation
307
open promoter complex characteristics
- 17bp segment = 1.6 DNA turns - lower G-C content is easier to unwind
308
prokaryotic transcription elongation mechanism
1) formation of transcription bubble 2) nucleotide addition cycle: events required for addition of a nucleotide to the RNA product to form a cyclic process
309
prokarytotic transcription bubble
- RNA polymerase unwinds DNA at the front, ssDNA enters active site - rewinds DNA at the back - RNA-DNA helix formed as RNA synthesis occurs with 3' end of RNA in active site - Phe in exit channel inserts into RNA-DNA helix to separate them, promoting exit of RNA strand
310
RNA-DNA helix characteristics
- ~8bps long = 1 double-helix turn - moves 170 angstroms/sec = 50nt/sec - rotates because of re/unwinding
311
nucleotide addition cycle important structures
- bridge helix - trigger loop - different conformations facilitate translocation and NAC
312
pre-insertion site in RNAP
- inactive site - revealed by streptolydigin: inhibits transcription by sitting in insertion site, but RNAP could still bind NTPs
313
NAC steps
catalysis: 1) open trigger loop accepts NTP into pre-insertion site 2) closure of trigger loop moves NTP into insertion site 3) catalytic incorporation releases pyrophosphate translocation equilibrium: 4) trigger loop opens 5) equilibrium formed between open with pre-translocated DNA, wedged with intermediate and open with post-translated DNA states 6) latter conformation allows another NTP to be accepted
314
general steps of transcription termination
1) ceasing of phosphodiester bond formation 2) RNA/DNA hybrid dissociation 3) rewinding of dissociated DNA strands 4) release of DNA by RNAP
315
two methods of termination in prokaryotes
1) intrinsic 2) protein-dependent
316
intrinsic termination signal in prokaryotes
- palindromic GC region separated by ~4A (forms U loop) - followed by 4+ U (coded for by DNA) - forms stem loop structure = RNAP stalls - A/U rich area is weaker, leads to RNAP falling off
317
protein-dependent termination signal in prokaryotes
- Rho factor recognizes Rho termination sites - used in rRNA synthesis: DNA template contains different Rho sites for 10S, 13S, 17S rRNA - experiment: depending on timing of Rho addition = different subunits
318
how does Rho cause termination?
- binds to C-rich site: Rho utilization or rut site on synthesized RNA - catches up to RNAP, causes unwinding of RNA/DNA helix = stalling + dissociation
319
Rho characteristics
- ATP-dependent helicase - hexamer
320
drugs targeting transcription
1) rifampicin 2) actinomycin 3) amanitin
321
rifampicin action
- binds RNA/DNA helix channel in prokaryotes (not eukaryotes) - stops initiation - no effect once elongation starts
322
actinomycin action
- intercalates with DNA - DNA is no longer an effective template - also affects DNA polymerase (replication)
323
steady-state level vs synthetic capacity of RNA
- synthetic capacity: cell's ability to synthesize - usually correlated, except in the case of mRNA: immediately processed and transcribed
324
prokaryotic post-transcriptional modification of rRNA and tRNA
- usually on one transcript - undergoes cleaving, processing (ex. CCA to tRNA), and modification (to bases) - other mRNA typically undergoes little to no modification after synthesis
325
rRNA/tRNA mRNA cleavage
1) RNase III: cuts out rRNA 2) M16, 5, 23 (specific varieties): trims rRNA 3) RNase P: 5' end of tRNA 4) RNase D: 3' end of tRNA
326
mRNA base modification examples in prokaryotes
- ribothymidylate, pseudouridylate - methylation ex. 6-dimethyladenine
327
prokaryotic vs eukaryotic transcription
1) spatial-temporal regulation: translation co-transcriptionally vs. must be exported out of nucleus (after fully processed) 2) processing: minimal vs extensive 3) number of RNAP: 1 vs 3 4) RNA subunits: 5 vs 7-10
328
characteristics of eukaryotic RNAP
1) all large proteins with 8-15 subunits 2) RNAP II has unique CTD with repeats of YSPTSPS 3) phosphorylation of CTD (S/T) regulates activity 4) different responses to alpha amanitin
329
eukaryotic RNAP I
1) location: nucleolus 2) cellular transcripts: 18S, 5.8S, 28S rRNA (one copy / primary transcript) 3) alpha amanitin effects: insensitive 4) promoters: A-rich upstream promoter element (UPE), ribosomal initiation element
330
eukaryotic RNAP II
1) location: nucleoplasm 2) cellular transcripts: mRNA precursors and snRNA (splicing) 3) alpha amanitin effects: strong inhibition, Kd = 10nm 4) promoters: TATA box, initiator element, downstream promoter element
331
eukaryotic RNAPIII
1) location: nucleoplasm 2) cellular transcripts: tRNA, 5S rRNA 3) alpha amanitin effects: inhibited by high conc., Kd = 1um 4) promoters: downstream promoters
332
important RNAPII subunits
- RBP1 contains CTD, homolog to beta prime subunit - RPB4: promoter recognition
333
alpha-amanitin mechanism
- inhibits open to closed trigger loop change: ribonucleotide cannot move to insertion site - inhibits wedged loop to open: RNAP cannot accept ribonucleotide
334
RNAP I promoter
core promoter: 1) -200 to -150: UPE 2) +1 ribosomal initiation element
335
RNAP II promoter
1) enhancer at least -1kB core promoter: 2) upstream TATA box -100 to -20 OR DPE around +30 3) ribosomal initiation element +1
336
RNAP III promoter
type 1: 5srRNA 1) downstream A and C block type 2: tRNA 1) downstream A and B block *A/B/C all ~11bp
337
TATA box mutations
- single base markedly impairs promoter activity
338
upstream enhancers in RNAPII promoter
1) CAAT box 2) GC box - usually for housekeeping genes - increase transcriptional frequency - bound by RPE3
339
DNA looping
- binding of transcription activators to enhancer or silencer - interaction with RNAP facilitated by DNA looping (to bring elements close together)
340
eukaryotic transcription initiation
1) TFIID (multi-subunit complex) recognizes TATA box via TATA box binding protein (TBP) 2) TFIIA stabilizes complex 3) TFIIF recruits RNAPII 4) TFIIB, TFIIE, TFIIH also recruited = basal transcription apparatus (BTA) aka pre-initiation complex 5) TFIIH (helicase) unwinds DNA and phosphorylates CTD 6) all except TFIIF dissociate 7) elongation begins: NAC
341
TFIID subunits
1) TBP 2) TAFs: recognize non-TATA elements, histone acetyltransferase activity
342
TFIIB function
recruits RNAPII and TFIIF, helps start-site selection
343
TFIIF function
- promoter targeting of RNAPII - destabilizes nonspecific RNAPII-DNA interactions
344
TFIIE function
- recruited by RNAPII - recruits and modulates TFIIH helicase, ATPase, kinase activities
345
TFIIH
- helicase activity - CTD phosphorylation via 2 cyclin:CDK pair subunits
346
negatively regulated gene
- transcription prevented by a repressor - transcribed in absence of active repressor
347
positively regulated gene
- transcription occurs in presence of activator - not transcribed in absence
348
repressors and activators characteristics
- allosteric proteins - bind ligands
349
positive transcriptional regulation methods
1) molecular signal causes binding of activator = transcription 2) molecular signal causes dissociation of activator = no transcription
350
negative transcriptional regulation methods
1) molecular signal causes dissociation of regulatory protein = transcription 2) molecular signal causes binding of regular protein = no transcription
351
operons
linearly organized: regulator gene: 1) promoter for regulator gene 2) regulator gene control sites: 3) promoter 4) operator (surrounding operon) structural genes: 5) ex. lactose operon: z, y, a
352
polycistronic
mRNA that encodes multiple proteins
353
lac operon components and functions
1) z = beta-galactosidase: lactose --> allolactose 2) y = permease: transports lactose in 3) a = transacetylase: acetylates lactose to be exported out
354
lactose vs allolactose
1-4 beta-glycosidic linkage vs 1-6
355
lac operon two control mechanisms
1) regulatory response to lactose 2) regulatory response to glucose
356
lac operon lactose regulation
1) lac repressor prevents transcription 2) allolactose inactivates repressor and allows transcription
357
lac repressor mechanism
1) binds DNA (operator regions) as dimer 2) dimers from both operator regions forms tetramer = DNA looping 3) blocks RNAP
358
lac repressor transient binding
- transient binding ensures transcription occurs at least once = basal levels of lac operon genes
359
lac repressor/DNA interactions
- lac repressor alpha helix binds major groove - H-bonding with AAs ex. Arg
360
lac repressor bound vs unbound
- lactose = unbound by allolactose = bound to DNA = very ordered + lactose = bound by allolactose = unbound to DNA = more disordered (conformational change)
361
results of lactose lac operon study
- adding lactose = beta-galactosidase and total bacterial protein (indicating growth) have direct relationship - removing lactose = plateau
362
accessory proteins for promoters
some require additional accessory proteins to speed up transcription, ex, catabolite activator protein (CAP) for lac operon
363
CAP characteristics
- dimer of 22.5 kD peptides - N-terminus binds cAMP - C-terminus binds DNA
364
lac operon glucose regulation
1) increase in glucose = decrease in cAMP and vice versa 2) if glucose decreases, cAMP increase and binds CAP 3) CAP can bind to DNA: upstream of RNAP binding site (-41, -61 or -71 bp) 4) assists formation of closed promoter complex
365
purpose of catabolite repression
- ensures that operons for metabolism of alternative energy sources are repressed until glucose exhausted
366
components of eukaryotic transcription machinery
1) activators 2) repressors 3) coactivators 4) basal factors
367
common DNA sequences bound by proteins
- two-fold axis of symmetry - palindromic - dimer binding - Glu/Asn often AT - Arg often CG
368
DNA-binding motifs
80% DNA-binding proteins have one of: 1) helix-turn-helix motif 2) zinc-finger 3) leucine zipper-basic region (bZIP)
369
helix-turn helix
- recognition helix binds major groove
370
zinc fingers
- 10x small domains with Zn coordination to 4 Cys or 2Cys/2His - key helix that binds DNA
371
leucine zippers
1) DNA binding region 2) 6-AA connector 3) leucine zipper: coiled coils held together by hydrophobic interactions between leucines (every 7th residue)
372
estrogen: example of hormonal control of transcription in humans
- cholesterol-derived hormone required for development and ovarian cycle - diffuses across cell membrane - ligand for DNA-binding protein (estrogen receptor)
373
estrogen receptor
- contain Zn fingers - bind at estrogen-receptor elements - three domains: transcription activation (variable), DNA binding (conserved, 66-68 residues), hormone binding (variable
374
estrogen receptor conformational change
- binding of estrogen causes helix 12 to fold up into side of receptor - does NOT alter DNA binding affinity of receptor - increases affinity of COACTIVATOR: P160 family = chromatin remodelling = increases transcription
375
drugs targeting estrogen receptor
- ex. tamoxifen - binds in ligand-binding pocket - prevents proper folding of helix 12, coactivator cannot bind - ER antagonist to treat ER+ breast cancers
376
types of cellular RNA
1) mRNA: template for protein synthesis 2) tRNA: carries AAs to site of protein synthesis on ribosome 3) rRNA: part of ribosome 4) other: components of ribonucleoproteins with variety of functions
377
rRNA splicing
1) RNAPI produces pre-RNA with 3 ribosomal RNAs: 18S, 5.8S, 28S, separated by spacer regions 2) nucleotide modification: methyl groups and pseudouridines 3) spacer regions cleaved out
378
rRNA splicing machinery
- rDNA with many branches of pre-rRNA - SSU (small subunit) processome (aka large ribonucleoprotein): at the ends for processing premRNA
379
tRNA splicing
1) RNAPIII produces tRNAs 2) 5' cleavage by RNAase P 3) 3' cleavage by RNase D 4) nucleotide modification 5) tRNA nucleotidyl transferase adds CCA to 3' end, releasing 3 PPi = AA attachment site 6) eukaryotes only: intron splicing near anticodon
380
eukaryotic mRNA delivery to ribosomes
- must be processed into mature mRNAs - must be transported from nucleus into cytosol
381
eukaryotic vs prokaryotic mRNAs
- eukaryotic are monocistronic
382
mRNA capping
- 7-methyl-G capped added to increase transcript stability and define start site for protein synthesis - occurs co-transcriptionally
383
mRNA capping steps
1) RNAP II produces mRNA 2) GTP added by guanylyl transferase: a) hydrolysis of terminal phosphate on 5' end of transcript b) diphosphate attacks alpha phosphate of GTP, PPi released = 5'-5' triphosphate linkage 3) methylation by RNA methyltransferase using S-adenosyl methionine: a) cap O: guanine methylated at position 7 b) cap 1: 2'OH of sugar c) cap 2: 2'OH of sugar
384
mRNA 3' polyadenylation steps
1) RNAP transcribes past consensus AAUAAA sequence (polyA addition site) 2) 10-35 nts past, RNAP stalls, resulting in cleavage and polyadenylation specificity factor (CPSF) recruitment 3) CPSF binds consensus sequence and invariant GU in mRNA = looping 4) cleavage factors (CFs) cleave downstream of consensus sequence 5) CFs and 3' fragment dissociate 6) poly(A)-adenylation protein (PAP) recruited to add 100-200 As 7) CPSF dissociates
385
timing of polyadenylation
closely linked to transcription termination
386
RNA editing
- change in nt sequence not as a result of splicing - deamination of C to U or A to I, changing coding possibilities in transcript - introduces diversity from same gene transcript
387
apolipoprotein
fatty acid and steroid transport protein
388
apolipoprotein RNA editing
1) in liver: unedited mRNA translated to ApoB-100 = LDL, VLDL surface 2) in small intestine: enzymatic deamination of CAA to UAA = early stop codon = no LDL receptor binding part = chylomicron surface
389
methods of increasing protein diversity by RNA editing
1) altering amino acid coding possibilities 2) introducing premature stop 3) changing splice site in transcript
390
organization of split eukaryotic genes
DNA: 1) promoter/enhancers 2) start of mRNA 3) exons and introns 4) poly-A addition signal RNA: 1) 5' UTR 2) exons and introns 3) 3' UTR mature mRNA 1) 7-mG cap 2) exons: as little as ~1/3 of DNA coding region! 3) 3'UTR with poly-A tail
391
alternative splicing
- more diversity of EXONs - 2^n possibilities, where n is the number of exons that can be alternatively spliced
392
RNA sequences in intron for splicing
1) 5' splice site: invariant GU 2) branch site: key A residue 3) pyrimidine tract: ~10nts 4) 3' splice site: invariant AG
393
transesterification reaction
- exchange of ester groups - requires no energy
394
two transesterification reactions in splicing
1) 2'OH on key A in branch site attacks phosphate of 5' splice site = precursor to lariat intermediate 2) 3'OH of exon attacks phosphate of exon 2 = spliced product + lariat form of intron
395
splicing branch point: lariat form of intron
adenine has 3 phosphodiester linkages: 1) 5' to 3' 2) 3' to 5' 4) 2' to 5' of guanine
396
snRNP
- small nuclear ribonucleoprotein particles - required for splicing - small RNA (100-200 bases) + ~10 proteins (some general and specific) - regions that bind RNA are protein-deficient to facilitate base-pairing - major snRNP species are abundant, >100 000 per nucleus
397
key snRNP in pre-mRNA splicing
1) U1: 5' splice site 2) U2: branch site 3) preformed complex of U4-6: 5' splice, recruitment of branch point to 5'splice site
398
splicing steps
1) U1 recognizes 5' splice site 2) ATP hydrolysis for U2 binding to branch site 3) ATP hydrolysis for U4-6 complex formation + ATP for binding 4) once U5 aligned at 5' splice site, ATP for U4 dissociation unmasks U6 activity = U2/U6 catalytic site forms across pre mRNA 5) U5 uses ATP to align 2’OH of the A branch site and 5’ intron splice site for first transesterification reaction 6) U5 uses ATP to align3’OH of exon 1 with the 3’ intron splice site for second transesterification 7) ATP releases U5/6/2, another to dissociate U2/6
399
spliceosome catalytic center
- U6/2 base pair - U2 base pairs with branch site = key A residue protrudes - dissociated by ATP dependent helicase
400
CTD recruits which proteins to pre-mRNA
1) capping enzymes 2) splicing components 3) endonuclease
401
how does CTD recruit proteins?
phosphorylation
402
CTD coupling transcription to premRNA processing mechanism
1) phosphorylated CTD has capping enzymes, splicing factors and polyadenylation factors (starting from end inwards) 2) capping 3) spliceosome recruited, splicing factors splice 4) poly(A) added
403
issue of complex RNA processing
difficult to trace mature mRNA back to original gene sequence
404
result of mutations that affect pre-mRNA splicing
50% normal RNA 50% abnormal RNA --> degraded = disease ex. beta globin subunit in thalassemia
405
thalassemia splicing
1) A-->G mutation in an intron, creating new 5' splice site 2) 50/50 chance to assemble on mutated or original splice site 3) if mutated: in-frame stop codon retained, leads to truncated protein
406
tissue-specific splicing and processing
- same gene with multiple poly(A) sites - recognition of site and alternative splicing due to different regulatory factors in tissues - ex. calcitonin/CGRP
407
calcitonin vs CGRP processing
in thyroid: - 1st poly(A) site recognized - CGRP removed - splicing: calcitonin exon kept in brain: - 2nd site recognized - CGRP gene kept - splicing: calcitonin removed
408
self-splicing introns vs spliceosome
- spliceosome more intricately regulated: influenced by snRNP availability, regulated by CTD of RNAP - self-splicing introns do not require proteins
409
types of self-splicing introns
Group 1: 1) GMP/DP/TP cofactor required 2) 3'OH attacks exon 1 5' splice site 3) 3'OH of exon 1 attacks exon 2 (5' end) 4) NO lariat formed Group 2: 1) key A (part of transcript) has 2'OH that attacks exon 1 5' splice site 2) 3'OH of exon 1 attacks exon 2 (5' end) 3) lariat formed
410
ribozyme classes
1) Group 1 introns: eukaryotic genomes (rRNA, tRNA) 2) Group 2 introns: mitochondrial genomes, lower eukaryotes 3) viral RNAs: a) hammerhead ribozyme: plant viruses b) hairpin ribozyme: tobacco plant virus c) hepatitis delta virus ribozyme 4) RNase P 5) spliceosome 6) ribosome
411
Group 1 intron mechanism
1) internal guide sequence (IGS) base pairs with exon 1 (3' region of upstream exon) 2) folding allows binding of G cofactor, aligned so that 3'OH attacks for first TE 3) 2nd TE, spliced RNA and LINEAR intron released
412
IGS characteristics
rich in G
413
self-splicing introns in tetrahymena
1) release of 414nt linear intron 2) intron self-splices 2x: -15, -4 = final L19 catalytic molecule
414
self-splicing RNA 3D structure
- complex, necessary for function
415
self-splicing intron experiments
1) magnesium/protein dependence 2) guanosine cofactor binding site 3) role of magnesium
416
magnesium/protein dependence experiment
1) 26S rRNA requires intervening sequence (IVS) removal for mature produce 2) conditions: a) negative control: unspliced version b) positive control: circular 414, linear L19 c) +/- Mg2+ with no treatment, pronase or proteinase K 3) results: Mg2+ required for splicing, occurred in presence of proteases = Mg-dependent, protein-independent
417
metal ions and self-splicing RNA
stabilizes structure
418
guanosine binding with group 1 intron
guanosine stabilized through triple-base pair formation with G264, C3111
419
guanosine binding site experiments
1) WT RNA a) + guanosine = most efficient b) + 2-aminopurine = most activity lost 2) mutant RNA: A264-C311 or A264-U311 a) + guanosine = limited activity b) + 2-aminopurine = some activity restored for double mutant
420
2-aminopurine as cofactor with mutated guanosine binding site
creates isosteric triple base pair = same # of electrons in same arrangement, but different atoms
421
L19 function
catalyzes nucleotidyl transfer reactions
422
L19 mechanism
1) (C)5 oligomer binds to IGS 2) TE reaction incorporates 1 C at 3' end, releasing (C)4 oligomer 3) another (C)5 oligomer binds IGS 4) TE reaction transfers C to make (C)6 5) L19 restored
423
Mg structural role in group 1 intron
1) coordinates with cofactor 2) stabilizes transition state of TE reaction (positive charge with 2 negative O's)
424
Mg functional role experiment
1) normal or mutated (S-link instead of O to phosphate) 2) Mg2+ (binds oxygen only) or Mn2+ (binds sulfur and oxygen = positive control) cofactor 3) results: a) normal + Mg2+ = splicing b) mutated + Mg2+ = no splicing c) normal + Mn2+ = splicing d) mutated + Mn2+ = splicing 4) conclusion: Mg at the active site is required for splicing
425
characteristics of viral ribosymes
- use metals or base functionalities to assist catalysis
426
translation definition
genomic sequences to protein sequences
427
ribosome subunit functions
large = catalyzes reaction + has exit channel for polypeptide, small = interacts with mRNA
428
active sites in ribosome
Exit: uncharged tRNAs exit Peptidyl: peptide bonds formed Aminoacyl: charged tRNAs enter *in both small and large subunit
429
protein synthesis direction
N to C terminal
430
tRNA role in translation
- deliver coded amino acids to ribosome - requires different sequences to transmit coding info - but similar structure to be used with equivalent efficiencies
431
genetic code determination
- examined which polypeptides were produced in response to synthetic RNA polymers with repeating sequences of three or four bases
432
genetic code features
1) used by all living organisms 2) codon = triplets of nts in mRNA 3) nt sequence written 5' to 3', codons translated in same direction 4) 3nts = one AA 5) synonyms = codons for same AA 6) most synonyms differ in only the last base 7) code is non-overlapping
433
genetic code patterns
- similar codons = similar properties - ex. U first base = hydrophobic
434
non-overlapping meaning
- every three nts = one AA - no overlap of reading frame = this would mean one mutation affects multiple codons vs just one
435
genetic code translation features
1) unambiguous 2) multiple codons for most AAs = degeneracy, helpful against mutations 3) first two nts specify AA, third is variable 4) similar codon = chemically similar AAs 5) only 61/64 are AAs, 3 are stop 6) no punctuation
436
polypeptide chain growth reaction (protein synthesis reaction aka transpeptidation)
1) N-terminal of AA in A site attacks C=O of peptide in P site 2) transition state 3) peptide bond formed, protons released --> H2O = dehydration reaction
437
probability of protein with no errors formula
p = (1-e)^n e = frequency of inserting n = number of AA residues
438
max error rate for large proteins
cannot exceed 10^-4, otherwise excessive production of potentially mutant proteins
439
codon-anticodon pairing
anticodon 3 pairs with codon 1, 2 with 2, 1 with 3
440
tRNA and tRNA synthetase interaction regions
1) position 73 2) acceptor stem 3) variable pocket 4) variable loop 5) variable stem loop 6) anticodon
441
tRNA sequence conservation
conserved = AA attachment site, TΨC loop variable = variable loop, anticodon, other nucleotides near anticodon
442
tRNA structure
1) L-shaped 2) 2 double helix segments from 4 base paired regions 3) bases in non-helical regions = H-bonding participants 4) CCA from 3' end 5) anticodon at other end of L-shape
443
how are tRNAs charged?
specific aminoacyl-tRNA synthetases covalently link AAs with tRNAs
444
tRNA charging mechanism (Class 1)
1) activated by adenylation: AMP added (requires 1 more ATP to regenerate ATP!) 2) 2'OH nucleophilic attack 3) transesterification to 3'-O aminoacyl tRNA
445
tRNA charging mechanism (Class 2)
1) adenylation 2) 3'OH nucleophilic attack *NO transesterification
446
adenylation of AA is analogous to...
charging of fatty acyl coA
447
tRNA nomenclature
1) aminoacyl-tRNA--amino acid--anticodon, ex. alanyl-tRNA = tRNA^Ala a) if wrong: Y-tRNA^Ala 2) aminoacyl-tRNA synthetase, ex. threonyl-tRNA synthetase
448
amino acid specificity for aminoacyl-tRNA synthetases
required for correct addition of AA
449
Thr-tRNA synthetase key residues
- Asp, 2xHis, Cys in active site interact with Thr with Zn2+ coordination
450
importance of OH for Thr-tRNA synthetase
- Zn2+ coordinates with OH and NH3 - Asp H-bonds to OH
451
mischarging of Thr-tRNA synthetase
- Ser common (also has OH) - Val has no OH, not mischarged
452
Thr-tRNA synthetase proofreading site
- incorporation of Ser flips CCA into editing site - hydrolysis to remove - analogous to DNAP proofreading
453
size discrimination proofreading by tRNA synthetases
- larger amino acids rejected from activation site - smaller mischarged amino acids accepted by editing site - ex. Ile-tRNA and Thr-tRNA both use this mechanism
454
discovery of aminoacylation specificity
- mutational studies in yeast, E. coli - different regions important in different tRNAs
455
alanyl-tRNA recognition by alanyl-tRNA synthetase
- relies on key G3:U70 microhelix in acceptor stem - mutation of G3:C70 to G3:U70 in Cys-tRNA leads to recognition by Ala-tRNA = mutant proteins
456
class I vs II tRNA synthetases interaction with tRNA
class 1: interacts with hairpin CCA on face of tRNA class 2: interacts with helical CCA on side of tRNA
457
class I vs II tRNA synthetase amino acids
- each 10/20
458
key differences between class I vs II tRNA synthetases
1) 2'OH vs 3'OH 2) different ATP binding (conformation) 3) monomeric vs dimeric
459
tRNA synthetases and isoacceptor tRNAs
- specificity but can recognize isoacceptors: different tRNAs that for the AA
460
prokaryotic vs eukaryotic ribosomes
1) entire ribosome: 70S (50/30) vs 80S (60/40) 2) large subunit: 23S rRNA, 5sRNA, 31 proteins vs. 5.8S, 28S, 5S, 40 proteins 3) small subunit: 21 proteins, 16S vs 30 proteins, 18S
461
ribosome protein composition
~1/3 protein, areas between subunits have no proteins for RNA binding
462
polysomes
- translation and transcription coupled in prokaryotes - many ribosomes on each mRNA transcript
463
what is NOT the start signal for translation?
first 3 nts of each mRNA
464
where is the start signal in translation?
- approximately 25nts away from 5' end - all mRNAs contain beginning and end signals
465
mRNA unique feature in prokaryotes
all are polycistronic/polygenic
466
shine-dalgarno sequences
- recognized by prokaryotic ribosomes - ~10 nts upstream - purine-rich - base pairs with 3' end of 16SrRNA in small subunit to establish correct reading frame
467
initiator tRNA in prokaryotes
- adds formylated Met (fMet) - different from internal Met tRNA
468
fMet tRNA formation
1) charged with unmodified Met by Met-tRNA synthetase 2) transformylated by Met-tRNAi^fMet formyl transferase: additional formyl group mimics peptide bond, allowing binding to P site instead of A
469
specificity of Met-tRNAi^fMet formyl transferase
does not interact with free Met or Met-tRNA^Met
470
general protein synthesis mechanism
1) tRNAi in P site 2) aminoacyl-tRNA binds in A site 3) peptide bond formation, leading to positional shift in acceptor arms (large subunit) 4) translocation to position next codon 5) uncharged tRNA dissociation from E site
471
translocation in prokaryotes requires...
elongation factor G, which uses GTP hydrolysis
472
protein sequence is coded by...
mRNA sequence only: if AA modified after adenylation and charging, the wrong AA will be incorporated
473
wobble hypothesis
- first two bases of codon and last two of anticodon = canonical WC pairing - last pair less stringent
474
inosine in wobble hypothesis
- appears in first position of many anticodons - can pair with U, C, A = steric freedom - ex. yeast tRNA^Ala anticodon = IGC, recognizes GCU, GCC, GCA
475
U base pairs with...
A or G
476
C base pairs with...
only G
477
A base pairs with...
only U
478
G base pairs with...
U or C
479
I base pairs with...
U, C or A
480
generalizations in codon reading
1) codons that differ in first bases must be recognizes by different tRNA 2) first base of anticodon determines whether tRNA reads 1, 2, or 3 codons
481
monitoring of codon-anticodon base pairing
- 16S rRNA - A1493, A1492 hydrogen bond with with first two codon spots, ensuring WC - G530 also involved - wobble site not proofread
482
initiation factors (IFs)
- aid assembly of 30S initiation complex, then 70S initiation complex - P-loop NTPases (G protein family) that use GTP
483
prokaryote translation initiation steps
1) IF-1/3 bind, IF3 prevents premature joining of 50S 2) mRNA-IF2-tRNAi^fMet bind with GTP in correct reading frame = 30S initiation complex 3) GTP hydrolysis = conformational change, releasing IF1/2/3 and allowing 50S association to form 70S initiation complex
484
elongation factors (EFs)
- deliver aminoacyl tRNAs to A site of ribosome - prokaryotes: Ef-Tu
485
purpose of Ef-Tu binding to aminoacyl tRNA
1) protects ester linkage from hydrolysis 2) GTP hydrolysis only occurs with appropriate codon-anticodon base pairing = fidelity, no hydrolysis means tRNA leaves A site 3) does not interact with fMet-tRNAf (whereas IF2 only recognizes this)
486
Ef-Tu cycling mechanism
1) EF-Tu-GTP binds aminoacyl tRNA 2) GTP hydrolysis if correct pairing, allowing tRNA to bind A site firmly 3) EF-Tu-GDP dissociates 4) Ef-Ts (GEF) regenerates EF-Tu-GTP
487
prokaryote translocation mechanism
1) EF-G binds 50S 2) GTP hydrolysis = conformational change 3) EF-G dissociates
488
EF-G structure
mimics EF-Tu bound amino-acyl tRNA complex, allowing it to fit easily near the A site
489
stop codon recognition
1) aminoacyl tRNA will not bind to A site 2) recognized by release factors (RFs)
490
translation termination reaction
termination factors contain H2O for hydrolysis of polypeptide from tRNA
491
termination factor association with H2O
- Gln, Gly, Gly interact with H2O
492
prokaryote termination mechanism
1) RF1 or RF2 recognizes stop codon, binds with RF3-GTP 2) hydrolysis of polypeptide = release from P site of ribosome 3) RF3 GTP hydrolysis = RF dissociation 4) RRF GTP hydrolysis releases entire translocation complex
493
RF-1 recognizes...
UAA, UAG
494
RF-2 recognizes...
UAA, UGA
495
total required ATP/GTP for protein synthesis
ex. n AA in length 1) charging tRNA: 2 ATP x n 2) initiation (IF2): 1 GTP 3) elongation + translocation (EF-Tu, EF-G): 2 GTP x n 4) termination (RF3): 1 GTP 5) ribosome dissociation (RRF): 1 GTP
496
eukaryotic vs prokaryotic translation initiation
- mostly differ in translation initiation - more IFs (eIFs) - no SD sequence - initiator tRNA is Met - precomplex assembled first to search for start - starts at first AUG near 5' end - circularization for scanning process
497
eukaryotic translation mechanism
1) eIF4F complex and 43S ribosomal (initiator) complex assembled 2) bind mRNA at 5' cap (eIF4F first through eIF4E recognition of m2G) 3) mRNA circularization 4) mRNA scanning with eIF4H (helicase) ATP hydrolysis 5) start codon recognition = GTP hydrolysis = 60S binding, eIFs dissociate
498
eIF4F complex
- heterotrimer: eIF4G bound to eIF4E and eIF4A
499
43S ribosomal complex
- 40S small subunit - many eIFs, including eIF2 - eIF2 bound to GTP, brings in Met-tRNAi^Met
500
Met-tRNAi^Met
different from internal Met, but not formylated!
501
eukaryotic mRNA circularization in translation
eIF4G serves as bridge: - N-terminal binds eIF4E and polyA binding proteins - eIF4E also bound to methyl-G cap
502
eukaryotic vs prokaryotic translocation
1) EF1alpha = EF-Tu 2) EF1betagamma = EF-Ts 3) EF2 mediates GTP driven translocation = EF-G
503
eukaryotic vs prokaryotic termination
- eRF1 recognizes all stop codons - no equivalent of RRF or RF3
504
antibiotics interfering with translation
- can be prokaryote or eukaryote specific or both - can inhibit different steps of translation
505
puromycin mechanism
- analog of terminal aminoacyl A residue of aminoacyl tRNA, allows fit into A site - has amide linkage instead of ester = no more ester to be nucleophilically attacked once incorporated - unstable in P site, leading to dissociation
506
streptomycin mechanism
- highly basic - interferes with initiator tRNA binding in prokaryotes
507
Diptheria toxin mechanism
1) cleaved into A and B fragments in cells 2) A fragment catalyzes covalent modification of diphthamide (modified His in eEF2) using NAD+ = ADP-ribose added 3) EF2 function blocked (no translocation)
508
ricin mechanism
1) heterodimeric catalytic A chain and B chain by S-S bond: released in cell cytoplasm 2) A chain (N-glycosidase) removes A4324 in 28S rRNA = abasic site 3) prevents elongation factor binding