lecture II: drug-target characterizations Flashcards

1
Q

What is needed to experimentally determine the affinity of a drug on a target?

A
  1. Pure drug
  2. Pure target
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How can a pure target be obtained?

A

Protein purification

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

Protein purification methods

A
  1. Protein expression in bacteria
  2. Chromatography
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Protein expression in bacteria steps

A
  1. cDNA encoding for protein of interest ligated into plasmid/vector for bacterial expression
  2. Transform bacteria
  3. Grow over night
  4. Harvest bacteria
  5. Cell lysis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Which method often uses protein expressed in bacteria?

A

Crystallography

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

Crystallography

A

Crystallography is the experimental science of determining the arrangement of atoms in crystalline solids.

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

Chromatography

A

Chromatography is a process for separating components of a mixture. To get the process started, the mixture is dissolved in a substance called the mobile phase, which carries it through a second substance called the stationary phase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

FPLC

A

Fast protein liquid chromatography

A typical laboratory FPLC consist of one or two high-precision pumps, a control unit, a column, a detection system and a fraction collector.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Types of chromatography

A
  1. Size exclusion
  2. Affinity
  3. Ion exchange
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Radioligand competition assay

A
  • competitive binding assay
  • measures affinity
  • method: A known antagonist (or ligand) for the target receptor is labelled with radioactivity and is added to cells or tissue such that it can bind to the receptors present. Once an equilibrium has been reached, the unbound ligands are removed by washing, filtration, or centrifugation. The extent of binding can then be measured by detecting the amount of radioactivity present in the cells or tissue, and the amount of radioactivity that was removed.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Radioligand competition assay evaluation

A

log([competing ligand]) vs. amount of radioligand bound

→gives IC50

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

IC50

A

Inhibitory concentration 50

Concentration at which the competing ligand displaces 50% of the radioligand.

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

Identification of same or different binding sites

A

Radiolabeled drug with known binding site (drug A) and addition of other drugs with unknown binding sites.

→ if the drug competes with drug A, then it has the same binding site
→ if the drug does not compete with drug A, then it has a different binding site

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Elution

A

Elution is the process of extracting one material from another by washing with a solvent; as in washing of loaded ion-exchange resins to remove captured ions.

→ essentially, a type of competition

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

Size exclusion chromatography

A

Size exclusion chromatography separates molecules based on their size by filtration through a gel. The gel consists of spherical beads containing pores of a specific size distribution. Separation occurs when molecules of different sizes are included or excluded from the pores within the matrix.

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

Size exclusion chromatography elution order

A
  • constant buffer flow & porous material
  1. Large molecules excluded from pores → elute first
  2. Small molecules diffuse in pores → elute later
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Gel filtration: 205 nm absorbance

A

Peptide bond

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Gel filtration: 280 nm absorbance

A

Tryptophan

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Affinity chromatography

A

To separate recombinant protein from other proteins by its biophysical traits.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Affinity chromatography: different traits

A
  1. Hydrophobicity
  2. Ligands
    →lectins, protein A/G, collagen
  3. Metals
    →IMAC
  4. Protein tags
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Protein tag examples

A
  1. His ∞ Ni-NTA
    →6 His in a row likes to bind to Ni
  2. GST ∞ glutathion-sepharose
  3. MBP ∞ amylose resin
    →larger protein tag, but it may help POI to remain in solution when in bacteria
  4. immunoprecipitation: myc, flag ∞ antibodies
    →from myc pro-oncogene expressed in our cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

SPR

A

Surface plasmon resonance

Surface plasmon resonance is a biophysical phenomenon happening when light strikes a gold layer at the interface of two media (glass and buffer) at the angle of total internal reflection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

SPR: Total angle of internal reflection

A

Angle of light at which total reflection occurs - light does not enter other media anymore

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

SPR steps

A
  • POI is immobilized on surface
  • Drug passes over chip surface in buffer solution

→when molecules/drugs are injected with buffer and bind to chip, it results in a change of mass on chip surface, subsequent change in diffractive index between media, which is a proportional change in diffraction angle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Evanescent wave

A

Evanescent waves are formed when sinusoidal waves are (internally) reflected off an interface at an angle greater than the critical angle so that total internal reflection occurs.
→the size of the evanescent wave changes as drug binds to protein on the chip, therefore the shadow/angle changes as well (proportional to mass change)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

SPR: light angle

A

Light is shone at the total reflection angle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

SPR: gold

A

Gold layer absorbs energy, resulting in decreased light intensity at a certain angle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

SPR sensorgram

A

Measures the resonance signal, and thus affinity

→ time vs. resonance signal

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Resonance signal

A

Change in angle of “shadow” in reflected light

→dependent on the mass found on the chip, which is then reflected in the shadow angle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

SPR sensorgram phases

A
  1. Buffer flow
    injection of drug
  2. Association of drug with POI till equilibrium →on rate only
  3. Equilibrium point
    wash off bound drug (buffer flow only)
  4. Dissociation of drug →off rate only
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

KA

A

Equilibrium constant KA describes association tendency

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

KA equation

A

KA = [LR] / ([L][R])

→low KA = low affinity
→unit: M^-1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

KD

A

Equilibrium constant KD describes dissociation tendency

→When drug is given at a concentration equal to its dissociation constant KD, 50% of the receptors will be occupied.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

KD equation

A

KD = ([L][R]) / [LR]

→low KD = high affinity
→unit: M

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

How to test specificity of drug binding?

A

Use the concept of “competition” to show specificity

→using SPR, inject drug+buffer, then inject buffer only as a control

36
Q

Target identification method

A

Use of trifunctional capture compounds, where you modify the drug to fish for protein-interaction partners

37
Q

Trifunctional capture compound components

A
  1. Capture compound
    →drug of interest
  2. Reactive azide group
    →forms a covalent bond upon UV light
  3. Biotin
    →for purification
38
Q

Avidin-Biotin

A

Naturally occurring strong interaction between avidin and biotin.

→interaction: KD ≈ 10^-14
→synthetic avidin will be used and bind to the biotin moiety will very high affinity, almost ensuring that our protein will be pulled out

39
Q

Biotin

A

Essential cofactor for a lot of enzymes.

40
Q

Avidin

A

Functions as an antibiotic in the eggs of birds, reptiles and amphibians.

41
Q

Target identification: trifunctional capture compound steps

A
  1. Add trifunctional compound to cells/tissue/mice…
  2. Crosslink compound to proteins by UV light (like a shot of the moment where it is bound)
  3. Purify trifunctional compound using biotin-streptavidin
  4. Protein digestion
  5. Perform mass spectrometry to identify proteins that were co-purified and thus qualify as potential drug-binding partners
42
Q

Protein digestion

A

Typically done trypsin (protease), which only specifically cleaves after Lysine (K) or Arginine (R)

43
Q

Peptide mass fingerprint

A

Each protein has its individual unique pattern of tryptic peptides.

44
Q

Mass spectrometry

A

Mass spectrometry is an analytical tool useful for measuring the mass-to-charge ratio (m/z) of one or more molecules present in a sample.

→ m/z vs. intensity

45
Q

Mass spectrometry: functional compounds

A
  1. Ion source
    →ionization
  2. Mass analyzer
    →ion separation
  3. Detector
    →ion detection
46
Q

Ionization methods

A
  1. Electrospray ionization
  2. Matrix-assisted laser desorption ionization
47
Q

Mass analyzer methods

A
  1. Time-of-flight (TOF)
  2. Quadrupole
  3. Ion trap
48
Q

Detector methods

A
  1. Electron multiplier
49
Q

Fingerprint analysis

A
  1. Generate list with exact peptide masses measured in spectrum
  2. Compare list to database
    →“in silico” cleavage of known proteins in a certain species
50
Q

Peptide validation by MS

A
  • Collision of peptides with other molecules (air) will break peptide bonds
    →N-terminal peptide fragment & C-terminal peptide fragment
  • Distance between two peaks usually mass of one amino acid
51
Q

Florescence

A

Fluorescence is the emission of light by a substance that has absorbed light or other electromagnetic radiation. It is a form of luminescence.

52
Q

Fluorophore

A

Molecules that absorb light, and emit light at longer wavelengths after a certain time.

53
Q

Two features of fluorophores

A
  1. Emission always at a higher wavelength
  2. Always mirror-imaged emission/excitation peaks
54
Q

Fluorescence measurement

A

Perpendicular to the light source

55
Q

Absorption measurement

A

Parallel to the light source

56
Q

Jablonski diagram

A

Diagram that illustrates the electronic states of a molecule and the transitions between them.
→Lengths of arrows correlate with energy – emitted photon has less energy than absorbed photon

57
Q

Short wavelength, _____ energy

A

high

58
Q

Long wavelength, _____ energy

A

low

59
Q

Stoke’s shift

A

The difference (in energy, wavenumber or frequency units) between positions of the band maxima of the absorption and emission spectra of the same electronic transition.

→explains why emission is always at a higher wavelength

60
Q

Non-radiative transition / Internal conversion

A

Solvents take energy.

→photons like to be excited before they drop, causing for a loss in energy
→may occur with interaction with water

61
Q

Fluorophore fluorescence lifetime

A

Time spent in the excited state
→E will be lost after a while

62
Q

Franck-Condon principle

A

During the transfer from the ground state to the excited state, it is most likely that transfer happens between the best overlapping vibrational wave functions.

→discrete positions for excitation and emission
→explains why there are always mirror-imaged excitation/emission peaks

63
Q

What may hinder ideal fluorescence spectra shape?

A
  • dipole moments
  • interactions with solvents
64
Q

Visible light spectrum

A

380 (violet) - 740 (red)

65
Q

Causes of radiative relaxation

A
  1. Internal conversion
  2. Quenching
  3. Chemical reactions
66
Q

How can you measure fluorescence?

A

96-well plate reader for absorption, fluorescence, (luminescence)
→use of excitation/emission filters to measure fluorescence

67
Q

When is a black 96-well plate used?

A

For fluorescent assays.

→black plates absorb light and reduce background and crosstalk

68
Q

When is a white 96-well plate used?

A

For luminescent assays.

→white plates reflect light and will maximize light output signal

69
Q

Applications for fluorescence

A
  1. Protein-protein interactions
  2. Quenching
  3. Mobility of proteins
  4. Processes at the plasma membrane
  5. High resolution microscopy
  6. Fluorescence anisotropy
70
Q

Applications for fluorescence: Protein-protein interactions methods

A
  1. FRET
  2. BRET
  3. Split-YFP system
71
Q

FRET

A

Fluorescence Resonance Energy Transfer.

→when emission spectrum of donor overlaps with excitation spectrum of acceptor, FRET will occur in close proximity of fluorophors (less than 10 nm)
→radiation of the donor will be transferred to the acceptor
→shine excitation light of donor and, if the two proteins are in close proximity, the emission of the acceptor will be the output
→use as a ruler to see how close proteins in sample are

72
Q

BRET

A

Bioluminescence energy transfer.

→exploits the naturally occurring phenomenon of dipole-dipole energy transfer from a donor enzyme (luciferase) to an acceptor fluorophore following enzyme-mediated oxidation of a substrate
→light emission through luciferases
→substrate luminol undergoes reaction with oxygen to release light
→donor fluorophore is not fluorophore, but rather a luminescent substrate
→when emitted light is in close proximity to a fluorophor that is being excited at 490 nm

73
Q

Quenching

A

Gain of fluorescence after enzymatic activity.

→quencher is not a fluorophore (it accepts light, but doesn’t do anything with it)
→excite fluorophore and a large proportion
of its E will be transferred to quencher, which will not do anything with that light, but then post-reaction you get fluorescence if the protease completed its function

74
Q

Applications of fluorescence: mobility of proteins method

A

Fluorescence recovery after photobleaching (FRAP)

75
Q

Applications of fluorescence: processes at the plasma membrane method

A

Total internal reflection (TIRF)

76
Q

Applications of fluorescence: high resolution microscopy method

A

Stimulated emission (STED microscopy)

77
Q

Fluorescence anisotropy

A

A measurement of how a molecule changes its orientation in space, with respect to the time between absorption and emission events.
→describes how much emitted light is still polarized
→indicator of how fast the protein moves with/without inhibitor
→depends on the direction of the fluorophore’s dipole

78
Q

Photoselection

A

Only the fluorophores that align to the orientation of the polarized light will get (best) excited by polarized light.

79
Q

Polarization filter with vertical passage

A

Allows unpolarized light (all directions) to be linear polarized light with only one plane of oscillation.

80
Q

Explain theory behind anisotropy.

A

See slides 57, 59, 61

81
Q

Possible movements of fluorophore

A
  1. Rotational diffusion of the fluorophore
  2. Mobility of protein segment
  3. Rotation of the whole protein
82
Q

Anisotropy measurement: slow fluorophore movement

A
  • polarized light emitted (parallel)
  • bound to something
  • high affinity
83
Q

Anisotropy measurement: fast fluorophore movement

A
  • depolarized light emitted (in all directions, more perpendicular)
  • unbound
  • low affinity
84
Q

Applications of anisotropy

A
  1. Compound screening assays
  2. Fluorophore label at selected positions in target – monitor mobility
  3. Use of tryptophan residues (intrinsically fluorescent)

→often used to measure binding affinity

85
Q

Advantage of anisotropy

A

It is a ratio measurement, thus values are intrinsically normalized