Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

ELECTROPHORESIS

SLAB (THIN LAYER GEL) AND


CAPILLARY METHODS

A. General Introduction
• Electrophoresis: a separation method based on
differential rate of migration of charged species in an
applied dc electric field
• Rate of migration
– Depends on charge and size
– Separation based on differences in charge-to-size
ratios
• High efficiency and resolution

1
Historical notes
• Initially developed by Arne Tiselius in the 1930’s
– Separated serum proteins
• Slab gel electrophoresis: developed in the 1950’s
• Capillary electrophoresis:
– developed in the 1980’s
– Narrow bore tubes used
• Applications of electrophoresis:
– Separation of proteins and nucleic acids (single
nucleotide differentiation capability!)
– The human genome Project
• Human DNA: ~three billion nucleotides

B. Principle and Theory of


Electrophoresis
• Basic requirements
– Conducting medium (aqueous buffer/ electrolyte/
run buffer)
– Applied Electric Field
– Positively charged species move to the cathode (-)
– Negatively charged species move to the anode (+)
• Two Basic Techniques
1. Free solution: narrow capillary bore (Instrumental)
2. Non-conducting matrix (Agarose, Polyacrylamide
gel/PAGE)
a. Joule heating is minimal
b. Sieving effect of gel allows separation of species with
same charge to size ratio if they have different sizes.

2
• Efficiency depends on:
– Electrophoretic mobility (µep)
– Electroosmotic flow of the bulk solution (EOF)
– Joule heating

B-1 Electrophoretic Mobility


• Ion placed in an electrical field (E) experiences
force Fef that is proportional to field strength and
the charge (q) on the ion (Fef=q.E)
• As the ion moves, a frictional force (ffr) opposes
the forward movement of the ion
• In a constant electrical field the velocity of particle
is constant and depends on the balance between
the two forces
• Electrophoretic mobility is the fundamental
parameter which determines the efficiency of
separation based on charge to size ratio (q/r)
• Change in pH effectively alters the charge on the
ions and their electrophoretic mobility.

3
Electrophoretic Mobility
Fef = q × E

F fr = 6 × π × η × r × v ep

η : vis cos ity


r : radius
v ep : electropho retic − migration − velocity

Fef = F fr

q×E
v ep =
6 × π ×η × r
v ep q q
µ ep = = = const ×
E 6 × π ×η × r r
µ ep : electropho retic − mobility

B-2 Joule Heating


• Ohmic/Joule heating
– heating occurs as charged particles move within the conducting
buffer upon application of an electrical field.
• Temperature gradient are generated leading to convective
flows within the electrolyte
• Convective flow leads to band broadening
• Increase in temperature can also damage
macromolecules.
• Method for decreasing joule heating:
– Decrease applied voltage- leads to long analysis time
– Dissipate heat- use thin gel or small diameter
capillaries which have large surface-to-volume ratio.
Their electrical resistance is high, thus current flow is
reduced for a given voltage.

4
B-3 ElectoOsmotic Flow (EOF)
• EOF refers to migration of the bulk liquid toward the cathode
• Origin: formation of a double layer at the wall of the capillary.
– Glass, fused silica, agarose exibit surface charges
– Silanol groups are deprotonated at pH higher than 4.
• Potential difference is established
• Positive ions in solution migrate to the wall. A double layer is developed at the
wall of the capillary.
• Stern layer (SL): layer of positive charge that compensate for the negative
charge on the surface
• Diffuse layer (DL): layer adjacent to the Stern layer: layer of mobile cations

SL Incomplete neutralization

B-3 ElectroOsmotic Flow (EOF)


• Upon application of the electrical field,
cations within the double layer are ε ×ς × E
attracted towards the cathode and drag v EOF =
4 × π ×η
the bulk solvent with them.
• EOF is in opposite direction to analyte ε : dielectric − cons tan t
electrophoretic flow (generally) ς : zeta − potential ( SL − DL)
• EOF is practically equal across the v EOF
capillary, thus band broadening is µ EOF =
E
minimal
• EOF are not reproducible thus leading v EOF = µ EOF × E
to irreproducible separation efficiencies
• EOF in GE is minimal or practically
inexistent

HPLC flow
EOF

5
Use of EOF in Capillary Electrophoresis

• If EOF is more important that


electrophoretic mobility, all analytes
++ _
are dragged by the bulk solvent ⊕ +
towards the negative electrode
(Mobile Phase??!!)
• Electropherogram -
--
• The order of elution is: fastest cation,
slower cations, neutrals (single zone),
slowest anion, faster anions ++

-
--

Control of EOF in Capillary Electrophoresis

• Work at low pH
• Dynamic coating of the channel walls
– With Polyethylene glycol (PEG) added to the buffer
– Polymer layer masks charges and suppress EOF

• Chemical modification
– With trimethylchlorosilane-TMCS bonds to the surface and reduces the
number of silanol groups- low EOF
– Sulfonic acid- high EOF
– Problem: long term stability
• Use additives to change the viscosity and zeta potential
– Hydroxy ethyl cellulose or polyvinyl alcohol increase viscosity of the run
buffer and thus reduce vEOF.
– Organic solvents: methanol (reduce) and acetonitrile (increase)
– Cationic surfactants such as dodecyl trimethyl ammonium bromide
(DoTAB) adsorb onto the capillary walls and thus change the surface
charge

6
B-4 Separation Efficiency and Resolution

• Efficiency and resolution depend on both the


electrophoretic flow and the EOF
• Apparent mobility is given by:
µ app = µ ep + µ EOF
• If EOF dominates, all analytes move towards
the cathode (-)

Efficiency and Resolution


• If EOF is more important that
electrophoretic mobility, all analytes are
dragged by the bulk solvent towards the
negative electrode (Mobile Phase??!!)
• Electropherogram
• The order of elution is:
1. fastest cation
2. slower cations
3. neutrals (single zone),
4. slowest anion
5. faster anions

( )
v = µ ep + µ EOF E = µ app
V
L
L L2
t = =
v µ appV

t : migration − time
L : length − of − capillary

7
Band Broadening
• Main contribution to band
broadening: longitudinal
diffusion
• Peak “dispersion” is 2 × D × L2
proportional to the diffusion σ = 2× D×t =
2
coefficient, D and the µ app × V
migration time of the
analyte
• In theory: efficiency superior L2 µ app × V
to LC. Calculated N at N= =
operating voltages are on σ2 2× D
the order of 106.
• In practice: joule heating,
sample injection and
adsorption of analyte to
matrix decrease efficiency

Resolution

• Resolution depends on
the difference in
electrophoretic mobility
– What is the equivalent in
LC?
1 1
• Resolution depends on Rs = ∆µep × V × ×
the applied voltage V, µapp 4× 2× D
the apparent
electrophoretic mobility
and the diffusion
coefficient D.
– What are the equivalents
of these terms in LC?
• Optimizing resolution:

8
C. GEL ELECTROPHORESIS
• Matrix: electrically non-conductive hydrogel containing buffer
– Agarose
– Polyacrylamide
• Advantages
– Porous gel acts as a sieve
– Gel limits diffusion of sample molecules
– EOF is suppressed
– Joule heating is suppressed
• Disadvantages
– Slow, labor intensive and not readily automated
• Techniques
– Native gel electrophoresis: analyte separated according to differences in
apparent mobility
– Sodium dodecylsulfate-polyacrylamide gel electrophoresis (SDS-PAGE):
analyte separated according to size
– Isoelectric Focussing (IEF)
– Two dimensional gel electrophoresis (2D-GE)

C-1 Instrumentation for Gel Electrophoresis


• Power Supply
– 200-500 V
– 400 µA -100 mA
• Electrophoresis Chamber with Buffer
Reservoir
• Electrodes in buffer reservoir
• Mini Gel 8 cm x 8 cm
• Larger gel 40 x 20 cm

9
Gel Media
• Agarose
– 1g in 50 ml (2%)
– Dissolve, heat, cool, pour in casting stand
– Large pore size (e.g. 150 nm for 1%): no
sieving
– Charged surfaces: EOF present

Polyacrylamide Gel
• Polyacrylamide
– Copolymerization of
acrylamide and N,N’-
methylene-bisacrylamide
– Initiator: TEMED
(N,N,N′,N′-
Tetramethylethylenedia
mine) and Ammonium
Persulfate(NH4)2S2O8 )
– Sieving effect present: pore
size depends on total gel
concentration (%T: 5 -20%)
and degree of cross linking
– SDS for denaturinf and
coating proteins
– Mercaptoethanol for
reducing dissulfide bonds

10
Sample Preparation
• Buffer:
– Tris (pH 8), Tris-glycine (pH 9.1)
– 50-100 mM
• Amount of sample: µg
• Sample volume: depends on size of well (
µL to mL)
• Denaturing agent: SDS
• β-mercaptomethanol: to reduce disulfide
bonds

Visualization and detection


• Staining
– Coomasie brillant blue
(alchoholic solution of dye)
– Silver nitrate
• Fluorescent reagents
– Ethidium Bromide for DNA

11
SDS PAGE

• Proteins are totally denatured and coated SDS


charge (negative): rod-like shaped
• Charge on protein depend on its size: constant
net charge per unit mass: same electrophoretic
mobility
• Separation based on size/ molecular weight
• Sample preparation: heating (90) in the
presence of SDS and β-mercaptoethanol
• Used to determine MW of proteins
• Molecular weight standards used

Isoelectric Focussing (IEF)


• Separation based on pI • Apply field:
• Agarose gel used – Negatively charged
• pH gradient from cathode to anode amphoyte will towards
• Several ampholytes with different pI the anode (lowest pI at
used (same concentration to maintain end).
conductivity constant) – Positively charged
• Gel immersed in medium pH mixture (1 ampholytes will move
– 2 % carrier ampholyte) towards the cathode
(highest pI at end).
• Most ampholytes are charged – Cathodic side becomes
• Anode compartment: low pH buffer in more acidic
(lower than the lowest pI) – Anodic side becomes
• High pH buffer in cathode more basic
compartment (higher the highest pI) – pH range determined
by choice of
ampholytes

12
2D GEL
• First dimension separation in single lane:
IEF
• Second dimension separation: SDS-PAGE
• 1000 to 2000 proteins can be separated

Capillary Electrophoresis Instrumentation

• Capillary
– Fused silica
– Diameter: 20 to 100µm
• Length 10 -100 cm
• Typical voltages: 10-30 kV
• Current 300µA
• Temperature control to control EOF and Joule
heating
• Buffer (10 – 100 mM): pH and conductivity control
• Detection: UV, fluorescence, refractive index, etc.

13
CE: Performance Characteristics
• EOF present
• Convective flows due to joule heating
• Excellent for both large and small
biomolecules (amino acids, peptides)
• Short separation time
• Small sample: nL
• Solvent consumption low: few mL
• Neutral analytes can be separated using
Micellar ElectroKinetic Chromatography
(MEKC).

• Automatic pressure or
electrokinetic sample injection
- Built-in capillary rinsing
system
- Convenient access to the
vials and capillary
- Possibility of visual check of
the position of the vials and
capillary rinsing
- Liquid cooling system in
CAPEL103 and in CAPEL-105,
CAPEL-105M
- Built-in monochromator (190 -
380 nm) in CAPEL-105 and
CAPEL-105M

14
Capillary Zone Electrophoresis (CZE)
• Controlled electroosmotic flow used
• Separates anions as well as cations
• Separation of cations: no coating of walls
– Positively charged “mobile phase” moves towards
cathode
• Separation of anions: treat walls with an alkyl
ammonium salt: reverse osmotic flow
– Negatively charged “mobile phase” moves
towards anode
• Apparent mobility

µ app = µ ep + µ EOF

Separation of 30 anions including glutamate


, galactarate, (2 – 10 ppm)

15
CZE of Anti-inflammatory drugs

• (1)naporexen
• (2)ibuprofen
• (3) tolmetin

CZE separation of protein mixture

1. Cytochrome c
12,400, pI = 10.7
2. Lysozyme, 14,100, pI
= 11.1
3. Trypsin, 24,000, pI =
10.1 pH = 2.7
4. Trypsinogen, 23,700,
pI=8.7
5. Trypsin inhibitor, 20,
100, pI= 4.5

16
Capillary Isoelectric Focussing (CIEF)
• Isoelectric focusing
– Capillary filled with ampholyte mixture
– Electric field applied: pH gradient develops within capillary
– Sample can be introduced with ampholyte mixture
• Capillary must be coated to suppress EOF
• Detection at a fixed point
– Mobilization of the focused bands is necessary for detection
• Mobilization Methods
– Hydrodynamic mobilization
• After focussing
• Capillary attached to a pressure or vaccum pump
– Electrophoretic (salt) mobilization
• After focussing
• Addition of salt to cathode buffer (NaCl): disrupts pH gradient
(proteins dragged towards the cathode)

CIEF of Proteins

17
Micellar Electrokinetic Chromatography
(EKC)/CE and LC
• Micelles included in run buffer
– SDS - (CMC: 8 mM) (25 – 150 mM used)
• Partitioning of analyte between core of micelles (hydrophobic) and
run buffer
• Both micelles and buffer are moved by applied electrical field
– Two mobile phases?!
• Neutral analytes are separated according to their hydrophobicity
• Elution in SDS
– Micelle is negatively charged: low apparent mobility due to
electrophoretic mobility towards the anode
– Hydrophilic substances elute with the buffer (t0)
– Very Hydrophobic substances which are completely solubilized by the
micelle elute with the micelles (tmc)
– Others elute between the two
• Applications:
– separation of amino acids, oligopeptides, nucleic acids, fatty acids,
steroids and pharmaceutical drugs

Capillary Gel Electrophoresis


• Gel
– anti-convective medium
– sieving medium
• Separation of analytes with similar mobilities
• Advantages
– Speed
– Efficiency
– Fully automated
– No casting of gels, no staining, no scanning!!! NO WORK!?
• Disadvantages
– No multisample capability
– No 2D capability
• Applications
– Separation of ssDNA, dsDNA, RNA
– Sizing of DNA fragments

18
CGE separation of SDS denatured proteins in
polyethylene glycol column

(1) lactalbumin
(2) Soybean trypsin inhibitor

(4) Ovalbumin

(5) Bovine serum albumin

(3)Carbonic anhydrase

(6) Phosphorylase

19

You might also like