• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/119

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

119 Cards in this Set

  • Front
  • Back

Electrophoresis

Migration of charged solutes or particles in an electrical field

Electrophoresis is a great method to

separating charged biomolecules

Components of electrophoresis

  • Driving force
  • Support Medium
  • Buffer
  • Sample
  • Detecting system

Charged particles migrate towards

the opposite charged electrode

Velocity of migration is controlled by

  • Net charge of the particle
  • size and shape of the particle
  • strength of the electrical field
  • chemical and physical properties of the supporting medium
  • Electrophoretic temperature

Cations migrate towards

the negatively charged cathode

Anions migrate toward

the positively charged anode

Neutral molecules

are not attracted to either electrode

Electrophoresis works because

a particle with an electrical charge moves in a viscous medium by the force of an electrical field

How is the driving force is counterbalanced

The frictional resistance that the molecule must overcome to migrate in the support medium

The velocity of a charged particle in a electrical field depends on:

  • (E) electrical field
  • (q) particle charge
  • inversely on counteraction forces

Counteraction drag generated by

viscous drag

Equation for movement of charged particles in an electrical field:

-V=Eq/f


V: velocity of the molecule


E: electrical field volts/cm


q: net molecule charge


f= friction coefficient , related to molecular mass and shape

Support matrix types that proteins are applied to:

At a single pH


  • Cellulose acetate
  • agarose
  • starch
  • polyacrylamide

Conventional electrophoresis is:

  • Performed on gel paper
  • minimal automation
  • long analysis times
  • detection performed post separation
  • low voltages (because of heat build up)

Electric current across the support causes the protein to:

migrate at a constant velocity, based on their surface charge, size, and shape

Power supply either run on:

Constant voltage


Constant power

Ohm's law

V=IR

Voltage

current (amps) X resistance

Constant voltage depends on:

q/f (charge/resistance)


migration rate is proportional to the molecular charge to mass ratio

Electrophoretic factors

  • Molecule charge
  • molecular size and shape
  • ionic strength
  • buffer pH
  • voltage
  • temperature
  • time
  • diffusion (when power is off)
  • wicking (evaporation)
  • matrix
  • electro-osmosis

Acidic pH will give a molecule a

positive charge

Alkaline pH will give a molecule a

negative charge

Peltier device

In automated electrophoresis:


conducts heat away from the gel to prevent protein denaturation

Why are high quality reagents needed

impurities may effect electrophoresis

Buffer is responsible for:

carrying the current

The buffer pH determines

The charge of the solutes and thus their migration

Buffer ionic strength

determines the electroendosomsis

What happens if the gel runs too long?

the solutes can run off the gel (except in IEF)

Tracking dye

bromophenol blue- small anionic compound moves the fastest

Band diffusion

separated bands tend to diffuse and broaden after the power supply is turned off

How to stop protein diffusion

fixing the gel

How to remove unbound stain

destain to produce a clear background

What specimen should be used

Serum- plasma has fibrinogen which produces an extra band

Support media

cellulose acetate or agarose gel

Current and volatge

constant

Types of stains

  • Colormiteric
  • Fluorescent
  • Enzymatic
  • Radioactive

Protein Dyes

  • Amido black
  • Ponceau S
  • Bromophenol blue
  • Coomassie brilliant blue

Silver stains

10-1000 times more sensitive than dyes:

Protein dye interactions depends on

  • Type of protein
  • Degree of protein denaturation

Destains typically used:

  • 7-10% acetic acid
  • water: methanol: acetic acid (5:5:1)

Quantitation

Performed by densitometry

What is a densitometer

reads electrophoresis pattern

Parts of densitometer

  • Light source
  • Filter system
  • photo-detector
  • converts electrical signal into numbers

The denser the band

the more light they absorb

Tracing of the pattern of a gel and

calculates the percentage of each fraction

In-gel kinase

Method to identify kinase substrate:


enzymes can still be active after electophoresis; the product of its substrate is visualizable

Ethididium bromide

interclataes between bases increases its fluorescent intensity

Visualizing nucleic acid stains

under uv light as red-orange stains on the gel

Autoradiography

  • In a darkroom the gel is covered with a heavy chromatography paper
  • Covered with plastic wrap
  • Placed on an x-ray film
  • triple sandwich is placed in a folder incubated at -70 to avoid band diffusion

Biotin

can be attached to large biomolecules; properties of the enzyme are measured

What pH is used for protein separation

over 8.0 for optimal separation:


proteins will carry a net negative charge

Endosmosis

movement of solvent and solutes in relation to the fixed support

Electroendosmosis

  1. Neg charge on gel
  2. Positive charge cations in the buffer move toward the cathode
  3. Uncharged or weakly negative charged molecules are carried toward the cathode by the electroendosmotic movement

What support media eliminate electroendosmosis?

Neutral support media:


agarose


acrylamide


Heat problems

  • Heat= volts X current X time
  • current meets electrical resistance which causes heat build up
  • Compensate by cooling gel or lower voltage

Wicking Problems

Heat can cause solvent to evaporate from the gel:


  • wicking of the buffer onto the gel from both compartments to replace the solvent that evaporate
  • flow of buffer from both directions affects migration

Types of electrophoresis

  • Paper
  • Starch gel
  • Agarose gel
  • Cellulose acetate
  • Polyacrylamide
  • disc gel
  • IEF, IEP, IFE, CE

Disc gel electrophoresis layers

  • Upper layer
  • Lower layer

Disc gel:


upper layer

stacking gel

Disc gel:


lower layer

resolving gel

Disc gel:


The buffers for two layers

they are different ionic strength

Disc gel:


Pore size of the two layers

  • Stacking gel: decreased concentration of acrylamide (larger pore size)
  • Resolving gel: increased percentage of aryclamide (smaller pore size)

The two layer format causes the proteins to:

form highly concentrated bands: allows for greater resolving power

PAGE

polyacrylamide gel electrophoresis

How does PAGE separate biomolecules

size sieving and charge; small molecules move faster

Chemical polymerization in PAGE is controlled by:

free radical initiator-catalyst system- TEMED/Ammonium persulfate

Photochemical polymerization is initiated by

riboflavin in the presence of UV radiation

The PAGE gel is formed by:

the free radical polymerization of acrylamide and the cross-linking agent N,N'-methylene-bisacrylamide

Acrylamide Reagents:

All are toxic:


  • Acrylamide
  • N,N,N,N'-tetramethylethylenediamine (TEMED)
  • N,N'-methyllene-bis-acrylamide
  • Ammonium persulfate

Resolving power of PAGE

Depends on the concentration of acrylamide and bisacrylamide


  • Lower concentration gives gels larger pore size
  • Hi conc gives gels smaller pore size

pH of PAGE

basic;


most biomolecules neg charged and will migrate to the anode

SDS-PAGE

Separates biomolecules based on size:


  • Proteins interact with the detergent of about 1%
  • Produces a linear peptide chain

SDS

Sodium dodecyl sulfate

SDS masks:

the charge of the protein by forming anionic complexes; give proteins a net negative charge per unit mass

SDS disrupts what bonding

hydrogen



blocks hydrophobic interactions and partially unfolds proteins (eliminating secondary and tertiary structure

SDS-Page can be further processed

Western blot

Nucleic acid gel:


Sample pretreatment

  • Nucleic acids are digesting into fragments of varying size with common ends
  • Samples are treated with a denaturant (urea or formamdie) to prevent denaturation
  • Bases are modified

Nucleic acid gel electrophoresis

Nucleic acid fragments with common ends are electrophoretically separated by PAGE:



Optimized by altering % acrylamide

Nucleic acid detection

  • Labeled with 32P or a fluorescent tag
  • Autoradiography: radioactive
  • Fluorimeter: fluorscent tag

Base pair range of DNA and RNA characterized

200-50,000 bp

DNA electrophoresis matrix

Agarose, seaweed extract; serves as a sieve



Density dependent on the concentration

DNA charge

Net negative charge (due to phosphate group)



Migrates towards the anode

Rate of DNA migration is dependent upon:

  • Size (bp length)
  • Shape
  • Agarose concentration
  • voltage
  • buffer composition
  • dye

DNA mobility

  • Supercoiled compact DNA migrates faster through the gel than linear or circular shapes
  • Shorter fragments move faster than longer fragment

DNA size determination

  • With linear DNA; migration rate is inversely related to the log of the DNA fragment length in bp

DNA visualization

  • Ethididium bromide- fluorescent dye

DNA fingerprint

the electrophoretic patter or restriction pattern is a DNA fingerprint of fragments separated by size

Pulsed field gel Electrophoresis (PFGE)

Separates large DNA (200-300kb)


  • Electric field changes repeatedly in direction and strenght
  • The electrical pulses cause the gel to become more fluid changing DNA conformation; the DNA "slithers" through the gel matrix

2-D electrophoresis

  • Different electrophoretic technique in each dimension
  • Proteins separated by different properties

2-D electrophoresis First dimension

Proteins are separated by charge in IEF

2-D electrophoresis second dimension

Proteins are separated by size in SDS-PAGE

2-D electrophoresis is used for

Proteome analysis;


Proteom complements expresses by a genom

Steps of 2-D electrophoresis

  1. Sample preparation
  2. First dimension isoelectric focusing
  3. Second dimension gel electrophoresis
  4. Staining
  5. Imaging analysis

Immunoelectrophoresis (IEP)

Two step process:


  • First step separates protein on agarose gel electrophoresis
  • Second step: proteins interact with specific antibodies
  • examine the proteins' antigenic properties

IEP- antibody interaction

Antibody is added to a trough cut in the gel;


diffuses through the gel to separated protein



If there is a reaction a visible insoluble precipitin is formed

IEP uses

Assesses protein:


  • Purity
  • Composition
  • Antigenic properties

Immunofixation electorphoresis (IFE) involves

protein electrophoresis followed by an immunoprecipitation



Wash stain and destain to visualize the protein:antibody complex

Ampholytes

synthesized polyamine carboxylic acids, multi-branched chained and deprotonated carboxylic acid groups

Properties of Carrier Ampholytes

  • Maintain pH due to have a good buffering capacity
  • Conduct electric current due to zwitterionic
  • Net zero mobility at their pI's
  • Soluble in water
  • Do not interfere with the sample or its detection

Use of Carrier ampholytes

  • Mixing a series of carrier ampholytes
  • Apply them to a gel matrix ( acrylamide), pH gradient is generated when a electrical current is applied to the gel

pH range of IEF is determined by

the ampholyte mixture

Amino acids are amphoteric because

the carry multiple charges:


acidic pH: positive charge


alkaline pH: negative charge



the net charge on a protein changes as the pH changes

What is IFE

electrophoresis in a pH gradient, separation base on pI

Isoelectric focusing

as a protein approach their pI they gradually become less charged

IEF set up

A solution of ampholytes is mixed with the gel prior to polymerization



When an electrical field is applied the ampholytes move their pI to create a constant pH gradient (creates a buffer zone)

Ampholytes movement

the move ahead faster than the proteins because they are smaller est the pH gradient ahead of the protein

IEF protein migration

As the protein migrates through the different pH zones, its net charge decreases



Once the protein has no charge it cannot move



Stops when it enters the pH zone that equals its isoelectric point

IEF


pH<pI

protein carries a net positive charge and migrates towards the cathode

IEF


pH>pI

proteins carry a net negative charge and migrate towards the anode

IFE


pH=pI

protein carries a net zero charge does not migrate

Protein bands in IEF

Bands are very sharp due to the small pH ranges



  • Relatively independent of electrophoresis duration
  • Balance between focusing and diffusion
  • Resolution will be within 0.2 pH units

IEF protein visualization

ampholytes must be removed to be stained:


Soak in 5% TCA solution

Added benefit of IEF

concentrates proteins because they are focused in a specific pH zone

IEF uses

  • Estimate protein purity
  • Determine protein identity by comparing migration with that of a known protein standard

Capillary Electrophoresis

Conventional Electrophoresis:


on gel or paper, minimal automation, long anaylsis, detection performed post separation, low voltages



Performed in capillary tubes, hi voltage can be used due to heat diffusion of the tubes

Advantages of CE

  • Automation
  • High voltage
  • rapid real time separation
  • in-line detection
  • high separation efficiency
  • Small sample size

CE uses

  • Narrow bore fused silicia capillaries filled with buffers
  • Sample loaded by replacing the buffer at the anode end
  • electro-osmotic flow; bulk of liquid to cathode

Applications of CE

  • Pharmaceuticals
  • DNA
  • Proteins, peptides
  • Clinical and forensic specimens
  • Agrochemicals
  • Natural products
  • Fine chemicals
  • biochemicals