You are on page 1of 64

Â

ultrafiltration
handbook
Tools and Techniques for
Life Science Research
Introduction
This handbook is written for research scientists who are using, or who are interested in using,
ultrafiltration (UF) technology in their work.

Our goal is to provide researchers with a handy reference piece that will allow them to optimize
their UF separations and provide them with purified and/or concentrated sample that is ready for
downstream experimentation.

The intention is not to make ultrafiltration experts out of research scientists—in fact, one of the
appealing benefits of the technology is its simplicity. However, having a basic understanding of
how ultrafiltration works, and how it can be applied to everyday research, enables the researcher
to select the right membrane or device for their particular experiment.

The information contained in this handbook is culled from decades of collective experience of
Millipore and Amicon applications scientists and technical service employees, as well as the
company’s library of technical publications on ultrafiltration, UF devices, and UF applications.
Millipore customers have also contributed data, helping to make this handbook a complete
resource for ultrafiltration.

Ultrafiltration has evolved significantly since it was first introduced in the 1970s, and it will
continue to evolve as life science research progresses. New device platforms and
new applications work have expanded the use of UF into new areas, such as genomics,
proteomics, drug compound screening, and small molecule assays.

To keep up with the latest developments, visit our web site: www.millipore.com.

The Protein Research Team


Millipore Corporation
Life Sciences Division
Table of Contents
Overview of Membrane Filtration Purification of Serum Peptides
for Biomarker Research with
M em b r a n e P rocesses . . . . . . . . . . . . . 2 Amicon Ultra Centrifugal Filters . . . . . .24
Ultrafiltration . . . . . . . . . . . . . . . . . . . . . .2 Rapid Antibody Concentration with
Microfiltration . . . . . . . . . . . . . . . . . . . . .2 Amicon Ultra Centrifugal Filters . . . . . .27
Reverse Osmosis . . . . . . . . . . . . . . . . . . .4 Removal of Unincorporated Label from
Labeled Protein with Amicon Ultra
R e c o ver y . . . . . . . . . . . . . . . . . . . . . . . . 4
or Centricon Centrifugal Filters . . . . . .28
Nominal Molecular Weight Limit . . . . . . .4
Urine Concentration with
Nucleotide Cut-off . . . . . . . . . . . . . . . . . .4 Amicon Ultra Centrifugal Filters . . . . . .30
Retention . . . . . . . . . . . . . . . . . . . . . . . .5 Affinity Purification with Ultrafree-MC
Concentration Polarization . . . . . . . . . . . .5 Centrifugal Filters . . . . . . . . . . . . . . . .32
Flux . . . . . . . . . . . . . . . . . . . . . . . . . . . .6 Use of Centrifugal Filter Devices
Effects of Operating Parameters on Flux . . .6 as an Alternative to Stirred Cells . . . . .35
Importance of Recovery . . . . . . . . . . . . . .6 Nucleic Acids . . . . . . . . . . . . . . . . . . . 36

Mode of Operation . . . . . . . . . . . . . . 7 Concentrating and Desalting DNA or


RNA with Microcon or Centricon
Normal vs. Tangential Flow Filtration . . . . .7
Centrifugal Filters . . . . . . . . . . . . . . . .36
Diafiltration . . . . . . . . . . . . . . . . . . . . . 7 Preparing Samples for Forensics
Dialysis vs. Diafiltration . . . . . . . . . . . . . .8 Identification Analysis with
Microcon Centrifugal Filters . . . . . . . . .39
Guide to Selecting Membranes PCR Purification with Montage PCR
and Devices Filter Units . . . . . . . . . . . . . . . . . . . . .40

Membrane Selection . . . . . . . . . . . . . 9 Preparation of Fluorescent DNA Probe


from Human mRNA or Total RNA
Types of Membranes . . . . . . . . . . . . . . . .9
with Microcon Centrifugal Filters . . . . .42
Membranes Organized by
Purification of In Vitro Synthesized mRNA
Typical Recoveries . . . . . . . . . . . . . . . .9
with Microcon or Centricon
Membranes Organized by Application . .10 Centrifugal Filters . . . . . . . . . . . . . . . .44
Device Selection . . . . . . . . . . . . . . . . 10 Quantitative Recoveries of Nanogram
Small Volume Devices . . . . . . . . . . . . . .12 Amounts of Nucleic Acids with
Microcon Centrifugal Filters . . . . . . . .46
Medium Volume Devices . . . . . . . . . . . .13
Effect of Centrifugal Ultrafiltration
Large Volume and TFF Devices . . . . . . . .14
on Large Fragment DNA Integrity . . . . .48
Stirred Cells . . . . . . . . . . . . . . . . . . . . .14
DNA Extraction from Agarose Gels
Specialty Devices and Kits . . . . . . . . . . .15 with Montage Gel Extraction Kit or
Ultrafree-DA Centrifugal Filters . . . . . .50
Protocols RNA Purification with Microcon or
P ro tein s . . . . . . . . . . . . . . . . . . . . . . . . 16 Centricon Centrifugal Filters . . . . . . . .52
Concentration, Desalting, and Enzyme Removal with Micropure-EZ
Buffer Exchange with Amicon Ultra Centrifugal Filters . . . . . . . . . . . . . . . .55
or Microcon Centrifugal Filters . . . . . . .16 H igh Th rou gh pu t Applicat ion s . . . . 58
Detergent Removal with Microcon, Using MultiScreen Filter Plates with
Amicon Ultra or Centricon Ultracel-10 Ultrafiltration Membrane . .58
Centrifugal Filters . . . . . . . . . . . . . . . .20
Two-Dimensional Electrophoresis Appendix
Sample Preparation with
Glossary . . . . . . . . . . . . . . . . . . . . . . .59
Amicon Ultra Centrifugal Filters . . . . . .22

1
Overview of Membrane Filtration

Membrane Processes Ultrafiltration is typically used to separate


proteins from buffer components for buffer
Ultrafiltration
exchange, desalting, or concentration. Ultrafilters
Ultrafiltration (UF) is the process of separating
are also ideal for removal or exchange of sugars,
extremely small particles and dissolved molecules
non-aqueous solvents, the separation of free from
from fluids. The primary basis for separation is
protein-bound ligands, the removal of materials of
molecular size, although in all filtration applications,
low molecular weight, or the rapid change of ionic
the permeability of a filter medium can be affected
and/or pH environment (see Figure 1.6, page 8).
by the chemical, molecular or electrostatic proper-
Depending on the protein to be retained, the most
ties of the sample. Ultrafiltration can only separate
frequently used membranes have a nominal molecular
molecules which differ by at least an order of
weight limit (NMWL) of 3 kDa to 100 kDa.
magnitude in size. Molecules of similar size can
Ultrafiltration is far gentler to solutes than
not be separated by ultrafiltration (see Figure 1.3).
processes such as precipitation. UF is more efficient
Materials ranging in size from 1K to 1000K
because it can simultaneously concentrate and
molecular weight (MW) are retained by certain
desalt solutes. It does not require a phase change,
ultrafiltration membranes, while salts and water will
which often denatures labile species, and UF can
pass through. Colloidal and particulate matter can
be performed either at room temperature or in a
also be retained. Ultrafiltration membranes can be
cold room.
used both to purify material passing through the
filter and also to collect material retained by the Microfiltration
filter. Materials significantly smaller than the pore Microfiltration (MF) is the process of removing
size rating pass through the filter and can be particles or biological entities in the 0.025 µm to
depyrogenated, clarified and separated from 10.0 µm range from fluids by passage through a
high molecular weight contaminants. Materials microporous medium such as a membrane filter.
larger than the pore size rating are retained by the Although micron-sized particles can be removed by
filter and can be concentrated or separated from use of non-membrane or depth materials such as
low molecular weight contaminants.

Reverse Osmosis Ultrafiltration Microfiltration Clarification


0.0001 µm 0.001 µm 0.01 µm 0.1 µm 1 µm 10 µm 100 µm

0.2 kDa 200 kDa 20,000 kDa

Red Blood Smallest


Sugars Proteins B. diminuta
Cell Visible Particle

Amino Acids
Carbon Black Yeast Pollens
Nucleotides

Oligo- Polio
Salts Mycoplasma E. coli Clouds Fog
nucleotides Virus

Human
Antibiotics Mammalian Virus Bacteria
Hair

Figure 1.1 Comparison of ultrafiltration with other commonly used membrane separation techniques.

2
those found in fibrous media, only a membrane filter The retention boundary defined by a membrane
having a precisely defined pore size can ensure filter can also be used as an analytical tool to
quantitative retention. Membrane filters can be used validate the integrity and efficiency of a system.
for final filtration or prefiltration, whereas a depth For example, in addition to clarifying or sterilizing
filter is generally used in clarifying applications filtration, fluids containing bacteria can be filtered
where quantitative retention is not required or as a to trap the microorganisms on the membrane surface
prefilter to prolong the life of a downstream for subsequent culture and analysis. Microfiltration
membrane. Membrane and depth filters offer certain can also be used in sample preparation to remove
advantages and limitations. They can complement intact cells and some cell debris from the lysate.
each other when used together in a microfiltration Membrane pore size cut-offs used for this type of
process system or fabricated device. separation are typically in the range of 0.05 µm
to 1.0 µm.

Figure 1.2 Ultrafiltration membranes vs. traditional microporous membranes


Ultrafiltration membranes generally Microporous membranes are
have two distinct layers: a thin generally rigid, continuous meshes
(0.1– 1.5 µm), dense skin with a of polymeric material with defined
pore diameter of 10 – 400 Å and pore sizes. They are used to retain
a more porous substructure. Any bacteria, colloids and particulates.
species capable of passing through Species are either retained on the
the pores of the skin (whose size is membrane surface or trapped
precisely controlled in manufacture) in its substructure.
can therefore freely pass the
Cross-section of ultrafiltration membrane. Cross-section of traditional micro-
membrane with skin and porous porous membrane with uniform
substructure. pore structure from top to bottom.

Figure 1.3 Fractionating samples using ultrafiltration membranes


A popular misconception is that one can pass a mixture of macromole- properties than the traditional evaluation with two or three protein
cules through an ultrafiltration membrane and separate all molecules species. The mixed dextran protocol uses a total of 11 dextran
whose molecular weights are larger than the membrane’s cut-off from standards of known molecular weight (180 to 1750K daltons) and the
those whose molecular weights are smaller than the cut-off. relative retention/passage of the mixture is determined after filtration
This expectation is based on a misunderstanding of the principles by analyzing the retentate and filtrate fractions using gel permeation
of membrane filtration. Most scientists believe that molecules smaller chromatography.
than the membrane rating will pass freely through to the filtrate while An examination of the curve for the Ultracel PL 300K NMWL
molecules larger than the membrane rating will be quantitatively membrane confirms that molecules larger that the 100K NMWL rating
retained. are retained at a level greater than 90%. However, molecules smaller
In fact, membrane ratings are based on retention and not on than the membrane rating do not exhibit free passage to the filtrate;
passage properties. The relative retention/passage properties for for example, a 75K kDa species is retained at a 83% level and a
a 100K NMWL rated membrane are illustrated below. These data 30K kDa species is 32% retained on this membrane.
were generated using Millipore’s mixed dextran rejection procedure. These smaller molecules are being retained by a membrane with
This protocol provides a more detailed look at membrane retention a higher retention rating because the retention/passage phenomenon
is a function of both the membrane and the solution being processed.
1.0
Buffer composition as well as solute concentration can have major
0.9
impacts on the retention/passage of molecules. For example, dilute
Rejection Coefficient

solutions of single solutes less than the NMWL ratings are more likely
Ultracel PL 300K
0.8
NMWL membrane
to appear in the filtrate than the same molecule in a more concen-
trated solution or in a complex mixture.
0.7
However, it may be possible to affect this type of separation
0.6
between two macromolecules if there is at least 10X difference in their
molecular weight and if there is an ultrafiltration membrane available
0.5 with a NMWL rating between the molecular weights of the two
10,000 100,000 1,000,000
proteins. These separations are also generally more successful if the
Molecular Weight (kDa)
protein solution is dilute and the separation is run at low pressure.
Mixed Dextran Rejection Profile

3
Reverse Osmosis teristics; sample concentration and composition;
Reverse osmosis (RO) separates salts and small operating conditions; and device or system
molecules from low molecular weight solutes configuration. Two membranes may have the same
(typically less than 100 daltons) at relatively NMWL but will exhibit different retention of
high pressures using membranes with NMWLs of molecules within a relatively narrow range of sizes.
1 kDa or lower. RO membranes are normally rated In addition, slender, linear molecules (e.g., nucleic
by their retention of sodium chloride while ultrafiltra- acids) may find their way through pores that will
tion membranes are characterized according to the retain a globular species of the same weight.
molecular weight of retained solutes. Millipore Retention can also be affected by hydration with
water purification systems employ both reverse counter-ions. Nevertheless, NMWL has proven to
osmosis membranes as well as ultrafiltration be an effective general indicator of membrane
membranes. Reverse osmosis systems are primarily performance for globular proteins.
used to purify tap water to purities that exceed When using membrane ultrafiltration for sample
distilled water quality. Ultrafiltration systems ensure concentration or desalting, care must be taken to
that ultrapure water is free from endotoxins as well select a membrane (or device) with a NMWL
as nucleases for critical biological research. appropriate for the application. Because there are
several considerations in determining whether a
Recovery given solute will or will not be retained by a
membrane of a specific cut-off, it is best to choose
The ultimate aim of ultrafiltration is to maximize
a device with cut-off at about one half of the
recovery of solutes of interest, but there are many
molecular weight of the protein to be concentrated.
membrane characteristics that affect that goal.
This maximizes protein recovery and minimizes
Factors affecting recovery include:
filtration time.
• Nominal molecular weight limit (NMWL)/
nucleotide cut-off (NCO) Nucleotide Cut-off (NCO)
• Retention
For most membranes, the NMWL is determined
• Concentration polarization
experimentally under a standard set of operating
• Flux
conditions. These analyses typically employ purified
Nominal Molecular Weight Limit globular proteins to serve as markers or indicators of
the retention characteristics of an ultrafiltration
A microfiltration membrane’s pore size rating,
membrane. Although this approach is useful for
typically given as a micron value, indicates that
choosing the appropriate NMWL for most protein
particles larger than the rating will be retained.
research applications, selection of a membrane with
Ultrafiltration membranes are rated according to
an appropriate NMWL membrane for nucleic acid
the nominal molecular weight limit (NMWL), also
or polysaccharide purification is considerably more
sometimes referred to as molecular weight cut-off
complex. By virtue of the rod-like three-dimensional
(MWCO). The NMWL indicates that most dissolved
structures of these molecules, these types of
macromolecules with molecular weights higher than
molecules require a tighter membrane (with a
the NMWL will be retained.
smaller cut-off) than do globular proteins of the
An ultrafiltration membrane with a stated NMWL
same molecular weight. It is therefore convenient
should retain (reject) at least 90% of a globular
to consider the membrane retention characteristics
solute of that molecular weight in daltons. However,
of nucleic acids as being related to their length
for a wider safety margin, the selected cut-off
(in nucleotides) rather than their molecular weight.
should be well below the molecular weight of the
Complicating matters even further are several
solute to be retained. When solutes are to be
additional factors that affect the recovery of nucleic
exchanged, the cut-off should be substantially
acid fragments from a membrane of a given
above that of the passing solute. A lower NMWL
NMWL. These factors include: the strandedness
increases rejection but decreases the filtration rate
of the DNA or RNA molecule; whether the DNA is
for the same membrane material.
linear, relaxed or supercoiled (for plasmid); the ionic
Retention and product recovery are a function
strength of the solvent; the velocity of the process
of a variety of other factors, including the molecular
stream over the membrane; and the nature of the
shape and size of the molecule; electrical charac-
driving force. The overall effect is that optimal

4
nucleic acid recovery is achieved in low salt buffers Millipore recommends the selection of a
run under conditions of relatively low velocity (e.g., membrane filter NMWL that is one half the size
low vacuum pressure or low g-force). The membrane of the molecule of interest. Other manufacturers
and protocol developed for the Montage® PCR may recommend a smaller differential between the
centrifugal filter device takes into account these size of the NMWL and the size of the molecule
conditions in order to provide for high recovery of but Millipore’s recommendation is designed to
small PCR products (e.g., ~150 base pairs [bp]) as provide maximum recovery. Please see additional
quickly as possible. For purifications that are driven information regarding membrane NMWL selection
by vacuum, significantly tighter membranes are on page 4.
typically required to obtain optimal recovery.
If the DNA sample is in the presence of high salt Concentration Polarization
(or the device is run at a higher-than-recommended Another factor affecting the retention characteristics
g-force), a significantly reduced DNA recovery may is the potential for membrane fouling, or concentra-
be observed. Under these conditions, higher DNA tion polarization. This occurs when there is an
or RNA recovery can be achieved by using a tighter accumulation of the retained solute on the surface
membrane. However, it will take significantly longer of the membrane. At high concentrations, a gel
to complete the purification. For applications such layer forms that can act as a secondary membrane
as PCR where removal of unincorporated single- (Figure 1.4). This may interfere with passage of the
stranded primers from double-stranded DNA molecules through the membrane and can adversely
fragments is required, the molecular weights of affect the flow rate. In addition, pH, buffer compo-
the primer and DNA fragment should differ by at nents, and concentration can result in a protein
least an order of magnitude for efficient separation. behaving in an anomalous manner in terms of its
Millipore offers devices that are specifically retention or passage by UF membranes.
designed for separating and concentrating genomic During concentration polarization, the gel layer
DNA and PCR products by ultrafiltration. on the membrane surface superimposes its own
rejection characteristics on those of the membrane.
Retention Usually, concentration polarization increases reten-
Retention, also sometimes called rejection, is a tion of lower-molecular weight species. A membrane
function of molecular size and shape. Nominal cut- with a 100K NMWL may reject 10 –20% of
off levels, defined with model solutes, are conven- albumin in a 0.1% solution of pure albumin.
ient indicators. Degree of hydration, counter ions, However, in the presence of larger solutes such as
and steric effects can cause molecules with similar IgG, it may reject 90% of the albumin. Concentration
molecular weights to exhibit very different retention polarization makes it very difficult to use UF for
behavior. Many biological macromolecules tend to solute fractionation unless the solutes to be separated
aggregate, or change conformation under varying differ in size by at least an order of magnitude.
conditions of pH and ionic strength, so that effective
size may be much larger than the “native” molecule,
causing increased rejection. Solute/solvent and
solute/solute interactions in the sample can also
change effective molecular size. For example, some
proteins will polymerize under certain concentration
and buffer conditions while others (e.g., heme
proteins) may break into corresponding subunits.
Ionic interactions or π – π stacking can cause small
molecules to behave similarly to molecules of
greater molecular weight. When this occurs, as in
the case of phosphate ions with a 500 NMWL
membrane, the small molecules may not effectively
permeate the membrane. Figure 1.4 Ultrafiltration separates proteins
from soluble salts. “Concentration polarization”
slows down filtration. The proteins form a gel layer
on the membrane surface.

5
Flux (UF Flow Rate) Many antifoams exhibit a phenomenon called
During ultrafiltration, it is important to balance speed “cloud point.” As temperatures increase, antifoam
with retention to obtain optimal performance. A comes out of solution, forming a second phase.
membrane’s flux is defined as the flow rate divided Increasing temperature above the cloud point
by the membrane area. Using membranes with causes flux to decrease.
higher NMWL ratings will increase the flow, but at pH
the same time lower the retention. A membrane Changing solution pH often changes molecular
should be selected for required rejection, consistent structure. This is especially true for proteins. At its
with desired flow rate. This is determined by surface isoelectric point, a protein begins to precipitate,
area, macrosolute type, solubility, concentration and causing a flux decrease.
diffusivity, membrane type, temperature effects on
viscosity and, to some extent, pressure. When Fouling
concentration polarization is rate-controlling, flux is Flux decrease due to concentration polarization
affected by solute concentration, fluid velocity, flow should not be confused with the effect of membrane
channel dimensions, and temperature. fouling. Fouling is usually the deposition and
accumulation of submicron particles and solute on
Effects of Operating Parameters on Flux the membrane surface and/or crystallization and
Pressure precipitation of smaller solutes on or within the pores
When ultrafiltering dilute protein solutions or colloid of the membrane. There may be a chemical
suspensions, flux will increase with increasing interaction with the membrane.
transmembrane pressure (TMP). These effects are
Importance of Recovery
most apparent when operating under controlled
positive pressure, such as when using a stirred cell. While rejection is used to characterize membrane
When the process is membrane-controlled (i.e., performance, it does not always directly correlate
when the resistance of the gel layer is much smaller with solute recovery from a sample or volume.
than that of the membrane), the flux-pressure Actual solute recovery—the amount of material
relationship is linear. When the process is controlled recovered after ultrafiltration—is generally based
by polarization (e.g., when the resistance of the on mass balance calculations.
gel layer is much larger than that of the membrane), In many cases, especially when working with
flux will reach a plateau and may actually decrease small samples of dilute, valuable solutions, the
with increases in pressure. degree of recovery of a target solute is vitally
important. In such cases, potential loss by non-
Concentration specific adsorption must be considered.
When concentration of the retained species is very Different membrane materials adsorb biomole-
low, flux is independent of concentration. As solute cules to varying degrees. Where maximum recovery
concentration rises during operation, increased is desired, the choice of a membrane with the least
viscosity and the polarization effect cause flux to non-specific adsorptivity is essential. Millipore’s
decrease. Ultracel® regenerated cellulose membranes were
Temperature specifically developed to minimize non-specific
adsorption.
Increasing the operating temperature normally
Since adsorption is a direct function of mem-
increases UF rates. A higher temperature increases
brane and device surface area, device size must be
solute diffusivity (typically 3 –3.5% per degree
considered when recovery is important. Small, dilute
Celsius for proteins) and decreases solution
samples should be concentrated with membranes of
viscosity. Common practice is to operate at the
minimal surface area, commensurate with achieve-
highest temperature tolerated by the solutes and
ment of reasonable flow rates. Millipore offers a
the equipment.
wide range of centrifugal devices, stirred cells, and
An exception to the rule is fermentation broth
tangential flow systems with an extensive choice of
concentration in the presence of some antifoams.
membrane areas and NMWLs.

6
Mode of Operation molecules are at least 10 times smaller than the
molecules to be retained and concentrated by the
The pressure required for ultrafiltration can be
membrane. Diafiltration is useful for sample
supplied in a number of different ways depending
desalting and buffer exchange.
on the product in use. For example, Millipore's
When diafiltration is used for sample desalting
small volume ultrafiltration products generally use
or buffer exchange, there is no resulting change in
centrifugal force. Pump pressure is used with the
buffer composition. A solution volume with 100 mM
tangential-flow filtration (TFF) products and com-
salt still contains 100 mM salt after the initial
pressed gas is utilized with the stirred cell products.
concentration spin. Rediluting the retentate with
In addition, Millipore provides multiwell ultrafiltration
water and spinning again effectively decreases
products that utilize vacuum and centrifugation.
the salt concentration of the sample by the con-
Normal vs. Tangential Flow Filtration centration factor of the ultrafiltration. For example,
Filtration can be broken down into two different if a 4,000 µL sample containing 100 mM salt is
operational modes: normal flow filtration (NFF) concentrated to 50 µL (80X) in an Amicon® Ultra
and tangential flow filtration (TFF). The difference centrifugal filter unit, rediluted with water to
in fluid flow between these two modes is shown 4,000 µL, and reconcentrated, the salt concen-
in Figure 1.5. tration will be reduced 80X to 1.25 mM.
To achieve more complete salt removal, multiple
Diafiltration concentration and redilution spins are required.
For most samples, two concentration/reconstitution
Millipore membranes provide an inexpensive means
cycles will remove about 99% of the initial salt
of separating macromolecular mixtures into size-
content.
graded classes either by direct ultrafiltration or by
With very small sample volumes, dilution of the
diafiltration. Diafiltration removes microsolutes by
sample before the initial concentration spin can
adding solvent to the solution being ultrafiltered
often decrease salt concentration to an acceptable
at a rate equal to the UF rate, independent of
level. For example, if a 200 µL sample containing
microspecies concentration. This rapid, efficient
100 mM salt is diluted to 4,000 µL before concen-
process washes microspecies from the solution at
tration in an Amicon Ultra centrifugal filter unit,
constant volume, thereby purifying the retained
the salt concentration in the 4,000 µL sample will
species. This process is most effective if the passing
be 5 mM. The concentrate will still contain 5 mM

Figure 1.5 Normal flow filtration (NFF) vs. tangential flow filtration (TFF)

In normal flow filtration (NFF), fluid is convected directly toward the often called dead-end filtration. However, the term “normal” indicates
membrane under an applied pressure. Particulates that are too large that the fluid flow occurs in the direction normal to the membrane
to pass through the pores of the membrane accumulate at the surface, so NFF is a more descriptive and preferred name. NFF can
membrane surface or in the depth of the filtration media, while smaller be used for sterile filtration of clean streams, clarifying prefiltration,
molecules pass through to the downstream side. This type of process is and virus/protein separations.
In tangential flow filtration (TFF), the fluid is pumped tangentially
along the surface of the membrane. An applied pressure serves to
Normal Flow Filtration Tangential Flow Filtration force a portion of the fluid through the membrane to the filtrate side.
Feed Flow Pressure Pressure As in NFF, particulates and macromolecules that are too large to pass
through the membrane pores are retained on the upstream side.
However, in this case the retained components do not build up at the
surface of the membrane. Instead, they are swept along by the
Feed Flow
tangential flow. This feature of TFF makes it an ideal process for finer
sized-based separations. Although TFF is more commonly associated
with large scale processing, centrifugal UF devices with vertical
membrane panels, such as Amicon Ultra devices, also benefit from a
Membrane Membrane TFF-like mode of separation, particularly in a swinging bucket rotor.
TFF is also commonly called cross-flow filtration. However, the term
Filtrate Filtrate “tangential” is descriptive of the direction of fluid flow relative to the
membrane, so it is the preferred name.

7
salt. If more complete salt removal is desired, a Dialysis vs. Diafiltration
re-dilution/spin cycle should be added. In this Dialysis is a traditional method for removing
example, if the original spin ended with 50 µL of microsolutes or exchanging solvents. It is a slow
retentate, redilution to 4,000 µL results in 0.06 mM diffusive process generally employing regenerated-
salt concentration. The sample can then be recon- cellulose tubing as the barrier membrane. In dialysis,
centrated to 50 µL in an Amicon Ultra centrifugal the process solution and exchange solvent are on
filter device. opposite sides of the semi-permeable barrier
Diafiltration can be a continuous or a discon- membrane through which permeating microsolutes
tinuous process. In continuous diafiltration, such diffuse. The permeation rate of solutes from sample
as in a stirred cell or a TFF device, the solution is to dialysate is a direct ratio to the solute concen-
maintained at a fixed volume while solvent flows tration and inversely proportional to the solutes’
continuously through the mixture. Salts and other molecular weight. Desalting by dialysis is time-
microsolutes are steadily removed by convective consuming and relatively inefficient at low
transport. Microsolute exchange can be accom- concentrations.
plished using the same principle. Constant operator Millipore’s Amicon centrifugal concentrators
attention is not required and the possibility of solute provide a fast, convenient, high-recovery alternative
denaturation by overconcentration is eliminated. to dialysis or precipitation without diluting samples.
In discontinuous diafiltration, such as in a centrifugal The relative merits of diafiltration and dialysis are
ultrafiltration device, salts and microsolutes are summarized in the Table 1.1.
removed by repeated concentration and dilution
(Figure 1.6).

100 µL Add 90 µL 100 µL


10 µL 10 µL
UF membrane Milli-Q® water

10X 10-fold 10X


concentration 90 µL dilution concentration 90 µL
of protein of protein

100 mM 100 mM 10 mM 10 mM
NaCl NaCl NaCl NaCl

Figure 1.6 Removing salts from retained solutes using diafiltration. The sodium chloride concentration is
reduced by dilution.

Table 1.1 Comparision of diafiltration and dialysis


Diafiltration Dialysis
Transport convective with solvent, Transport diffusion-controlled,
independent of microsolute composition. dependent on type of microsolute.
Rapid rate. Fractional removal Slow transport. Lower efficiency with
independent of content. decreased microsolute concentration.
Ultrafiltration rate reduced with decreased Marked temperature dependence
temperature (net effect not as marked). (reduced transport at lower temperature).
At elevated macrosolute content, Microsolute transport relatively unaffected
ultrafiltration rate reduced. by macrosolute content.
Minimal exchange solvent required; Frequent dialysate change. Recirculation
easily contained in reservoir. about bags to maximize transport.
Simple automation with endpoint Automation possible with complex equipment.
control apparatus.

8
Guide to Selecting Membranes and Devices
Millipore offers a complete range of centrifugal and particulate materials. Durapore membranes are
devices used for sample concentration, purification, extremely hydrophilic, and they provide the lowest
and desalting or buffer exchange of soluble macro- binding of proteins and other biologicals of all
molecules. Millipore products are also available for commercially available microporous membranes.
applications such as cleaning up PCR reactions,
separating protein-bound from free ligands, Membranes Organized by
removing restriction enzymes, and recovering Typical Recoveries
oligonucleotides from agarose gels. This section For globular proteins, there is a good correlation
provides information to aid in choosing the correct between molecular weight and Stoke’s radius.
product for a particular application. This usually allows one to predict the recovery
of a protein based on its molecular weight if a
Membrane Selection membrane with the same nominal molecular weight
Millipore offers three distinct types of membranes to rating is used (see typical recovery of a panel of
choose from. This section will describe these three protein solutes in Table 2.1). However, in order to
types of membranes and then provide information accommodate the wide range of potential protein
about choosing the correct membrane based on solutes with different tertiary structures, we suggest
typical recoveries or application. initially using the “rule of two” to ensure optimal
recovery.
Types of Membranes
Rule of Two
Ultracel Ultrafiltration Membrane
For Ultracel (regenerated cellulosic) membranes,
To concentrate or desalt dilute solutions, use Millipore recommends using a membrane with
Ultracel series regenerated cellulose ultrafiltration a NMWL at least two times smaller than the
membranes. The hydrophilic, tight microstructure molecular weight of the protein solute that one
of Ultracel membranes assures the highest possible intends to concentrate.
retention with the lowest possible adsorption of
protein, DNA or other macromolecules. Rule of Three
For Biomax (polyethersulfone) membranes for
Biomax® Ultrafiltration Membrane
stirred cells and TFF, Millipore recommends using
To concentrate or desalt higher volumes of more
concentrated samples (recommended for protein
concentrations greater than 1.0 mg/mL), use Table 2.1 Membrane selection by recovery
Biomax polyethersulfone (PES) ultrafiltration mem- Cytochrome c BSA IgG
branes. Biomax membranes are recommended for NMWL (12.4 kDa) (67 kDa) (156 kDa)
samples such as serum, plasma, or conditioned 3K Q Q Q
tissue culture media.
10K P Q Q
Durapore® Microporous Membrane 30K • Q Q
To clarify biological samples, recover DNA from 50K • P Q
agarose gels, retain chromatography resins or 100K • • P
suspended solid media, use Durapore hydrophilic
PVDF microporous membranes. Durapore mem- Q Recommended (>90% recovery)
P Typically >90% recovery, depends on solute
branes allow all soluble protein and nucleic acids to
• Not recommended
pass, retaining sub-cellular fragments, whole cell

9
a membrane with a NMWL at least three times Device Selection
smaller than the molecular weight of the protein
See Table 2.3 to determine which centrifugal
solute that one intends to concentrate.
product to use for protein concentration based on
Membranes Organized by Application initial sample molecular weight and volume.

See Table 2.2 to determine which centrifugal


product to use based on application and membrane.

Table 2.2 Membrane selection by application Ultrafiltration Membranes Microporous Specialty


Membranes Devices
Type* Molecular Weight
(NMWL) Pore Size (µm)

Montage Gel Extraction


Montage PCR
Micropure-EZ
Ultracel
Biomax

100K

0.45
0.65
10K
30K
50K

0.1
0.2

5.0
3K
5K
Protein concentration • • • • • • • •
Protein purification/desalting/buffer exchange • • • • • • •
Desalting of column fractions • • • • • • •
Protein isolation from cell lysates • • • •
Peptide concentration/desalting/buffer exchange • • •
Antibody concentration • • • •
Virus concentration or removal • • • •
Nucleic acid concentration/desalting/buffer exchange • • • • • • •
Oligonucleotide concentration/desalting/buffer exchange • •
PCR cleanup • • • • •
Remove linkers prior to cloning • • •
Remove labeled nucleotides • • • •
Antibody purification from hybridoma cells • • • •
Rapid restriction mapping • •
Clarify samples of particulate prior to HPLC • •
Clarification of cell lysates and tissue homogenates • • • • •
Cell harvesting • • • • •
Natural product screening • • • • • • • •
Restriction enzyme removal •
Bound vs. free drugs from serum/plasma (protein removal) • • •
DNA/RNA recovery from polyacrylamide gel • •
DNA recovery from agarose gel •
Oligonucleotide recovery from polyacrylamide gel • •
Removal of unincorporated label (e.g., fluorescein)
from protein • • • • • • •
Removal of imidazole from His-tag fusion protein • • • • • • •
*Selection of ultrafiltration membrane: Ultracel regenerated cellulose membrane is ideal for protein samples and nucleic acids.
Biomax polysulfone membrane is ideal for complex samples (e.g., serum).

10
Table 2.3 Protein concentration devices by filtration capacity

Filtration Capacity
Millipore Device 0.5 mL 2 mL 4 mL 15 mL 20 mL 70 mL 1L 2L 10 L ≥ 10 L

Small Volume Filtration Devices

Microcon® Centrifugal Filters*


Ultrafree ® 0.5 Centrifugal Filters
Ultrafree-MC Centrifugal Filters
MultiScreen® Filter Plate with Ultracel Membrane**
Medium Volume Filtration Devices

Centricon ® Centrifugal Filters


Ultrafree-CL Centrifugal Filters
Amicon Ultra-4 Centrifugal Filters
Amicon Ultra-15 Centrifugal Filters
Centriprep® Centrifugal Filters
Centriplus ® Centrifugal Filters
Centricon Plus-20 Centrifugal Filters
Large Volume Filtration Devices

Centricon Plus-70 Centrifugal Filters


Amicon Stirred Cells

Series 8000, High Output, Solvent-Resistant Stirred Cells


Tangential Flow Filtration (TFF)

Pellicon® XL 50 Ultrafiltration Devices


Prep/Scale® Spiral Wound UF Modules
Pellicon-2 Ultrafiltration Modules
*Bold type indicates recommended devices.
**96-well plate. Volume per well.

11
Small Volume Devices
Ultrafiltration Devices MultiScreen Filter Plates with
Ultracel-10 Membrane
Microcon Centrifugal Filters Maximum initial volume: 500 µL per well
Maximum initial volume: 500 µL NMWL: 10K
Typical final volume: 5 µL MultiScreen Filter Plates with Ultracel-10
NMWLs: 3K, 10K, 30K, 50K, 100K Membrane are the first automation-
Microcon centrifugal filter devices, the lab compatible, high throughput ultrafiltration
standard, simply and efficiently concentrate plates for protein purification. The 10K
and desalt macromolecular solutions using NMWL Ultracel regenerated cellulose ultrafiltration membrane
any centrifuge that can accept 1.5 mL tubes. The device’s low- provides low non-specific binding and high protein recovery
adsorption components and its Ultracel-YM membrane, together (>95% retention of Cytochrome c).
with the patented invert recovery spin, combine to yield unusually
high recovery rates—typically > 95%, with concentration factors
as high as 100X. The invert spin also permits the recovery of Microfiltration Devices
low volumes.
Ultrafree-MC Centrifugal Filters
Ultrafree-0.5 Centrifugal Filters Maximum initial volume: 400 µL
Maximum initial volume: 500 µL Typical final volume: 5 µL
Typical final volume: 50 µL Pore sizes: 0.1 µm, 0.22 µm, 0.45 µm,
NMWLs: 5K, 10K, 30K, 50K, 100K 0.65 µm, 5.0 µm
Ultrafree-0.5 centrifugal filter devices Ultrafree-MC centrifugal filter units are
process aqueous biological solutions disposable devices used for sample
using any centrifuge that accommodates clarification with high recovery of biological solutions in the
2.2 mL centrifuge tubes. The device’s vertical Biomax PES mem- 0.05 to 0.5 mL range. Ultrafree-MC units are available in a
brane configuration, parallel to the direction of the centrifugal range of microporous and ultrafiltration membranes and are for
force, reduces concentration polarization. This allows for the single use only. They fit standard microcentrifuge, fixed-angle
highest possible flow rates, even in solutions with high levels of rotors capable of holding 1.5 mL or 20 mm outer diameter (O.D.)
particles. Concentrated protein is retrieved by pipette from a tubes. Ultrafree-MC centrifugal filter units provide fast filtration and
concentrate “pocket” located below the membrane surface. highly reproducible performance.

12
Medium Volume Devices
Ultrafiltration Devices Amicon Ultra-15 Centrifugal Filters*
Maximum initial volume: up to 15 mL
Amicon Ultra-4 Centrifugal Filters* Typical final volume: 200 µL
Maximum initial volume: 4 mL NMWLs: 5K, 10K, 30K, 50K, 100K
Typical final volume: 50 µL Amicon Ultra-15 centrifugal filters contain a
NMWLs: 5K, 10K, 30K, 50K, 100K high recovery Ultracel membrane and are
Amicon Ultra centrifugal filters are the premier used to concentrate biological samples, to
tool for protein concentration, desalting, and purify macromolecular components found in tissue culture extracts
removing macromolecules and precipitates. or cell lysates, and for desalting and buffer exchange. The devices
The devices combine Ultracel low-binding can concentrate and desalt 15 mL in as few as 10 minutes.
ultrafiltration membrane with a vertical housing for fast sample
processing and typical sample recoveries of >90%. For volumes Centricon Plus-20 Centrifugal Filters
up to 4 mL, the Amicon Ultra-4 device provides fast ultrafiltration Maximum initial volume: 20 mL
with the capability for high concentration factors. Typical final volume: 200 µL
NMWLs: 5K, 10K, 30K, 100K
Centricon Centrifugal Filters Centricon Plus-20 centrifugal filter units are
Maximum initial volume: 2 mL disposable, single-use devices designed for
Typical final volume: 25 µL rapid processing of aqueous biological
NMWLs: 3K, 10K, 30K, 50K, 100K solutions. The devices can concentrate most 20 mL solutions
Centricon centrifugal filter devices provide down to a minimum volume of 200 µL.
fast, efficient concentration and desalting of
macromolecular solutions by ultrafiltration
through low-adsorption, hydrophilic Microfiltration Devices
Ultracel-YM regenerated cellulose membranes. Designed for use
Ultrafree-CL Centrifugal Filters
in centrifuges with fixed-angle rotors, they can provide up to
Maximum initial volume: 2 mL
80-fold sample enrichment with minimal solute loss by adsorption.
Typical final volume: 10 µL
Centriprep Centrifugal Filters Pore sizes: 0.1 µm, 0.22 µm, 0.45 µm,
Maximum initial volume: 15 mL 0.65 µm, 5.0 µm
Typical final volume: 700 µL Ultrafree-CL centrifugal filter units are
NMWLs: 3K, 10K, 30K, 50K disposable, single-use devices used to process aqueous biological
Centriprep centrifugal filter devices are solutions. Ultrafree-CL units can be used in standard, variable-
disposable ultrafiltration devices used for speed centrifuges with fixed-angle rotors capable of holding
concentrating and desalting high solute 15 mL tubes. A complete device consists of a filter cup and a
biological samples. These complete, filtrate collection tube with cap. Ultrafree-CL units are available in
ready-to-use ultrafiltration devices are designed for operation in a range of microporous and ultrafiltration membranes.
most centrifuges that can accommodate 50 mL centrifuge tubes.
Centriprep devices are best suited for applications in which
concentration factors are so high that they might affect protein
precipitation or aggregation.

Centriplus Centrifugal Filters


Maximum initial volume: 15 mL
Typical final volume: 500 µL
NMWLs: 3K, 10K, 30K, 50K, 100K
The disposable Centriplus centrifugal filter
device is used for the concentration and
desalting of macromolecular solutions with up
to 100-fold sample enrichment with minimal
loss of solute. These devices are designed for use in most centrifuges
that can accommodate 50 mL centrifuge tubes, with swinging-
bucket or fixed-angle rotors. *Amicon Ultra-4 and 15 are the first centrifugal devices to combine
high speed and high recovery. They are highly recommended for
most protein concentration and desalting applications.

13
Large Volume and TFF Devices
Ultrafiltration Devices Prep/Scale Spiral Wound
Ultrafiltration Modules
Centricon Plus-70 Centrifugal Maximum initial volume: 2 L to 10 L
Filters NMWLs: 1K, 3K, 5K, 10K, 30K, 50K,
Maximum initial volume: 70 mL 100K, 300K
Typical final volume: 350 µL Prep/Scale modules are used in conjunc-
NMWLs: 5K, 10K, 30K, 100K tion with a peristaltic pump, flexible tubing
Centricon Plus-70 centrifugal filter units and feed solution in order to economically
concentrate most 70 mL solutions down to prepare laboratory scale samples up to small production volumes
350 µL in just 25 minutes, making them a convenient alternative by tangential flow filtration. Highly reliable and consistent,
to dialysis, lyophilization, precipitation techniques, or stirred cells. Prep/Scale modules offer cassette-like efficiencies with
Samples are typically concentrated in the 50X to 200X range, reproducible separations performance.
depending on the sample type and starting sample volume.
Pellicon 2 Modules
Volumes >10 L
Ta n g e n t i a l F l o w F i l t r a t i o n ( T F F )
NMWLs: 1K, 3K, 5K, 8K, 10K, 30K,
Pellicon XL Devices 50K, 100K, 300K, 500K, 1000K
Maximum initial volume: 2 L Pellicon 2 modules offer true linear
NMWLs: 5K, 8K, 10K, 30K, 50K, scalability from laboratory size cartridges
100K, 300K, 500K, 1000K to industrial cartridge assemblies for processing thousands of liters.
Pellicon XL devices are small volume Pellicon 2 modules facilitate predicatable and faster scale-up with
tangential flow filtration (TFF) devices less effort and are available with membranes to match virtually
offering performance, ease of use, and any separation challenge.
linear scalability for pharmaceutical and
biotechnology development applications.
Pellicon XL devices are available in a wide selection of membrane
types and cut-offs. An optional graduated 100 mL reservoir is
available for simple small-volume separations.

Stirred Cells
Maximum initial volume: 2,000 mL Series 8000 Stirred Cells
Typical final volume: 3 mL • Five different sizes handle volumes from 3 mL to 400 mL
NMWLs: 1K, 3K, 10K, 30K, 50K, 100K, 300K, 500K • High flow rates with solutions up to 10% macrosolute
Stirred Cells are ideal for concentration, diafiltration and buffer concentration
exchange of macromolecule solutions including proteins, enzymes,
antibodies and viruses. They are capable of rapid concentration High-Output Stirred Cells
or salt removal followed by concentration in the same unit. • For 150 mm disc filters
Millipore offers three types of stirred cells: • Large membrane area

Solvent-Resistant Stirred Cells


• For 47 and 76 mm disc filters
• Borosilicate glass cylinder and PTFE components
for broad compatibility

Series 8000, High-Output and Solvent-Resistant Stirred Cells

14
Specialty Devices and Kits
Montage DNA Gel Extraction Kit Micropure ® -EZ Centrifugal Filters
and Ultrafree-DA Centrifugal Filters Maximum initial volume: 250 µL
Designed to extract DNA fragments that are Holdup volume: < 5 µL
100 to 10,000 bp in size; 0.45 µm Durapore membrane NMWLs: 3K, 10K, 30K, 50K, 100K
The Montage DNA Gel Extraction Kit is a fast, effective solution Micropure-EZ centrifugal filters provide an
for fully functional DNA recovery from agarose gel slices. In one easy, rapid means of removing restriction
10-minute spin, the and other enzymes from double-stranded
agarose gel material (ds) DNA. In a single 30-second spin, enzymes from the reaction
containing the DNA mix are selectively adsorbed by the membrane and DNA is
of interest is frag- recovered in a vial—undiluted and enzyme-free. If concentration
mented and com- or desalting of the sample is required, a Microcon centrifugal
pressed to extrude filter device may be substituted for the filtrate vial, providing for
the DNA that is ready DNA purification, concentration and desalting in a single unit.
for sequencing or DNA recovery is not affected by the binding capacity of an
cloning. The Montage Gel Extraction kit contains all the necessary adsorption matrix. Micropure-EZ devices will deliver a minimum
consumables and the kit protocol is optimized for use with of 85% recovery of dsDNA; higher recoveries can be achieved
standard agarose gels (<1.25% concentration). Ultrafree-DA by adding a rinsing step.
centrifugal devices available separately.
Centrilutor Micro-Electroeluters
Montage PCR Centrifugal Filters Maximum initial volume: 2 mL
Maximum initial volume: 500 µL Typical final volume: 25 µL
Typical final volume: 100 µL NMWLs: 3K, 10K, 30K, 50K, 100K
NMWLs: 3K, 10K, 30K, 50K, 100K The Centrilutor Micro-Electroeluter
The Montage PCR filter unit is a convenient elutes proteins from gel slices and then
method for single-sample PCR purification. removes the buffer and concentrates the
The high-performance device purifies PCR sample using Centricon centrifugal filter devices. This fast, easy
products from salts and primers in a single way to recover 1– 25 µg of protein from gel slices requires
centrifugation step. Purified samples are no additional handling of the sample once the gel slices are
ready for downstream applications with no placed into the eluter.
additional purification steps.

15
Protocols • Proteins

Concentration, Desalting, and Buffer Exchange


with Amicon Ultra or Microcon Centrifugal Filters

Introduction Two examples below demonstrate the use of


Amicon Ultra devices for high protein and
Amicon centrifugal devices from Millipore are
enzymatic activity recovery.
ideal for removal or exchange of salts, sugars,
nucleotides, and non-aqueous solvents, as well as
Method
other materials of low molecular weight. They also
serve to separate free from bound species. 1. Select the device with the appropriate
Millipore centrifugal concentrators provide fast, NMWL and volume for the application.
convenient, high-recovery alternatives to dialysis 2. Add the sample to the reservoir of the centrifugal
and ethanol precipitation. Sample dilution, often device.
associated with spin columns, is not a problem. 3. If the sample is smaller than the maximum
Salt transfer across the membrane is efficient and volume, it can be diluted up to the maximum
independent of microsolute concentration or size. volume before the first centrifugation step.
Millipore’s Amicon Ultra-4 and -15 centrifugal This will help increase the salt removal.
filters are designed for high speed with high 4. Centrifuge at the specified g-force for the
recovery. The devices incorporate low-binding recommended amount of time.
Ultracel regenerated cellulose ultrafiltration mem- 5. Remove the initial filtrate from the filtrate tube
brane for sample concentration and purification of and set aside.
solutions containing dilute or purified protein solutes,
6. Add enough buffer or water to the device to
antigens, antibodies, enzymes, or microorganisms.
bring the sample volume up to 4 or 15 mL.
Their speed and excellent recovery
7. Centrifuge again.
make them ideal for desalting and buffer exchange
applications. One of the most common applications 8. Set aside the filtrate.
for Amicon Ultra devices is concentration and 9. Recover the concentrated, de-salted sample.
desalting of column fractions during protein NOTE: Both of the filtrates should be retained until
purification by various chromatography methods. the concentrated sample has been analyzed.

Table 3.1 Removal of sodium chloride and recovery of protein with Amicon Ultra-15 device
Cytochrome c Cytochrome c BSA BSA IgG
0.25 mg/mL 0.25 mg/mL 1 mg /mL 1 mg/mL 1 mg/mL
NMWL 5 kDa 10 kDa 30 kDa 50 kDa 100 kDa
Spin % Protein % NaCl % Protein % NaCl % Protein % NaCl % Protein %NaCl % Protein % NaCl
Recovery Removal Recovery Removal Recovery Removal Recovery Removal Recovery Removal
1 94.5 97.2 97.3 97.9 96.0 98.2 98.9 97.1 99.9 97.7
2 92.6 99.9 95.6 99.9 94.4 99.9 92.4 99.9 97.1 99.5

Three Amicon Ultra-15 devices of each cut-off were tested with 15 mL of solute. 500 mM NaCl was added to each solution. Each spin was
performed at 4000 x g for 30 minutes. After the first spin, the retentate was brought up to 15 mL with ultrapure water from a Milli-Q® (Millipore)
system. OD readings were taken at 410 nm for Cytochrome c and 280 nm for BSA and IgG.

16
Results As the results show in Tables 3.1– 3.4 , the
efficient design of the Millipore devices allowed
The transfer of salts across a membrane filter is
> 90% of the salt to be removed during the first
independent of sample concentration or size. There
centrifugation step. Typically, only one subsequent
is no change in the composition of the buffer when
centrifugation step was needed to increase the
desalting using ultrafiltration. For example, a solution
typical salt removal to 99% with > 90% recovery of
containing 500 mM salt still contains that concen-
the sample.
tration after the initial centrifugation. Adding another
Protein purification by chromatography usually
volume of salt-free buffer or water to the retentate
involves the collection of multiple column fractions,
and centrifuging again will reduce the salt concen-
with only some of those fractions containing the
tration. This process, known as diafiltration, can be
protein of interest. After the fractions are combined,
repeated to achieve maximum salt removal.
a protein concentration step is often required for
Diafiltration can also be used when it is desirable to
protein storage, or concentration with buffer
have the sample in a different buffer. The sample is
exchange may be needed for downstream
concentrated and then repeatedly diluted with the
separations.
desired buffer and concentrated again.

Table 3.2 Removal of sodium chloride and recovery of protein with Amicon Ultra-4 device
Cytochrome c Cytochrome c BSA BSA IgG
0.25 mg/mL 0.25 mg/mL 1 mg /mL 1 mg/mL 1 mg/mL
NMWL 5 kDa 10 kDa 30 kDa 50 kDa 100 kDa
Spin % Protein % NaCl % Protein % NaCl % Protein % NaCl % Protein %NaCl % Protein % NaCl
Recovery Removal Recovery Removal Recovery Removal Recovery Removal Recovery Removal
1 100.0 95.2 97.0 93.0 96.4 96.4 98.5 92.1 100.0 99.2
2 95.8 97.9 95.6 99.5 93.7 99.7 99.3 99.7 99.4 99.7

Three Amicon Ultra-4 devices of each cut-off were tested with 4 mL of solute. 500 mM NaCl was added to each solution. Each spin was performed
at 4000 x g for 10 minutes. After the first spin, the retentate was brought up to 4 mL with ultrapure water from a Milli-Q (Millipore) system. OD
readings were taken at 480 nm for Cytochrome c and 280 nm for BSA and IgG.

Table 3.3 Removal of sodium chloride and recovery of IgG with Centricon Plus-20 filter device
with Ultracel-PL 30 membrane
Spin % NaCl % IgG
Number Remaining Recovered
1 1 —
2 0.4 89.8

19 mL of 1 mg/mL bovine IgG in 500 mM NaCl was concentrated to 150 µL. Sample was spun twice at
2000 x g in a swinging bucket rotor at 25 °C for 20 minutes.

Table 3.4 Removal of riboflavin and recovery of IgG with Microcon filter device with Ultracel-YM
membrane
Spin % Riboflavin % IgG
Number Remaining Recovered
1 9 95
2 2 93
3 <1 93
4 <1 92

500 µL of a 50:50 mixture of riboflavin and IgG were spun in a Microcon 3K NMWL device at 12,000 x g for 75
minutes at room temperature in 55° angle rotor. After the initial spin, the retentate was twice diluted with 500 µL of
PBS and spun again. After each spin, concentration of riboflavin and IgG in the filtrate and retentate were monitored.

17
Concentration of Indoleamine concentrate the IDO fractions from an initial
concentration of ~ 0.5 mg/mL to a final concentra-
2,3-Dioxygenase
tion of 10 mg/mL. Samples were analyzed by
Courtesy of Eduardo Vottero,
SDS-PAGE using a 12.5% polyacrylamide gel
University of British Columbia
(Figure 3.1). In addition, it was shown that no IDO
Indoleamine 2,3-dioxygenase (IDO; MW 48,000) activity loss was observed after concentration
is a heme-containing enzyme that is the first and using an Amicon Ultra device.
rate-limiting enzyme in human tryptophan metabo-
lism. IDO processes 98% of the total tryptophan Concentration of PKR
available in the human body and is critical in
and Buffer Exchange
suppression of immunoresponse by blocking
Courtesy of Peter A. Lemaire and
T-lymphocyte proliferation locally [Swanson et al,
Dr. James Cole, University of Connecticut
Am J Respir Cell Mol Biol [manuscript in prepara-
tion] (2003); Sarkhosh et al, J Cell Biochem 90, Human protein kinase R (PKR) is one of the major
206 (2003); Mellor et al, J Immunol 171, (2003)]. proteins induced by interferon as part of the host
Recombinant IDO was expressed in E. coli BL21 defense against viral infection1–4. PKR is synthesized
(DE3) cells utilizing the pET 28a (+) vector system. in a latent form and is activated by autophosphory-
In this system, a hexahistidyl tag was fused to full- lation induced upon binding dsRNA. Once
size IDO at the N-terminus with a spacer sequence phosphorylated, active PKR phosphorylates the
and a thrombin cleavage site. The protein was eukaryotic translation initiation factor elF2α leading
purified by conventional His-tag purification methods to a block in protein synthesis in virally infected
and eluted with imidazole. The histidine tag was cells. PKR has been implicated as a participant in
removed by thrombin cleavage. Final purification various signal transduction pathways associated
was done by gel filtration chromatography G-75. with cellular processes including transcription7–9,
Amicon Ultra-15 centrifugal devices were used to differentiation10, apoptosis11, splicing14 and
transformation5,6. However, difficulties in purifying
PKR in large amounts has limited rigorous biophys-
ical characterization of the mechanisms of PKR
activation. A high-yield prokaryotic expression
system has been developed for PKR, and PKR has
40 kDa
been purified using three chromatography steps on
Agarose-Heparin, Agarose-Poly (I), Poly (C) and
35 kDa
Sephacryl S-200 gel filtration columns. After the
last step, PKR-containing column fractions were
pooled and concentrated using Amicon Ultra-15
30K NMWL devices. The concentration step was
necessary for long-term protein storage. Table 3.5
Figure 3.1 SDS-PAGE of purified indoleamine shows the protein recovery results obtained after
2,3-dioxygenase before and after concentration four concentrations. Over 90% recovery was
using Amicon Ultra-15 centrifugal devices. obtained and no protein loss to the filtrate was
observed.

Table 3.5 PKR concentration results


Spin 1 Spin 2 Spin 3 Spin 4
Starting volume (mL) 15 15 15 15
Starting concentration (mg/mL) 0.229 0.229 0.169 0.169
Volume of retentate (µL) 590 490 203 225
Total amount of PKR (mg)
Before UF 3.4350 3.4350 2.5320 2.5320
After UF 3.1005 3.1548 2.4081 2.3983
Filtrate 0.0000 0.0000 0.0000 0.0000
% Recovery 90.26 91.84 95.11 94.72

18
Amicon Ultra-15 30K NMWL devices were also 3. Lebleu B, et al. Interferon, double-stranded RNA,
used for exchanging buffer for PKR autophosphory- and protein phosphorylation. Proc Natl Acad Sci
lation (activation) assay. 200 µL of 6.301 mg/mL USA 1976;73(9):3107–11.
PKR in Protein Storage buffer (20 mM HEPES, 4. Samuel CE. Mechanism of interferon action:
1 M NaCl, 10 mM β-mercaptoethanol, 0.1 mM phosphorylation of protein synthesis initiation
factor eIF-2 in interferon-treated human cells by
EDTA, 10% glycerol, pH 7.5) was diluted to 15 mL
a ribosome-associated kinase processing site
with Phosphorylation Buffer (20 mM HEPES, 50 mM
specificity similar to hemin-regulated rabbit
KCl, 5 mM MgCl2, 0.1 mM EDTA, 1 mM DTT, reticulocyte kinase. Proc Natl Acad Sci USA
pH 7.5) and re-concentrated three times using 1979;76(2):600–4.
Amicon Ultra devices at 3000 x g for 20 minutes at 5. Koromilas AE, et al. Malignant transformation by
4 °C. The filtrates from the three steps were pooled a mutant of the IFN-inducible dsRNA-dependent
and the total amount of protein in all samples was protein kinase. Science 1992;257:1685–9.
determined by UV absorption A280. 6. Meurs E, et al. Tumor supressor function of
Protein activity was tested by autophosphory- interferon-induced double-stranded RNA
lation assay. The protein in the storage buffer was activated protein kinase. Proc Natl Acad Sci
supplemented with 5 mM MgCl2 and the samples USA 1993;90:232–6.
were allowed to undergo autophosphorylation at 7. Wong AH, et al. Physical association between
30 °C for 20 minutes in the presence of 3 mM ATP STAT1 and the interferon-inducible protein kinase
and 3 µCi [γ32P] ATP. The activity was determined PKR and implications for interferon and double-
stranded RNA signaling pathways. EMBO J,
by autoradiography and quantified by liquid
1997;16(6):1291–304.
scintillation counting. As shown in Figure 3.2, the
8. Cuddihy AR, et al. Double-stranded-RNA-
activity of PKR when no buffer exchange was only
activated protein kinase PKR enhances tran-
6% of that when buffer exchange step was per-
scriptional activation by tumor suppressor p53.
formed. Hence, the UF successfully exchanged the Mol Cell Biol 1999;19(4):2475–84.
buffer while maintaining the activity of the protein.
9. Demarchi F, Gutierrez MI, Giacca M. Human
immunodeficiency virus type 1 Tat protein
References activates transcription factor NF-kappaB through
1. Samuel CE. Antiviral actions of interferon. the cellular interferon-inducible, double-stranded
Interferon-regulated cellular proteins and their RNA-dependent protein kinase, PKR. J Virol
surprisingly selective antiviral activities. 1999; 73(8):7080–6.
Virology 1991;183(1):1–11. 10. Petryshyn R, et al. Effect of interferon on protein
2. Hovanessian AG. The double stranded RNA- translation during growth stages of 3T3 cells.
activated protein kinase induced by interferon: Arch Biochem Biophys 1996;326(2):290 –7.
dsRNA-PK. J Interferon Res 1989;9(6):641– 7. 11. Barber GN. Host defense, viruses and apop-
tosis. Cell Death Differ 2001;8(2):113–26.
12. Tan SL, Katze MG. The emerging role
of the interferon-induced PKR protein kinase as
100 a apoptotic effector: A new face of death?
J Interferon Cytokine Res 1999;19:543–54.
Relative Activity of PKR

80 13. Balachandran S, et al. Activation of the dsRNA-


dependent protein kinase, PKR, induces apop-
60 tosis through FADD-mediated death signaling.
EMBO J, 1998;17(23):6888–902.
40
14. Osman F, et al. A cis-acting element in the
3'-untranslated region of human TNF-alpha
20
mRNA renders splicing dependent on the
activation of protein kinase PKR. Genes Dev
0
No Buffer 1999;13(24):3280–93.
Buffer Exchange
Exchange

Figure 3.2 Comparison of PKR autophosphoryla-


tion with buffer exchange by ultrafiltration and
without buffer exchange.

19
Protocols • Proteins

Detergent Removal with Microcon,


Amicon Ultra or Centricon Centrifugal Filters

Introduction Method and Results


Microcon, Amicon Ultra and Centricon centrifugal In a series of studies, Millipore researchers used
filters are efficient laboratory tools for removing small Total Organic Carbon (TOC) analysis to measure
molecules from solutions of proteins or nucleic acids. detergent removal by Microcon, Amicon Ultra and
Often, the molecule to be removed is one of a Centricon concentrators after a single centrifugation
number of commonly used detergents or protein spin (note that complete detergent removal generally
solubilizing agents. The chemical nature of most requires 3–5 spins). As the results in the Tables 4.1
detergents allows for micelle formation above a and 4.2 indicate, detergent removal depends both
critical concentration limit (Critical Micelle on the original detergent concentration and the
Concentration, CMC). Micelle formation results in NMWL of the centrifugal units. All measurements
aggregation of the detergent and leads to gross shown were made using detergent/distilled water
changes in molecular structure. This affects the solutions.
amount of the detergent that can be removed from a NOTE: Temperature, the presence of salts in
solution by centrifugal devices with specific nominal the solution, and/or macromolecule/detergent
molecular weight limit (NMWL) membranes. interactions may lower the CMC for a particular
For example, the monomer of Triton® X-100 detergent. Use the tables only as general
has a molecular weight of 500–650 daltons. guidelines in assessing the efficiency of detergent
Triton X-100 should pass readily through the removal with the Microcon, Amicon Ultra and
10,000 NMWL membrane in an Amicon Ultra Centricon devices.
device. However, at concentrations above
0.01% (0.2 mM), Triton X-100 forms micelles
composed of approximately 140 monomeric units.
During ultrafiltration, the micelles behave like
70,000–90,000 dalton globular proteins.
As a result, more than 90% of Triton is retained by
the ultra-filtration membrane. Therefore, above the
CMC of Triton X-100, an Amicon Ultra-4 100K
NMWL concentrator would be required to remove
the detergent effectively.

20
Table 4.1 Percent detergent removal after one spin with Centricon and Table 4.2 Percent detergent removal
Microcon centrifugal devices after one spin with Amicon Ultra-4
centrifugal devices
NMWL NMWL
Detergent 3kDa 10 kDa 30 kDa 50 kDa 100 kDa Detergent 10 kDa 30 kDa
SDS SDS
0.01% > 90% > 90% > 90% > 90% > 90% 0.1% 95% 98%
0.1% > 90% > 90% > 90% > 90% > 90% 1% 38% 48%
1% 40 – 89% 40 – 89% 40 – 89% 40 – 89% 40 – 89% 5% 94% 95%
5% < 40% < 40% < 40% 40 – 89% 40 – 89% Tween-20
NaDeoxycholate 0.1% 30% 42%
0.1% > 90% > 90% > 90% > 90% > 90% 1% 28% 35%
1% > 90% > 90% > 90% > 90% > 90% 5% 82% 77%
5% 40 – 89% 40 – 89% 40 – 89% > 90% > 90% Triton X-100
CAPS 0.1% 3% 47%
5% > 90% > 90% > 90% > 90% > 90% 1% 2% 20%
CPCl1 5% 2% 20%
0.01% > 90% > 90% > 90% > 90% > 90% CHAPS
0.1% 40 – 89% 40 – 89% 40 – 89% 40 – 89% 40 – 89% 0.1% 90% 96%
1% < 40% < 40% < 40% < 40% 40 – 89% 1% 66% 93%
5% < 40% < 40% < 40% < 40% 40 – 89% 5% 38% 73%
TDMABr2
0.1% > 90% > 90% > 90% > 90% > 90%
1% < 40% < 40% < 40% 40 – 89% > 90%
5% < 40% < 40% < 40% < 40% > 90%
Digitonin
0.01% > 90% > 90% > 90% > 90% > 90%
0.1% 40 – 89% 40 – 89% 40 – 89% 40 – 89% 40 – 89%
1% < 40% < 40% < 40% < 40% < 40%
Tween®-20
0.01% < 40% < 40% 40 – 89% 40 – 89% > 90%
0.1% < 40% < 40% < 40% 40 – 89% > 90%
1% < 40% < 40% < 40% < 40% 40 – 89%
5% < 40% < 40% < 40% < 40% < 40%
Triton X-100
0.01% 40 – 89% 40 – 89% 40 – 89% 40 – 89% > 90%
0.10% < 40% < 40% < 40% < 40% > 90%
1% < 40% < 40% < 40% < 40% 40 – 89%
5% < 40% < 40% < 40% < 40% < 40%
CHAPS
0.10% > 90% > 90% > 90% > 90% > 90%
1% 40 – 89% 40 – 89% > 90% > 90% > 90%
5% < 40% < 40% 40 – 89% > 90% > 90%
1Cetylpyridinium chloride
2Tetradecyltrimethylammonium bromide

21
Protocols • Proteins

Two-Dimensional Electrophoresis Sample Preparation


with Amicon Ultra Centrifugal Filters
Two-dimensional electrophoresis (2DE) is one of the Usually protein concentration is achieved by
most commonly used methods in proteome analysis. protein precipitation with acetone or TCA. The
Briefly, proteins are separated by their isoelectric disadvantage of protein precipitation is that some
point (first dimension separation) followed by SDS- of the proteins become insoluble and can not be
PAGE separation by molecular weight (second resolubilized in IPG buffer. Another disadvantage is
dimension separation). Although it is a powerful that many salts become insoluble in acetone and
method for simultaneously displaying hundreds of precipitate along with the proteins.
proteins, 2DE presents a challenge for sample Ultrafiltration (UF) can achieve protein concen-
preparation. Salts and ionic detergents are common tration and desalting in one step. Figures 5.1 and
chemicals contaminating biological samples that 5.2 present two examples of protein preparation
are not compatible with 2DE separation; however for 2DE by acetone precipitation and ultrafiltration.
they are often required to solubilize proteins from Both examples demonstrate that UF provides more
cells and tissues. The high concentration of salts efficient salt removal and allows better separations
combined with the relatively low protein content and improved resolution of protein spots in
make samples completely unsuitable for isoelectric two-dimensional electrophoresis.
focusing.

Figure 5.1 Two-dimensional electrophoresis of yeast cell lysate prepared by acetone precipitation (left) and
ultrafiltration (right). Sacharomyces cerevisiae strain s288c was grown to log phase. The cells were pelleted
and resuspended in Cellular and Organelle Membrane Solubilizing Reagent from the ProteoPrep™ kit (Sigma)
and lysed by sonication. Cellular debris was removed by centrifugation and the supernatants were reduced
and alkylated with 5 mM tributylphosphine and 10 mM acrylamide. Lysates were acetone-precipitated to
remove residual Tris and alkylating reagent or were filtered through Amicon Ultra-4 10K NMWL devices.
Samples were redissolved in ProteomIQ™ Resuspension Reagent (Proteome Systems), focused in broad range
(pH 3 –10) immobilized pH gradients (IPGs) on the IsoelectrIQ™ IEF instrument. Second dimension gels were
6–15% polyacrylamide gradient GelChips™ (Proteome Systems) run on an ElectrophoretIQ™ 2D instrument
(Proteome Systems). Data courtesy of Dr. G. Smejkal, Proteome Systems, Inc., Woburn, MA, USA.

22
Figure 5.2 Two-dimensional electrophoresis of endothelial cell lysates prepared by acetone precipitation (left)
and ultrafiltration with Amicon Ultra devices (right). Whole cell lysates of endothelial cells were prepared in
7 M urea, 2 M thiourea, 4% CHAPS, 10 mM DTT, 20 mM Tris buffer. The samples were very dilute and presum-
ably contained DNA fragments, salts and lipids. 300 mg of the total protein was either acetone-precipitated or
desalted in Amicon Ultra devices. After acetone precipitation with four volumes of cold acetone, samples were
incubated overnight at – 20 °C, precipitated by centrifugation, rinsed with cold acetone, and pelleted again.
The resulting pellet was resuspended in rehydration buffer (7 M urea, 2 M thiourea, 4% CHAPS, 10 mM DTT,
20 mM Tris). Alternatively, samples were filtered through Amicon Ultra-4 10K NMWL devices after being
mixed with nine volumes of 20 mM Tris HCl, pH 7. The sample was concentrated to approximately 40 µL
and adjusted to 350 µL with rehydration buffer. The proteins were separated by IEF in 18 cm IPG Drystrips™,
pH 3–10 (Amersham). A second dimension separation was performed in 10% SDS-PAGE gel. The gels
were silver stained and scanned. Data courtesy of Dr. Leonid Kryazhev, Genome Quebec, Montreal, Canada.

23
Protocols • Proteins

Purification of Serum Peptides for Biomarker Research


with Amicon Ultra Centrifugal Filters

Introduction for mass spectrometry analysis than the acetonitrile


precipitation method.
A biomarker can be defined as a molecule that
indicates an alteration in physiology. Biomarkers
Methods
play an essential role in the drug discovery and
development process. They provide powerful clues Preparation of Serum Peptides by UF and SPE
to genetic susceptibility, disease progression, and One milliliter of human serum, with or without
predisposition, as well as drug response and the acetonitrile, was filtered using Amicon Ultra-4
physiological and metabolic profiles of diseases 10K NMWL centrifugal devices. The devices were
and drug responses. Biomarkers can also provide centrifuged in a swinging bucket rotor for 15 to 30
valuable diagnostic and prognostic information that minutes at 3000 x g. Ten microliters of the filtrate
can facilitate personalized medicine. Peptide and was acidified with 5 µL of 1% TFA, concentrated
protein patterns have been linked to ovarian cancer, with ZipTip®µC18 pipette tips. Co-elution was
breast cancer, prostate cancer and astrocytoma1–5. performed directly onto a MALDI target with 2 µL of
Most diagnostic tests are based on blood or ∂-cyano-4-hydroxycinnamic acid matrix (5 mg/mL
urine analysis5. Serum is a key source of putative in 50% acetonitrile, 0.1% TFA). If acetonitrile was
protein biomarkers, and, by its nature, can elucidate added to the serum prior to the filtration, the samples
organ-confined events. Use of mass spectrometry were briefly evaporated in a Speed-Vac™ centrifuge
coupled with bioinformatics has been demonstrated to remove solvent before ZipTip purification.
as being capable of distinguishing between serum
Peptide Analysis by Mass Spectrometry
protein pattern signatures in late-stage and early-
stage ovarian cancer patients6. Peptide-containing ultrafiltrates from cell lysates or
One of the major impediments to the discovery human serum were acidified with 1% TFA and
of new biomarkers is the presence of salts, proteins, concentrated on ZipTipµC18 or ZipTipSCX pipette
and lipids in plasma or serum that makes it difficult tips following the procedure outlined in the user
to detect and analyze peptides by mass spectrom- guide. All samples were overlaid with 1 µL of
etry. When untreated serum is spotted onto a ∂-cyano-4-hydroxycinnamic acid matrix (5 mg/mL
MALDI-TOF plate, it does not produce any useable in 50% acetonitrile, 0.1% TFA) and analyzed on
signal in mass spectrometry. Multiple protocols have Voyager-DE™ Workstation (Applied Biosystems)
been developed to extract and enrich peptides from in linear mode.
tissues and body fluids, such as batch reversed phase
Preparation of Serum Peptides
chromatography over C18 resin and extraction with
by Acetonitrile Precipitation
0.1% trifluoroacetic acid (TFA) or 50% acetonitrile to
Acetonitrile was added to human serum at a
selectively precipitate large proteins while enhancing
1:1 ratio (v:v), and samples were centrifuged to
the solubility of smaller proteins and peptides.
precipitate larger proteins. The supernatant was
Ultrafiltration has previously been reported as
dried in a Speed-Vac centrifuge, resuspended in
a sample preparation tool to prepare low molecular
0.1% TFA, and then desalted and concentrated
weight fractions for biomarker analysis7–10. In this
with ZipTipµC18 pipette tips. Co-elution was
study we show that ultrafiltration in combination with
performed directly onto a MALDI target with
solid phase extraction (SPE) on C18 resin can be a
2 µL of ∂-cyano-4-hydroxycinnamic acid matrix
convenient and efficient method for serum peptide
(5 mg/mL in 50% acetonitrile, 0.1% TFA).
purification. The approach provides more peptides

24
Results supernatant after 50% acetonitrile precipitation;
and (C) serum ultrafiltrate processed with an Amicon
While human serum contains numerous peptides
Ultra-4 30K NMWL device. We observed higher
and small proteins, they are not accessible by direct
quality spectra and an increased number of MALDI-
mass spectrometry analysis. Even after reverse
TOF detected peptides if the serum was ultrafiltered.
phase concentration and desalting, only a few
Further improvement of spectra can be
peptides are detectable in the mass spectrum
achieved by reverse phase chromatography,
(data not shown). This can be explained by the
which concentrates and desalts the peptides.
high concentration of proteins and lipids competing
The ZipTipµC18 pipette tip is a convenient and
with the peptides to bind to the resin (Figure 6.1A).
efficient tool for micro-scale sample preparation
One of the common methods for serum peptide
prior to mass spectrometry. Figure 6.2 shows the
preparation for mass spectrometry is acetonitrile
MALDI-TOF spectra of rat serum peptides prepared
fractionation, where the addition of 50 – 70%
with ZipTipµC18 pipette tip out of (A) straight serum,
acetonitrile precipitates larger proteins, while the
(B) 50% acetonitrile supernatant and (C) serum
peptides stay soluble in the supernatant. Another
processed by ultrafiltration. The last method provided
way to produce relatively protein-free filtrates is
stronger MALDI-TOF signal, higher signal-to-noise
ultrafiltration. Figure 6.1 presents the MALDI-TOF
ratio and double the number of detected peptides.
spectra of (A) straight rat serum; (B) serum

A B C

Figure 6.1 MALDI-TOF spectra of (A) unprocessed rat serum; (B) serum peptides in 50% acetonitrile supernatant; and (C) serum ultrafiltrate
processed with Amicon Ultra-4 30K NMWL centrifugal device.

Figure 6.2 MALDI-TOF spectra of rat serum


peptides after concentration and desalting by A B
reversed phase chromatography: (A) rat serum
processed with ZipTipµC18 pipette tip; (B) serum
supernatant after 50% acetonitrile precipitation
processed with ZipTipµC18 pipette tip; (C) 30K
serum ultrafiltrate processed with ZipTipµC18
pipette tip; and (D) the same as (C) but 20%
acetonitrile was added to the serum prior to
ultrafiltration.

C D

25
We have also investigated whether the addition References
of acetonitrile to serum prior to ultrafiltration improves 1. Ardekani AM, Liotta LA, Petricoin III. Expert
the detection of serum peptides (MALDI spectrum Rev Mol Diagn 2002;2:12.
shown in Figure 6.2D). Further improvement of the 2. Carter D, Douglass JF, Cornellison CD, Retter
spectrum was attained, especially in the m/z 1000 MW, Johnson JC, Bennington AA, Fleming TP,
area of the spectrum. Reed SG, Houghton RL, Diamond TS, Vedvick
TS. Biochemistry 2002;41:6714.
Conclusion 3. Wellmann A, Wollscheid V, Lu H, Ma ZL,
For the analysis of serum peptides, complexity Albers P, Schutze K, Rohde V, Behrens P,
reduction by eliminating higher molecular weight Dreschers S, Ko Y, Wernert N. Int J Mol Med
2002;9:341.
proteins is critical for high resolution mass spec-
trometry. Efficient separation of peptides from the 4. Petricoin EF, Ardekani AM, Hitt BA, Levine PJ,
Fusaro VA, Steiberg SM, Mills GB, Simone C,
majority of proteins and salts can be achieved by
Fishman DA, Kohn EC, Liotta LA. Lancet 2002;
sample ultrafiltration. We have shown the effective
359:572.
use of Amicon Ultra-4 30K NMWL centrifugal
5. Bischoff R, Luider TM. J Chrom B 2004;803:
devices for the preparation of peptides. Other
27–40.
molecular weight cut offs can be utilized depending
6. Stevens EV, Liotta LA, Kohn EC. J Gynecol
on the desired range of peptides. The method can
Cancer 2003;13:133– 9.
be used directly in combination with ZipTipµC18
7. Schulz-Knappe P, Schrader M, Standker L,
pipette tips for peptide identification by MS/MS or
Richter R, Hess R, Jurgens M, Forssmann W-G.
as a first step prior to further surface-mediated J Chromatogr A 1997;776:125–132.
enrichment using SELDI-TOF methods.
8. Basso D, Valerio A, Seraglia R, Mazzza S,
Piva MG, Greco E, Fogar P, Gall N,
Pedrazzoli S, Tiengo A, and Plebani M.
Pancreas 2002;24:8–14.
9. Prazeres S, Santos MA, Ferreira HG, Sobrinho
LG. Clin Endocrinol (Oxf) 2003;58:686–90.
10. Tirumalai RS, Chan KC, Prieto DA, Issaq HJ,
Conrads TP, Veenstra TD, Mol Cell Proteomics
2003;10:1096 –103.

26
Protocols • Proteins

Rapid Antibody Concentration


with Amicon Ultra Centrifugal Filters
Ultrafiltration offers a fast and convenient way to
concentrate antibodies purified from serum, ascites
40 12
fluid, or hybridoma supernatants. The traditional

IgG Concentration (mg/mL)


purification protocol for immunoglobulins includes PROSEP-A
10
30 PROSEP-G
affinity binding to Protein A or G chromatography

IgG Volume (mL)


8
media, washing unbound proteins, and eluting with
a low pH buffer. Subsequently, purified antibodies 20 6

are often too dilute for their intended purpose or 4


for long-term storage. In addition, harsh elution 10
2
conditions sometimes require buffer exchange to
preserve protein activity. 0 0
0 3 10 15 20
Dialysis is often used for antibody concentration
Centrifugation Time (min)
and buffer exchange. However, ultrafiltration provides
a quick, alternative method to concentrate and
Figure 7.1 Concentration of rabbit IgG with Amicon Ultra-15 devices. The
diafilter immunoglobulins with up to 99% recovery IgGs were purified using Montage PROSEP-A or PROSEP-G Antibody Purification
and one-step salt removal (see Concentration, Kits. The lines show IgG volume reduction, while the bars show a proportional
Desalting, and Buffer Exchange with Amicon Ultra increase in IgG concentration after 20 minutes of centrifugation time.
and Microcon Centrifugal Filters, page 16).
To demonstrate the suitability of Amicon Ultra
devices for concentrating purified IgG, rabbit Figure 7.2 kDa

serum was processed with Montage PROSEP-A SDS-PAGE gel of purified rabbit 116
IgG before and after concentration
and PROSEP-G Antibody Purification Kits (Millipore)
with Amicon Ultra-15 devices.
and then concentrated with Amicon Ultra-15 30K 66
55
NMWL devices. Figure 7.1 shows a decrease in Lane 1: MW standards Heavy
Lanes 2, 3: PROSEP-A-purified IgG Chain
the retentate volume proportional to the increase in
before concentration 36
antibody concentration. A twenty-minute centrifuga-
(lane 2) and after
Light
tion resulted in > 95% recovery of immunoglobulins. concentration (lane 3)
21 Chain
Figure 7.2 shows an SDS-PAGE gel of purified Lanes 4, 5: PROSEP-G-purified IgG
14
rabbit IgGs before and after ultrafiltration. before concentration
(lane 4) and after
concentration (lane 5)
Protein Load: 5 µL in lanes 2 and 4;
1 µL in lanes 3 and 5 1 2 3 4 5

27
Protocols • Proteins

Removal of Unincorporated Label from


Labeled Protein with Amicon Ultra or
Centricon Centrifugal Filters

Introduction Results
Centrifugal devices containing ultrafiltration Figure 8.1 shows the SDS-PAGE gel of FITC-labeled
membranes are ideal for the removal or exchange BSA before and after each of four diafiltration
of salts, sugars, nucleotides, non-aqueous solvents, cycles. Unincorporated FITC is clearly visible in the
and other materials of low molecular weight. starting material and in the first filtrate. After only
They also serve to separate free from bound one cycle of ultrafiltration, the majority of the free
species. One of the applications is removal of label is removed. Subsequent cycles of filtration
unincorporated label in protein labeling applications result in BSA that is virtually free of unincorporated
where Millipore centrifugal devices provide fast, FITC.
convenient, high-recovery alternatives to gel Fluorescence measurement offers a more sensitive
filtration. Sample dilution, often associated with method to monitor the ultrafiltration process. Figure
gel filtration, is not a problem. A few rounds of 8.2 shows the change in fluorescence of filtrate and
diafiltration can efficiently remove unincorporated retentate during four cycles of FITC-BSA diafiltration.
label. Two examples below demonstrate the use of The results indicate that almost 80% of free FITC
Millipore centrifugal ultrafiltration devices for can be removed after the first ultrafiltration and
removal of unreacted fluorescent and isotope labels. three rounds result in 98% removal. Therefore,
ultrafiltration can be used to clean up protein
Method for Removal labeling reactions as a viable alternative to gel
of FITC from FITC-BSA filtration. Each ultrafiltration using Amicon Ultra-4
1. Bovine serum albumin was labeled with devices is accomplished in 10 –15 minutes and
fluorescein-isothiocyanate (FITC); the free allows high recovery of target protein. While gel
unincorporated label was not removed. filtration results in diluted protein fractions, ultra-
filtration offers the additional advantage of concen-
2. Two mL of FITC-labeled BSA solution at
trating protein while removing unincorporated label.
0.5 mg/mL were loaded into two Amicon
Ultra-4 10K NMWL devices and centrifuged
Purification of Radiolabeled
at 3,000 x g for 10 minutes.
Tumor Necrosis Factor
3. Retentates (about 50 µL each) were re-diluted
to 2 mL with water and centrifuged again. Courtesy of Mathew L. Thakur, Ph.D., Thomas
This step was repeated twice. Jefferson University Hospital, Philadelphia, PA
Tumor necrosis factor (TNF) is a low-MW protein
4. After each ultrafiltratation, the retentate
(approximately 17 kDa) predominantly derived from
and filtrate were analyzed by SDS-PAGE
macrophages (TNF-alpha) or activated lymphocytes
and their fluorescence measured on a
(TNF-beta). Different cell types, normal and
SpectraFLUOR™ plate reader (Tecan) at
malignant, possess receptors for TNF. The finding
excitation 485 nm and emission 530 nm.
that TNF can destroy tumors in vivo, even in the
5. The SDS PAGE gel was scanned on a
absence of the direct lytic effect of neoplastic cells,
Storm® (GE Healthcare) fluorescence scanner.
has led researchers to the hypothesis that TNF

28
Figure 8.1
60,000
SDS-PAGE of FITC-labeled BSA
Lane 1: Starting material 50,000 Retentate
Lane 2: Retentate after first ultrafiltration

(Aribitrary Units)
Filtrate
40,000

Fluorescence
Lane 3: Retentate after 2 rounds of ultrafiltration
Lane 4: Retentate after 3 rounds of ultrafiltration 30,000
Lane 5: Retentate after 4 rounds of ultrafiltration
20,000
Lane 6: Filtrate after first ultrafiltration
Lane 7: Filtrate after 2 rounds of ultrafiltration 10,000
Lane 8: Filtrate after 3 rounds of ultrafiltration
0
Starting Spin 1 Spin 2 Spin 3 Spin 4
material

Figure 8.2 Fluorescence measurements of the filtrates and retentates after each
FITC-BSA of four cycles of FITC-labeled BSA ultrafiltration. Free label was transferred through
the 10,000 NMWL membrane while labeled BSA was retained and concentrated.
All retentates and filtrates were volume adjusted to 2 mL prior to the measurement.

FITC Table 8.1 Comparison of Centricon centrifugal device


and gel filtration for purification of Tc-99m TNF
1 2 3 4 5 6 7 8
Centricon Sephadex
Device Column
TNF Recovery 83 ± 2% 77.3 ± 8.5%
Time 70 minutes 40 minutes
labeled with short-lived gamma-emitting radio-
Volume 75 µL 9 mL
nuclides may be useful in scintigraphic imaging
of certain tumors.
In order to prepare radiolabeled TNF, ten
micrograms of purified TNF were labeled with
metastable isotope Tc-99m. Centricon 3K NMWL
microconcentrators and Sephadex™ G-25 gel
filtration columns were used to separate Tc-99m-TNF
from unbound Tc-99m. Human serum albumin and
lysozyme were added as a carrier in both cases.
The results of this experiment indicate that
Centricon 3K NMWL concentrators may be used
to purify proteins of small molecular weight from
unbound radioactivity without excessive loss of
proteins. When the quantity of such a protein is
limited, the addition of some carrier molecule after
radiolabeling may be necessary. While eliminating
more than 98% of the unbound radioactivity,
Centricon 3K NMWL units concentrate the protein
with high recovery and without loss, denaturation
or excessive dilution.

29
Protocols • Proteins

Urine Concentration with


Amicon Ultra Centrifugal Filters

Introduction Method
The measurement of specific proteins in urine is 1. Determine the total protein in a 24-hour urine
important for the diagnosis and management of specimen.
disease states. In most cases, the content of these 2. Fill Amicon Ultra-4 device with 4 mL of urine.
proteins in urine is too low to be detected and 3. Centrifuge at 3400 x g for 30 – 45 minutes
needs to be concentrated. Amicon Ultra-4 devices (approximately 25 – 50 µL concentrate volume).
can be used to concentrate urine samples prior to This produces up to a 160-fold increase in
clinical laboratory analyses. For example, patients concentration.
with multiple myeloma exhibit a proliferation of one
4. Insert a pipetter into the bottom of the filter
antibody-producing plasma cell, which leads to
unit and withdraw the concentrated sample.
excess production of free immunoglobulin light
5. Perform agarose electrophoresis on the
chains known as Bence-Jones proteins. After sample
concentrate to quantitate light chains and
enrichment in the Amicon Ultra-4 10K NMWL
identify other proteins. Determine the
device, immunofixation electrophoresis can be used
percentage of light chains with respect to
to identify free light chains (Bence-Jones proteins) in
the total number of components in the urine.
urine by forming a light chain-antibody complex.
Then multiply the percentage of light chains
Also, agarose electrophoresis can be used to
by the total 24-hour protein concentration
quantitate light chains and identify additional
(grams per 24-hour volume).
low molecular weight proteins such as albumin,
α-1 globulins, transferrin and IgG that can be 6. Perform immunofixation electrophoresis on
present in renal tubular disorders. Ultrafiltration of the concentrate to identify light chains.
urine samples in Amicon Ultra-4 devices provides
reproducible, high sample recovery for electro- Acknowledgements
phoretic analyses, usually in 45 minutes or less. Research using Amicon Ultra devices for urine
concentration in this protocol was conducted by
Materials Mark Merchant, Ph.D. at Helena Laboratories,
• Amicon Ultra-4 device, 4 mL, 10K NMWL Beaumont, TX.
• Centrifuge with fixed-angle or swinging bucket
rotor capable of 3400 x g
• Kit for microprotein determination (i.e. Sigma
# 610-A/Brilliant blue G/Coomassie™ blue)
• Pipetter with 200 µL tip
• Electrophoresis (agarose gel) and immunofixation
equipment with apparatus and reagents

30
References Additional Notes
1. Tietz N., Clinical Guide to Laboratory Tests, 1. Amicon Ultra devices can also be used to
2nd ed. Philadelphia:W.B. Saunders; concentrate serum, plasma and cerebrospinal
1990;362–363.
fluid for similar analyses. A concentration of
2. Kahns L, Clinical Chemistry 1991; approximately 20 mg/mL is required in order to
37:1557–1558.
detect free light chains from diseased patients
3. Cleveland Clinic homepage. Accessed by agarose electrophoresis. Detection by
July 2002. www.clevelandclinic.org/
immunofixation electrophoresis is 10 times more
myeloma/DiagnosisAndTreatmentOf
sensitive than by agarose electrophoresis.
MultipleMyeloma.html
2. Normal heterogeneous immunoglobulins may
4. Christenson RH, et al. Clinical Chemistry
1983;29(6):1028–1030. also be seen in urine concentrate with immuno-
fixation electrophoresis. This “ladder effect” is
5. Christenson RH, and Russell ME. Clinical
Chemistry 1985;31(6):973. comprised of microheterogenous light chains.
Bence-Jones proteins may be within this ladder.
To verify the presence of Bence-Jones proteins
requires additional analysis by two-dimensional
electrophoresis.
3. If there is excess antigen, dilution of the
concentrate will be required until equilibrium is
achieved between the antigen (Bence-Jones
protein) and the antibody.
4. Millipore also offers static concentrators
(Minicon® devices) for concentration of
Bence-Jones protein in urine.

31
Protocols • Proteins

Affinity Purification with


Ultrafree-MC Centrifugal Filters

Introduction Although the method is relatively simple, care must


be taken not to remove the chromatography resin
Affinity interaction chromatography is often the
when pipetting off the supernatant.
single most effective step in any protein purification
Pre-packed mini-spin columns are a convenient
procedure. Up to 95% purity can be achieved in
tool for small-scale protein purification. They are
one step, depending on the nature of the interaction
operated by centrifugation and usually require
and the starting composition of the protein solution.
less than an hour for the whole procedure.
Well known examples of highly specific affinity
Another alternative for small-scale purification
interactions include antibodies and protein A/G;
are centrifugal devices with microporous membrane,
multiple histidine tags and nickel; streptavidin and
such as Ultrafree-MC centrifugal devices. The
biotin; antibodies and antigens; and many others.
sample can be added to the filter basket and mixed
Less specific interactions are also used for enrich-
for the needed residence time and then centrifuged.
ment or depletion protocols, including albumin
The process removes the interstitial liquid but does
depletion on cibacron blue resin, glycoprotein
not dehydrate the beads. Washing and elution can
enrichment on concavalin A resin, and capture of
also be performed in a similar manner and are more
nucleic acid-binding proteins on heparin resin.
effective due to the efficient removal of buffer
Affinity chromatography is often employed in
and/or eluant.
the small-scale batch mode as a quick method for
Ultrafree-MC centrifugal filter units with
microgram-scale protein purification. The typical
microporous membrane (Figure 9.1) come with low
protocol involves:
protein-binding Durapore PVDF membrane in five
1. Pipetting a small volume of affinity resin into
different pore sizes from 0.1 to 5.0 µm. Affinity
a microfuge tube that contains the sample
resin can be loaded into the filter basket and the
2. Vortexing the tube for a few minutes
device used as a “home-made” mini-spin column.
3. Centrifuging the resin to the bottom We show the applicability of the device for
4. Pipetting off the supernatant purification of rabbit IgG on PROSEP-A resin and
5. Washing a few times (using steps 2 and 3) His-tagged C-RP protein on three different
6. Eluting with a small amount of eluant commercial metal-chelate resins.

Materials
• Ultrafree-MC 0.45 µm centrifugal devices
(Millipore cat. no. UFC3 0HV 00)
• PROSEP-A high capacity resin
(Millipore cat. no. 1131 118 26)
• Rabbit serum Gibco
(Invitrogen lot no. 1132782)
Figure 9.1
• Micro-centrifuge Biofuge® Pico
Ultrafree-MC
centrifugal (Heraeus instruments)
filter unit • Jouan CR1822 fixed angle rotor centrifuge

32
• Xcell SureLock™ Mini-cell vertical electrophoresis Method for His-tagged
system (Invitrogen cat. no. EI0001)
C-RP Purification
• NuPage® NOVEX Bis-Tris 4 –12%, 1 mm thick,
Solutions
15 well SDS gels, (Invitrogen cat. no. NP0323)
• Lysis buffer:
• NuPage Sample Reducing agent (10X)
50 mM sodium phosphate, 300 mM sodium
(Invitrogen cat. no. NP009)
chloride,10 mM immidazole, pH 7
• NuPage SDS Sample Buffer (4X)
• Binding buffer:
(Invitrogen cat. no. NP007)
50 mM sodium phosphate, 300 mM sodium
• SimplyBlue™ SafeStain Coomassie G-250 stain chloride, 10 mM immidazole, pH 7
(Invitrogen cat. no. LC6060)
• Wash buffer:
50 mM sodium phosphate, 300 mM sodium
Method for IgG Purification chloride, 20 mM immidazole, pH 7
Solutions • Elution buffer:
• PROSEP-A binding buffer A: 50 mM sodium phosphate, 300 mM sodium
1.5 M Glycine/NaOH, 3 M NaCl, pH 9.0 chloride, 250 mM immidazole, pH 7
• PROSEP-A elution buffer B2: • 1 mg/mL Lysozyme stock
0.2 M Glycine/HCl, pH 2.5 • Benzonase
• PROSEP-A neutralization buffer:
1 M Tris/HCl, pH 9.0 Procedure
1. Recombinant proteins were expressed in
Procedure Escherichia coli.
1. 200 mg of PROSEP-A media were placed in 2. Cells were prepared at a 10X concentration
Ultrafree-MC 0.45 µm filter basket. using lysis buffer. Lysozyme was added to a
2. The columns were equilibrated with 400 µL of concentration of 0.1 mg/mL. To reduce the
binding buffer A and centrifuged for 1 minute viscosity, benzonase was added to the lysate.
at 100 x g. The lysates were clarified by centrifugation.
3. 200 µL of rabbit serum were diluted 1:1 3. 200 µL of the 50% resin slurry were added to
with binding buffer and the entire volume the Ultrafree-MC device and the residual fluid
was loaded into the spin column containing was removed by centrifugation for 1 minute
PROSEP-A resin. at 500 x g.
4. Devices were placed on a shaker for 15 minutes 4. The resin was equilibrated with 500 µL of
at room temperature and centrifuged at 100 x g binding buffer and centrifuged for 2 minutes
for 5 minutes. Flow-through was collected for at 500 x g.
future analysis. 5. 500 µL of the clarified lysate were added
5. Three consecutive washes of 400 µL each to the resin.
were performed by adding 400 µL of binding 6. The His-tagged proteins were bound
buffer A and centrifuging at 2,000 x g for for 30 minutes with light agitation.
2 minutes each.
7. The lysate was removed by centrifugation
6. 200 µL of elution buffer B2 were added and at 500 x g for 10 minutes.
centrifuged for 2 minutes at 2,000 x g.
8. The resin was washed with 500 µL of wash
7. 26 µL of neutralization buffer were added to buffer for 5 minutes with agitation. The wash
each collection tube. A second elution was solution was removed by centrifugation for
collected after repeating the same process 5 minutes at 500 x g. This step was repeated
one more time. two more times.
9. 250 µL of elution buffer were added to the
Ultrafree-MC device and mixed for 5 minutes.
Purified protein was recovered by centrifugation
at 500 x g for 1 minute.

33
1 2 3 4 5 6 7 8
Results
Figure 9.2 Rabbit IgG kDa
purification on PROSEP-A resin 200
The results of purifying rabbit IgG using Ultrafree-MC
in Ultrafree-MC devices. centrifugal devices are shown in Figure 9.2. The
116.3
Lane 1: Molecular weight 97.4 device was challenged with approximately 14 mg
66.3
standards of total protein, with an estimated IgG content of
55.4
Lane 2: Rabbit serum 1.5 – 2 mg. The original serum, flow-through, three
Lane 3: Flow through washes and two eluted fractions were analyzed by
Lanes 4 – 6: Three consecutive 36.5

washes
31.0 SDS-PAGE. The total amount of purified IgG, as
Lanes 7, 8: Eluted IgG from 21.5
estimated by OD280, was 1.2 mg and 1.1 mg
two devices on two devices processed in parallel. The whole
14.4
procedure was completed in less than 1 hour.
6.0
This method can be useful for monitoring the titer
of antigen-specific antibodies after immune
activation, or whenever small amounts of IgG
need to be purified.
Figure 9.3 His-tagged 1 2 3 4 5 6 7 Figure 9.3 shows the results of His-tagged
kDa
protein purification using
200 protein purification in Ultrafree-MC devices using
Ultrafree-MC devices.
Ni-NTA agarose and BD Talon™ resin. Two
Lane 1: Molecular weight 116.3

standards
97.4 recombinant proteins were purified: C-RP, high-
66.3 RT66
Lane 2: E. coli lysate 55.4 expressing protein of 26 kDa; and RT66, low-
expressing C-RP expressing protein of 66 kDa. As seen in Figure 9.3,
protein 36.5 both purifications resulted in high purity proteins.
Lane 3: E. coli lysate 31.0

expressing RT66 C-RP The amount of proteins purified out of 500 µL of


21.5
protein lysate was 32 – 35 µg for C-RP protein and 8 µg
Lanes 4 , 5: Proteins purified 14.4 for RT66.
on Ni-NTA resin
The data show that affinity batch purification
Lanes 6, 7: Proteins purified 6.0
on BD Talon resin can be effectively performed on a small scale
using Ultrafree-MC devices loaded with resin.
The process combines the high efficiency of batch
binding and washing with the handling convenience
of a mini-spin column. With minimal hands-on time,
the method provides flexibility of resin to lysate
ratio and binding conditions, independent of
centrifugation speed and rotor angle. This method
is applicable to recombinant protein purification,
antibody purification and immunoprecipitation.

34
Protocols • Proteins

Use of Centrifugal Filter Devices


as an Alternative to Stirred Cells

Introduction The eluted proteins were concentrated from


volumes of 50 0 –1000 mL to 5 –10 mL using a
Stirred cell devices have been successfully used to
Centricon Plus-70 centrifugal filter device in a
purify proteins from large volumes of solution for
swinging bucket rotor at 3000 x g for 10 minutes
many years. However, the need for assembly and
(60 mL per filter unit). Buffer exchange was also
cleaning between uses, and the lengthy separation
performed subsequently using the same units in
times, can present difficulties for active, or under-
tissue culture grade sodium azide-free PBS. The
staffed, laboratories.
concentrated samples were then filter-sterilized
Millipore’s large volume centrifugal filter devices
under sterile conditions using a 0.2 µm pore filter.
come preassembled and ready to use. Individual
devices are available for processing 20 or 70 mL
Results
volumes. For larger volumes, multiple devices can be
spun simultaneously. Spin times are typically Spectrophotometric evaluation of alpha-gamma
measured in minutes. And unlike stirred cells, fusion protein recovery using Centricon Plus-70
centrifugal devices run unattended. There is no need devices showed a result of 18.8 mg/L of super-
to refill a reservoir and, therefore, less risk of natant. HPLC trace analysis revealed the chro-
adventitious contamination. matogram shown in Figure 10.1.
The following protocol demonstrates the use
of Centricon Plus-70 centrifugal filter devices to Acknowledgement
purify and concentrate proteins from large volumes Protocol courtesy of GKT School of Biomedical
of solution. Sciences, London.

Abstract
This study aims to achieve purification and concen-
tration of a fusion protein composed of the alpha
140
and gamma subunits of the human high affinity IgE
120
receptor. The alpha-gamma fusion protein is purified
Absorbance Units

and then coupled to Sepharose® (GE) beads to 100


produce an affinity column for isolating human IgE 80
antibodies.
60

Method 40

The alpha and gamma subunit components of the 20

high affinity IgE receptor were transfected into 0


mouse myeloma cell line NS0 and the secreted 0 5 10 15 20 25 30

fusion protein was purified on a Protein G column. Time (min)

Figure 10.1 HPLC profile of alpha-gamma fusion component of the human FcεRI

35
Protocols • Nucleic Acids

Concentrating and Desalting DNA or RNA


with Microcon or Centricon Centrifugal Filters

Introduction which often denatures labile species. DNA and


RNA samples with starting concentrations as low as
Centrifugal filter devices serve as powerful tools in
5 ng/mL can be routinely concentrated in minutes
molecular biology applications, such as DNA or
with 99% recovery of starting material, and without
RNA concentration and desalting procedures.
the use of co-precipitants. Centrifugal concentrator
Ultrafiltration (UF) is a pressure-driven, convective
devices are ideal for separating high and low-
process that uses semipermeable membranes to
molecular weight species. Ultrafiltration can also
separate species by molecular size and shape.
be used to change solvents by diafiltration. In this
UF is highly efficient, allowing for concentration
process, the sample is concentrated, then diluted
and desalting at the same time. Unlike the use of
to the original volume with the desired buffer and
chemical precipitation methodologies (i.e., ethanol,
concentrated again, thus “washing out” the original
phenol/chloroform), there is no phase change,
solvent.
Millipore ultrafiltration membranes are character-
ized with well-defined, globular solutes (proteins).
Table 11.1 Nucleotide cut-off guidelines for Microcon and Centricon
centrifugal devices (based on >90% recovery of nucleic acids) Typically, the nominal molecular weight limit
(NMWL) for a membrane is the point at which over
Single-Stranded* Double-Stranded
NMWL Nucleotide Cut-off (bp) Nucleotide Cut-off (bp) 90% of a solute with that molecular weight will be
retained. To process DNA or RNA, the membrane
3K 10 10
needs to be characterized according to the number
10K 30 20
of nucleotides in the fragment. Polynucleotides (DNA
30K 60 50 and RNA) have tertiary structures that are ordinarily
50K 125 100 more extended than those of typical globular
100K 300 125 proteins of similar size. Millipore has determined
nucleotide cut-offs (based on the number of bases
*Single-stranded nucleic acids with extensive secondary structure will be better
retained than those without.
or base pairs in a fragment of DNA or RNA) that
correspond to the NMWL of each of their low
binding membranes. The nucleotide cut-off (NCO)
indicates the fragment length of single- or double-
Table 11.2 Desalting and recovery with Centricon 30K NMWL devices
stranded DNA or RNA that one would expect to
Spin Number % DNA Recovered % CsCl Removed recover at 90% efficiency with a unit of the named
1 94 91.5 NCO. It is best to choose the NCO with about half
2 91 98.9 the length of the fragment of interest. For example,
3 96 99.9 selecting a 30K NMWL membrane for a 50 base
pair fragment generally results in 90% product
Using 1 mL of 25 µg/mL E. coli DNA in 6 M CsCl, sample was repeatedly
recovery. A 10K NMWL membrane would provide
concentrated to 0.1 mL. Sample spun at 2000 x g in a 45 degree fixed angle rotor.
The concentrated sample was reconstituted to original 1 mL volume by adding
for closer to 100% recoveries but would take much
50 mm Tris. After three spins, CsCl concentration was reduced by four orders of longer to process the sample. For nucleic acid
magnitude. Equivalent results can be obtained with the Microcon 30K NMWL filter samples >500 base pairs, a 100K NMWL
unit (as well as other NMWLs). membrane is appropriate.

36
Table 11.1 offers guidelines for DNA/RNA Figure 11.1 shows that both recovery and
retention based on the nucleotide content of single- biological activity are similar when DNA is concen-
and double-stranded pieces. For example, more trated with Millipore’s high recovery centrifugal filter
than 90% of a single-stranded 30-mer will typically devices versus conventional techniques. However,
be retained by a Microcon 10K NMWL or a the efficiency and immediate capability to further
Centricon 10K NMWL centrifugal filter device. utilize the nucleic acids without a subsequent
Concentrating dilute DNA solutions is a key step desalting step is a significant advantage when
for many subsequent preparative and analytical using Millipore centrifugal devices.
procedures. For example, standard plasmid
preparations involving cesium chloride, equilibrium Methods
centrifugation, and gel filtration yield DNA in large Microcon Device
volumes that require concentrating prior to precipita- 1. Select a Microcon unit with nucleotide cut-off
tion. DNA concentration is also necessary in equal to or smaller than the molecular size
purifying restriction fragments from gels. of the nucleic acid you want to retain (refer
There are three major techniques currently to Table 11.1).
available for concentrating nucleic acids:
2. Insert Microcon sample reservoir into one of the
• repeated extractions with n-butanol
two vials provided for each unit.
• adsorption to ion exchange resin, followed
3. To concentrate (without affecting salt concen-
by high salt elution
tration): Pipette up to 500 µL of DNA or RNA
• lyophilization
sample into the reservoir. Spin for recommended
The first method has the disadvantage that
time, not exceeding recommended g-force
n-butanol concentrates all solutes, including salt,
guidelines shown in Table 11.3.
which tends to co-precipitate with DNA upon
addition of ethanol. The second method, ion
exchange, aside from requiring buffers of various
ionic strengths, yields DNA in a high salt solution. Table 11.3 Recommended g-force and spin time for Microcon devices

The third method, lyophilization, increases the Maximum Spin Time Spin Time
G-Force (min.) (min.)
concentration of buffer components, which can
NMWL Rating at 4 °C at 25 °C
result in degradation of nucleic acids.
3K 14,000 185 95
Ultrafiltration membranes retain DNA or RNA but
are permeable by smaller ionic buffer components. 10K 14,000 50 35
Ultrafiltration alone does not change buffer com- 30K 14,000 15 8
position. The salt concentration in a sample 50K 14,000 10 6
concentrated by Microcon or Centricon centrifugal 100K 500 25 15
filter devices will be the same as in the original
sample. For desalting, the concentrated sample is
diluted with water or buffer to its original volume Figure 11.1 Retention of labeled DNA fragments 1 2 3
and spun again in a process called diafiltration. by Centricon 10K and 30K NMWL devices.
1631
This removes the salt by the concentration factor of Two 1 µL aliquots of 32P-labeled pBR322 DNA
the ultrafiltration. fragments were diluted to 1.0 mL using TE buffer
For example, if a 500 µL sample containing (10 mM Tris-HCl, 1 mM EDTA, pH 8.0). They
were subsequently concentrated to 40 µL with the
100 mM salt is concentrated to 25 µL (20X
Centricon devices. 20 µL of each concentrate were 517/506
concentration factor), 95% of the total salt in the analyzed on a 2.5% agarose gel. The intensities of 396
sample will be removed. The salt concentration in the autoradiographic bands were compared to bands 344
298
the sample will remain at 100 mM. Rediluting the from a 1 µL aliquot of the solution of labeled DNA
221/220
sample to 500 µL will bring the salt concentration fragments that had not been concentrated by the
devices. The gel was dried and autoradiographed. 154
to 5 mM. Concentrating to 25 µL once more will
remove 99% of the original total salt. The concen- Lane 1: Not concentrated using Centricon device
75
trated sample will now be in 5 mM salt. For more Lane 2: Concentrated with Centricon 10K NMWL device
Lane 3: Concentrated with Centricon 30K NMWL device
complete salt removal, an additional redilution and
spinning cycle will remove 99.9% of the initial
salt content (see Table 11.2).

37
4. To exchange salt: Add the proper amount of Centricon Device
appropriate the diluent to bring the concentrated 1. Select a Centricon unit with a nucleotide cut-off
sample to 500 µL. Spin for the recommended equal to or smaller than the molecular size of
time, not exceeding g-force shown in Table 11.3. the nucleic acid you want to retain (refer to
To achieve lower salt concentration, repeat the Table 11.1).
entire step as necessary. NOTE: Do not let
2. To concentrate (without affecting salt concentra-
filtrate vial overfill.
tion): Pipette up to 2 mL of DNA or RNA sample
5. Remove reservoir from vial and invert into a new into the reservoir. Spin for the recommended time,
vial (save filtrate until sample has been analyzed). not exceeding g-force shown in Table 11.4.
6. Spin for 2 minutes at 500 –1000 x g to recover 3. To exchange salt: Add the proper amount of
nucleic acid in the vial. appropriate the diluent to bring the concentrated
7. Remove reservoir. Cap vial to store. sample to 2 mL. Spin for the recommended time,
not exceeding g-force shown in the Table 11.4.
To achieve lower salt concentration, repeat the
Table 11.4 Recommended g-force and spin time for Centricon devices entire step as necessary. NOTE: Do not let
Maximum Spin Time (min.) filtrate vial overfill.
NMWL G-Force Rating at 25 °C
4. Remove reservoir from filtrate vial and invert unit
3K 7,500 120 (save filtrate until sample has been analyzed).
10K 5,000 60 5. Spin for 2 minutes at 300 –1000 x g to recover
30K 5,000 30 nucleic acid in the retentate vial.
50K 5,000 15 6. Remove reservoir. Cap retentate vial to store.
100K 1,000 30

38
Protocols • Nucleic Acids

Preparing Samples for Forensics Identification


Analysis with Microcon Centrifugal Filters
Centrifugal filter devices with ultrafiltration (UF) remove inhibitors that can adversely affect PCR
membranes are used by forensics laboratories to amplification. In addition, Chelex extraction of small
isolate genomic DNA for human identification. The bloodstain samples often results in DNA concentra-
source materials are typically blood stains and/or tions below detection limits. Concentrating extracted
other bodily fluids obtained from crime scenes, samples with Microcon 100K NMWL devices
criminal suspects, or human remains. improves the success rate of STR amplification and
The isolated genomic DNA is used to identify yields a full STR allele profile even with minute blood
individual persons based on their patterns of Short or semen stains. According to the New York City
Tandem Repeats. STRs are polymorphic DNA loci Department of Forensic Biology, several of the peaks
that contain repeated DNA sequences. Because the shown in Figure 12.1 would have been below the
repeats can vary from two to seven bases in length, detection threshold and identification would have
many different alleles are possible for each locus. been negative or inconclusive without sample
The parallel analysis of 9 to 13 polymorphic cleanup using Microcon devices.
STR loci can unequivocally identify an individual.
Several suppliers offer STR assay kits. Each Figure 12.1
supplier offers several versions of their kits with Genotype analysis of a
different protocols, modes of operation, and semen sample before
detection systems to suite the user’s needs. and after concentration
by a Microcon
Millipore’s UF-based centrifugal devices are
centrifugal filter.
specified in several of these protocols (AmpFLSTR® Electropherogram A
Profiler, Applied Biosystems; GenePrint® STR and shows a partial profile
GenePrint Fluorescent STR Systems, Promega). with a high molecular
weight loci in each
color missing.
Use of UF-based Centrifugal
Electropherogram B
Devices in New York City shows a full profile and
Depending on the type of evidence under investiga- higher peak heights.
A
tion, the Department of Forensic Biology in New Amounts amplified were
0.62 ng for electro-
York City had been preparing genomic DNA by
pherogram A and 1 ng
either (1) organic extraction (phenol/chloroform/ for electropherogram B.
isoamylalcohol) followed by alcohol precipitation or The sample was
(2) direct lysis in the presence of Chelex® beads extracted with Chelex
(Bio-Rad) with no further purification. beads. The peaks are
labeled with allele
The Department of Forensic Biology’s laboratory
name, size in base
has replaced the ethanol precipitation step with
pairs, and fluorescent
Microcon centrifugal filters with a 100K NMWL peak height. The
ultrafiltration membrane. The centrifugal devices improvement in results
provide a faster way to concentrate the purified is due not only to the
DNA without the use of toxic chemicals. increased DNA input
but also to the
The laboratory also uses the Microcon
concentration step
100K NMWL device after Chelex extraction to B following extraction.

39
Protocols • Nucleic Acids

PCR Purification with


Montage PCR Filter Units

Introduction Primer DNA


Salt
After polymerase chain reaction (PCR)1, amplified dNTP

DNA must be separated from excess reaction


components that can interfere with subsequent
manipulations such as cloning or sequencing.
The Montage PCR filter unit is a single-use device
that simplifies the purification of PCR products.
It concentrates amplified DNA and removes primers
and unincorporated dNTPs, while providing
excellent capacity, high recovery, and high purity. Step 1 Step 2 Step 3
The method is quick and highly reproducible, which
makes it ideal for processing one-up or multiple
samples in middle-throughput operations. 5. Add 20 µL purified water or TE buffer to sample
reservoir.
Materials 6. Invert the reservoir and spin at 1000 x g for
• Variable speed microcentrifuge 2 minutes to retrieve the purified PCR (step 3).
• Montage PCR filter unit
• Purified water (such as Milli-Q water) or TE buffer Results
• PCR reaction (aqueous phase) Montage PCR filter units were used to purify 100 µL
PCR reactions according to the specified protocol.
Procedure After recovering the DNA fragments (n = 10) using
1. Insert the Montage PCR sample reservoir into a reverse spin, the samples were separated by
one of the two vials provided. agarose gel electrophoresis. Recoveries of the
2. Fill the reservoir with 300 µL purified water or TE various PCR products were determined by
buffer. Add 100 µL PCR reaction to the reservoir densitometry (Figure 13.1).
(step 1). Smaller volumes of PCR product may The Montage PCR filter unit is a convenient
be used, but the volume should be adjusted to a method for single-sample PCR purification. The
final volume of 400 µL. high-performance device purifies PCR products in a
single centrifugation step. Purified samples are ready
3. Spin the Montage PCR device at 1000 x g
for downstream applications with no additional
for 15 minutes (step 2). NOTE: For optimal
purification steps.
recovery, do not centrifuge longer than the
specified 15 minutes or greater than 1000 x g.
References
4. To recover DNA, remove the sample reservoir
1. PCR is covered by U.S. patents issued to
from filtrate collection vial and place in a
Hoffmann-LaRoche, Inc.
clean vial.

40
100%

80%
Percent Recovery

60%

40%

20%

0%
137 bp 301 bp 500 bp 657 bp 1159 bp

Figure 13.1 Purification of PCR products using Montage PCR Filter Units

Figure 13.2 Typical electropherogram shows an 1159 bp PCR product purified with a Montage PCR filter unit.
Note the uniform signal intensity and long read lengths. Purified samples are ready for cloning or sequencing with
no additional purification steps.

41
Protocols • Nucleic Acids

Preparation of Fluorescent DNA Probe from Human


mRNA or Total RNA with Microcon Centrifugal Filters

Introduction Method
Microarrays are efficient tools that enable the high 1. To anneal primer, mix 2 µg of mRNA or
throughput identification of genes that are differen- 50 –100 µg total RNA with 4 µg of a regular
tially regulated in response to disease, drugs or or anchored oligo-dT primer in a total volume
other stimuli. With the completion of several key of 15.4 µL as shown in Table 14.1.
genome sequencing projects, scientists now have 2. Heat to 65 °C for 10 minutes and cool on ice.
the ability to custom design DNA microarrays 3. Add 14.6 µL of reaction mixture each to Cy3
specific to their research interests. The recent and Cy5 reactions as shown in Table 14.2.
advances in robotics, bioinformatics and detection
4. Incubate at 42 °C for 1 hour.
technologies have greatly simplified the manufacture
5. Add 1 µL SSII (RT booster) to each sample.
and analysis of microarrays. However, the successful
Incubate for an additional 0.5 –1 hours.
application of microarray technology requires highly
purified, fluorescently labeled cDNA probes. The 6. Degrade RNA and stop reaction by addition
application of ultrafiltration technology to this 15 µL of 0.1 N NaOH, 2 mM EDTA and
challenge has resulted in a robust, efficient and incubate at 65 – 70 °C for 10 minutes.
rapid method for the generation of high quality If starting with total RNA, degrade for
fluorescently labeled probes suitable for use with 30 minutes instead of 10 minutes.
microarrays. The protocol below describes a 7. Neutralize by addition of 15 µL of 0.1N HCl.
method for the generation and purification of 8. Add 380 µL of TE (10 mM Tris, 1 mM EDTA)
fluorescently labeled probes using a Microcon to a Microcon 30K NMWL device column.
30K NMWL centrifugal filter device. Next add the 60 µL of Cy5 probe and the
60 µL of Cy3 probe to the same Microcon
device. NOTE: If re-purification of Cy dye
flow-through is desired, do not combine
probes until Wash 2.
9. Wash 1: Spin column for 7 – 8 minutes at
14,000 x g.
10. Wash 2: Remove flow-through and add
450 µL TE and spin for 7– 8 minutes at
14,000 x g. It is a good idea to save the
flow trough for each set of reactions in a
separate microcentrifuge tube.

42
11. Wash 3: Remove flow-through and add 12. Invert the Microcon device into a clean tube
450 µL 1X TE, 20 µg of Cot1 human DNA and spin briefly at 14,000 RPM to recover
(20 µg/µL, Gibco-BRL), 20 µg polyA RNA the probe.
(10 µg/µL, Sigma, #P9403) and 20 µg tRNA 13. Select the appropriate row from Table 14.3.
(10 µg/µL, Gibco-BRL, #15401-011). Adjust the probe volume to the value indicated
Spin 7–10 min. at 14,000 x g. Look for in the “Probe and TE” column.
concentration of the probe in the Microcon 14. For final probe preparation add 4.25 µL 20X
device. The probe usually has a purple color SSC and 0.75 µL 10% SDS. When adding
at this point. Concentrate to a volume of the SDS, be sure to wipe the pipette tip with
less than or equal to the volume listed in clean, gloved fingers to remove excess SDS.
the “Probe and TE” column in Table 14.3. Avoid introducing bubbles and never vortex
These low volumes are attained after the after adding SDS.
center of the membrane is dry and the probe
The probe is now ready for hybridization.
forms a ring of liquid at the edges of the
membrane. Make sure not to dry the
Acknowledgements
membrane completely.
Protocol courtesy of Patrick Brown, Max Diehn,
and Ash Alizadeh, Stanford University School
of Medicine

Table 14.1 Preparation of primer


Primer Cy3 Cy5 Notes
mRNA (1 µg/µL) x µL y µL 2 µg of each if mRNA; 50 –100 µg if total RNA
Oligo-dT (4 µg/µL) 1 µL 1 µL Anchored: 5'-TTT TTT TTT TTT TTT TTT TTV N-3'
Purified H2O (DEPC) to 15.4 µL to 15.4 µL
Total volume 15.4 µL 15.4 µL

Table 14.2 Preparation of probe


Final
Reaction Mixture Volume Unlabeled dNTPs Volume Concentration
5X first-strand buffer* 6.0 µL dATP (100 mM) 25 µL 25 mM
0.1 M DTT 3.0 µL dCTP (100 mM) 25 µL 25 mM
Unlabeled dNTPs 0.6 µL dGTP (100 mM) 25 µL 25 mM
Cy3 or Cy5 (1 mM, Amersham) 3.0 µL dTTP (100 mM) 10 µL 10 mM
Superscript® II (200 U/µL, Gibco-BRL) 2.0 µL Purified H2O 15 µL
Total volume 14.6 µL Total volume 100 µL

*5X first-strand buffer: 250 mM Tris-HCl (pH 8.3), 375 mM KCl, 15 mM MgCl2

Table 14.3 Final probe preparation


Cover Slip Size Total Hybrid Probe and TE 20X SSC* 10% SDS
(mm) Volume (µL) (µL) (µL) (µL)
22 x 22 15 12 2.55 0.45
22 x 40 25 20 4.25 0.75
22 x 60 35 28 5.95 1.05

*20X SSC: 3.0 M NaCl, 300 mM NaCitrate (pH 7.0)

43
Protocols • Nucleic Acids

Purification of In Vitro Synthesized mRNA with


Microcon or Centricon Centrifugal Filters

Introduction Methods
In vitro transcription reactions employing T3, T7 or RNA Transcription
SP6 phage-encoded RNA polymerases are widely For our studies we chose plasmid pGEM-luc
used to synthesize RNA from recombinant vectors containing the luciferase gene (luc) in the center of
containing appropriate promoters. Production of a multiple cloning cassette of the pGEM-11Zf (-)
large amounts of specific RNA is valuable in the plasmid (Promega). DNA template was linearized
preparation of hybridization probes and in vitro with XhoI, followed by enzyme and salt removal by
translation studies; in the synthesis of ribozymes, diafiltration in Microcon 100K NMWL devices.
rRNA, SRP, antisense RNA and substrates for RNA Linearized template was transcribed, using
splicing; and in RNA-protein interaction studies. MEGAscript® kit (Ambion) according to the recom-
Centricon and Microcon centrifugal filters are mended protocol. After the reaction was completed
well suited for the purification of radiolabeled RNA (3 to 4 hours), template DNA was degraded with
transcripts1. Ultrafiltration can simultaneously and DNase I and the reaction mix added to a Microcon
efficiently remove unincorporated ribonucleotides 30K NMWL device filled with 450 µL of water.
and salts from the transcripts and concentrate the The device was spun for 20 minutes at 12,000 x g
RNA. RNA molecules retain their integrity and are in a temperature-controlled centrifuge at 4 °C.
recovered with high yields. Purified, concentrated RNA was recovered by
Purity of a transcript is especially important when inverted spin. For the preparation of capped
it is used in in vitro translation systems. Trace amounts transcript, cap analog m7G (5’) ppp (5’) G
of ethanol, phenol, salts or excess cap analog used (New England Biolabs, Inc.) was included in the
during the synthesis of capped mRNA can cause a transcription reaction and the level of GTP reduced
dramatic decrease in translation efficiency. (4:1 ratio of cap analog to GTP). To purify the
After the transcription reaction is complete, transcript by phenol/chloroform extraction, the
template DNA is usually degraded by the addition reaction mix was diluted with water and a one-tenth
of DNase I. The RNA is purified by two phenol/ volume of ammonium acetate stop solution was
chloroform extractions followed by ethanol pre- added. The mixture was extracted once with
cipitation. Other, less popular methods are gel phenol/chloroform, followed by chloroform
purification (used predominantly when separation extraction. RNA was precipitated with isopropanol
of full-length transcript from shorter RNAs is and the pellet resuspended in distilled water.
important, e.g., ribonuclease protection assays) Alternatively, LiCl precipitation solution (one-half
or LiCl precipitation. volume) was added to the reaction mix, followed
A series of experiments was performed in our by incubation at minus 20 °C for 1 hour. RNA was
laboratory to determine the effectiveness of using pelleted by centrifugation and dissolved in water.
Centricon and Microcon devices to purify in vitro Size and integrity of the in vitro transcription products
synthesized mRNA and in vitro translation studies. were assessed by running an aliquot of the purified
Results indicate that ultrafiltration can efficiently RNA transcript on a formaldehyde/formamide
remove inhibitory contaminants from mRNA agarose gel. Ethidium bromide was added to the
preparations, leading to increased translational RNA before lading on the gel to stain the RNA
efficiencies. sample and keep background fluorescence low2.

44
Translation In Vitro References
In vitro translations were performed in the Flexi™ 1. Krowczynska AM. bioSolutions 1993;2(1):1–2.
Rabbit Reticulate Lysate System (Promega) according 2. Ogretmen B, Ratajczak H, Kats A, Stark BC.
to standard luciferase RNA translation conditions Biotechniques 1993;14(6):932-5.
with minor modifications (Rnasin Ribonuclease 3. Polayes D. Focus 1991;13(4):130 –2.
inhibitor was omitted and 35S-methionine added). 4. Dasso MC, Jackson RJ. Nucl. Acid. Res.
Results of translation were analyzed by determina- 1989;17:3129.
tion of percent incorporation of 35S-methionine and
fold stimulation, compared to controls without RNA.
Minimum acceptable stimulation was 8-fold.
1 2 3 4 5
Figure 15.1 Comparison of pGem-luc transcript
Results purification methods. Transcripts were synthesized in
20 µL reactions. After DNase I treatment, RNA was
Aliquots of RNA transcript purified by different
purified from the reaction mix. 500 ng of purified
methods (ultrafiltration, phenol extraction and RNA were run on 1% agarose/formaldehyde gel.
LiCl precipitation) were run on a denaturing
Lane 1: 0.16 –1.77 kb RNA ladder (Gibco BRL)
agarose/formaldehyde gel. Results are shown in
Lane 2: RNA purified in Microcon 30K NMWL device
Figure 15.1. The banding pattern of the 1.7kb RNA
Lane 3: RNA purified in Centricon 100K NMWL device
transcripts is identical regardless of purification
Lane 4: RNA purified by phenol extraction
method. Similar results were obtained in the case of
Lane 5: RNA purified by LiCl precipitation
capped transcript (results not shown).
The effect of increasing the mRNA concentration
on the translational efficiencies was examined.
At low mRNA levels, the capped luc mRNA was Figure 15.2 Effect of
pGem-luc RNA concen- 140
translated three times more efficiently than the
tration on in vitro trans- 120
uncapped mRNA (Figure 15.2). At higher mRNA lation. Increasing amounts
100
levels, the translation of both transcripts was of uncapped Luc RNA
cpm x 104

80
comparable. Similar behavior was observed with transcripts and capped
CAT mRNA3. Even relatively high levels of mRNA Luc RNA were used in 60
Capped pGem-luc RNA

did not cause the decrease in translational efficien- translation reactions. 40 pGem-luc RNA
Incorporation of
cies noted by other groups4. This result could be 35S-methionine was
20

attributed in part to the lack of inhibitory contami- determined by TCA


0
0 0.5 1.0 1.5 2.0
nants in the mRNA preparation. precipitation. Both RNA µg RNA/50 µL Reaction
We also checked the effect of the RNA transcripts were purified
clean-up method on in vitro translational efficiency. with Microcon 30K
NMWL devices.
For details of various procedures, see the Methods
section. RNA purified by each of the methods
(1 µg) was translated and results showing total
35S-methionine incorporation are presented in
Table 15.1 Effects of RNA clean-up method on translation efficiency
Table 15.1. While there were no observable 35S-methionine
RNA Total Incorporation
differences between these RNAs by gel electro-
RNA 1 972,000
phoresis analysis (Figure 15.1), RNA purified by
ultrafiltration gave twice the translation efficiencies RNA 2 514,000
of phenol-extracted RNA. RNA 3 776,000

The RNA transcripts were cleaned using DNase I incubation followed by purification
in Centricon 100K NMWL devices (RNA 1), phenol/chloroform extraction (RNA 2),
or precipitation with 7.5 M LiCl (RNA 3). 1 µg of each RNA was used to program
50 µL in vitro translation reaction, using the Flexi Rabbit Reticulocyte Lysate System.
35S-methionine incorporation was determined by TCA precipitation. Data show the

average of three independent experiments.

45
Protocols • Nucleic Acids

Quantitative Recoveries of Nanogram Amounts


of Nucleic Acids with Microcon Centrifugal Filters

Introduction were added to each tube. The content was well


mixed and incubated for the specified period of
Molecular cloning experiments often require
time at –20 °C or –70 °C (dry ice). The solutions
concentration of nanogram quantities of DNA.
were then centrifuged at 12,000 x g in a fixed-
When dealing with such small amounts of nucleic
angle microcentrifuge at 4 °C for 15 minutes. The
acids, the use of coprecipitants (tRNA or purified
supernatants were carefully removed and pellets
glycogen) is essential for effective DNA recovery1.
resuspended in 50 µL of TE buffer. The radioactivity
However, when carrier tRNA cannot be used (for
in the precipitates and supernatants was then
example, when DNA is to be labeled with 32P dATP
determined by counting Cherenkov radiation.
and polynucleotide kinase), the only method of
The percentage of recovered DNA was calculated
recovering DNA as a precipitate in ethanol has
from precipitates and the initial counts. Each data
been the ultracentrifugation method developed by
point in the tables represents the average of at least
Shapiro2. Ultrafiltration, which has been traditionally
three samples.
used to concentrate and desalt protein samples
To precipitate the oligomer, 60 µL of TE buffer
simultaneously, can be an efficient alternative to
containing a given amount of 25-mer and 5 ng of
ethanol precipitation.
radiolabeled tracer was supplemented with 240 µL
In this experiment, three popular ethanol precipi-
of 5 M ammonium acetate and 750 µL of EtOH.
tation protocols for DNA and oligonucleotides are
Subsequent steps were identical to those described
compared with ultrafiltration in Centricon and
for DNA.
Microcon centrifugal filter devices. For a detailed
discussion on the effects of incubation time, Ultrafiltration
temperature and centrifugation parameters on
A solution of DNA (500 µL) was placed into a
ethanol precipitation, see Reference 3.
Microcon 30K NMWL unit and spun at 12,000 x g
for 10 minutes. A 500 µL solution of oligonucleo-
Methods tides in TE buffer was placed into a Microcon 3K
Plasmid pBR322 was digested with EcoR I and the NMWL unit and spun for 45 minutes at 12,000 x g.
3’ recessed termini were filled-in with alpha 32P The retentates were recovered by inverting the units
dATP, using Klenow fragment of DNA polymerase I. and centrifuging at 500 –1000 x g for 2 minutes.
A 25 nucleotide mixed base oligomer was radio- While concentrating the oligomer samples, it is
labeled by phosphorylation with bacteriophage T4 important to avoid high salt concentrations which
polynucleotide kinase. promote binding of single-stranded nucleic acids
to the cellulose-based ultrafiltration membrane.
Ethanol Precipitation
The radioactivity in the retentates and filtrates
All DNA precipitations were performed in 250 µL
was determined by counting Cherenkov radiation.
volume. Each tube contained a given amount of
Data represent averages of six samples each.
DNA, supplemented with 10 ng of radioactively
labeled pBR322 in 0.3 M sodium acetate buffer.
To precipitate the DNA, 2.5 volumes of 95% EtOH

46
Results can significantly improve recovery. In contrast to
EtOH precipitation, ultrafiltration using Microcon or
Standard methods for ethanol precipitation of
Centricon devices delivered almost 100% recovery
nucleic acids and ultrafiltration are compared in
with concentrations as low as 10 ng/mL and took
Tables 16.1 and 16.2. Use of Centricon 30K
only 10 minutes. The ultrafiltration devices were
NMWL and Centricon 3K NMWL (data not shown)
found to concentrate and desalt nucleic acids
devices gave results similar to those obtained with
effectively in one step resulting in high recoveries
Microcon devices. Recovery of DNA was assessed
and providing a quick, alternative to ethanol
at close to 100%, indicating that no significant
precipitation.
amount of nucleic acid was lost to the membrane
or device due to adsorption.
References
In contrast, DNA recovery using EtOH precipi-
1. Wallace DM. Precipitation of Nucleic Acids.
tation varied from as little as 14% as in the case
Methods in Enzymology 1987;152:41–8.
of DNA (10 ng/mL) precipitated for 15 minutes
2. Shapiro DJ. Quantitative Ethanol Precipitation
at – 70 °C to a maximum of 76% after overnight
of Nanogram Quantities of DNA and RNA.
incubation at – 20 °C. Anal Biochem 1981;110:229–31.
3. Crouse J, Amorese D. Ethanol Precipitation:
Discussion Ammonium Acetate as an Alternative to
Poor recovery of nucleic acids at very low concen- Sodium Acetate. Focus 1987;9(2):3–5.
tration with ethanol precipitation may be partially
due to the fact that small amounts of DNA do not
adhere well to the tube walls following sedimenta-
tion unless high g forces (ultracentrifugation) are
employed. The yield of DNA incubated at – 70 °C
is slightly reduced, in agreement with previous
studies. Also an overnight precipitation at – 20 °C

Table 16.1 Efficiency of different methods for concentration of DNA


DNA Concentration
10 ng/mL 25 ng/mL 50 ng/mL 250 ng/mL 1000 ng/mL
Method % Recovery % Recovery % Recovery % Recovery % Recovery
EtOH, –70 °C, 15 min 14 15 23 52 55
EtOH, –20 °C, 30 min 13 20 25 60 72
EtOH, –20 °C, 18 hr 31 45 45 76 67
Microcon 30K NMWL Device 95 95 98 98 99

Table 16.2 Efficiency of different methods for concentration of oligonucleotides


Oligomer Concentration
10 ng/mL 25 ng/mL 50 ng/mL 250 ng/mL 1000 ng/mL
Method % Recovery % Recovery % Recovery % Recovery % Recovery
EtOH, –70 °C, 15 min 4 4 5 6 33
EtOH, –20 °C, 30 min 6 5 6 4 33
EtOH, –20 °C, 18 hr 17 20 15 30 58
Microcon 3K NMWL Device 93 94 93 95 95

47
Protocols • Nucleic Acids

Effect of Centrifugal Ultrafiltration on


Large Fragment DNA Integrity

Introduction To be useful, centrifugation must not cause


breakage of the DNA during the ultrafiltration
Centrifugal ultrafiltration provides a fast and easy
procedure. This article summarizes the results of a
method for the concentration and desalting of
series of experiments designed to evaluate the effect
biological molecules. Previous reports document
of g forces on various samples of high molecular
the use of ultrafiltration for desalting samples of
weight DNA during centrifugal ultrafiltration in
nucleic acids1, removing excess primers from PCR2
Microcon or Centricon units.
reactions3, or concentrating DNA/RNA samples
without the need to use ethanol precipitation4.
Methods
Centricon and Microcon filters offer a
convenient, reproducible means for centrifugal DNA Ladder
ultrafiltration of samples from 50 µL to 2 mL. They As a model system to check for the introduction
insure high sample recovery with their patented of single-strand nicks during the ultrafiltration spin,
inverted recovery spin. supercoiled Ladder DNA (Gibco-BRL, Gaithersburg,
The analysis of complex genomes depends on MD) which ranges in size from 2 to 8 kb was used.
the ability of the researcher to prepare pure, high DNA diluted with TE buffer was loaded into
molecular weight DNA. When preparing high Microcon 30K NMWL and Centricon 30K NMWL
molecular weight DNA samples for cosmid cloning devices. The Microcon units were spun for 7 minutes
or other applications, it is important to treat the at 12,000 x g, and the Centricon units were spun
DNA gently to avoid shearing or other damage for 30 minutes at 5,000 x g. The concentrated
to the sample. As virtually all protocols for the DNA sample (retentate) was diluted again with
preparation of high molecular weight DNA require, TE buffer (to 500 µL or 2 mL, depending on the
at some point, buffer exchange and/or concentra- device used) and spun once more, as described
tion, centrifugal ultrafiltration can provide an above. The dilution step was repeated a third time.
efficient alternative to standard procedures. After the third concentration spin, the retentate was
collected by placing the sample reservoirs upside
down in new vials and spinning the units for
1 2 3 4 1 minute at 1,000 x g.
Figure 17.1 Supercoiled DNA Ladder.
The concentrated DNA was run on a 0.9%
Lane 1: Starting material
SeaKem® GTG agarose gel (FMC) and stained
Lanes 2, 3: Retentate from DNA ladder spun 3 times
at 12,000 x g in Microcon 30K NMWL with ethidium bromide to monitor sample integrity
device (Figure 17.1). DNA bands in lane 1 (starting
Lane 4: Retentate from DNA ladder spun 3 times material) are indistinguishable from the bands on
at 5,000 x g in Centricon 30K NMWL lanes 2– 4, which were spun repeatedly in the
device
ultrafiltration devices. There is no evidence that any
supercoiled DNA was converted to the relaxed or
linear forms during the concentration procedure,
which would be the case had single-strand nicks
been introduced during centrifugation.

48
Discrete Size Plasmids Conclusions
The next set of experiments monitored the effect of DNA samples of up to 49 kb were concentrated
centrifugal ultrafiltration on single population, repeatedly without any loss of sample integrity.
discrete size plasmids. The plasmids used were Some loss of integrity was observed with a 125 kb
pBR322 (4,361 bp; New England Biolabs, Inc.), sample, although it was not complete and repre-
pSPT18 (3,104 bp; Boehringer Mannheim) and sents a small percentage of the total DNA in the
pXTl (10,400 bp; Stratagene). Samples (1 µg) of sample. For large fragments of DNA, centrifugal
each plasmid were spun in Microcon 30K NMWL ultrafiltration provides a fast and efficient method to
or Centricon 30K NMWL units, as described concentrate or desalt the sample. It results in high
previously. The starting material and retentates recovery of intact product.
were run on a 1% agarose gel and stained with
ethidium bromide. References
The results are shown in Figure 17.2. As in the 1. Takagi S, Kimura M, Katsuki M.
case of the DNA Ladder, the concentrated samples BioTechniques 1993;14(2):218–21.
(lanes 2, 3, 5, 6, 8, and 9) appear to be identical 2. PCR is covered by U.S. patents issued to
with their corresponding starting material. Again, Hoffmann-LaRoche, Inc.
there is no evidence of conversion of the supercoiled 3. Sheng N, Zhang J, Whitton JL, McKee T.
form to the relaxed form after exposure to g forces BioTechniques 1993;14(5):781–4.
of 5,000 x g for 90 minutes and 12,000 x g for 4. Ruano G, Pagliaro EM, Schwartz TR, Lamy K,
21 minutes during the concentration spins. Messina D, Gaensslen RE, Lee HC.
BioTechniques 1992;13(2):266–74.
Genomic Size DNA
To monitor the integrity of molecules in the size range
of genomic DNA after centrifugation, lambda DNA
(49 kb; Boehringer Mannheim, Indianapolis, IN) Figure 17.2 Specific plasmid DNA

and Bsu36 l digested BacPAK6 DNA (125 kb) was Lane 1: pBR322 starting material 1 2 3 4 5 6 7 8 9
Lane 2: pBR322 spun in Microcon
used. The samples were diluted and centrifuged as
30K NMWL device
described above. The retentates and starting Lane 3: pBR322 spun in Centricon
material were run on a 1% agarose gel in a 30K NMWL device
CHEF-DR® II pulsed field electrophoresis system
Lane 4: pSPT18 starting material
(Bio-Rad, Richmond, CA). Electrophoresis was Lane 5: pSPT18 spun in Microcon
performed at 200 V at 14 °C in 0.5X TBE with 30K NMWL device
ramped pulse from 1 to 6 seconds over 14 hours. Lane 6: pSPT18 spun in Centricon
The results with the lambda DNA mimic those 30K NMWL device
of the other samples run in these sets of experiments. Lane 7: pXT1 starting material
No adverse effects are noted after spinning the Lane 8: pXT1 spun in Microcon 30K NMWL device
Lane 9: pXT1 spun in Centricon 30K NMWL device
DNA at g forces up to 12,000 x g in the ultrafiltra-
tion units. However, the larger BacPAK®6 DNA does
show some degradation after ultrafiltration at both
5,000 and 12,000 x g (Figure 17.3, smearing in Figure 17.3 Large fragment DNA 1 2 3 4 5 6
lanes 5 and 6). Although a large percentage of the
Lane 1: Lambda DNA starting material
sample appears to be intact, there was loss of Lane 2: Lambda DNA spun in
integrity of the BacPAK6 DNA sample after the Microcon 30K NMWL device
concentration procedure. Lane 3: Lambda DNA spun in
Centricon 30K NMWL device
Lane 4: BacPAK DNA starting material
Lane 5: BacPAK DNA spun in
Microcon 30K NMWL device
Lane 6: BacPAK DNA spun in
Centricon 30K NMWL device

49
Protocols • Nucleic Acids

DNA Extraction from Agarose Gels with Montage


Gel Extraction Kit or Ultrafree-DA Centrifugal Filters

Introduction Materials
The Ultrafree-DA device is designed to recover • Microcentrifuge
100 to 10,000 bp DNA from agarose gel slices in • Pre-assembled Ultrafree-DA centrifugal filter
one 10-minute spin. It consists of a pre-assembled device or Montage Gel Extraction Kit
sample filter cup with an agarose gel nebulizer and • Modified TAE* electrophoresis buffer (40 mM
a microcentrifuge vial. The device uses gel compres- Tris-acetate, pH 8.0, 0.1 mM Na2EDTA)
sion to extract DNA from the agarose. Centrifugal • SeaKem agarose (FMC BioProducts) or
force collapses the gel structure, drives the agarose equivalent
through a small orifice in the gel nebulizer and • Long-wavelength UV lamp
captures the resultant gel slurry in the sample filter • Scalpel or razor blade
cup. As the agarose is compressed at 5,000 x g,
DNA is extruded from the gel's pores. The gel Procedure
matrix is retained by the microporous membrane, 1. Electrophorese 30 µL of PCR product or other
and the DNA passes freely through the membrane. DNA through a <1.25% ordinary agarose gel,
DNA can then be recovered in the filtrate vial. prepared in modified TAE buffer with ethidium
The Montage Gel Extraction Kit consists of bromide (0.5 µg/mL).
50 Ultrafree-DA centrifugal filters as well as a 2. Locate the band of interest with a long wave-
modified TAE buffer that allows the casting and length UV lamp or transilluminator. With a razor
running of the gel from which the DNA fragment blade or scalpel, cut out the slice of agarose
is to be extracted. (<100 µL or 100 mg) containing the band of
DNA prepared with the Ultrafree-DA centrifugal interest. Trim any excess agarose away from
filter requires no further purification for most applica- band.
tions, including cloning and radioisotopic or
3. Place the gel slice into the gel nebulizer/sample
fluorescent DNA sequencing. Since agarose gel
filter cup/filtrate vial assembly and seal the
electrophoresis has high resolving power, the small
device with the cap attached to vial.
and large non-specific amplification products that
frequently interfere with cloning and sequencing
after PCR (polymerase chain reaction) are com-
pletely removed from the product.

*Modified TAE is recommended rather than TBE for the


following reasons: (1) TBE buffer strongly inhibits DNA
sequencing reactions while modified TAE buffer does
not. (2) Modified TAE has 0.1 mM Na2EDTA while
regular TAE has 1.0 mM Na2EDTA. The EDTA level at
0.1 mM Na2EDTA will not interfere with the magnesium
concentration in sequencing reactions and other
downstream enzymatic treatments, many of which are
dependent on magnesium.

50
4. Spin at 5,000 x g for 10 minutes.
Centrifugation forces the agarose through the Table 18.1 Effect of gel disruption on typical DNA recoveries from
gel nebulizer, converting it to a fine slurry that is agarose gels
captured by the sample filter cup. Extruded % DNA Recovered
DNA in electrophoresis buffer passes through % DNA Recovered from Gel Disrupted
DNA Size (bp) from Intact Gel by Gel Nebulizer
the microporous membrane in the sample filter
100 74 78
cup and collects in the filtrate vial.
400 39 ND
5. DNA in the filtrate is now ready for sequencing
or cloning without further purification. Discard 700 43 71
the gel nebulizer and sample filter cup and store 1000 55 77
the DNA in the capped filtrate vial. 2027 * 47
4361 14 35
Results 9416 * 32
Gel compression is a quick and easy technique for 23130 * 29
recovering DNA from an agarose gel slice.
* = Not detectable

51
Protocols • Nucleic Acids

RNA Purification with Microcon


or Centricon Centrifugal Filters

Introduction Methods
When working with RNA, introduction of RNase RNA Sample Integrity
contamination during sample preparation is of major The primary objective of the study was to determine
concern. Sample yield and integrity can impact the the effect of centrifugation on RNA integrity. The
efficiency of subsequent translations, hybridizations, second objective was to determine any gross
protein binding, antisense, or ribozyme studies. changes in the RNA that could be a result of contact
A series of experiments was performed to determine of the sample with the ultrafiltration devices
the effectiveness of using Amicon centrifugal ultra- themselves. An RNA transcript was made, using
filtration devices from Millipore to concentrate and MEGAscript T7 RNA polymerase (Ambion) and
diafilter RNA samples. The results indicate remarkably DNA template Riboprobe® Gemini (Promega)
high RNA recoveries and low adsorption losses— according to manufacturer’s protocols. This transcript
especially with prior membrane passivation. served as the starting RNA sample for the experi-
ments discussed below.
Figure 19.1 is an autoradiogram of labeled
RNA. It compares an untreated sample to samples
that were concentrated in Microcon 100K NMWL
and 30K NMWL devices, used directly as supplied.
The Microcon 100K NMWL unit was spun at a
force of 3,000 x g and the Microcon 30K NMWL
unit at 12,000 x g. As is evident from the auto-
radiogram, both unfiltered samples are virtually
indistinguishable from the starting material.
Similar results were obtained with Centricon units,
Figure 19.1 Concentration of RNA 1 2 3 spun at 1,000 x g (Centricon 100K NMWL unit)
transcript. RNA is intact and recovered or 5,000 x g (Centricon 30K NMWL unit).
1.4 kb
with high efficiency.
Lane 1: Starting material Diafiltration of RNA Samples
Lane 2: RNA concentrated in a Experiments were run to determine the RNA recovery
Microcon 100K NMWL device 0.68 kb
and diafiltration efficiency of Centricon devices.
Lane 3: RNA concentrated in a
Centricon 100K NMWL units were used to remove
Microcon 30K NMWL device
unincorporated, radiolabeled ribonucleotides from
the RNA transcript. Two mL of distilled water were
Table 19.1 TCA and total counts of unpurified and purified RNA samples placed into the Centricon sample reservoir, then
90 µL of the unpurified RNA transcript were added.
Unpurified Centricon-purified
Transcript Transcript The device was spun for 30 minutes at 1,000 x g,
TCA counts 70,000 68,500 then inverted and spun for 2 minutes at 1,000 x g
to collect the concentrated RNA (retentate). The
Total counts 136,500 69,000
retentate was diluted 1:10 with water. Total and
TCA precipitable counts were measured.

52
If the Centricon device effectively removed the
unincorporated ribonucleotides, Total and TCA 100%
counts should be close in value. TCA counts of the
retentate sample and of the unpurified transcript 80%

RNA Recovered
(starting material) should also be similar if ultra-
60%
filtration resulted in high recovery of the transcript.
Average values (n = 3) of the Total and TCA counts
40%
appear in Table 19.1. For the retentate sample, the
Total and TCA counts are similar, indicating > 95% 20%
removal of the nucleotides. TCA counts of the samples
indicate that RNA recovery was also above 95%. 0%
0.025 0.25 0.5 1.0 5.0 10.0
RNA Concentration (µg/mL)
RNA Recovery
RNA recovery is a function of the initial RNA concen- Microcon Microcon Microcon Centricon
tration and the buffer salt concentration. Figure 19.2 Filtrate Membrane Retentate Retentate

shows RNA transcript recovery in Microcon 100K


NMWL and Centricon 100K NMWL units as a
Figure 19.2 RNA recovery in Centricon and Microcon concentrators. RNA
function of RNA concentration. For all concentrations
was concentrated as described in text. Cherenkov counts of Centricon retentate
evaluated, retentate recovery is above 85%, even and Microcon retentate, filtrate and membrane (n=3 units) were compared to total
at an initial concentration as low as 25 ng/mL. starting counts.
Figure 19.2 also displays the material monitored in
the filtrate and on the membranes of the Microcon
units. (Membranes were removed and counted 100%
without further treatment.) The amount of RNA in the
filtrate does not vary significantly as a function of 80%
concentration. The RNA recovered on the mem-
RNA Recovered

branes varies from <1% at high initial concentrations 60%

to 4% at the lowest concentration tested.


40%
Millipore’s Amicon centrifugal ultrafiltration
devices contain low-binding cellulosic Ultracel-YM
20%
membranes. Nitrocellulose is commonly used to
immobilize RNA, generally in high salt concentra- 0%
tions. Figure 19.3 shows RNA recovery as a 10 100 200 500
NaCl Concentration (mM)
function of buffer salt concentration. The data
show that both retentate and total recoveries fall as Filtrate Membrane Device Retentate
salt concentration increases. An increase in the
amount of material on the membrane is also seen
Figure 19.3 Effect of increasing buffer salt concentration on RNA recovery
(from 2% to 6%) as the salt concentration increases
with Centricon devices (two left bars at each concentration) or Microcon devices
from 10 to 500 mM. (two right bars at each concentration). Radiolabeled RNA samples were spun at
Similar trends are seen in the use of Microcon 1,000 x g for 30 minutes in a Centricon device or at 3,000 x g for 15 minutes
100K NMWL devices, except that retentate in a Microcon device. Retentate was collected by inverting the unit and spinning
recoveries fall to 60% at 500 mM salt. It was for 2 minutes (Centricon devices) or 1 minute (Microcon devices) at 1,000 x g.
Cherenkov counts of retentate, filtrate, membrane, and device (Microcon units
possible to count the devices themselves in addition
only) were compared to total starting counts.
to the filtrate and membrane. The results show a
slight increase in counts on the membrane with
increasing salt concentration. Significantly more
(from 9% to 17%) material remained on the device
as salt concentration increased.

53
Passivation increased RNA recovery to 80%,
100% regardless of initial salt concentration (Figure 19.4).
This is attributed to a decrease in the amount of
80% RNA adhering to the device. When the devices
RNA Recovered

were passivated, RNA loss due to adsorption was


60%
reduced to 2%, regardless of salt concentration.
40%
Conclusion
20% The results shown here demonstrate that RNA
samples can be concentrated or diafiltered with
0%
Centricon and Microcon concentrators without loss
10 100 200 500
NaCl Concentration (mM) of RNA integrity. RNA recovery in the retentate is
typically > 85%. As the salt concentration of the
Filtrate Membrane Device Retentate buffer system increases, RNA retentate recovery
decreases, principally due to RNA adhering to the
device rather than to the membrane. RNA recovery
Figure 19.4 Improvement in RNA recoveries using Microcon devices (n= 6)
which were pre-treated with a 5% SDS solution. Samples were run as described in from solutions with high initial salt concentrations
Figure 19.3. can be significantly improved by pre-treating the
devices with a 5% SDS solution.

54
Protocols • Nucleic Acids

Enzyme Removal with Micropure-EZ


Centrifugal Filters

Introduction Figure 20.1 Operation of Micropure-EZ device


The Micropure-EZ device is a convenient device
for removing enzymes from double-stranded (ds)
DNA ( 20 bp to >50,000 bp) solutions in a single
DNA Enzyme Salt
centrifugation. It can be used to remove enzymes Remove Enzyme
Micropure-EZ Restriction and other
whenever heat inactivation or phenol/chloroform enyzmes are absorbed
by Micropure-EZ.
extraction is impossible or impractical. After an dsDNA passes freely.
enzyme reaction, the modified DNA solution
Concentrate DNA
containing dilute protein is simply pipetted into a Remove Enzyme DNA is retained by
Micropure-EZ
Restriction and other
Micropure-EZ device that has been inserted into a enyzmes are absorbed Microcon
ultrafilter in Microcon.
Salts pass freely.
Assembly
vial (which is supplied) and centrifuged for 1 minute by Micropure-EZ.
dsDNA passes freely.
at 12,000 –14,000 x g. The DNA passes freely
Vial
into the vial (typically with 85– 90% DNA recovery) Recover
≥ 85% DNA
while the enzyme remains bound to the proprietary
membrane in the Micropure-EZ device. Removal
is defined as the complete absence of detectable
enzyme activity or, as in the isolated case of 30 seconds 3 minutes
T4 polynucleotide kinase, inconsequential residual
activity (<0.08%). For simultaneous concentration
of the enzyme-free DNA, place the Micropure-EZ
device into a Microcon microconcentrator (Figure
Method
20.1) before centrifuging again. The purified DNA
In order to ensure that each enzyme assay was
is suitable for cloning or for other enzymatic
detecting trace amounts of restriction enzymes in
manipulation.
Micropure-EZ filtrates, serial dilutions of Bgl I
While most enzymes were removed by the
(representing 2 units, 0.4 units, 0.08 units, and
Micropure-EZ device (Table 20.1, on page 57),
0.016 units of total activity in 10 µL) were carried
Millipore felt it was equally important to notify
out in a reaction mix* prepared with either sterile
customers of several enzymes that were not removed
DI water (sdH2O) or with sdH2O that had been
(Table 20.2). Millipore scientists were very careful to
filtered through Micropure-EZ and then incubated.
rule out enzyme inhibition as a mechanism. The
Comparison of the resultant restriction digests after
sensitivity of each assay to detect trace amounts
agarose gel electrophoreses verified that aqueous
of restriction enzymes in Micropure-EZ filtrates
extractables from Micropure-EZ devices did not
was determined by carrying out a dilution series
inhibit the enzyme. Also, these standard curves
(this is the method used by restriction enzyme
determined the sensitivity of the enzyme assay
manufacturers to estimate total activity).
(i.e., the theoretical amount of residual enzyme
activity that could be detected, if present).

*15.8 µL of sdH2O, 2 µL of 10X NEBuffer 2,


0.2 µL of 100X BSA, and 2 µL of Bgl I (10 units/µL)

55
To stress test Micropure-EZ devices sufficiently was observed against the decoy pBR322 DNA,
with enzymes and DNA, several extreme operating as expected during its brief exposure to Bgl I
conditions were used consistently. Micropure-EZ (particularly evident in lane 14). However, the
devices were challenged with 50 units of Bgl I in pUC19, which was added directly to the filtrate
the presence of decoy DNA (1 µg of pBR322) and and incubated, appeared completely intact
5 µg of bovine serum albumin (BSA). Devices were (lanes 11–14). Since the standard curve indicated
spun at 14,000 x g for 30 seconds. The filtrates that 0.08 units would be detected if it were present,
were then assayed for residual enzyme activity by we calculated that at least 99.8% of Bgl I was
adding 1 µL of pUC19 DNA and 0.5 µL of 100X removed and/or inactivated. Removal, rather than
BSA to the filtrate, mixing thoroughly and incubating inactivation, is the probable mechanism by which
at 37 °C for 1 hour. After a brief centrifugation to Micropure-EZ devices operate, since in separate
collect the condensate at the bottom of the micro- experiments we were unable to detect any enzyme
centrifuge tubes, 1 µL of 0.5 M disodium EDTA (irrespective of activity) in Micropure-EZ filtrates
and 10 µL of 5X loading buffer were added to using very sensitive HPLC methods (data not shown).
stop the reaction.
The negative control was a portion of DNA Summary
master mix. Control digests of pBR322 DNA and Enzyme removal and excellent DNA recovery are
pUC19 DNA were carried out separately. accomplished with Micropure-EZ devices in a single
60-second spin without any pre-wetting, binding,
Results and Discussion washing or elution step. Cumbersome phenol/
The Bgl I standard curve made with the Micropure- chloroform extraction and the associated hazardous
EZ-filtered sdH2O was indistinguishable from the waste accumulation are avoided. An additional line
standard curve made with untreated sdH2O. The of evidence for the suitability of Micropure-EZ
plasmid DNA (1 µg of pUC19 and 1 µg of pBR322) devices in molecular biology applications is that
was cut to completion by 2 units of Bgl I after one restriction digest purified with Micropure-EZ devices
hour at 37 °C (Figure 20.2, lanes 1 and 6). None alone are readily ligated and cloned into plasmid
of the enzymes shown in Tables 20.1 or 20.2 was DNA (M.H.A.L., unpublished observations).
measurably inhibited by Micropure-EZ-filtered Micropure-EZ devices used alone or with Microcon
sdH2O. The standard curves indicated that as little devices permit rapid and efficient sequential
as 0.08 units of Bgl I could be detected by this enzymatic DNA manipulations.
assay (lanes 3 and 8).
The devices removed the 50 units of Bgl I as
evidenced by an absence of detectable activity in
the filtrates. In the lanes corresponding to the Bgl I
challenged Micropure-EZ inserts, some activity

Figure 20.2 Analytical agarose gel, demonstrating the detection limits for Bgl I and Bgl I removal by Micropure-EZ devices.
Lanes 1–4: Serial dilutions of BgI I representing 2 units, 0.4 units, 0.08 units, and 0.016 units of activity (respectively) against the DNA
master mix prepared in non-filtered sdH2O
Lane 5: Undigested DNA master mix used as substrate for the standard curve containing pUC19 and pBR322 DNA
Lanes 6–9: Inhibition control: serial dilutions of BgI I representing 2 units, 0.,4 units, 0.08 units, and 0.016 units of activity (respectively)
against the DNA master mix prepared in Micropure-EZ-filtered sdH2O
Lane 10: Molecular weight standard: 1 kb DNA ladder (Gibco-BRL, Gaithersburg, MD)
Lanes 11–14: Filtrates incubated with
pUC19 DNA after challenging
Micropure-EZ devices with
50 units of BgI I, BSA and
decoy pBR322 DNA
Lane 15: Uncut pBR322
Lane 16: BgI I–cut pBR322
Lane 17: Uncut pUC19
Lane 18: BgI I–cut pUC19 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18

56
Table 20.1 Enzymes removed by Micropure-EZ devices

In tests, all enzymes were removed from solutions containing DNA or RNA (in the case of RNase A). Five µg of bovine serum
albumin were also present during removal of restriction enzymes. Removal was indicated by undetectable or inconsequential
enzyme activity in filtrate (< 0.08% residual in the case of T4 polynucleotide kinase).
Enzyme Challenge Enzyme Challenge Enzyme Challenge
AMV reverse transcriptase 50 U Apa I 100 U Mlu I 50 U
Calf intestinal alkaline 10 U BamH I 100 U Nco I 50 U
phosphatase Bcl I 50 U Nde I 100 U
DNase I (bovine pancreas) 10 U Bgl I 50 U NgoM I 50 U
Exonuclease III (E. coli) 100 U BsiW I 70 U Nhe I 25 U
MMLV reverse transcriptase 600 U BssH II 20 U Not I 50 U
Mung bean nuclease 50 U BstN I 50 U Nru I 50 U
Proteinase K (Amresco) 5 µg Dpn I 100 U Pst I 100 U
T4 DNA ligase 2,000 U* EcoR I 100 U Sac I 100 U
T4 DNA polymerase 15 U Hae III 100 U Sac II 100 U
T4 polynucleotide kinase** 50 U Hinc II 50 U Sal I 100 U
Taq DNA polymerase 5U Hind III 100 U Sca I 50 U
Terminal deoxynucleotidyl 45 U Hpa I 25 U Sph I 25 U
transferase Kpn I 50 U Sst I 50 U
Acc I 50 U Mbo I 25 U Xho I 100 U
*New England Biolabs unit definition
**T4 polynucelotide kinase is not recommended with oligonucleotides (dsDNA only). The results suggest that this kinase mediates the binding of
oligos to the membrane in Micropure-EZ device, causing oligo loss.

Table 20.2 Enzymes not removed by Micropure-EZ devices

In tests, Micropure-EZ did not remove the indicated number of units of the listed enzymes. It may be effective in removing a lower
number of units.
Enzyme Challenge Enzyme Challenge Enzyme Challenge
Bacterial alkaline phosphatase 0.6 U ApaL I 10 U Msp I 100 U
DNA polymerase I (Klenow) 20 U Bgl II 50 U Pvu I 25 U
Exonuclease I 50 U BsoB I 50 U Pvu II 50 U
Pfu DNA polymerase 2.5 U Cla I 25 U Sau3A I 20 U
Vent™ DNA polymerase 4U Eae I 15 U Sfi I 50 U
Shrimp alkaline phosphatase 1U EcoR V 50 U Sma I 25 U
RNase A (bovine pancreas) 1 µg Hinf I 50 U Xba I 50 U
T4 RNA ligase is not recommended. Results suggest this ligase mediates binding of nucleic acids to the membrane in Micropure-EZ causing sample loss.

57
Protocols • High Throughput

High Throughput Applications

Nucleic Acid Purification


and Concentration
AN1040EN00
The MultiScreen Filter Plate with Ultracel-10
membrane can be used to purify and concentrate
single-stranded oligonucleotides and double-
stranded DNA fragments, as well as plasmid and
genomic DNA.

Concentration of Proteins
in Cell Lysate
Figure 1. MultiScreen
AN1424EN00
Filter Plate with Ultracel-10
ultrafiltration membrane The MultiScreen filter plate with Ultracel-10
membrane can be used to concentrate whole cell
lysates without loss of protein and with high
reproducibility across the plate. Applications include
The MultiScreen filter plate with Ultracel-10 parallel protein purification, protein concentration
membrane provides a new method for high and buffer exchange in cell lysates for subsequent
throughput sample prep. The ultrafiltration-based separation or assaying.
filter plate is designed for use with centrifugation
and is compatible with standard microtiter plates, Purification of Serum Peptides
instrumentation, and liquid handling equipment. for Biomarkers Research
All of the publications summarized below AN2010EN00
can be supplied by your local Millipore office or This study is a high throughput version of the
downloaded from www.millipore.com/ultracel. application nate on page 24 of the Protocols
section of this handbook.

Enzymatic Activity Recovery


AN2011EN00
This study demonstrates the use of the MultiScreen
plate with Ultracel-10 membrane for concentrating
alkaline phosphatase without loss of biological
activity and with high reproducibility across
the plate.

58
Glossary
Asymmetric Membrane1 Fouling Retention Factor1 (rF)
Membrane constituted of two or more struc- Irreversible decline in membrane flux due to Parameter defined as one minus the ratio
tural planes of non-identical morphologies. deposition and accumulation of submicron of permeate concentration to the retentate
particles and solutes on the membrane concentration of a component. Note:
Batch Process
surface. Also, crystallization and precipita- r F = 1 – [p]/[r] where [p] = concentration
A fixed volume of solution contained in a tion of small solutes on the surface and in the of solute in permeate, and [r] = concentra-
tank to which the concentrate is returned pores of the membrane. Not to be confused tion of solute in retentate.
during the process. with concentration polarization.
Reverse Osmosis1
Composite Membrane1 Membrane1 Liquid-phase pressure-driven separation
Membrane having chemically or structurally Structure having lateral dimensions much process in which applied transmembrane
distinct layers. greater than its thickness, through which pressure causes selective movement of solvent
Concentration Polarization mass transfer may occur under a variety of against its osmotic pressure difference.
Accumulation of rejected solute on the driving forces.
Tangential Flow Filtration (TFF)
membrane surface. Depends on interactions Molecular Weight Cut-off (MWCO) Flow through a membrane device in which
of pressure, viscosity, crossflow (tangential) See Nominal Molecular Weight Limit. the fluid on the upstream side moves parallel
velocity, fluid flow conditions, flow channel
to the membrane surface.
conditions and temperatures. Nanofiltration1
Pressure-driven membrane-based separation Transmembrane Pressure (TMP)
Crossflow (Tangential Flow)
process in which particles and dissolved The driving force in ultrafiltration. In a stirred
Solution flows across (tangential to) a mem- molecules smaller than about 2 nm are cell, equivalent to gas pressure. In centrifugal
brane surface. Facilitates back diffusion of rejected. devices, it is related to g-force. In a flowing
solute from that surface into the bulk solution,
system, TMP decreases as the stream moves
counteracting concentration polarization. Nucleotide Cut-off (NCO)
from inlet to outlet. Average TMP =
The number of nucleotides in a DNA
Diafiltration [(Pin + Pout)/2] – Ppermeate
fragment (single- or double-stranded) at
Removal of small components from the which 90% of the fragment is retained by Ultrafiltration1
retained species during ultrafiltration by the membrane. Pressure-driven membrane-based separation
adding water or buffer solution to the
process in which particles and dissolved
retentate. See page 7 for further discussion. Nominal Molecular Weight Limit
molecules smaller than 0.1 µm and larger
(NMWL)
Dialysis than about 2 nm are rejected.
The molecular weight at which at least 90%
Diffusive transport of ions or other small of a globular solute of that MW is retained Yield
molecules through a membrane barrier by the membrane. Amount of species recovered at the end of
that contacts solvent on both sides.
the process as a percentage of the amount
Permeate (Filtrate, Ultrafiltrate)
Downstream1 present in the feed solution.
The solution passing through the membrane,
Side of a membrane from which the containing solvent and solutes not retained
permeate emerges. by the membrane. References
Feed (Sample) 1. Terminology for membranes and
Plugging membrane processes (IUPAC
The starting solution (sometimes the solution Accumulation of debris on the fluid flow path, Recommendations 1996). IUPAC,
remaining upstream of the membrane). restricting or blocking flow. Journal of Membrane Science.
Fluid Velocity Rejection 1996;120(1):149–59.
The flow rate of solution across the mem- The fraction of solute held back by the
brane surface in cross (tangential) flow. membrane. Can be measured at any point in
Related to hydraulic pressure drop. the process or averaged over the run.
Flux Retentate (Reject Stream, Concentrate)
The filtration rate through the membrane per The solution containing the retained (rejected)
unit area. species.

59
Acknowledgements
Millipore wishes to acknowledge the efforts of the following employees in developing
this reference book:

Elena Chernokalskaya, Ph.D.


Manager, Proteomics Applications

Mark Kavonian
Worldwide Group Product Manager
Protein Research Products

Richard Leary
Technical Service Specialist

Jack T. Leonard, Ph.D.


R&D Director, Life Science Research Applications

Peter Rapiejko

Millipore also wishes to acknowledge the following individuals for their contributions:
Eduardo Vottero, University of British Columbia
Peter A. Lemaire and Dr. James Cole, University of Connecticut
Gary Smejkal, Proteome Systems, Woburn, MA
Leonid Kryazhev, Genome Quebec, Montreal, Canada
Mathew L. Thakur, Thomas Jefferson University Hospital, Philadelphia, PA
Mark Merchant, Helena Laboratories, Beaumont, TX
Department of Forensic Biology, New York City
Patrick O. Brown, Max Diehn, Ash Alizadeh, Stanford University School of Medicine
Dr. Sophia N. Karagiannis, GKT School of Biomedical Sciences, London

60
Patent
PCR is covered by US patents issued to Hoffmann-LaRoche, Inc.

Trademarks
Millipore, Amicon, Biomax, Centricon, Centrilutor, Centriplus, Centriprep, Durapore, Immobilon,
Microcon, Micropure, Milli-Q, Minicon, Montage, MultiScreen, Pellicon, Prep/Scale, PROSEP, Ultracel,
Ultrafree, Zip, and ZipTip are trademarks of Millipore Corporation.
ProteoPrep is a trademark of Sigma-Aldrich Biotechnology LP.
ProteomIQ, ElectrophoretIQ, IsoelectrIQ, and GelChips are trademarks of Proteome Systems.
SpectraFLUOR is a trademark of Tecan Group AG.
DryStrips, Sephadex, Sephacryl, Sepharose, and Storm are trademarks of Amersham Biosciences AB.
CHEF-DR II and Chelex are trademarks of Bio-Rad Laboratories.
MEGAscript is a trademark of Ambion Inc.
GenePrint, Flexi and Riboprobe are trademarks of Promega Corporation.
SeaKem is a trademark of FMC.
AmpFLSTR and Voyager-DE are trademarks of Applera Corporation or a subsidiary.
Triton is a trademark of Union Carbide Chemicals & Plastics Technology Corporation.
Biofuge is a trademark of Kendro Laboratory Products, Gmbh.
NuPage, SimplyBlue, Superscript and Xcell Surelock are trademarks of Invitrogen Corporation.
Qiagen is a trademark of Qiagen Gmbh.
Speed Vac is a trademark of Thermo Savant.
BacPAK and Talon are trademarks of Becton, Dickinson and Company.
Vent is a trademark of New England Biolabs, Inc.
Coomassie is a trademark of Imperial Industries, PLC.
Tween is a trademark of Atlas Powder Company.
Additional Tools for Life Scientists
From Amicon® centrifugal devices to Mill-Q® water systems
Â
to Zip® micro-SPE technology, no one offers a broader range
of sample preparation tools than Millipore.

Blotting
Proteins
Bookmarks
The life sciences change every day. And so does
Immobilon™ are the most widely used PVDF
millipore.com. We’re continually adding new content
membranes for western, dot/slot, and other
and new functionality.
protein blots. Request a free copy of our
56-page Protein Blotting Handbook at
Millipore life sciences home page
www.millipore.com/forms.nsf/eblotting.
www.millipore.com
Nucleic acids (on the banner, under “Applications”)
Immobilon-Ny+ are positively charged nylon
membranes that provide reliable transfer, On-line ordering
hybridization, and reprobing with maximum www.millipore.com/purecommerce
sensitivity and minimal background for Southern
blotting, northern blotting, and colony/plaque lifts. Order tracking
www.millipore.com/MilliTrak
Sterile Filtration and
Chromatography Sample Prep
Membranes and devices for
sterile and non-sterile filtration
Millipore offers hundreds of membrane-based
devices for sterile filtration, chromatography sample
prep, and almost any other application in the life
sciences laboratory.

DNA Sequencing
Kits and devices for nucleic acid
sample prep Technical Support
Montage® life science kits use Millipore’s Millipore Technical Service Specialists support
size-exclusion technology for high quality ultrafiltration and many other life science applica-
plasmid mini-preps and PCR and sequencing tions, including blotting, DNA sequencing sample
reaction cleanup. prep, sterile filtration, and MS sample prep.
To contact a Specialist, call your local office
Laboratory Water Systems or submit a question at www.millipore.com/
Pure water solutions techservice. To access our library of frequently
Millipore provides total solutions from bench-top asked questions, go to www.millipore.com/faqs.
systems to custom-engineered purification chains
for laboratory buildings. You’ll find Millipore water
systems installed in over 70,000 laboratories
worldwide supplying pure water for electrophoresis,
PCR, chromatography, and other life science
applications.

Lit. No. TP0040EN00 Rev.– Printed in U.S.A. 04-337 12/04


© 2004 Millipore Corporation, Billerica, MA 01821 U.S.A.
All rights reserved.

You might also like