Magnetic techniques for the isolation and purification of proteins and peptides

Magnetic techniques for the isolation and purification of proteins and peptides

Ivo Safarik*1,2 and Mirka Safarikova1

Address:

1Laboratory of Biochemistry and Microbiology, Institute of Landscape Ecology, Academy of Sciences, Na Sadkach 7, 370 05 Ceske Budejovice, Czech Republic and

2Department of General Biology, University of South Bohemia, Branisovska 31, 370 05 Ceske Budejovice, Czech Republic

Email: Ivo Safarik* - ivosaf@yahoo.com; Mirka Safarikova - mirkasaf@uek.cas.cz

* Corresponding author

Abstract

Isolation and separation of specific molecules is used in almost all areas of biosciences and biotechnology. Diverse procedures can be used to achieve this goal. Recently, increased attention has been paid to the development and application of magnetic separation techniques, which employ small magnetic particles. The purpose of this review paper is to summarize various methodologies, strategies and materials which can be used for the isolation and purification of target proteins and peptides with the help of magnetic field. An extensive list of realised purification procedures documents the efficiency of magnetic separation techniques.

Introduction

Isolation, separation and purification of various types of proteins and peptides, as well as of other specific mole­cules, is used in almost all branches of biosciences and biotechnologies. Separation science and technology is thus very important area necessary for further develop­ments in bio-oriented research and technology. New sep­aration techniques, capable of treating dilute solutions or solutions containing only minute amounts of target mol­ecules in the presence of vast amounts of accompanying compounds in both small and large-scale processes, even in the presence of particulate matter, are necessary.

In the area of biosciences and biotechnology the isolation of proteins and peptides is usually performed using vari­ety of chromatography, electrophoretic, ultrafiltration, precipitation and other procedures, affinity chromatogra­phy being one of the most important techniques. Affinity ligand techniques represent currently the most powerful tool available to the downstream processing both in term of their selectivity and recovery. The strength of column affinity chromatography has been shown in thousands of successful applications, especially in the laboratory scale. However, the disadvantage of all standard column liquid chromatography procedures is the impossibility of the standard column systems to cope with the samples con­taining particulate material so they are not suitable for work in early stages of the isolation/purification process where suspended solid and fouling components are present in the sample. In this case magnetic affinity, ion-exchange, hydrophobic or adsorption batch separation processes, applications of magnetically stabilized fluid-ized beds or magnetically modified two-phase systems have shown their usefulness.

The basic principle of batch magnetic separation is very simple. Magnetic carriers bearing an immobilized affinity or hydrophobic ligand or ion-exchange groups, or mag­netic biopolymer particles having affinity to the isolated structure, are mixed with a sample containing target

compound(s). Samples may be crude cell lysates, whole blood, plasma, ascites fluid, milk, whey, urine, cultivation media, wastes from food and fermentation industry and many others. Following an incubation period when the target compound(s) bind to the magnetic particles the whole magnetic complex is easily and rapidly removed from the sample using an appropriate magnetic separator. After washing out the contaminants, the isolated target compound(s) can be eluted and used for further work.

Magnetic separation techniques have several advantages in comparison with standard separation procedures. This process is usually very simple, with only a few handling steps. All the steps of the purification procedure can take place in one single test tube or another vessel. There is no need for expensive liquid chromatography systems, cen­trifuges, filters or other equipment. The separation process can be performed directly in crude samples containing suspended solid material. In some cases (e.g., isolation of intracellular proteins) it is even possible to integrate the disintegration and separation steps and thus shorten the total separation time [1]. Due to the magnetic properties of magnetic adsorbents (and diamagnetic properties of majority of the contaminating molecules and particles present in the treated sample), they can be relatively easily and selectively removed from the sample. In fact, mag­netic separation is the only feasible method for recovery of small magnetic particles (diameter ca 0.1 - 1 цт) in the presence of biological debris and other fouling material of similar size. Moreover, the power and efficiency of mag­netic separation procedures is especially useful at large-scale operations. The magnetic separation techniques are also the basis of various automated procedures, especially magnetic-particle based immunoassay systems for the determination of a variety of analytes, among them pro­teins and peptides. Several automated systems for the sep­aration of proteins or nucleic acids have become available recently.

Magnetic separation is usually very gentle to the target proteins or peptides. Even large protein complexes that tend to be broken up by traditional column chromatogra­phy techniques may remain intact when using the very gentle magnetic separation procedure [2]. Both the reduced shearing forces and the higher protein concentra­tion throughout the isolation process positively influence the separation process.

Separation of target proteins using standard chromatogra­phy techniques often leads to the large volume of diluted protein solution. In this case appropriate magnetic parti­cles can be used for their concentration instead of ultrafil-tration, precipitation etc. [3].

The purpose of this review is to summarize various meth­odologies and strategies which can be employed for the isolation and purification of target proteins and peptides with the help of magnetic materials. An extensive list of realised purification procedures documents the efficiency of magnetic separation techniques. All these information will help the scientists to select the optimal magnetic material and the purification procedure.

Necessary materials and equipment

The basic equipment for laboratory experiments is very simple. Magnetic carriers with immobilized affinity or hydrophobic ligands, magnetic particles prepared from a biopolymer exhibiting affinity for the target compound(s) or magnetic ion-exchangers are usually used to perform the isolation procedure. Magnetic separators of different types can be used for magnetic separations, but many times cheap strong permanent magnets are equally effi­cient, especially in preliminary experiments.

Magnetic carriers and adsorbents can be either prepared in the laboratory, or commercially available ones can be used. Such carriers are usually available in the form of magnetic particles prepared from various synthetic poly­mers, biopolymers or porous glass, or magnetic particles based on the inorganic magnetic materials such as surface modified magnetite can be used. Many of the particles behave like superparamagnetic ones responding to an external magnetic field, but not interacting themselves in the absence of magnetic field. This is important due to the fact that magnetic particles can be easily resuspended and remain in suspension for a long time. In most cases, the diameter of the particles differs from ca 50 nm to approx. 10 |im. However, also larger magnetic affinity particles, with the diameters up to millimetre range, have been suc­cessfully used [4]. Magnetic particles having the diameter larger than ca 1 |im can be easily separated using simple magnetic separators, while separation of smaller particles (magnetic colloids with the particle size ranging between tens and hundreds of nanometers) may require the usage of high gradient magnetic separators.

Commercially available magnetic particles can be obtained from a variety of companies. In most cases pol­ystyrene is used as a polymer matrix, but carriers based on cellulose, agarose, silica, porous glass or silanized mag­netic particles are also available. Examples of magnetic particles used (or usable) for proteins and peptides sepa­ration can be found elsewhere [5-7].

Particles with immobilised affinity ligands are available for magnetic affinity adsorption. Streptavidin, antibodies, protein A and Protein G are used most often in the course of protein and peptides isolation. Magnetic particles with above mentioned immobilised ligands can also serve as generic solid phases to which native or modified affinity ligands can be immobilised (e.g., antibodies in the case of immobilised protein A, protein G or secondary antibod­ies, biotinylated molecules in the case of immobilised strep tavidin).

Also some other affinity ligands (e.g., nitrilotriacetic acid, glutathione, trypsin, trypsin inhibitor, gelatine etc.) are already immobilised to commercially available carriers. To immobilise other ligands of interest to both commer­cial and laboratory made magnetic particles standard pro­cedures used in affinity chromatography can be employed. Usually functional groups available on the sur­face of magnetic particles such as -COOH, -OH or -NH2 are used for immobilisation, in some cases magnetic par­ticles are available already in the activated form (e.g., tosy-lactivated, epoxyactivated etc).

In the laboratory magnetite (or similar magnetic materials such as maghemite or ferrites) particles can be surface modified by silanization. This process modifies the sur­face of the inorganic particles so that appropriate func­tional groups become available, which enable easy immobilisation of affinity ligands [8]. In exceptional cases enzyme activity can be decreased as a result of usage of magnetic particles with exposed iron oxides. In this case encapsulated microspheres, having an outer layer of pure polymer, will be safer.

Biopolymers such as agarose, chitosan, kappa carrageenan and alginate can be easily prepared in a magnetic form. In the simplest way the biopolymer solution is mixed with magnetic particles and after bulk gel formation the mag­netic gel formed is mechanically broken into fine particles [9]. Alternatively biopolymer solution containing dis­persed magnetite is dropped into a mixed hardening solu­tion [4] or water-in-oil suspension technique is used to prepare spherical particles [10].

Basically the same procedures can be used to prepare mag­netic particles from synthetic polymers such as polyacryla-mide, poly(vinylalcohol) and many others [11].

In another approach used standard affinity or ion-exchange chromatography material was post-magnetised by interaction of the sorbent with water-based ferrofluid. Magnetic particles accumulated within the pores of chro­matography adsorbent thus modifying this material into magnetic form [12,13]. Alternatively magnetic Sepharose or other agarose gels were prepared by simple contact with freshly precipitated or finely powdered magnetite [ 12,14].

Magnetoliposomes (magnetic derivatives of standard liposomes), either in the original form or after immobili­zation of specific proteins, have the potential for the separation of antiphospholipid antibodies [15], IgG antibodies [16] and other proteins of interest [17].

Recently also non-spherical magnetic structures, such as magnetic nanorods have been tested as possible adsorb­ent material for specific separation of target proteins [18].

Magnetic separators are necessary to separate the mag­netic particles from the system. In the simplest approach, a small permanent magnet can be used, but various mag­netic separators employing strong rare-earth magnets can be obtained at reasonable prices. Commercial laboratory scale batch magnetic separators are usually made from magnets embedded in disinfectant-proof material. The racks are constructed for separations in Eppendorf micro-tubes, standard test tubes or centrifugation cuvettes, some of them have a removable magnetic plate to facilitate easy washing of separated magnetic particles. Other types of separators enable separations from the wells of micro titra-tion plates and the flat magnetic separators are useful for separation from larger volumes of suspensions (up to approx. 500 - 1000 ml). Examples of typical batch mag­netic separators are shown in Fig. 1.

Flow-through magnetic separators are usually more expensive, and high gradient magnetic separators (HGMS) are the typical examples. Laboratory scale HGMS is composed from a column packed with fine magnetic grade stainless steel wool or small steel balls which is placed between the poles of an appropriate magnet. The suspension is pumped through the column, and magnetic particles are retained within the matrix. After removal the column from the magnetic field, the particles are retrieved by flow and usually by gentle vibration of the column.

For work in dense suspensions, open gradient magnetic separators may be useful. A very simple experimental set­up for the separation of magnetic affinity adsorbents from litre volumes of suspensions was described [19].

Currently many projects require the analysis of a high number of individual proteins or variants. Therefore, methods are required that allows multiparallel processing of different proteins. There are several multiple systems for high throughput nucleic acid and proteins preparation commercially available. The most often used approach for proteins isolation is based on the isolation and assay of 6xHis-tagged recombinant proteins using magnetic beads with Ni-nitriloacetic acid ligand [20]. The commercially available platforms can be obtained from several compa­nies such as Qiagen, USA (BioRobot and BioSprint series), Tecan, Japan (Te-MagS) or Thermo Electron Corporation, USA(KingFisher).

Figure 1

Examples of batch magnetic separators applicable for magnetic separation of proteins and peptides. A: Dynal MPC -S for six microtubes (Dynal, Norway); B: Dynal MPC - 1 for one test tube (Dynal, Norway); C: Dynal MPC - L for six test tubes (Dynal, Norway); D: magnetic separator for six Eppendorf tubes (New England BioLabs, USA); E: MagneSphere Technology Magnetic Separation Stand, two position (Promega, USA); F: MagnaBot Large Volume Magnetic Separation Device (Promega, USA); G: MagneSphere Technology Magnetic Separation Stand, twelve-position (Promega, USA); H: Dynal MPC - 96 S for 96-well microtitre plates (Dynal, Norway); I: MagnaBot 96 Magnetic Separation Device for 96-well microtitre plates (Promega, USA); J: BioMag Solo-Sep Microcentrifuge Tube Separator (Polysciences, USA); K: BioMag Flask Separator (Polysciences, USA); L: Mag-neSil Magnetic Separation Unit (Promega, USA); M: MCB 1200 processing system for 12 microtubes based on MixSep process (Sigris Research, USA); N: PickPen magnetic tool (Bio-Nobile, Finland). Reproduced with the permission of the above men­tioned companies; the photos were taken from their www pages.

 

Basic principles of magnetic separations of proteins and peptides

Magnetic separations of proteins and peptides are usually convenient and rapid. Nevertheless, several hints may be helpful to obtain good results.

Proteins and peptides in the free form can be directly iso­lated from different sources. Membrane bound proteins have to be usually solubilized using appropriate deter­gents. When nuclei are broken during sample preparation, DNA released into the lysate make the sample very vis­cous. This DNA may be sheared by repeated passage up and down through a 21 gauge hypodermic syringe needle before isolation of a target protein. Alternatively, DNase can be added to enzymatically digest the DNA.

Magnetic beads in many cases exhibit low non-specific binding of non-target molecules present in different sam­ples. Certain samples may still require preclearing to remove molecules which have high non-specific binding activity. If preclearing is needed, the sample can be mixed with magnetic beads not coated with the affinity ligand. In the case of immunomagnetic separation, magnetic beads coated with secondary antibody or with irrelevant anti­bodies have been used. The non-specific binding can also be minimised by adding a non-ionic detergent both in the sample and in the washing buffers after isolation of the target.

In general, magnetic affinity separations can be performed in two different modes. In the direct method, an appropri­ate affinity ligand is directly coupled to the magnetic par­ticles or biopolymer exhibiting the affinity towards target compound(s) is used in the course of preparation of mag­netic affinity particles. These particles are added to the sample and target compounds then bind to them. In the indirect method the free affinity ligand (in most cases an appropriate antibody) is added to the solution or suspen­sion to enable the interaction with the target compound. The resulting complex is then captured by appropriate magnetic particles. In case antibodies are used as free affinity ligands, magnetic particles with immobilised sec­ondary antibodies, protein A or protein G are used for capturing of the complex. Alternatively the free affinity ligands can be biotinylated and magnetic particles with immobilised streptavidin or avidin are used to capture the complexes formed. In both methods, magnetic particles with isolated target compound(s) are magnetically sepa­rated and then a series of washing steps is performed to remove majority of contaminating compounds and parti­cles. The target compounds are then usually eluted, but for specific applications (especially in molecular biology, bioanalytical chemistry or environmental chemistry) they can be used still attached to the particles, such as in the case of polymerase chain reaction, magnetic ELISA etc.

The two methods perform equally well, but, in general, the direct technique is more controllable. The indirect procedure may perform better if affinity ligands have poor affinity for the target compound.

In most cases, magnetic batch adsorption is used to per­form the separation step. This approach represents the simplest procedure available, enabling to perform the whole separation in one test-tube or flask. If larger mag­netic particles (with diameters above ca 1 цш) are used, simple magnetic separators can be employed. In case mag­netic colloids (diameters ranging between tens and hun­dreds of nanometres) are used as affinity adsorbents, high-gradient magnetic separators have usually to be used to remove the magnetic particles from the system.

Alternatively magnetically stabilised fluidised beds (MSFB), which enable a continuous separation process, can be used. The use of MSFB is an alternative to conven­tional column operation, such as packed-bed or fluidised bed, especially for large-scale purification of biological products. Magnetic stabilisation enables the expansion of a packed bed without mixing of solid particles. High col­umn efficiency, low pressure drop and elimination of clogging can be reached [21,22].

Also non-magnetic chromatographic adsorbents can be stabilized in magnetically stabilized fluidized beds if suf­ficient amount of magnetically susceptible particles is also present. The minimum amount of magnetic particles nec­essary to stabilize the bed is a function of various param­eters including the size and density of both particles, the magnetic field strength, and the fluidization velocity. A variety of commercially available affinity, ion-exchange, and adsorptive supports can be used in the bed for contin­uous separations [23].

Biocompatible two phase systems, composed for example from dextran and polyethylene glycol, are often used for isolation of biologically active compounds, subcellular organelles and cells. One of the disadvantages of this sys­tem is the slow separation of the phases when large amounts of proteins and cellular components are present. The separation of the phases can be accelerated by the addition of fine magnetic particles or ferrofluids to the system followed by the application of a magnetic field. This method seems to be useful when the two phases have very similar densities, the volumetric ratio between the phases is very high or low, or the systems are viscous. Mag­netically enhanced phase separation usually increases the speed of phase separation by a factor of about 10 in well-behaved systems, but it may increase by a factor of many thousands in difficult systems. The addition of ferrofluids and/or iron oxide particles was shown to have usually no influence on enzyme partioning or enzyme activity [24,25].

Table 1: Examples of proteinases and peptidases purified by magnetic techniques

Purified enzyme

Source

Magnetic carrier

Affinity ligand

Further details

Reference

Aminopeptidase

Arabidopsis

Amine-terminated magnetic beads

N-1 -Naphthylphthalamic acid

KCl gradient elution

[54]

Angiotensin- converting enzyme

Pig lung membranes

Dynabeads

Polyclonal antibodies

 

[57]

Bromelain

Commercial preparation

Polyacrylic acid - iron oxide magnetic nanoparticles

 

Elution with KCl solution

[59]

Caspase(recombinant, histidine-tailed)

Human cells

Magnetic agarose

Ni-NTA

Elution with SDS-PAGE buffer

[60]

Chymotrypsin

Commercial preparation

Magnetic chitosan beads

 

Elution with N-acetyl-D- tryptophan

[62]

N Ia-protease(recombinant, histidine- tagged)

Plum Pox Virus

Magnetic core and nickel- silica composite matrix

Ni2+

Elution with imidazole containing buffer

[36]

Proteinases

Commercial sources

Magnetic cross-linked erythrocytes

 

Elution with low pH buffer

[46]

Proteinase, bacterial (Savinase)

Bacillus clausii

Silanized magnetite particles

Bacitracin

 

[84]

Trypsin

Porcine pancreatin

Silanized magnetite particles

p-Aminobenzamidine

Elution with low pH buffer

[50]

 

Porcine pancreatin

Magnetic polymer particles

Soybean trypsin inhibitor

Elution with low pH solution

[86]

 

Commercial preparation

Silanized ferrite powder

Soybean trypsin inhibitor

 

[87]

 

Commercial preparation

Magnetic к-carrageenan particles

Soybean trypsin inhibitor

Elution with low pH solution, MSFB

[88] [89]

 

Commercial preparation

Magnetic polyacrylamide beads

Soybean trypsin inhibitor

Magnetically stabilized fluidized beds

[90]

 

Commercial preparation

Magnetic chitosan particles

Aprotinin

Elution with low pH solution

[91]

 

Commercial preparation

Magnetic cross-linked erythrocytes

 

Elution with low pH buffer; separation from large volume sample

[19]

Urokinase

Crude urokinase preparation

Magnetic dextran, agarose, polyvinyl alcohol, polyhydroxyethyl methacrylate microspheres

p-Aminobenzamide, L- arginine methyl ester, guanidine hexanoic acid or guanidine acetic acid

 

[93]

 

Proteins and peptides isolated using magnetic techniques have to be usually eluted from the magnetic separation materials. In most cases bound proteins and peptides can be submitted to standard elution methods such as the change of pH, change of ionic strength, use of polarity reducing agents (e.g., dioxane or ethyleneglycol) or the use of deforming eluents containing chaotropic salts. Affinity elution (e.g., elution of glycoproteins from lectin coated magnetic beads by the addition of free sugar) may be both a very efficient and gentle procedure.

Examples of magnetic separations of proteins and peptides

Magnetic affinity and ion-exchange separations have been successfully used in various areas, such as molecular biol­ogy, biochemistry, immunochemistry, enzymology, ana­lytical chemistry, environmental chemistry etc [26-29]. Tables 1, 2, 3, 4, 5, 6, 7, 8, 9 show some selected applica­tions of these techniques for proteins and peptides isolation.

In the case of proteins and peptides purifications, no sim­ple strategy for magnetic affinity separations exists. Vari­ous affinity ligands have been immobilised on magnetic particles, or magnetic particles have been prepared from biopolymers exhibiting the affinity for target enzymes or lectins. Immunomagnetic particles, i.e. magnetic particles with immobilised specific antibodies against the target structures, have been used for the isolation of various anti­gens, both molecules and cells [5] and can thus be used for the separation of specific proteins.

Table 2: Purification of lysozyme by magnetic techniques

Purified enzyme

Source

Magnetic carrier

Affinity ligand

Further details

Reference

Lysozyme

Hen egg white

Magnetic chitin

 

Elution with 0.01 M HCl

[71]

 

Hen egg white

Magnetic acetylated chitosan

 

Elution with 0.01 M HCl

CT

 

Commercial preparation

Magnetic poly(2- hydroxyethyl methacrylate)

Cibacron Blue F3GA

Elution with 1 M KSCN

[72]

 

 

Magnetic chitosan beads

 

Magnetically stabilized fluidized bed

[73]

 

Ornithodoros moubata

Magnetic chitin

 

Elution with alkaline, high salt buffer

[74]

 

Commercial preparation

Magnetic cross-linked polyvinyl alcohol

Cibacron blue 3GA

Elution with high salt buffer

[52]

 

 

Magnetite - polyacrylic acid nanoparticles

 

Ion-exchange separation

[75]

 

 

Magnetic cross-linked polyvinylalcohol beads

 

Adsorption study

[76]

 

Commercial preparation

Magnetic agarose beads

Cibacron blue 3GA

Magnetically stabilized fluidized bed

[77]

 

 

Magnetic chitosan

Cibacron blue 3GA

Study of adsorption properties

[78]

 

Commercial preparation

Ferrofluid modified sawdust

 

Elution with 0.5 M NaCl

[79]

 

Commercial preparation

Nano-sized magnetic particles

 

Elution with NaH2PO4 and NaSCN

[80]

Lysozyme (recombinant, histidine-tailed)

T4

BioMag, amine terminated

Iminodiacetic acid charged with Cu2+

Elution with low pH buffer

[81]

Table 3: Examples of polysaccharide and disaccharide hydrolases purified by magnetic techniques

Purified enzyme

Source

Magnetic carrier

Affinity ligand

Further details

Reference

a-Amylases

Porcine pancreas,

Magnetic alginate

 

Elution with 1 M

И

 

Bacillus subtilis, wheat germ

beads

 

maltose

 

 

Bacillus amyloliquefaciens, porcine pancreas

Magnetic alginate microbeads

 

Elution with 1 M maltose

[10]

P-Amylase

Sweet potato

Magnetic alginate beads

 

Elution with 1 M maltose

[55]

P-Galactosidase

Escherichia coli homogenate

Silanized magnetite

p-Aminophenyl-P-D- thiogalactopyranoside

Elution with borate buffer, pH 10

[58]

P-Galactosidase (fusion protein comprising the DNA-binding lac repressor)

Bacterial lysate

Magnetic beads

DNA containing Escherichia coli lac operator

Elution with lactose analogue

[64]

Glucoamylase

Aspergillus niger

Magnetic alginate beads

 

Elution with 1 M maltose

[55]

Pectinase

Commercial preparation

Magnetic alginate beads

 

 

[82]

Pullulanase

Bacillus acidopullulyticus

Magnetic alginate beads

 

Elution with 1 M maltose

[55]

Table 4: Examples of other enzymes purified by magnetic techniques

Purified enzyme

Source

Magnetic carrier

Affinity ligand

Further details

Reference

Alcohol dehydrogenase

Yeast homogenate

Magnetic cross-linked polyvinylalcohol

Cibacron blue 3GA

Elution with high salt buffer

[52]

 

Saccharomyces cerevisiae extract

 

PEG with bound Cibacron blue

Magnetic two-phase system

[53]

Aldolase (recombinant, histidine tagged)

Pea

Magnetic core and nickel-silica composite matrix

Ni2+

Elution with imidazole containing buffer

[36]

Angiol-TEM-P- lactamase

Escherichia coli cells extracts

Magnetic agarose beads

Iminodiacetic acid charged with Zn2+

Elution with low pH buffer

[56]

Asparaginase

Escherichia coli homogenate

Magnetic polyacrylamide gel particles

D-Asparagine

Elution with D- asparagine solution

[58]

Carbonic anhydrase

Model mixture

Magnetic agarose beads

Sulfanilimide

Elution with high salt buffer

[14]

Catalase

Bovine liver, commercial preparation

Magnetic poly(EGDMA-MAH) beads

Fe3+

Elution with NaSCN solution

[61]

Cytochrome c

Horse, Candida krusei

Amine terminated iron oxide particles

Iminodiacetic acid charged with Cu2+

Binding studies

[63]

 

Commercial preparation

Au@magnetic particles

 

MALDI MS analysis

[31]

 

Horse heart

Magnetic agarose beads

Iminodiacetic acid charged with Cu2+

Elution with EDTA containing buffer

[56]

 

Bovine heart

Magnetic ion- exchange particles

 

Protein binding studies

[12]

Glucose-6-phosphate dehydrogenase

 

Ferrofluid modified Sepharose 4B

ADP

 

[65]

 

Saccharomyces cerevisiae extract

 

PEG with bound Cibacron blue

Magnetic two-phase system

[53]

Hexokinase

Escherichia coli homogenate

 

PEG with bound Cibacron blue

Magnetic two-phase system

[53]

Lactate dehydrogenase

Beef heart

Ferrofluid modified Sepharose 4B

AMP

Elution with 1 mM NADH

[13]

 

Porcine muscle

Magnetic agarose beads

Reactive Red 120

Column elution with NaCl gradient

[66]

Lactoperoxidase

Sweet whey

Magnetic cation exchanger

 

HGMS

[67,68]

Luciferase (histidine- tagged)

Escherichia coli homogenate

MagneHis™ system

Ni2+

 

[69,70]

Phosphatase, alkaline

Human placenta

Dynabeads M-450

Specific antibody

Activity of bound enzyme measured

[83]

Phosphatase, alkaline(fusion protein comprising the DNA- binding lac repressor)

Bacterial lysate

Magnetic beads

DNA containing Escherichia coli lac operator

Elution with lactose analogue

[64]

Phosphofructokinase

Saccharomyces cerevisiae extract

 

PEG with bound Cibacron blue

Magnetic two-phase system

[53]

6-Phosphogluconate dehydrogenase

 

Ferrofluid modified Sepharose 4B

ADP

Elution with 1 mM NADP

[13]

Thioredoxin(recombinant, histidine-tagged)

Escherichia coli

Magnetic agarose

Ni-NTA

Elution with imidazole containing buffer

[20]

tRNA methionyl synthetase(recombinant, histidine-tagged)

Escherichia coli

MagneHis™ system

 

Rapid detection and quantitation of isolated protein

[85]

Uricase (recombinant, histidine-tailed)

Bacillus

Ion-chelating magnetic agarose beads

Ni2+

Elution by cleavage with proteinase K

[92]

 

Magnetic separation procedures can be employed in sev­eral ways. Preparative isolation of the target protein or peptide is usually necessary if further detailed study is intended. In other cases, however, the magnetic separa­tion can be directly followed (after elution with an appro­priate buffer) with SDS electrophoresis. Magnetically separated proteins and peptides can also be used for fur­ther mass spectroscopy characterization [30,31]. The basic principles of magnetic separations can be used in the course of protein or peptide determination using various types of solid phase immunoassays. Usually immu-nomagnetic particles directly capture the target analyte, or magnetic particles with immobilised streptavidin are used to capture the complex of biotinylated primary antibody and the analyte. The separated analyte is then determined (usually without elution) using an appropriate method. A combination of magnetic separation with affinity capil­lary electrophoresis is also possible [32].

Enzyme isolation is usually performed using immobilised inhibitors, cofactors, dyes or other suitable ligands, or magnetic beads prepared from affinity biopolymers can be used (see Tables 1, 2, 3, 4).

Table 5: Examples of antibodies purified by magnetinetic techniques

Purified antibody

Source

Magnetic carrier

Affinity ligand

Further details

Reference

Anti-BODIPY- fluorescein antibodies

 

Magnetoliposomes

BODIPY-fluorescein

 

[94]

Anti-DNA antibody

Systemic lupus erythematosus patient plasma

Magnetic poly(2-hydroxyethyl-methacrylate) beads

DNA

Desorption with 1 M NaSCN solution

[95]

Anti-human chorionic gonadotropin antibody

Murine ascites supernatants

Magnetic cellulose beads

Human chorionic gonadotropin

 

[96]

Antibody (from rat)

Sample from affinity chromatography

Dynabeads M-280

Sheep anti-rabbit IgG

Antibody concentration

[3]

Antibody

Rabbit serum

Dynabeads M-280

Sheep anti-rabbit IgG

Elution with 0.5 M acetic acid

[97]

Monoclonal antibodies

Mouse hybridoma culture broth

Magnetite particles

Protein A

 

[98]

Anti-bovine serum albumin antibodies

 

Thermosensitive magnetic microspheres

Bovine serum albumin

Immobilization by the carbodiimide method

[99]

Immunoglobulin G, human

Commercial preparation

Magnetic poly(ethylene glycol dimethacrylate-N-methacryloly-L-histidine-methylester) beads

 

Elution with 1 M NaCl

[100]

Immunoglobulin G

Blood serum

Carboxyl-terminated magnetic particles

MproteinAG

 

[101]

IgE antibodies

Allergic patients sera

Magnetoliposomes

Antigenic proteins

 

[16]

Murine anti-fibroblast growth factor receptor 1 IgM

Ascites

Polystyrene magnetic beads

Rat anti-mouse IgM monoclonal antibody

 

[102]

Genetic engineering enables the construction of gene fusions resulting in fusion proteins having the combined properties of the original gene products. To date, a large number of different gene fusion systems, involving fusion partners that range in size from one amino acid to whole proteins, capable of selective interaction with a ligand immobilized onto magnetic particles or chromatography matrices, have been described. In such systems, different types of interactions, such as enzyme-substrate, receptor-target protein, polyhistidines-metal ion, and antibody-antigen, have been utilized. The conditions for purifica­tion differ from system to system and the environment tolerated by the target protein is an important factor for deciding which affinity fusion partner to choose. In addi­tion, other factors, including protein localization, costs for the affinity matrix and buffers, and the possibilities of removing the fusion partner by site-specific cleavage, should also be considered [33,34]. As an example, isola­tion of recombinant oligohistidine-tagged proteins is based on the application of metal chelate magnetic adsorbents [35,36]. This method has been used success­fully for the purification of proteins expressed in bacterial, mammalian, and insect systems.

Antibodies from ascites, serum and tissue culture superna-tants can be efficiently isolated using magnetic particles with immobilized Protein A, Protein G or anti-immunoglobulin antibodies. Protein A, isolated from Staphylo-coccus aureus, binds the Fc region of IgG of most mammalian species with high affinity, leaving antigen specific sites free. Protein G, isolated from Streptococcus sp., reacts with a larger number of IgG isotypes. It has a higher binding affinity to immunoglobulins than Protein A, however, it also interacts with the Fab regions of IgG, although the affinity is ten times lower than for the Fc region [37].

Table 6: Examples of DNA/RNA/oligonucleotide/aptamer binding proteins purified by magnetic techniques

Purified protein

Source

Magnetic carrier

Affinity ligand

Further details

Reference

CUG binding proteins

fibroblasts

Dynabeads M-280 streptavidin

Biotinylated(CUG)10

Elution with 1 M NaCl

[103]

Transcription factor x

Saccharomyces cerevisiae

Dynabeads M-280 streptavidin

Biotinylated tRNAGlu gene fragment

Elution with high salt buffer

[104,105]

DNA-binding proteins

Crude tissue extract

Magnetic phospho cellulose particles

 

 

[106]

DNA-binding proteins

Escherichia coli

Magnetic phospho cellulose particles

 

 

[107]

DNA-binding proteins

HeLa nuclear extracts

Dynabeads M-280 streptavidin

Biotin-labelled DNA fragment

Elution with 2 M NaCl

[108]

Vaccinia virus early transcription factor

Vaccinia virions

Dynabeads M-280 streptavidin

Biotinylated double- stranded DNA

Elution with high salt buffer

[109]

Ecdysteroid receptor

Drosophila melanogaster nuclear extract

Dynabeads M-280 streptavidin

Biotinylated double- stranded oligonucleotide

Elution with 0.4 M KCl

[110]

NanR protein(recombinant)

Escherichia coli

U.MACS streptavidin MicroBeads

Biotin-labelled DNA fragment

Elution with 1 M NaCl

[111]

p27

Rabbit hepatocytes

Dynabeads M-280 streptavidin

Guanine-rich single- stranded DNA

Elution with NaCl solution

[112]

Pigpen protein

Endothelial cells

Magnetic streptavidin beads

Biotinylated aptamer

Elution with 1 M NaCl

[113]

RNA binding proteins

Saccharomyces cerevisiae

U.MACS streptavidin MicroBeads

Biotin-labelled RNA probe

Elution with 1 M NaCl

[114]

Single-stranded telomere binding protein (sTBP)

Nuclei from vertebrate tissues

Dynabeads M-280 streptavidin

Biotinylated single stranded TTAGGGn repeats

Elution with high salt buffer

[115]

Transcription proteins

Human myeloid cells

Dynabeads M-280 streptavidin

Biotinylated serum inducible element (hSIE)

Elution with high salt buffer

[116]

Transcription factor yRF-l

Human monocytes and epidermal cells

Dynabeads M-280 streptavidin

Biotinylated DNA containing yRF-l sequences

Elution with 0.6 M KCl

[117]

Protein factor MS2

Murine skeletal myotubes

Dynabeads

Double-stranded DNA

Elution with 100 mM sodium acetate, pH 4.2

[118]

Guide RNA binding protein

Trypanosoma brucei mitochondria

Dynabeads M-450 goat anti-mouse IgG

Monoclonal antibody

Elution with low pH buffer cont. SDS

[119]

RNA binding proteins

Pollen grains

Streptavidin MagneSphere particles

Biotinylated oligonucleotides

Elution with SDS buffer

[120]

DNA binding protein

Schistosoma mansoni

Dynabeads M-280 streptavidin

Biotinylated DNA

Elution with sodium acetate buffer

[121]

ssDNA binding proteins

Transfected mouse fibroblasts

Dynabeads anti-rabbit IgG

Rabbit antibody

Indirect method

[122]

Tenascin-C

Glioblastoma cells

Dynabeads streptavidin

Biotinylated aptamer

Elution with high salt buffer

[123]

Thermostable brain factor (ThBF)

Rat brain

Streptavidin magnetic particles

Biotinylated oligonucleotides

Elution with 0.7 M KCl

[124]

TTF1 protein

Escherichia coli lysate

Dynabeads M-280 streptavidin

Biotinylated aptamer

Elution with DNase

[125]

Antiphospholipid antibodies were success­fully isolated using magnetoliposomes [15].

Aptamers are DNA or RNA molecules that have been selected from random pools based on their ability to bind other molecules. Aptamers binding proteins can be immobilised to magnetic particles and used for isolation of target proteins.

Table 7: Purification of albumin and haemoglobin by magnetic techniques

Purified protein

Source

Magnetic carrier

Affinity ligand

Further details

Reference

Albumin, bovine serum

Commercial preparation

Magnetic agar beads

Cibacron blue3GA

Adsorption experiments

[126]

 

Commercial preparation

Magnetic cross-linked polyvinylalcohol

Cibacron blue3GA

Adsorption experiments

[76,127]

 

 

Magnetic chitosan microspheres

Cibacron blue3GA

 

[78]

 

Commercial preparation

Magnetic poly(glycidyl methacrylate-triallyl isocyanurate-divinylbenzene) particles

 

Anion exchange separation

[128]

 

Commercial preparation

Magnetic poly(ethylene glycol dimethacrylate-co-N- methacryloyl-(L)- histidine methylester) microbeads

Cu2+

Elution with 1.0 M NaSCN

[129]

Albumin, human serum

Commercial preparation

Magnetic poly(2- hydroxyethylmethacrylate) beads

Iminodiacetic acid charged with Cu2+

Elution with 1.0 M NaSCN

[130]

 

Human plasma

Magnetic poly(2-hydroxyethyl methacrylate) beads

Cibacron blue F3GA

Elution with 0.5 M NaSCN

[131]

 

Commercial preparation

Magnetic particles covered with thermosensitive polymer

-

Desorption by decreasing temperature

[132,133]

Albumin, human serum (recombinant, FLAG tagged)

Yeast cells

Magnetic glass beads

Anti-FLAG antibody

Elution with EDTA containing buffer

[1]

Glycated haemoglobin

Human blood

Magnetic poly(vinyl alcohol) beads

m-Aminophenyl- boronic acid

Elution with sorbitol

[138]

Haemoglobin

Bovine, commercial preparation

Amine terminated iron oxide particles

Iminodiacetic acid charged with Cu2+

Elution with imidazole containing buffer

[63]

Haemoglobin A1cHumanblo

Human blood

Magnetic particles isolated from Magnetospirillum magneticum AMB-1

m-Aminophenyl- boronic acid

used for affinity immunoassay

[150]

DNA/RNA binding proteins (e.g., promoters, gene regula­tory proteins and transcription factors) are often short­lived and in low abundance. A rapid and sensitive method, based on the immobilization of biotinylated DNA/RNA fragments containing the specific binding sequence to the magnetic streptavidin particles, can be used. The bound DNA/RNA binding proteins are usually eluted with high salt buffer or change of pH [38].

Other types of proteins were isolated using specific affin­ity-based procedures. For example, plasminogen immobi­lized on magnetic particles was used to separate scrapie and bovine spongiform encephalopathy associated prion protein PrPSc from its conformer which is a cellular pro­tein called PrPC. In fact, plasminogen represents the first endogenous factor discriminating between normal and pathological prion protein. This unexpected property may be exploited for diagnostic purposes [39,40].

Magnetic separation was also successfully used for the recovery of proteins expressed in the form of inclusion bodies, involving at first chemical extraction from the host cells, then adsorptive capture of the target protein

onto small magnetic adsorbents, followed by rapid collection of the product-loaded supports with the aid of high gradient magnetic fields [41].

A new approach for analytical ion-exchange separation of native proteins and proteins enzymatic digest products has been described recently [31]. Magnetite particles were covered with a gold layer and then stabilized with ionic agents. These charged stabilizers present at the surface of the gold particles are capable of attracting oppositely charged species from a sample solution through electro­static interactions. Au@magnetic particles having nega­tively charged surfaces are suitable probes for selectively trapping positively charged proteins and peptides from aqueous solutions. The species trapped by the isolated particles were then characterized by matrix-assisted laser desorption/ionization mass spectrometry (MALDI MS) after a simple washing.

Magnetic solid phase extraction (MSPE) enables to pre-concentrate target analytes from larger volumes of solutions or suspensions using relatively small amount of magnetic specific adsorbent. Up to now this procedure was used for preconcentration of low-molecular weight xenobiotics [42,43] but using suitable magnetic adsorbents the MSPE could be used to preconcetrate target proteins and peptides as well.

Table 8: Examples of other proteins purified by magnetic techniques

Purified protein

Source

Magnetic carrier

Affinity ligand

Further details

Reference

Aprotinin

Bovine pancreatic powder

Magnetic chitosan particles

Trypsin

Elution with low pH buffer

[134]

Concanavalin A

Jack bean extract

Magnetic particles

Dextran

 

[68,135]

Solanum tuberosum lectin

Potato tuber

Magnetic chitosan

 

Elution with low pH buffer

[136]

Green fluorescent protein (histidine tagged)

 

Magnetic nanoparticles

Ni-NTA

Elution with imidazole containing buffer

[137]

SIRT2 protein(recombinant, histidine tailed)

Human

Magnetic agarose beads

Ni-NTA

Elution with imidazole containing buffer

[139]

Elongation factor(recombinant, histidine tailed)

Caenorhabditis elegans

Magnetic agarose beads

Ni-NTA

Elution with imidazole containing buffer

[140]

Protein A

Recombinant Escherichia coli

Magnetic Eudragit

Human IgG

Magnetic two-phase system

[141]

Tumor necrosis factor(TNF)

 

Dynabeads M-280

Mouse monoclonal antibody

Solid phase immunoassay

[142]

Anti-MUC1 diabody fragment

Recombinant Escherichia coli

Magnetic agarose beads

Specific peptide

 

[143]

MHC class II molecules

MDCK cells

Dynabeads M-450 rat anti-mouse IgG 1

Specific antibodies

Elution with SDS-PAGE buffer

[144]

Lamin B3

Xenopus egg extracts

Dynabeads

Specific antibodies

Elution with 6 M urea

[44]

6x-His-tagged proteins

Human fibroblasts

Magnetic agarose beads

Ni-NTA

Elution with imidazole containing buffer

[145]

Estrogen receptor

Adipose tissue

Dynabeads M-280 streptavidin

Biotinylated monoclonal mouse anti-human estrogen receptor antibody

Indirect method

[146]

Thiol-reactive chromatin restriction fragments

Mouse fibroblasts

Mercurated agarose magnetic beads

p- Hydroxymercuribenzoat e

Elution with 0.5 M NaCl and 20 mM dithiothreitol

[147]

L1 coat protein

Human papillomavirus

Magnetic polyglutaraldehyde particles

Iminodiacetic acid charged with Cu2+

Elution with imidazole containing buffer

[41]

Insulin receptor

Rat muscle or liver extract

Dynabeads M-450

Anti-P5 antibody

SDS PAGE analysis

[148]

Stat3

DER cells

Dynabeads

Biotinylated tyrosine phosphorylated peptides

SDS PAGE analysis

[149]

Transferrin receptor

Human

Dynabeads M-450 sheep anti-mouse IgG 1

Anti-human transferrin receptor monoclonal antibody

SDS analysis

[151]

Prion protein PrPSc

Brain extract

Dynabeads M-280 tosyl activated

Plasminogen

SDS analysis

[39,40]

Biotinylated proteins from extracellular matrix

Bipolaris sorokiniana

Dynabeads

Streptavidin

SDS analysis

[152]

Cryoprotectin

Leaves of cold- acclimated cabbage(Brassica oleracea)

Dynabeads-protein A

Specific antibody

 

[153]

Prostate specific antigen

Serum from a prostate cancer suffering patient

Streptavidin-coated magnetic beads

Biotinylated monoclonal antibody

Elution with low pH solution

[154,155]

Estrogen receptor

In vitro translation

Magnetic beads

Antibody

Elution with SDS buffer

[156]

VHDL receptor

Helicoverpa zea

Streptavidin-coated magnetic beads

VHDL-biotin ligand

 

[157]

Fructosyllysine-specific binding protein

U937 cells

Dynabeads M-280 tosylactivated

Poly- L- lysi ne-gl ucose conjugate

Two proteins isolated

[158]

Ubiquitin (histidine tagged)

 

Nickel-gold nanorods

 

Elution with acidic buffer

[18]

 

Table 9: Examples of peptides purified by magnetic techniques

Purified peptide

Source

Magnetic carrier

Affinity ligand

Further details

Reference

Biotinylated peptides

Model mixtures

Dynabeads M-280 streptavidin

Streptavidin

Used in MALDI-TOF mass analysis

[159]

(His)6-Ala-Tyr-Gly

Synthetic peptide

Dynabeads M-280 tosyl activated

Aminocaproic nitrilotriacetic acid charged with Ni2+

Elution with imidazole solution

[160]

Synthetic pentapeptides against fructose-1,6- biphosphate aldolase

Synthetic mixture

Streptavidin-coated magnetic beads

Biotin labelled fructose-1,6-biphosphate aldolase of T. brucei

Pentapeptides were anchored on polystyrene beads

[161]

Tryptic digest products of cytochrome c

Trypsin digested cytochrome c

Au@magnetic particles

-

Ion-exchange separation followed by MALDI MS analysis

[31]

Glutathione

 

Gold and iron oxide nanocomposites

 

 

[162]

Nisin Z

Lactobacillus lactis

EDC activated magnetic beads

Anti-nisin antibody

Elution with 6 M urea

[163]

 

Sometimes the removal of certain proteins will reveal functions involving the depleted proteins or will help in the course of subsequent protein isolation. As an example, Dynabeads have been used to remove involved proteins from Xenopus egg extracts for analyses of the cell mitosis mechanisms [44,45]. Rapid removal of contaminating proteolytic enzymes from the crude samples could increase yields of sensitive proteins due to the limitation of their pro teolysis [46].

A combination of mechanical cell disintegration and mag­netic batch affinity adsorption was used to simplify the isolation of intracellular proteins. Magnetic glass beads were used because of their hardness and rigidity [1].

An example of quite different protein purification strategy can also be mentioned. Proteins associated with the endo-cytic vesicles of Dictyostelium discoideum were separated after magnetic isolation of the vesicles that was accom­plished by feeding the amoebae with dextran-stabilized iron oxide particles. The cells were broken, the labelled vesicles were magnetically separated and then disrupted to release proteins which were resolved by SDS-PAGE. After „in-gel" digestion with endoproteinase Lys-C or Asp-N the generated peptides were used for amino acid sequencing. This strategy allowed the identification of the major protein constituents of the vesicles [47]. Analogous procedure was used for the separation and study of perox-isomes proteins when at first peroxisomes were separated using magnetic beads with immobilized specific antibodies and then the protein content of the separated peroxi­somes was analysed [48].

Conclusions

Standard liquid column chromatography is currently the most often used technique for the isolation and purifica­tion of target proteins and peptides. Magnetic separation techniques are relatively new and still under develop­ment. Magnetic affinity particles are currently used mainly in molecular biology (especially for nucleic acids separa­tion), cell biology and microbiology (separation of target cells) and as parts of the procedures for the determination of selected analytes using magnetic ELISA and related techniques (especially determination of clinical markers and environmental contaminants). Up to now separations in small scale prevail and thus the full poten­tial of these techniques has not been fully exploited.

It can be expected that further development will be focused at least on two areas. The first one will be focused on the laboratory scale application of magnetic affinity separation techniques in biochemistry and related areas (rapid isolation of a variety of both low- and high-molec­ular weight substances of various origin directly from crude samples thus reducing the number of purification steps) and in biochemical analysis (application of immu-nomagnetic particles for separation of target proteins from the mixture followed by their detection using ELISA and related principles). Such a type of analysis will enable to construct portable assay systems enabling e.g. near-patient analysis of various protein disease markers. New methodologies, such as the application of chip and microfluidics technologies, may result in the develop­ment of magnetic separation processes capable of mag­netic separation and detection of extremely small amount of target biologically active compounds [49].

In the second area, larger-scale (industrial) systems are believed to be developed and used for the isolation of bio­logically active compounds directly from crude culture media, wastes from food industry etc., integrating three classical steps (clarification, concentration and initial purification) into a single unit operation [50]. It is not expected that extremely large amounts of low cost prod­ucts will be isolated using magnetic techniques, but the attention should be focused onto the isolation of minor, but highly valuable components present in raw materials. Of course, prices of magnetic carriers have to be lowered and special types of low-cost, biotechnology applicable magnetic carriers and adsorbents prepared by simple and cheap procedures have to become available. The existence of inexpensive and effective magnetic separators enabling large-scale operations is necessary, as well.

In the near future quite new separation strategies can appear. A novel magnetic separation method, which uti­lizes the magneto-Archimedes levitation, has been described recently and applied to separation of biological materials. By using the feature that the stable levitation position under a magnetic field depends on the density and magnetic susceptibility of materials, it was possible to separate biological materials such as haemoglobin, fibrin-ogen, cholesterol, and so on. So far, the difference of mag­netic properties was not utilized for the separation of biological materials. Magneto-Archimedes separation may be another way for biological materials separation [51].

It can be expected that magnetic separations will be used regularly both in biochemical laboratories and biotech­nology industry in the near future.

Acknowledgements

The research is a part of ILE Research Intention No. AV0Z6087904. The work was supported by the Ministry of Education of the Czech Republic (Project No. ME 583) and Grant Agency of the Czech Academy of Sciences (Project No. IBS6087204).

References

  1. Schuster M, Wasserbauer E, Ortner C, Graumann K, Jungbauer A, Hammerschmid F, Werner G: Short cut of protein purification by  integration  of cell-disrupture  and  affinity extraction. Bioseparation 2000, 9:59-67.
  2. Hofmann I, Schnolzer M, Kaufmann I, Franke WW: Symplekin, a constitutive  protein  of karyo-  and  cytoplasmic  particles involved in mRNA biogenesis in Xenopus laevis oocytes. M ol В/о/Cell2002, 13:1665-1676.
  3. AlcheJD, Dickinson K: Affinity chromatographic purification of antibodies to  a biotinylated fusion  protein  expressed  in Escherichia coli. Protein Expr Purif 1998, 12:1 38-143.
  4. Teotia S, Gupta MN: Purification of alpha-amylases using mag­netic alginate beads. Appl Biochem Biotechnol 2001, 90:21 1-220.
  5. Safarik I, Safarikova M: Use of magnetic techniques for the iso­lation of cells.J Chromatogr B Biomed Sci Appl 1999, 722:33-53.
  6. Sinclair B: To bead or not to bead: Applications of magnetic bead technology. Scientist 1998, 12:17.
  7. Bruce IJ, Taylor J, Todd M, Davies MJ, Borioni E, Sangregorio C, Sen T: Synthesis, characterisation and application of silica-mag­netite nanocomposites. J Magn Magn Mater 2004, 284:145-160.
  8. Weetall  HH,  Lee MJ: Antibodies immobilized on  inorganic supports. Appl Biochem Biotechnol 1989, 22:3 1 1 -330.
  9. Safarik I, Safarikova M: Batch isolation of hen egg white lys-ozyme with magnetic chitin. J Biochem Biophys Methods  1993, 27:327-330.
  10. Safarikova M, Roy I, Gupta MN, Safarik I: Magnetic alginate micro-particles  for  purification  of a-amylases. J  Biotechnol 2003, 105:255-260.
  11. I 1. Tanyolac D, Ozdural AR: A new low cost magnetic material: magnetic polyvinylbutyral microbeads. React Funct Polym 2000, 43:279-286.
  12. Nixon L, Koval CA, Noble RD, Slaff GS: Preparation and charac­terization of novel magnetite-coated ion-exchange particles. Chem Mater 1992,4:117-121.
  13. I 3. Mosbach K, Andersson L: Magnetic ferrofluids for preparation of magnetic polymers and their application in affinity chromatography. Nature 1977, 270:259-261.
  14. Hirschbein BL, Whitesides GM: Affinity separation of enzymes from mixtures containing suspended solids. Comparisons of magnetic and nonmagnetic techniques. Appl Biochem Biotechnol 1982,7:157-176.
  15. Rocha FM, de Pinho SC, Zollner RL, Santana MHA: Preparation and characterization of affinity magnetoliposomes useful for the detection of antiphospholipid antibodies. J Magn Magn Mater 2001,225:101-108.
  16. Zollner TCA, Zollner RD, de Cuyper M, Santana MHA: Adsorption of isotype "E" antibodies on affinity magnetoliposomes. J Dis­persion Sci Technol 2003, 24:615-622.
  17. Bucak S, Jones DA, Laibinis PE, Hatton TA: Protein separations using colloidal magnetic nanoparticles. Biotechnol Progr 2003, I 9:477-484.
  18. Lee KB, Park S, Mirkin CA: Multicomponent magnetic nanorods for  biomolecular separations. Angew Chem - Int Edit 2004, 43:3048-3050.
  19. Safarik I, Ptackova L, Safarikova M: Large-scale separation of mag­netic bioaffinity adsorbents. Biotechnol Lett 2001, 23:1953-1956.
  20. Schafer F, Romer U, Emmerlich M, Blumer J, Lubenow H, Steinert K: Automated  high-throughput  purification  of 6xHis-tagged proteins. J Biomol Tech 2002, 1 3:1 3 1 -142.
  21. Lochmuller CH, Ronsick CS, Wigman LS: Fluidized-bed separa­tors reviewed: a low pressure drop approach to column chromatography. Prep Chromatogr 1988, 1:93-108.
  22. Burns MA, Graves  DJ: Continuous affinity chromatography using a magnetically stabilized fluidized bed. Biotechnol Progr 1985, 1:95-103.
  23. 23.     Chetty AS, Burns MA: Continuous protein separations in a magnetically  stabilized  fluidized  bed   using   nonmagnetic supports. Biotechnol Bioeng 1991, 38:963-971.
  24. Wikstrom P, Flygare S, Grondalen A, Larsson PO: Magnetic aque­ous two-phase separation: a new technique to increase rate of phase-separation, using dextran-ferrofluid or larger iron oxide particles. Anal Biochem 1987, 167:331-339.
  25. Larsson P-O: Magnetically enhanced phase separation. Meth Enzymol 1994,228:1 12-1 17.
  26. Safarik I, Safarikova M: Biologically active compounds and xeno-biotics: Magnetic affinity separations. In Encyclopedia of Separa­tion Science Edited by: Wilson ID, Adlard RR, Poole CF, Cook MR. London: Academic Press; 2000:2163-2170.
  27. Safarik I, Safarikova M: Overview of magnetic separations used in biochemical and biotechnological applications. In  Scientific and Clinical Applications of Magnetic Carriers Edited by: Hafeli U, Schutt W, Teller J, Zborowski M. New York and London: Plenum Press; 1997:323-340.
  28. Safarikova M, Safarik I: The application of magnetic techniques in biosciences. Magn Electr Sep 2001, 10:223-252.
  29. Saiyed ZM, Telang SD, Ramchand CN: Application of magnetic techniques in the field of drug discovery and biomedicine. BioMagn Res Technol 2003, 1:2.
  30. Yaneva M, Tempst P: Affinity capture of specific DNA-binding proteins for mass spectrometric  identification. Anal Chem 2003, 75:6437-6448.
  31. Teng CH, Ho KC, Lin YS, Chen YC: Gold nanoparticles as selec­tive and concentrating probes for samples in MALDI MS analysis. Anal Chem 2004, 76:4337-4342.
  32. 32.     Heegaard NHH, Nilsson S, Guzman NA: Affinity capillary electro-phoresis:   important  application   areas  and  some   recent developments. J Chromatogr B 1998, 715:29-54.
  33. Nilsson J, Stahl S, LundebergJ, Uhlen M, Nygren PA: Affinity fusion strategies for detection, purification, and immobilization of recombinant proteins. Protein Expr Purif 1997, 1 1: 1 -16.
  34. Kobs G: Finding the right protein purification system. Cell Notes 2004, 9:2-5.
  35. 35.     Gaberc-Porekar V,  Menart V:   Perspectives  of immobilized-metal affinity chromatography. J Biochem Biophys Methods 2001, 49:335-360.
  36. 36.     Frenzel A, Bergemann C, Kohl G, Reinard T: Novel purification system   for   6xHis-tagged   proteins   by   magnetic   affinity separation.J Chromatogr B 2003, 793:325-329.
  37. 37.    Widjojoatmodjo MN, Fluit AC, Torensma R, Verhoef J: Compari­son of immunomagnetic beads coated with protein A, pro­tein  G,  or  goat  anti-mouse   immunoglobulins. J  Immunol Methods 1993, 165:1 1-19.
  38. Biomagnetic  Techniques  in  Molecular Biology.  Information Booklet, Dynal, Oslo, Norway 1998.
  39. Fischer MB, Roeckl C, Parizek P, Schwarz HP, Aguzzi A: Binding of disease-associated prion protein to plasminogen. Nature 2000, 408:479-483.
  40. Maissen M, Roeckl F, Glatzel M, Goldmann W, Aguzzi A: Plasmino­gen binds to disease-associated prion protein of multiple species. Lancet 2001, 357:2026-2028.
  41. Heeboll-Nielsen A, Choe WS, Middelberg APJ, Thomas ORT: Effi­cient inclusion body processing using chemical extraction and  high  gradient  magnetic  fishing.  Biotechnol Progr  2003, 19:887-898.
  42. Safarikova M, Safarik I: Magnetic solid-phase extraction. J Magn Magn Mater 1999, 194:108-1 12.
  43. Safarikova M, Safarik I: Magnetic solid-phase extraction of tar­get analytes from large volumes of urine. Eur Cells Mater 2002, 3(Suppl 2): 192-195.
  44. Goldberg M, Jenkins H, Allen T, Whitfield WG, Hutchison CJ:Xeno-pus lamin B3 has a direct role in the assembly of a replication competent nucleus: evidence from cell-free egg extracts. J Cell Sci 1995, 108:3451-3461.
  45. Bell P, Scheer U: Prenucleolar bodies contain coilin and are assembled in Xenopus egg extract depleted of specific nucle-olar proteins and U3 RNA.J Cell Sci 1997, 1 10:43-54.
  46. Safarik I, Safarikova M: Isolation and removal of proteolytic enzymes with magnetic cross-linked erythrocytes. J Magn Magn Mater 2001, 225:169-174.
  47. Adessi C, Chapel A, Vincon M, Rabilloud T, Klein G, Satre M, Garin J: Identification of major proteins associated with Dictyostel-ium discoideum endocytic vesicles.J Cell Sci 1995, 108:333 1 -3337.
  48. 48.     Luers GH, Hartig R, Mohr H, Hausmann M, Fahimi HD, Cremer C, Volkl A:  Immuno-isolation of highly purified peroxisomes using  magnetic   beads  and  continuous  immunomagnetic sorting. Electrophoresis 1998, 1 9:1205-1210.
  49. Brzeska M, Panhorst M, Kamp PB, Schotter J, Reiss G, Puhler A, Becker A, Bruckl H: Detection and manipulation of biomole-cules by magnetic carriers. J Biotechnol 2004, 1 12:25-33.
  50. Hubbuch JJ, Thomas ORT: High-gradient magnetic affinity sep­aration of trypsin from porcine pancreatin. Biotechnol Bioeng 2002,79:301-313.
  51. Hirota N, Kurashige M, Iwasaka M, Ikehata M, Uetake H, Takayama T, Nakamura H, Ikezoe Y, Ueno S, Kitazawa K: Magneto-Archimedes separation and its application to the separation of biological materials. Physica B 2004, 346-347:267-271.
  52. 52.    Tong XD, Xue B, Sun Y: A novel magnetic affinity support for protein   adsorption   and   purification.  Biotechnol  Progr  2001, 17:1 34-1 39.
  53. Flygare S, Wikstrom P, Johansson G, Larsson PO: Magnetic aque­ous two-phase separation in preparative applications. Enzyme Microb Technol 1990, 12:95-103.
  54. Murphy AS, Hoogner KR, Peer WA, Taiz L: Identification, purifi­cation,  and  molecular  cloning  of N-1-naphthylphthalmic acid-binding plasma membrane-associated aminopeptidases from Arabidopsis. Plant Physiol 2002, 128:935-950.
  55. 55.    Teotia S, Gupta MN: Magnetite-alginate beads for purification of some   starch   degrading   enzymes.   Mol  Biotechnol  2002, 20:231-237.
  56. Abudiab T, Beitle RR: Preparation of magnetic immobilized metal affinity separation media and its use in the isolation of proteins.J Chromatogr A 1998, 795:21 1-217.
  57. Barnes K, Murphy LJ, Turner AJ: Immunoseparation of mem­brane peptidases from pig lung membranes using magnetic beads. Biochem Soc Trans 1994, 22:S451-S451.
  58. Dunnill P, Lilly MD: Purification of enzymes using magnetic bio-affinity materials. Biotechnol Bioeng 1974, 16:987-990.
  59. Chen D-H, Huang S-H: Fast separation of bromelain by poly-acrylic acid-bound iron oxide magnetic nanoparticles. Process Biochem 2004, 39:2207-221 1.
  60. Himeji D, Horiuchi T, Tsukamoto H, Hayashi K, Watanabe T, Harada M: Characterization of caspase-8L: a novel isoform of cas-pase-8 that behaves as an inhibitor of the caspase cascade. Blood 2002, 99:4070-4078.
  61. Akgol S, Denizli A: Novel metal-chelate affinity sorbents for reversible use in catalase adsorption.JMol CatalB- Enzym 2004, 28:7-14.
  62. Ghosh  M, Tyagi  R,  Gupta  MN:  Preparation  of trypsin  free chymotrypsin. Biotechnol Tech 1995, 9:149-152.
  63. O'Brien SM, Thomas ORT, Dunnill P: Non-porous magnetic che-lator supports for protein recovery by immobilised metal affinity adsorption.J Biotechnol 1996, 50:13-25.
  64. Ljungquist C,  Lundeberg J,  Rasmussen AM,  Hornes  E, Uhlen M: Immobilization and recovery of fusion proteins and B-lym-phocyte cells using magnetic separation. DNA Cell Biol 1993, 12:191-197.
  65. Griffin T, Mosbach K, Mosbach R: Magnetic biospecific affinity adsorbents for immunoglobulin and enzyme isolation. Appl Biochem Biotechnol 1981, 6:283-292.
  66. Ennis MP, Wisdom GB: A magnetizable solid phase for enzyme extraction. Appl Biochem Biotechnol 1991, 30:155-164.
  67. Justesen SFL, Nielsen AH, Thomas ORT: High gradient magnetic fishing for the isolation of high-value proteins from sweet whey. In: Danish Biotechnology Conference VII: Vejle, Denmark. 2001.
  68. Heeboll-Nielsen A, Justesen S, Thomas ORT: Product recovery for crude bioprocess liquors by high gradient magnetic fishing. In: 1 0th European Congress on Biotechnology: Madrid, Spain. 2001.
  69. Betz N: Efficient purification of His-tagged proteins from insect and mammalian cells. Promega Notes 2004, 87:29-32.
  70. Betz N: Purifying His-tagged proteins from insect and mam­malian cells. Cell Notes 2004, 9:6-9.
  71. Safarik I: Magnetic biospecific affinity adsorbents for lysozyme isolation. Biotechnol Tech 1991, 5:1 1 1 -1 14.
  72. Odabasi M, Denizli A: Cibacron blue F3GA incorporated mag­netic poly(2-hydroxyethyl methacrylate) beads for lysozyme adsorption.J Appl Polym Sci 2004, 93:719-725.
  73. Goto M, Imamura T, Hirose T: Axial dispersion in liquid magnet­ically stabilized fluidized beds.J Chromatogr A 1995, 690:1 -8.
  74. Kopacek P, Vogt R, Jindrak L, Weise C, Safarik I: Purification and characterization of the lysozyme from the gut of the soft tick Ornithodoros moubata. Insect Biochem Mol Biol 1999, 29:989-997.
  75. Liao MH, Chen DH: Fast and efficient adsorption/desorption of protein by a novel magnetic nano-adsorbent. Biotechnol Lett 2002,24:1913-1917.
  76. Xue B, Sun Y: Protein adsorption equilibria and kinetics to a poly(vinyl alcohol)-based magnetic affinity support.J Chroma­togr A 2001, 921:109-1 19.
  77. 77.    Tong XD, Sun Y: Application of magnetic agarose support in liquid   magnetically   stabilized   fluidized   bed   for   protein adsorption. Biotechnol Progr 2003, 19:1721 -1727.
  78. Yu YH, Xue B, Sun Y, He BL: The preparation of chitosan affinity magnetic nanoparticles and their adsorption properties for proteins. Acta Polym Sinica 2000:340-344.
  79. Safarik I, Safarikova M, Weyda F, Mosiniewicz-Szablewska W, Slaw-ska-Waniewska A: Ferrofluid-modified plant-based materials as adsorbents for batch separation of selected biologically active compounds and xenobiotics.J Magn Magn Mater in press.
  80. Peng ZG, Hidajat K, Uddin MS: Adsorption and desorption of lys­ozyme on nano-sized magnetic particles and its conforma-tional changes. Colloid Surf B - Biointerfaces 2004, 35:169-174.
  81. O'Brien SM, Sloane RP, Thomas ORT, Dunnill P: Characterisation of non-porous magnetic chelator supports and their use to
  82. recover polyhistidine-tailed T4 lysozyme from a crude E. coli
  83. extract.J Biotechnol 1997, 54:53-67.
  84. 82.    Tyagi R, Gupta MN: Purification and immobilization of Aspergil-lus   niger   pectinase   on    magnetic   latex   beads.    Biocatal Biotransform 1995, 12:293-298.
  85. 83.     Hendrix PG, Hoylaerts MF, Nouwen EJ, Van de Voorde A, De Broe ME: Magnetic beads in suspension enable a rapid and sensi­tive     immunodetection     of    human     placental     alkaline phosphatase. EurJ Clin Chem Clin Biochem 1992, 30:343-347.
  86. HubbuchJJ, Matthiesen DB, HobleyTJ, Thomas ORT: High gradient magnetic separation versus expanded bed adsorption: a first principle comparison. Bioseparation 2001, 10:99-1 12.
  87. 85.     Engel L, Kar S, Johnson T: Rapid detection and quantitation of His-tagged  proteins  purified  by MagneHis™   Ni-particles. Promega Notes 2003, 84:27-30.
  88. Khng HP, Cunliffe D, Davies S, Turner NA, Vulfson EN: The synthe­sis of sub-micron magnetic particles and their use for prepar­ative purification of proteins. Biotechnol Bioeng 1998, 60:419-424.
  89. 87.    Halling PJ, Dunnill P: Recovery of free enzymes from product liquors by bio-affinity adsorption: Trypsin binding by immo­bilised   soybean   inhibitor.   European  J   Appl   Microbiol    1979, 6:195-205.
  90. Lochmuller CH, Wigman LS: Affinity separations in magnetically stabilized fluidized beds. Synthesis and performance of pack­ing materials. Sep Sci Technol 1987, 22:21 1 1-2125.
  91. Lochmuller CH, Wigman LS, Kitchell BS: Aerosol-jet produced, magnetic carrageenan-gel particles: a new affinity chroma-tography matrix.J Chem Technol Biotechnol 1987, 40:33-40.
  92. 90.    Cocker TM, Fee CJ, Evans RA: Preparation of magnetically sus­ceptible polyacrylamide/magnetite beads for use in magnet­ically  stabilized   fluidized   bed   chromatography.   Biotechnol Bioeng 1997,53:79-87.
  93. 91.    An XN, Su ZX: Characterization and application of high mag­netic   property  chitosan   particles. J  Appl  Polym   Sci   2001, 81:1 175-1181.
  94. Nishiya Y, Hibi T, Oda JL: A purification method of the diagnos­tic enzyme Bacillus uricase using magnetic beads and non­specific protease. Protein Expr Purif 2002, 25:426-429.
  95. Dong YS, Liang F, Jin HX, Xi Q, Chang JH: Preparation of novel magnetic affinity adsorbents and application for purification of urokinase. Chem J Chin Univ - Chin 2002, 23:101 3-1017.
  96. Dumitrascu G, Kumbhar A, Zhou WL, Rosenzweig Z: The use of derivatized magnetoliposomes for extraction of antibodies from aqueous solutions. IEEE Trans Magn 2001, 37:2932-2934.
  97. 95.    Odabasi  M,  Denizli  A:  Polyhydroxyethylmethacrylate-based magnetic    DNA-affinity    beads    for   anti-DNA    antibody removal from systemic lupus erythematosus patient plasma. J Chromatogr B 2001, 760:137-148.
  98. Li X, Li CX, He BL: Preparation and application of magnetic affinity adsorbent based on magnetic cellulose bead. Chem J Chin Univ - Chin 1998, 19:994-999.
  99. 97.    Quitadamo IJ, Kostman TA, Schelling ME, Franceschi VR: Magnetic bead   purification   as   a   rapid   and   efficient   method   for enhanced antibody specificity for plant sample immunoblot-ting and immunolocalization. Plant Sci 2000, 1 53:7-14.
  100. Shinkai M, Kamihira M, Honda H, Kobayashi T: Rapid purification of monoclonal antibody with functional magnetite particles. Kag Kog Ronbunshu 1992, 1 8:256-259.
  101. Kondo A, Kamura H, Higashitani K: Development and applica­tion of thermo-sensitive magnetic immunomicrospheres for antibody purification. Appl Microbiol Biotechnol 1994, 41:99-105.
  102. Ozkara S, Akgol S, Canak Y, Denizli A: A novel magnetic adsorb­ent for immunoglobulin-G purification in a magnetically sta­bilized fluidized bed. Biotechnol Progress 2004, 20:1 169-1 175.
  103. Meng ZH, Lin B, Xie YH, Zhang KQ: Cloning and expression of fused Fc-binding protein (SPA-SPG) and its application in purification of IgG. Progr Biochem Biophys 2004, 31:146-149.
  104. Quitadamo IJ, Schelling ME: Efficient purification of mouse anti-FGF receptor IgM monoclonal antibody by magnetic beads. Hybridoma 1998, 17:199-207.
  105. 103.   Bhagwati S, Ghatpande A, Leung B: Identification of two nuclear proteins which bind to RNA CUG repeats: Significance for myotonic    dystrophy.   Biochem   Biophys   Res    Commun    1996, 228:55-62.
  106. Gabrielsen OS, Hornes E, Korsnes L, Ruet A, Oyen TB: Magnetic DNA affinity purification of yeast transcription factor x - a
  107. new purification principle for the ultrarapid isolation of near homogeneous factor. Nucleic Acids Res 1989, 1 7:6253-6267.
  108. Gabrielsen OS, Huet J: Magnetic DNA affinity purification of yeast transcription factor. Meth Enzymol 1993, 21 8:508-525.
  109. 106.   Hollung K, Gabrielsen OS, Jakobsen KS: Enrichment of DNA-binding   proteins   from   crude   tissue   for  electrophoretic mobility   shift   assay   using   magnetic   phospho   cellulose particles. Nucleic Acids Res 1994, 22:3261-3262.
  110. 107.   Risoen PA, Struksnes K, Myrset AH, Gabrielsen OS: One-step mag­netic  purification  of recombinant  DNA-binding   proteins using magnetizable phosphocellulose. Protein Expr Purif 1995, 6:272-277.
  111. Zeng GC, Gao LY, Xia T, Tencomnao T, Yu RK: Characterization of the  5'-flanking fragment of the  human  GM3-synthase gene. Biochim Biophys Acta 2003, 1625:30-35.
  112. Gershon PD, Moss B: Early transcription factor subunits are encoded by vaccinia virus late genes. Proc Natl Acad Sci USA 1990,87:4401-4405.
  113. I 10. Ozyhar A, Gries M, Kiltz HH, Pongs O: Magnetic DNA affinity purification of ecdysteroid receptor. J Steroid Biochem Mol Biol 1992,43:629-634.
  114. Kalivoda KA, Braun KW, Vimr ER: Rapid purification of a prokaryotic regulatory protein with U.MACS™ Streptavidin MicroBeads. MACS&more 2003, 7:8-9.
  115. I 12. Rahat MA, Fry M: Purification and characterization of p27, a protein from hepatocyte chromatin. Evidence suggesting that it binds selectively to guanine-rich single-stranded DNA. FEBS Lett 1993, 334:60-64.
  116. I 1 3. Blank M, Weinschenk T, Priemer M, Schluesener H: Systematic evolution of a DNA aptamer binding to rat brain tumor microvessels. Selective targeting of endothelial regulatory protein pigpen. J Biol Chem 2001, 276:16464-16468.
  117. I 14. Albig A: Isolation of mRNA binding proteins using the U.MACS streptavidin kit. MACS&more 2001, 5:6-7.
  118. I 15. McKay SJ, Cooke H: A protein which specifically binds to single stranded TTAGGGn repeats. Nucleic Acids Res 1992, 20:1387-1391.
  119. I 16. Chakraborty A, White SM, Schaefer TS, Ball ED, Dyer KF, Tweardy DJ: Granulocyte colony-stimulating factor activation of Stat3a and Stat3(3 in immature normal and leukemic human myeloid cells. Blood 1996, 88:2442-2449.
  120. I 17. Feghali CA, Wright TM: Ligand-dependent and ligand-inde-pendent activation of the transcription factor Rf-I in a cell-free system. Biochem J 1995, 310:461-467.
  121. I 18. Ren LF, Chen H, Sternberg EA: Tethered bandshift assay and affinity purification of a new DNA-binding protein. Biotech-niques 1994, 16:852-855.
  122. I 19. Allen TE, Heidmann S, Reed R, Myler PJ, Goringer HU, Stuart KD: Association of guide RNA binding protein gBP21 with active RNA editing complexes in Trypanosoma brucei. Mol Cell Biol 1998, 18:6014-6022.
  123. Honys D: Isolation of proteins comprising native gene-specific messenger ribonucleoprotein particles using paramagnetic beads. Plant Sci 2001, 161:605-61 1.
  124. Fantappie MR, Correa-Oliveira R, Caride EC, Geraldo EA, Agnew A, Rumjanek FD: Comparison between site-specific DNA binding proteins of male and female Schistosoma mansoni. Comp Bio­chem Physiol B - Biochem Mol Biol 1999, 124:33-40.
  125. 122.   Kelm RJ Jr, Cogan JG, Elder PK, Strauch AR, Getz MJ: Molecular interactions between single-stranded DNA-binding proteins associated with an essential MCAT element in the mouse smooth   muscle   alpha-actin   promoter. J  Biol   Chem   1999, 274:14238-14245.
  126. Daniels DA, Chen H, Hicke BJ, Swiderek KM, Gold L: A tenascin-C aptamer identified by tumor cell SELEX: Systematic evolu­tion of ligands by exponential enrichment. Proc Natl Acad Sci USA 2003, 100:15416-15421.
  127. Skala-Rubinson H, Vinh J, Labas V, Kahn A, Tuy FPD: Novel target sequences for Pax-6 in the brain-specific activating regions of the rat aldolase C gene.J Biol Chem 2002, 277:47190-47196.
  128. Murphy MB, Fuller ST, Richardson PM, Doyle SA: An improved method for the in vitro evolution of aptamers and applica­tions in protein detection and purification. Nucleic Acids Res 2003, 31 :e 1 10.
  129. Tong XD, Sun Y: Agar-based magnetic affinity support for pro­tein adsorption. Biotechnol Progr 2001, 17:738-743.
  130. Xue B, Tong XD, Sun Y: Characterization of PVA-based mag­netic affinity support for protein adsorption. Sep Sci Technol 2001,36:2449-2461.
  131. Xue B, Sun Y: Fabrication and characterization of a rigid mag­netic  matrix for protein  adsorption. J Chromatogr A  2002, 947:185-193.
  132. Akgol S, Turkmen D, Denizli A: Cu(II)-incorporated, histidine-containing,  magnetic-metal-complexing  beads  as  specific sorbents for the metal chelate affinity of albumin. J Appl Polym Sci 2004, 93:2669-2677.
  133. 130.   Odabasi M, Uzun L, Denizli A: Porous magnetic chelator support for   albumin   adsorption   by   immobilized   metal   affinity separation. J Appl Polym Sci 2004, 93:2501 -2510.
  134. Odabasi M, Denizli A: Cibacron Blue F3GA-attached magnetic poly(2-hydroxyethyl methacrylate) beads for human serum albumin adsorption. Polym Int 2004, 53:332-338.
  135. Ding XB, Sun ZH, Zhang WC, Peng YX, Wan GX, Jiang YY: Adsorp-tion/desorption of protein on magnetic particles covered by thermosensitive polymers. J Appl Polym Sci 2000, 77:2915-2920.
  136. DingXB, SunZH, Wan GX, Jiang YY: Interactions between ther­mosensitive magnetic polymer microspheres and proteins. Acta Polym Sinica 2000:9-1 3.
  137. 134.  An XN, Su ZX, Zeng HM: Preparation of highly magnetic chi-tosan   particles  and  their  use  for affinity  purification  of enzymes. J Chem Technol Biotechnol 2003, 78:596-560.
  138. Heeboll-Nielsen A, Dalkiaer M, Hubbuch JJ, Thomas ORT: Super-paramagnetic adsorbents for high-gradient magnetic fishing of lectins  out  of legume  extracts.  Biotechnol  Bioeng  2004, 87:31 1-323.
  139. Safarikova M, Safarik I: One-step partial purification of Solanum tuberosum tuber lectin using magnetic chitosan particles. Bio­technol Lett 2000, 22:941 -945.
  140. Xu CJ, Xu KM, Gu HW, Zhong XF, Guo ZH, Zheng RK, Zhang XX, Xu B: Nitrilotriacetic acid-modified magnetic nanoparticles as a general agent to bind histidine-tagged proteins. J Am Chem Soc 2004, 126:3392-3393.
  141. Muller-Schulte D, Brunner H: Novel magnetic microspheres on the basis of poly(vinyl alcohol) as affinity medium for quanti­tative detection of glycated hemoglobin. J Chromatogr A 1995, 71 1:53-60.
  142. Dryden SC, Nahhas FA, NowakJE, Goustin A-S, Tainsky MA: Role for human SIRT2 NAD-dependent deacetylase activity in control of mitotic exit in the cell cycle. Mol Cell Biol 2003, 23:3173-3185.
  143. Ohtsuki T, Sakurai M, Sato A, Watanabe K: Characterization of the interaction between the nucleotide exchange factor EF-Ts from nematode mitochondria and elongation factor Tu. Nucleic Acids Res 2002, 30:5444-5451.
  144. Suzuki M, Kamihira M, Shiraishi T, Takeuchi H, Kobayashi T: Affinity partitioning of protein A using a magnetic aqueous two-phase system. J Ferment Bioeng 1995, 80:78-84.
  145. Liabakk NB, Nustad K, Espevik T: A rapid and sensitive immu-noassay for tumor necrosis factor using magnetic monodis-perse polymer particles. J Immunol Methods 1990, 134:253-259.
  146. Zhang ZR, O'Sullivan DA, Lyddiatt A: Magnetically stabilised flu-idised bed adsorption: practical benefit of uncoupling bed expansion from fluid velocities in the purification of a recom-binant protein from Escherichia coli. J Chem Technol Biotechnol 1999,74:270-274.
  147. Simonsen A, Stang E, Bremnes B, Roe M, Prydz K, Bakke O: Sorting of MHC class II molecules and the associated invariant chain (Ii) in polarized MDCK cells.J Cell Sci 1997, 1 10:597-609.
  148. Mikaelsdottir EK, Valgeirsdottir S, Eyfjord JE, Rafnar T: The Icelan­dic founder mutation BRCA2 999del5: analysis of expression. Breast Cancer Res 2004, 6:R284-R290.
  149. Welter BH, Price TM: Magnetic separation to concentrate the estrogen receptor from adipose tissue for western analysis. Biotechniques 1999, 27:282-286.
  150. 147.   Chen-Cleland TA, Boffa LC, Carpaneto EM, Mariani MR, Valentin E, Mendez  E, Allfrey VG: Recovery of transcriptionally active chromatin restriction fragments by binding to organomer-curial-agarose magnetic beads. A rapid and sensitive method for monitoring changes in higher order chromatin structure during  gene  activation   and  repression. J  Biol  Chem   1993, 268:23409-23416.
  151. Knutson VP, Donnelly PV, Balba Y, Lopez-Reyes M: Insulin resist­ance is mediated by a proteolytic fragment of the insulin receptor. J Biol Chem 1995, 270:24972-24981.
  152. Chakraborty A, Dyer KF, Cascio M, MietznerTA, Tweardy DJ: Iden­tification of a novel Stat3 recruitment and activation motif within the granulocyte colony-stimulating factor receptor. Blood 1999,93:15-24.
  153. Tanaka T, Matsunaga T: Detection of HbA(1 c) by boronate affin­ity immunoassay using bacterial magnetic particles. Biosens Bioelectron 2001, 16:1089-1094.
  154. Karlsson GB, Platt FM: Analysis and isolation of human transfer-rin receptor using the OKT-9 monoclonal antibody cova-lently cross-linked  to  magnetic  beads. Anal Biochem   1991, 199:219-222.
  155. Apoga D, Ek B, Tunlid A: Analysis of proteins in the extracellu­lar matrix of the plant pathogenic fungus Bipolaris sorokini-ana using 2-D gel electrophoresis and MS/MS. FEMS Microbiol Lett 2001, 197:145-150.
  156. Hincha DK, Neukamm B, Sror HAM, Sieg F, Weckwarth W, Ruckels M, Lullien-Pellerin V, Schroder W, Schmitt JM: Cabbage cryopro-tectin is a member of the nonspecific plant lipid transfer pro­tein gene family. Plant Physiol 2001, 1 25:835-846.
  157. Peter J, Unverzagt C, Lenz H, Hoesel W: Purification of prostate-specific antigen from human serum by indirect immunosorp-tion and elution with a hapten. Anal Biochem 1999, 273:98-104.
  158. Peter J, Unverzagt C, KroghTN, Vorm O, Hoesel W: Identification of precursor forms of free prostate-specific antigen in serum of prostate cancer patients by immunosorption and mass spectrometry. Cancer Res 2001,61:957-962.
  159. Yang Y-S, Yang M-CW, Wang B, Weissler JC: BR22, a novel pro­tein, interacts with thyroid transcription factor-1 and acti­vates the human surfactant protein B promoter. Am J Respir Cell Mol Biol 2001, 24:30-37.
  160. Persaud DR, Yousefi V, Haunerland N: Efficient isolation, purifica­tion, and characterization of the Helicoverpa zea VHDL receptor. Protein Expr Purif 2003, 32:260-264.
  161. Salazar R, Brandt R, Kellermann J, Krantz S: Purification and char­acterization of a 200 kDa fructosyllysine-specific binding pro­tein from cell membranes of U937 cells. Glycoconjugate J 2000, 17:713-716.
  162. Girault S, Chassaing G, Blais JC, Brunot A, Bolbach G: Coupling of MALDI-TOF mass analysis to the separation of biotinylated peptides by magnetic streptavidin  beads. Anal Chem   1996, 68:2122-2126.
  163. Ji Z, Pinon DI, Miller LJ: Development of magnetic beads for rapid and efficient metal-chelate affinity purifications. Anal Biochem 1996,240:197-201.
  164. 161.  Samson   I,   Rozenski J, Vanaerschot A,  Samyn   B, Vanbeeumen J, Herdewijn P: Screening of a synthetic pentapeptide library composed  of D-amino  acids  against  fructose-1,6-biphos-phate aldolase. Lett Pept Sci 1995, 2:259-260.
  165. Seino S, Kinoshita T, Otome Y, Nakagawa T, Okitsu K, Nakayama T, Sekino T, Niihara K, Yamamoto TA: Radiation-induced synthesis of Au/Fe oxide nanocomposite particles for magnetic sepa­ration of biomolecules. J Ceram Process Res 2004, 5:1 36-1 39.
  166. Prioult G, Turcotte C, Labarre L, Lacroix C, Fliss I: Rapid purifica­tion of nisin Z using specific monoclonal antibody-coated magnetic beads. Int Dairy J 2000, 10:627-633.