7
Chromatography: Separation Techniques in Biology

W John Lough and Mark Carlile

School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Chester Road, Sunderland, SR1 3SD, UK

7.1 Introduction to Chromatographic Separation

Chromatography is a term that is used to describe a group of separation techniques that utilise the distribution of molecules between a stationary phase and a mobile phase [1]. Separation is achieved by exploitation of a molecule's physicochemical properties that promote it to interact with the stationary phase or stay within the mobile phase. Interactions with the stationary phase promote a slower passage through the separation system. It is this distribution and the modulation of the distribution between these phases that makes chromatography a very powerful and versatile separation technique. Different types of molecule have different distribution constants and hence come off the column at different times. Thus, they are separated from one another before being measured. Importantly, this means that quantitative determinations based on chromatography can be optimised to give good specificity. In an analytical method validation to demonstrate that an analytical method is fit for its intended purpose, accuracy, precision, linearity, limit of quantitation and limit of detection may be evaluated, but it is the test for specificity that is often the most challenging of the validation tests [2]. Therein lies perhaps the main reason for the very widespread use of chromatography in biology.

The field of ‘biology’ is vast and complex. Within this there are a large and very diverse number of applications for which the power of chromatography may be effectively exploited. Consequently, the entire range of manifestations of ‘chromatography’ can find some use in biology. However, especially in the purification and analysis of intact, large biological molecules, liquid column chromatography (Figure 7.1) is by far the most common form of chromatographic separation used [3,4]. However, the application of gas‐phase chromatographic separation is also becoming more common, especially for the analysis and profiling of cellular metabolism [5].

A typical modern system for high-performance liquid chromatography (HPLC) with arrows pointing the mobile phase reservoirs, detector, column compartment, mobile autosampler, and pumping system.

Figure 7.1 A typical modern system for high‐performance liquid chromatography (HPLC). The instrument shown is also suitable for uHPLC in which higher pressures are used in obtaining good efficiency in short run times on columns containing sub‐2‐μm particles. Older systems had clearly identifiable pumps, detectors, etc., but, to reduce their laboratory footprint, modules of newer systems are stacked. Systems used for chromatography of biological samples may be biocompatible (e.g. an inert titanium/PEEK flow path not only reduces interactions of proteins with metallic surfaces but is also suitable for use with harsh salt conditions and de‐salting prior to MS detection).

The most common forms of liquid chromatographic separation (Table 7.1) are referred to as modes of chromatography, which differ in terms of their stationary phase chemistry and physical properties and the associated mobile phase (one stationary phase can be used for more than one mode of chromatography). The stationary phase is commonly regarded as the solid matrix (usually particles/beads) and its associated surface chemistry. However, this is almost always porous and the ‘stationary phase’ might better be regarded as the porous matrix and the stagnant mobile phase therein (making constituting up to 90% of the total ‘stationary phase’). The solid matrix is typically composed of regular beads of <1–100 μm in diameter. The beads are either xerogels (e.g. Sephadex™: able to shrink and swell dependent on the mobile phase) and aerogels (e.g. silica, porous glass: volume is independent of the mobile phase). Silica finds greater use for modern quantitative analysis (cf. preparative/purification) where there has been adaptation and modification of technology common in high performance liquid chromatography (HPLC) of small molecules. The choice of composition of the mobile phase is dependent on the mode of the separation method and the nature of the samples applied.

Table 7.1 Different modes of liquid chromatographic separation commonly used in the analysis of biological samples.

Physicochemical property Mode of chromatography Pressure format References
Size and shape Gel filtration/size exclusion separation for proteins and nucleic acids HIGH
LOW
[68]
Net charge Anion and cation exchange chromatography for proteins.
Water softening and demineralisation.
pH quenching
HIGH
LOW
[4, 9, 10]
Isoelectric point Chromatofocusing of proteins MEDIUM [11,12]
Hydrophobicity Hydrophobic interaction and reversed‐phase chromatography of proteins, nucleic acids and small molecule drugs HIGH
LOW
[13,14]
Biological function Affinity chromatography for proteins and nucleic acids.
Immobilised ligand chromatography for proteins and nucleic acids. Immunoadsorption chromatography for proteins
HIGH
LOW
[1517]
Carbohydrate content Lectin affinity chromatography HIGH [18]
Free thiols Chemisorption (covalent) chromatography applied to proteins HIGH [19,20]
Metal binding Immobilised metal ion affinity chromatography for proteins HIGH
LOW
[21,22]
Chiral Typically separation of pharmaceutical drug enantiomers but also biomolecules (e.g. d, l‐ amino acids were amongst the first analytes to be enantio‐resolved and there is a revival of interest in the determination of trace d‐ enantiomer content in natural amino acids) HIGH [2327]
Ion pair Used to separate charged substances using reversed‐phase HPLC. Works through the addition of mobile phase additives. Used to separate small molecule drug, metabolites, and nucleic acids HIGH [2830]

7.1.1 The Theory of Chromatographic Separation

The mobile phase flow and analyte separation when applied to chromatography can be explained by several main mathematical relationships and physical parameters. Essentially a chromatogram, the signal response product of a chromatographic separation, is a plot of analyte concentration versus eluent volume or, less fundamentally, time (Figure 7.2). The retention time (t r ) or volume for a particular analyte is an important parameter for characterising the separation. Retention time is dependent on the volumetric flowrate. However, the retention time also depends on the dimensions of the column used. Therefore, the retention time is usually reported as a normalised retention quantity known as the capacity factor (or retention factor) (k). Selectivity (or separation factor) (α) is the ratio of the retention (capacity) factors (k) of two peaks. The efficiency (N) is reported as the number of theoretical plates for a column. Traditionally, a ‘plate’ (the name is taken from the ‘plates’ used in fractional distillation towers used in the petroleum industry) is defined as a hypothetical separation layer within the column in which the two phases establish equilibrium with each other. The higher the number of plates the more efficient the separation will be. The concept of a layer in which equilibrium is established is a rather abstract one in that chromatography is a dynamic process and equilibrium is never reached. For simplicity it may be useful to note that analytes elute from the chromatography column as a peak with a Gaussian distribution and the plate height is related to the variance of that peak. The Gaussian distribution arises from the band broadening processes that take place in the column, giving rise to random error in the time taken for molecules of a single analyte to elute from the column. Resolution (R) is defined as the separation of two analyte peaks, taking into account the individual peak widths. The main mathematical relationships used in chromatographic separation are shown below:

equation

where

  • k is the retention factor
  • t r is the retention time for a particular analyte (e.g. r = 1 for the first peak)
  • t 0 is the column void volume (solvent front)
  • N is the number of theoretical plates
  • w n is the width of an individual peak
  • R s is the resolution of two individual peaks
A chromatographic separation of a mixture of two analytes depicting a fluctuating curve with unretained peaks, peak 1, peak 2, and injection point being marked. Peaks 1 and 2 have widths of w1 and w2, respectively.

Figure 7.2 Chromatographic separation of a mixture of two analytes showing the measurements that need to be made in order to calculate capacity factor, selectivity, efficiency and resolution.

The above equation for resolution is used for the actual measurement of resolution from a chromatogram. This should not be confused with an alternative equation for resolution, which may be obtained by substituting the other three equations into the equation for resolution.

The Purnell equation [31] describes the resolution of two similar peaks under chromatographic separation taking into account the selectivity, capacity and plate number:

equation

The Purnell equation is an extremely useful relationship that can be used to gain a deeper insight into what qualitatively is obvious. Better resolution may be had by having narrower peaks (better efficiency) and by having a larger ratio of capacity factors (selectivity). As the Purnell equation also indicates, better resolution may be had by increasing the capacity factor. However, not only is this at the expense of increasing analysis time but there is little benefit in terms of increasing resolution for k > 10.

The separation of two analytes (Figure 7.2) is only possible if their retention times differ by virtue of having different distribution coefficients between the mobile phase and stationary phase. While the principal goal in this case is to obtain reliable baseline resolution in as short a time as possible, an associated goal is to obtain this not just through good peak efficiency but also through good peak symmetry; i.e. there may be deviations from the anticipated symmetrical Gaussian peak shape in the form of fronting and/or tailing. There are several main causes for non‐Gaussian peak shapes, which include: column stationary phase deterioration, column under‐ or overpacking, mobile phase impurities or loss of liquid handling control and/or efficiency [32,33]. Peak width is usually measured at both the baseline inflection points and at half of the maximum signal height (Figure 7.2). By monitoring peak width over repeated separations, both column and system performance may be determined. For example, if a column is still being conditioned with the sample there will be a reduction in peak width over time. Also of note is the fact that as the concentration of an analyte in the sample increases there is usually a consequent broadening of the peak (mass overloading). Other factors leading to an increased peak width are dilute sample injections over long periods of time (usual in preparative protein chromatography and volume overloading), very shallow or elongated gradient elution profiles, mobile phase impurities or incorrect make‐up and/or problems with liquid handling.

Gradient elution is in contrast to isocratic elution. When eluting analytes in chromatographic separations, there are three distinct types of elution (Figure 7.3) available. Isocratic elution utilises one mobile phase consistently across the whole separation. Isocratic elution is used in size exclusion chromatography but is rarely used when other modes of liquid chromatography that are used for the separation of large biologicals. This applies in particular to preparative liquid chromatography. Gradient elution utilises two (or very occasionally more) mobile phases and over a period of time gradually mixes the phases so that the composition changes from the initial composition to a new composition. For the majority of liquid separations, the gradient is linear. Step elution, like gradient elution, utilises two (or more) mobile phases, but rather than gradually changing the composition it uses a series of ‘step‐up’ transitions to achieve a new mobile phase composition. In the analysis of small molecules a gradient is commonly used to shorten run times by bringing strongly retained sample constituents off the column more quickly. In preparative work on large biologicals the application of a gradient is more commonly used to facilitate separation and aid resolution of closely related analytes. Step elution is used to generate fast but crude ‘cuts’ of adsorbed species from the column. It is common to use step elution after the mobile phase composition needed for a specific analyte elution has been characterised via gradient elution.

3 Graphs depicting the standard elution profiles used in liquid chromatograph for isocratic, gradient, and step (left-right) displaying a horizontal line, an ascending line, and an ascending step curve, respectively.

Figure 7.3 Standard elution profiles used in liquid chromatography.

In considering the theory of chromatography in the context of biological applications it must be remembered that it is not just about manipulating retention, selectivity and efficiency to obtain resolution of large molecules. Biological samples, particularly those containing small molecules, can be very complex, say several hundreds of thousands of components compared to maybe tens of components in a small molecule drug sample. A real problem can be that, even with very narrow peaks, there are simply too many sample constituents for them all to be resolved in a given run time. In other words, there is insufficient peak capacity. Temperature, flow rate, use of smaller particles, etc., can be used to increase peak capacity. However, for a step improvement it is necessary to use multidimensional chromatography where the peak capacity is the product of the peak capacity of each dimension. For this reason, comprehensive 2D‐LC (very small volumes of eluent from the first column are all chromatographed on a second column with different selectivity in real time) is attracting great interest [34]. In the meantime, in the quantitative determination of complex biological samples it is almost the norm to add another dimension when using a single column through the use of a mass spectrometer as the detector. Mass spectrometry in its own right is a powerful tool in the analysis of biologicals (Chapter ) but while the focus here is more on the actual chromatography of biological molecules, including for preparative purposes, it cannot be emphasised enough that liquid chromatography in combination with mass spectrometric detection is very powerful for the characterisation and quantitative determination of biologicals.

7.1.2 Chromatography for Biological Samples

Biological samples are, by their very nature, complex, containing impurities that are analyte‐related (minority) and analyte‐unrelated (majority). Analytical and preparative separation methods need to be robust in terms of their reproducibility and, for preparative work, throughput (multiple injections/loadings), while maintaining a selectivity for a physicochemical property for the analyte. Chromatography as a separation technique can provide this selectivity requirement even when applied to heterogeneous sample mixtures from a variety of sources.

Chromatography can also be used in‐line with other preparative and analytical techniques since it can provide the analyte in an eluent that is suitable for further downstream separation and analysis (for example spectrophotometry, electrophoresis, circular dichroism, nuclear magnetic resonance [NMR] and mass spectrometry).

However, the dynamic nature of biological samples can be a technical challenge when applying any form of separation technique. Ensuring that the sample applied to the chromatographic technique is indeed representative of the true biological system from which the sample was taken needs to be considered at all stages of sample preparation through to data analysis. When applied correctly, chromatography can be non‐destructive, modulated in terms of physical properties (pH, temperature, conductivity), have efficient sample processing times, and take advantage of computation integration and processing methods to circumvent many of these challenges.

7.1.3 Biological Sample Preparation for Chromatography

While there are chromatographic applications for isolating [35] and characterising intact cells [36], in just about all cases for chromatographic separation of biological samples some initial form of cellular disruption is required [37]. Disruption methods will release various compartmentalised hydrolases (phosphatases, glycosidases and proteases) that can alter the composition of the lysate [38]. In all modern experimental approaches, it is essential to preserve sample composition from the point of generation to analysis. Indeed, managing sample complexity can be an added challenge when working with biological material [39,40]. Even the most sophisticated modern instrument cannot produce meaningful data without some form of rational sample preparation. Biological samples, irrespective of their origin, will contain a heterogeneous mixture of small and macromolecular species (Table 7.2), all of which will behave differently when applied to chromatographic separation.

Table 7.2 General consideration and techniques for chromatographic separation of biological samples.

Sample type Description Technical considerations
Protein and peptides Large bio‐macromolecule consisting of one or more chains of amino acids joined via peptide bonds. Proteins are found in all forms of life and have structural and catalytic functions. Peptides are short chains of 2 (di‐peptide) to 20–30 amino acids in length.

The structure of a protein directs its function and so maintenance of the 3D conformation of a protein is important when preparing a sample. Proteins can be found in both soluble and insoluble forms depending on their function.
  • The amino acid sequence of a protein dictates its physicochemical properties. Proteins and peptides are sensitive to extremes of temperature and pH and need to be buffered to stay in solution. Each protein will have an isoelectric point at which it will have an equal number of positive and negative charges on its constituent amino acids and will precipitate out of solution.
  • Proteins are susceptible to denaturation when exposed to chaotropes and/or reducing agents. Increasing concentrations (or prolonged exposure) of oxidising agents may lead to changes in the hydrophobicity profile of the protein and eventual unfolding.
  • Recombinant proteins have a tendency to aggregate if generated using strong induction conditions that overwhelm the protein folding capacity of the expression system (example T7 promoter induction in E. coli strains).
  • UV spectrophotometric analysis of proteins is routinely carried out at 280 nm.
  • Proteins are stable when frozen.
[4143]
Nucleic acids and oligonucleotides Large bio‐macromolecule consisting of one or two strands of nucleotides joined by phosphodiester bonds. Nucleic acids carry the genetic information for all known forms of life.

DNA: Is a stable duplex of two strands of deoxyribonucleotides (Adenine, Thymine, Guanine and Cytosine). The stable structure is formed by the stacking of the nucleotide bases and the exclusion of water to the phosphate backbone on the outside of the duplex.

RNA: Can be found in both single‐stranded and duplex conformations. RNA consists of strands of ribonucleotides and the base Thymine is replaced with Uracil.

Oligonucleotides are short double‐ or single‐stranded nucleic acids that can be composed of ribo‐ or deoxyribonucleotides or unnatural nucleotide monomers.
  • The nucleotide sequence carries information that can be turned into a protein sequence or directs the formation of a structural, regulatory or catalytic RNA. Care must be taken not to modify both the base sequence or the structure of any isolated samples.
  • DNA has a maximal absorbance at 260 nm and has hypochromic properties, wherein, as it denatures, the absorption at 260 nm increases.
  • DNA is a relatively stiff molecule with a high axial ratio leading to an increase in sample viscosity at high concentrations.
  • In strong acids and high temperature nucleic acids are hydrolysed to their constituent nucleotides. More dilute acid treatment leads to the breakage of glycosidic bonds in the purine nucleotides, leading to the sample becoming apurinic.
  • Addition of alkali to DNA will change the tautomeric state of the bases and results in denaturation through loss of hydrogen bonding. Addition of alkali to RNA will lead to hydrolysis of the nucleotide strand.
  • As with all polymers, nucleic acids can be separated based upon their size and indeed sizing is used routinely for characterising both DNA and RNA preparations.
  • Phenol‐chloroform extraction of nucleic acids will isolate DNA and/or RNA while denaturing any bound or associated proteins. Ethanol can be used to precipitate‐out nucleic acids from the aqueous layer of the extraction, when the Na+ ion concentration is >0.3 M, therein concentrating them from relatively dilute samples. There are numerous proprietary methods available for isolation of nucleic acids.
  • Nucleic acids are stable when frozen. Ideally DNA should be stored in a lyophilised form.
  • DNA can be a major contaminant in protein isolation and poses an obstacle to sample processing due to its viscosity.
Lipids A diverse family of biological macromolecules with mostly hydrophobic properties. Lipids can be long chained ‘fatty’ acids or shorter hydrocarbon chains with polar phosphate, saccharide groups or even proteins attached.
Lipids are a major energy storage molecule in the cell and the main constituent of cell membranes. Steroids are also part of the lipid family.
  • Due to their hydrophobicity, lipids are poorly soluble in aqueous solutions but very soluble in organic solvents (e.g. acetone, chloroform, ether). Lipids added to aqueous buffer will form micelles.
  • When working with lipids in the lab it is usually important to dissociate the lipid from other cellular components (usually protein).
  • Care must be taken not to hydrolyse or oxidise the lipids in the sample by controlling the level and activity of co‐purified hydrolase enzymes and reactive oxygen species.
  • Lipids are stable when frozen but temperatures below –30 °C need to be ensured to stop acylation and hydrolysis. Slow thawing can have similar effects on lipid samples.
[44]
Carbohydrates Carbohydrates are a diverse family of biomolecules containing carbon, hydrogen and oxygen. The monomeric unit of carbohydrates is the saccharide. Carbohydrates are commonly referred to as sugars.
Within biological systems, carbohydrates are used for their energy storage and structural properties.
  • As with proteins and lipids, carbohydrates can be found in both soluble and insoluble forms. As with proteins and lipids, care must be taken to not change the carbohydrate analyte profile of the sample by chemical and/or enzymatic hydrolysis and microbial spoilage.
  • Exhaustive ethanol partition and extraction has been applied to carbohydrate isolation, but there is a technical movement to derivatisation and direct analysis.
  • Elevated temperatures will promote hydrolysis of carbohydrate samples and it is common to use temperatures in excess of 40 °C when carrying out an isolation. Other methods utilise acid hydrolysis to extract carbohydrates for analysis.
  • Carbohydrates are stable when frozen.
[4547]
Clinical samples Clinical (patient‐derived) samples will contain a mixture of all biological macromolecules and bio‐inorganic analytes in different proportions dependent on the source.
It should be noted that the generation of clinical samples usually indicates some form of perturbed physiological process associated with the patient and the sample.

Common clinical samples are:
  • Whole blood and blood fractions
  • Tissue biopsies
  • Urine samples
  • Tissue swabs
  • DNA isolates
  • Maintaining sample integrity is essential when dealing with any clinical isolate so as to preserve any biomarkers and generate a true representation of a pathological state. It is difficult to generate a holistic analytical profile for all analytes in a clinical sample, but it is sometimes the subtle interactions and fluctuations between analytes that provide the most insight into a biological system or process.
  • Controlling and accounting for variability in clinical analysis is important. Patient‐derived variability: will be present due to genetic makeup, normal biological variability and environmental and physiological factors specific to the patient. Pre‐analytical variability: will be derived from sample preparation and storage. Analytical variability: will arise from changes in analytical method performance – can be accounted for through the use of technical standards and replicates.
[39,48,49]

At the start of the sample preparation phase, there are several important considerations that need to be taken into account since they impact the sample preparation strategy:

  • Measurement: The application of a simple measurement like UV‐spectrophotometry may still incur problems if the sample does not have a chromophore or if the chromophore is masked by sample impurities leading to coelution and/or signal broadening. The application of more sophisticated measurement techniques like mass spectrometry [5] or NMR spectroscopy [50] may require the removal of specific sample constituents prior to their use, not to mention other associated technical considerations (Table 7.3).
  • Quantitation: Accurate quantitation is needed for all chromatographic separations. The level of quantitation can range from basic peak area determination to more complex comparisons with internal standards to generate accurate titre values for the analyte and in some cases the main impurity peaks.
  • What is the sample composition and how much is known about it? The organic versus inorganic, solid versus soluble sample composition is by far the most important property of the sample to consider prior to chromatographic separation. Of course, it goes without saying that if the sample is solid or a mixture of solid and liquid then exactly how subsampling is carried out is a major consideration. Failure to account for sample composition can lead to poor chromatographic separation and even failure of the chromatographic media and liquid pumping systems. The sample volume and viscosity will affect the solubility of many analytes and this may change as a sample is stored based on the environmental conditions.

Table 7.3 Detection methods used with chromatographic separations.

Detection classification Description and technical considerations References
General detectors UV/visual light absorbance: Visible light detection (400–750 nm) and the more commonly used ultraviolet detection (100–400 nm; e.g. proteins absorb strongly at 280 nm by virtue of aromatic amino acids such as tryptophan) are a routine detection method used by biological sample analysis. The detector measures the absorbance of light by chromophores in the sample. For most applications UV/Vis detection obeys the Beer Lambert law but may show a non‐linear relationship at high analyte concentrations. Many detectors will allow multiple wavelengths to be monitored. Diode array detectors allow scanning of wavelength ranges and can generate 3D plotting of the detection signal. Picogram per ml detection levels are achievable.
Refractive index: Involves measuring the change in refractive index of the column eluate. As an analyte passes through the detector the refractive index will change compared to the mobile phase. The greater the refractive index the greater the difference between the sample and mobile phase. Thus, the sensitivity will be higher for the higher difference in refractive index between the sample and mobile phase. The refractive index is said to be the only universal detection method for chromatography. Used in the analysis of carbohydrates and small molecules. Samples do not need a chromophore.
Evaporative light scattering: Causes vaporisation of the eluate combined with quantification of the analyte by light scattering. The eluate emerging from the column is combined with a flow of air or nitrogen to form an aerosol – the eluent is evaporated from the aerosol by passage through an evaporator and the emerging dry particles of analyte are irradiated with a light source and the scattered light detected by a photodiode. The intensity of the scattered light is determined by the quantity of analyte present and its particle size. Samples do not need a chromophore.
[51,52]




[53]





[54,55]
Selective detectors Fluorescence: Can be applied to samples that fluoresce – a major limitation of its use (although many samples can be derivatised to induce fluorescence). Fluorescent detection is extremely sensitive down to the femtogram per ml level.
Conductivity: Measures electrical impedance of the column eluate, which will change as analytes elute from the column. Its application is not common for biological samples but can be developed for protein separation. However, the sensitivity for protein is relatively low at the microgram per ml level.
Electrochemical: Selective detection of electroactive analytes. Amperometric and coulometric forms of detection can be used. Analytes in the eluate flow through the flow cell, wherein molecules at the electrode surface undergo either an oxidation or a reduction, resulting in a current flow between the two electrodes. Sensitivity can be at the nanogram per ml level.
Flame photometric: Applied in gas chromatographic separation for looking at residual metals such as tin, boron, arsenic and chromium as well as for sulphur and phosphorous containing compounds. Samples are carried in a hydrogen/air flame. Detection is via chemiluminescene at specific wavelengths, which when passed into a photomultiplier gives an electrical signal that can then be measured. Sensitivity can be at the picogram per ml level for some applications.
[42,56,57]




[58]
Structure determining detectors (hyphenated analysis) Combining sample separation with identification and/or structural deduction is a very powerful analytical method combination that is applied in the analysis of many biological samples.
Mass spectrometry (MS): The analyte is introduced into a mass spectrometer after elution wherein its mass is determined. The mobile phase is mostly removed before the sample is introduced to the spectrometer. Detection can be via by total ion current (TIC) or selected ion monitoring (SIM). Mass spectrometry is able to distinguish analytes in overlapping peaks via ion monitoring as long as the analytes have a unique molecular ion or fragment ion. Mass spectrometry is very sensitive down to the femtogram per ml range for most analytes.
Fourier transform inferred (FTIR) spectroscopy: Infrared spectroscopy arises because different analytes produce different spectral fingerprints when exposed to infrared radiation. The Fourier transform converts the detector output to an interpretable spectrum that provides structural insights. Sensitivity is in the nanogram per ml range.
Nuclear magnetic resonance (NMR) spectroscopy: The sample is transferred directly into the NMR in the eluate solvent. The sample can be transferred during the chromatographic separation and the NMR spectra are then acquired either in on‐flow mode (continuously, while the chromatography is running), alternatively the NMR spectra are acquired under static conditions, requiring ‘parking’ of the analytes before analysis. Successful application has been seen with pharmaceutical analysis. Selectivity is in the femtogram per ml range.


[59,60]

[61,62]

[63]

Only after addressing these considerations can an appropriate strategy for sample pre‐treatment be determined.

7.1.4 Extraction and Cleanup Methods for Chromatographic Sample Preparation

Through sample extraction, it is possible to change the sample composition to suit the chromatographic separation method and/or reduce the sample complexity quite significantly. Common sample extraction methods used for biological samples prior to chromatographic separation (Table 7.4) and subsequent sample cleanup techniques that can be applied to an extracted sample (Table 7.5) can, in many cases, be applied in series to yield a sample that is ‘clean enough’ to be further processed via chromatographic separation.

Table 7.4 Sample extraction methods commonly used prior to chromatographic separation of biological samples.

Extraction method Description and technical considerations References
Solid–liquid extraction The sample is placed in a sealed container and solvent is added that dissolves/extracts/leaches the analyte of interest. The solution is then separated from the solid by filtration or centrifugation. Particularly useful for extracting small molecules from biological samples. [6466]
Liquid–liquid extraction Also known as solvent extraction or partitioning. Analytes in the sample are separated or portioned between two immiscible liquids (usually polar water and a non‐polar organic solvent) based on their relative solubility in each of the liquids. Particularly useful for extracting small molecules, pharmaceutical compounds and secondary metabolites. [67,68]
Homogenisation Biological tissue: Sample is placed in a blender or a mechanical homogeniser containing a solvent or aqueous buffer. The sample is homogenised to a homogeneous finely ‘chopped’ state. The liquid or suspended solids can be removed (filtration/centrifugation) for further workup.
Microbial cultures: The culture is first harvested via centrifugation to yield a cell pellet. The pellet is then resuspended in a cold aqueous buffer and before mechanical homogenisation at high pressure (> 600 bar/8700 psi). Homogenisation is aided through the use of detergents. It is usual to carry out 3–5 passes through the homogeniser using optical density and/or microscopy to monitor cell lysis.
[6971]
Sonication The use of sound waves to vigorously agitate particles in a solution. The sound waves can be applied via the use of a probe immersed into the sample or through sound waves being applied to a sample container immersed in an ultrasonic bath. Particularly useful for resuspending solid samples (e.g. protein aggregates) and lysing small bacterial culture volumes, although care must be taken to not denature protein and nucleic acid analytes of interest. Also, can be used to generate emulsions and nano‐emulsions with more homogeneous characteristics. [7275]

Table 7.5 Sample ‘clean‐up’ methods used prior to chromatographic separation.

Clean‐up method Description and technical considerations References
Filtration Sample is passed through a paper or membrane filter (or SPE cartridge) to remove suspended particulates; 0.2 μm filtration is recommended for all samples prior to chromatographic separation to ensure sample homogeneity and to protect the stationary phase. Various filter modalities are available, ranging from charged membranes to depth filters designed to remove specific particulate sizes. Note that filter flushing may be needed to recover valuable samples from filter media in some cases.
Ultrafiltration can be used to concentrate samples and also remove specific impurities of a set MW by using filter membranes with pores of a specific molecular weight cut‐off (MWCO). This technique is available in centrifugation‐tube format.
[76]
Centrifugation and sedimentation Centrifugation: Sample is separated by centrifugal force to remove particulates to yield a clarified supernatant and a particulate pellet. Used to prepare cell‐free samples and to clarify protein and nucleic acid preparations. Often used as an alternative to filtration for smaller biological samples.
Sedimentation: The sample is allowed to settle under gravity undisturbed. Sedimentation is a slow process and the rate of sedimentation is dependent on Stokes's radius.
[77]
Solid‐phase extraction (SPE) A form of chromatography itself – in SPE the sample is passed through a small disposable cartridge or column filled with a stationary phase that is able to adsorb the analyte of interest while allowing contaminating analytes to pass through with little or no adsorption. Used frequently in an ion exchange (IEX) ‘charged’ format to remove contaminating proteins from test solutions. [ 1, 28,78]
Lyophilisation Also known as freeze drying. An aqueous sample is frozen followed by controlled water removal via sublimation under vacuum. Particularly useful for thermally sensitive analytes (e.g. proteins) and samples containing volatile compounds (which will be removed during the drying). Can be used to concentrate an analyte from a large volume for subsequent reconstitution in a chromatography‐appropriate buffer or solution. [ 10,79,80]
Evaporation Liquid is removed from the sample by gentle heating at atmospheric pressure, usually with flowing air (or under N2 gas). A vacuum can be applied to remove liquids with a low volatility. Generally used on small samples under centrifugal force to retain sample analytes. [81,82]
Dialysis Analytes move between two aqueous liquid phases across a semi‐permeable membrane. Analytes transfer from one liquid to the other, based on differential concentration. Adaptations to this technique utilise membranes that have specific MWCO pores for more controlled analyte processing. Generally used to remove proteins from samples and for buffer exchange. [83,84]
Precipitation Analytes can be ‘brought‐out’ of solution by modulation of their physicochemical properties. A common method for preparing proteins is to change the pH of the protein solution. As the pH approaches the isoelectric point of the protein it will precipitate out of solution and can be sedimented and harvested via centrifugation. An alternative method for protein precipitation is through the addition of ammonium sulphate, which will remove water from around the protein and cause it to precipitate out of solution. Both of these methods can be used to remove impurities while maintaining the analyte of interest in solution. [85,86]

7.1.5 Protein Separation

As alluded to earlier, applications of chromatography to ‘biology’ are many and varied. However, the analysis of proteins has always been of paramount importance. Biochemists have been studying proteins since the early nineteenth century and the efficient chromatographic separation techniques in use today were developed in the early 1940s and 1950s and porous organic polymers were developed as part of the Manhattan Project (to develop the first atomic bomb!). Tiselius developed the basic chromatographic principles of chromatographic frontal analysis (continuous feed of the sample solution; the first constituent that breaks through may be obtained pure) in the early 1940s and applied a refined set of these techniques to the analysis of ovalbumin, serum albumin and immunoglobulins in the 1950s. In the subsequent six decades many different forms of chromatographic separation for protein preparation (purification) and analysis (concentration and structural determination) have been developed.

7.2 General Considerations for Protein Separation by Chromatography

The first consideration for protein separation by chromatography is to be aware of problems specific to proteins that are not encountered in liquid chromatography of small molecules. The most obvious of these is size. In recent research, very small particles with no pores have been used. That notwithstanding, large pore particles (usually greater than 300 Å) must be used. The size is also responsible for slow diffusion in the mobile phase and slow mass transfer in and out of the stationary phase. The former can be an advantage at very low flowrates but the latter is more prevalent at normal flowrates and contributes to poor efficiency. Proteins may be denatured and so in preparative work it is prudent to work at close to natural conditions using aqueous buffers at pH not too far away from 7.4. It is best to avoid polar organic solvents such as methanol and acetonitrile. The use of very non‐polar stationary phases is also inadvisable given that proteins can denature the surface of these or even may adsorb irreversibly. Lastly, large proteins are heterogeneous and, in a small way, this contributes to band broadening.

Proteins have four main physicochemical properties that can be exploited by four different modes of chromatographic separation (Tables 7.1 and 7.2):

7.2.1 Size/Shape – Exploited by Size Exclusion Chromatography (SEC)

In size exclusion chromatography (SEC) (Figure 7.4a and b) the molecular sieving properties of porous materials are used to separate proteins based upon their size and shape. Traditionally in SEC, a column of microparticulate cross‐linked copolymers (styrene or divinylbenzene) and with a narrow range of pore sizes is in equilibrium with the mobile phase. More recently, it is common to use silica‐based particles typically with an immobilised diol surface for protein SEC. Large analytes are excluded from the pores and pass through the spaces between the particles and are the first analytes to elute from the column. Smaller analytes are distributed between the mobile phase inside and outside the particles and will therefore take longer to pass through the column. The smallest analytes are the last to elute from the column. The mobile phase within a particle is available to an analyte that is able to penetrate the particle. The distribution of an analyte in a SEC column is determined by the total volume of mobile phase inside and outside the particles. For a given type of particle, the distribution coefficient (K d ) of a particular analyte between the inner and outer mobile phase is a function of its molecular size. If the analyte is large and completely excluded from the mobile phase within the particle, K d = 0. If the analyte is sufficiently small to gain complete access to the inner mobile phase, K d = 1. However, a variation in pore size will mean that the K d values vary between 0 and 1. It is this complete variation of K d between these two limits that makes it possible to separate analytes within a narrow molecular size range on a given particle type. It should be noted that since the protein separations take place before t 0, SEC columns are usually longer and wider than those for other modes of chromatography.

Graph of AU vs. time displaying a fluctuating curve with arrows marking the Mab aggregates, Mab dimer, low molecular weight impurities, and buffer/excipients.

Figure 7.4 (a) Size exclusion chromatography – there should be no adsorption/desorption on and from the solid matrix. Differences in residence times in the column arise from the ‘size’ of the molecules, small molecules totally permeating the pores of the particles where flow is diffusion controlled and hence taking longer to traverse the column. They elute at t 0 with all the separations taking place before t 0. (b) Monitoring aggregation and impurities of monoclonal antibodies using size exclusion chromatography. Experimental conditions: column – Agilent Bio SEC‐3300 Å, 7.8 × 300 mm; mobile phase: 150 mM phosphate, pH 7; flowrate: 1.0 ml/min; temperature: ambient; sample: monoclonal antibody (10 μl, 5 mg/ml).

The sizing properties of SEC make it a very powerful technique for characterising proteins in their native conformation, but SEC can also be applied for desalting protein preparations and concentration of particular analytes. The application of SEC has also been successful for separating nucleic acids [ 6,87], viral particles [88], large molecular weight formulation products [89] and multicomponent cellular assemblies [90]. For all of the applications, good knowledge of common SEC stationary phase polymers and the associated technical considerations (Table 7.6) is important.

Table 7.6 Chromatographic stationary phase description and technical considerations for the preparation and analysis of proteins.

Mode Stationary phase description and technical considerations
Size exclusion chromatography (SEC) Low pressure stationary phase polymers: Dextran (Sephadex, Sephacryl), Agarose (Sepharose) and Polyacrylamide (Bio‐gels)
High pressure stationary phase polymers: Polyvinyl chloride (Fractogel) and Dextran (Superdex)
SEC resins are prone to compression and sometimes difficult to pack. For some applications, fine resolution is only obtained with excessively long columns (30–100 cm), which can be prohibitive on many systems. SEC columns should be cleaned and stored in 200 mM sodium acetate, 20% v/v ethanol. 0.5 M NaOH can be used for brief robust cleaning if needed.
Ion exchange chromatography (IEX) Anion exchangers:
Strong: Trimethylaminomethyl: ‐O‐CH2N+(CH3)3 (cellulose matrix) Triethylaminoethyl: ‐O‐CH2CH2N+(CH2CH3)3 (dextran or cellulose matrix)
Weak: Aminoethyl: ‐O‐CH2CH2N+H3 on an agarose matrix Diethylaminoethyl: ‐O‐CH2CH2N+(CH2CH3)2H (cellulose matrix)
Cation exchangers:
Strong: Sulpho: −SO3 , O‐CH2SO3 , O‐CH2CH2CH2SO3 (cellulose, dextran, or polystyrene matrix)
Weak: Carboxymethyl: O‐CH2COO (cellulose or dextran matrix)
It is important that IEX columns are adequately equilibrated prior to sample application so as to bind and prepare all available functional groups with a counter ion (for proteins this would be Na+Cl‐) and to apply a set mobile phase pH across the entire column. IEX can be used in gradient or step elution modes and elution can be via pH changes or counterion concentration changes. A final high ionic strength wash after the elution step will allow the column to be cleaned and regenerated for subsequent reuse. 0.5 M NaOH can be used for brief robust cleaning. IEX columns should be stored in 20% v/v ethanol between uses.
Hydrophobic interaction chromatography (HIC) Common HIC and RPC stationary phase chemistries:
Illustration displaying a triangular ascending shade from hydrophilic to hydrophobic with formulas of ether, polypropylene glycol, phenyl, butyl, hexyl, and C18 at the bottom.
The performance of HIC separation is mostly dependent on the properties of the protein and the stationary phase chemistry. The stronger the ligand hydrophobicity and/or the more surface hydrophobicity on the protein the stronger the interaction. However, these interactions are also affected by the presence of contaminating analytes, detergents, pH and the ligand density/degree of substitution. The dynamic binding of the analyte and the subsequent resolution is determined by the flowrate, salt concentration and mobile phase properties. Complete elution of bound species can be a problem if a very hydrophobic ligand is matched with a high salt concentration in the injection sample. Selecting a less hydrophobic ligand or a less substituted matrix can help solve this problem. Temperature can have an effect on some complex sample separations also. Columns must not be left with bound analytes and can be fully eluted and cleaned by 1 M NaOH and are usually stored in 20% v/v ethanol between uses.
Affinity chromatography Established affinity chromatography applications:
  • Purification of kinase and dehydrogenase enzymes through the immobilisation of nucleotides (5′‐AMP and 2′5′‐ADP)
  • Purification of heparin binding proteins (growth factors, DNA, lipoproteins, membrane receptors) via the immobilisation of heparin
  • Isolation and identification of DNA binding proteins through the immobilisation of DNA (specifically promoter sequences and regulatory elements)
  • Isolation of fatty acid binding proteins through the immobilisation of fatty acids
  • Isolation of immunoglobulins through the immobilisation of Protein A and Protein G ligands
  • Isolation of mRNA through the immobilisation of poly‐Thymine
  • Isolation of recombinant proteins through the engineering of fusion protein partners and metal affinity binding motifs

Elution of the bound protein (or other analyte) needs to be complete to ensure performance upon reuse. Elution is usually through the application of high ionic strength buffers (1.5 M KCl or 2 M NaCl) or changes to the pH of the mobile phase that change the surface charge of the protein and allow it to dissociate from the column. Cleaning of the resin can be carried out using detergents, chaotropes in low or high pH buffers containing 1.5 M NaCl. Strong acids and bases should not be applied to affinity resins so as to maintain the ligand integrity. Storage should be in 20% v/v ethanol.

7.2.2 Charge – Exploited by Ion Exchange Chromatography (IEX)

This form of chromatographic separation relies on charge–charge interactions between the analyte and the stationary phase. Ion exchange chromatography (IEX) (Figure 7.5a and b) is a very versatile form of chromatography that can be applied over a wide range of biomolecules, synthetic polymers and small molecules. Both cation and anion exchanger stationary phases have a high binding capacity and their retention and resolution can be modulated through the ionic strength and pH of the mobile phase. The exchangers are either acidic (cation exchanger, negatively charged) or basic (anion exchanger, positively charged). Analytes are exchanged on the stationary phase through ionic interactions (a positively charged analyte interacting with a negatively charged stationary phase functional group, and vice versa). IEX can be carried out using either a strong or weak exchanger, wherein the strength of the exchanger refers to the extent to which the ionisation state of the functional groups varies with pH. It is not the strength of the interaction between the stationary phase and the analyte. The selection of either a weak or strong exchanger (Table 7.6) depends on the nature of the separation. If the separation is using a wide pH range (gradient pH‐based elution) then a strong exchanger is preferable as it ensures that retention via ion exchange will take place at both extremes of pH. Weak anion exchangers should be used when the binding capacity of the stationary phase is known to change as the pH of the mobile phase varies. In most IEX method development exercises it is advisable to select and screen strong exchangers first before moving to a weak exchanger to, if required, introduce an additional element of selectivity.

Graph of mAU vs. minutes displaying 2 fluctuating curves for pH and salt, with an inset graph at the top right displaying two fluctuating curves.

Figure 7.5 (a) Ion‐exchange chromatography – typically immobilised quaternary ammonium ions might be used for a strong anion exchanger and immobilised sulphonic acid groups for a strong anion exchanger. Retention of a particular protein will depend on its pI and the mobile phase pH, which can be used to manipulate selectivity. Retention can also be manipulated by the ionic strength of the aqueous buffer mobile phase, increasing numbers of competing ions and reducing the retention of analyte ions. For gradient elution the mobile phase changes from one with low ionic strength to one with high ionic strength. (b) Comparison of salt gradient v pH gradient in ion‐exchange chromatography showing that the outcomes are complementary. Column: Proteomix SCX‐NP5, 5 μm, 4.6 × 250 mm PEEK; mobile phase: a 10 mM phosphate, pH 7.0 B: A + 0.5 M NaCl; flowrate: 1.0 ml/min; detection: UV, 280 nm column temperature: 25 °C; samples: NIST mAb injection, 60 μg (pI 9.18, in 12.5 mM histidine, pH 6.0).

For protein separation via IEX the isoelectric point (pI, wherein the protein has no overall net charge) of the protein of interest should be obtained as this will allow an appropriate exchanger and mobile phase to be selected. This is particularly important if a pH gradient is being used instead of a buffer gradient. At a pH lower than the pI, the protein will have a net positive charge (suitable for cation exchange), while at a pH above the pI the protein will have a net negative charge (suitable for anion exchange). However, the stability of the protein as pH varies and the physicochemical properties of any putative impurities in the sample should also be considered so as to ensure that chromatograms obtained are representative of the initial sample. IEX may also be successfully employed for the separation of nucleic acids [91], drugs and impurities [10].

7.2.3 Hydrophobicity – Exploited by Hydrophobic Interaction Chromatography (HIC) and Reversed‐Phase Chromatography (RPC)

Hydrophobic interaction chromatography (HIC) (Figure 7.6a and b) has been applied for protein separation for over 30 years [92,93]. Separation is based on the adsorption of hydrophobic areas on the surface of an analyte on to the stationary phase (or, in some models, partition into a less polar area of mobile phase close to the stationary phase). For proteins the majority of hydrophobic residues are buried in the centre of the protein protected from the aqueous environment of the surface; indeed, it is the formation of this hydrophobic core that drives protein folding in globular proteins. Hydrophilic amino acids on the protein surface are covered with an ordered layer of water. Hydrophobic ‘patches’ on a protein surface can be exposed by treating the protein with salt ions (usually ammonium sulphate), which preferentially take up the ordered water molecules and allow the hydrophobic areas to be exploited for capture and separation activities. Interactions are via weak van der Waals forces between the protein and the stationary phase. However, protein–protein interactions are possible via the exposed hydrophobic areas and may cause aggregation of the protein and eventual precipitation. HIC can be applied in both low pressure protein capture (selective initial isolation step) and high pressure analysis formats. For low pressure chromatography, HIC makes an ideal intermediate or polishing step (further treatment of the ‘captured’ protein) in protein purification. The binding of proteins to an HIC stationary phase is normally in high salt concentrations and elution proceeds via the gradual decrease in salt concentration. This gradual change in salt concentration will cause a rehydration of the protein surface and a loss of interaction with the stationary phase. Changing the mobile phase pH can also be used to elute HIC separations, but the resulting separation can be quite variable for complex samples as this changes the separation to a more mixed mode (HIC:IEX) format and requires specialised stationary phase chemistries, which must have good batch‐to‐batch reproducibility. When applied in an HPLC format, HIC can be used to separate very closely related analytes using very shallow gradients and small resin beads or particle sizes (≤5 μm). The stationary phases commonly used for HIC are naturally hydrophobic, containing groups such as butyl, octyl and phenyl attached to a matrix, which promotes more protein–matrix interaction rather than protein–protein interactions. This quality of HIC makes it very versatile and able to accommodate high loading capacities. Common stationary phases include Phenyl Sepharose and Phenyl‐5PW for low pressure HIC and Bio‐Gel TSK Phenyl and Spherogel TSK Phenyl for HIC HPLC.

Graph of mAU vs. minutes displaying 3 fluctuating curves labeled G1, G2, and G3, with a four-column tabular representation located at the top right.

Figure 7.6 (a) Hydrophobic interaction chromatography – as in reversed‐phase chromatography, retention is based on hydrophobic interaction/partition but in this case the stationary phase is less non‐polar so less retentive so that an aqueous mobile phase with little or no polar–organic component must be used to obtain retention, thereby avoiding denaturing the protein analytes. Retention can be manipulated by the ionic strength of the aqueous buffer mobile phase, increasing numbers of competing ions effectively, making the mobile phase more polar and driving hydrophobic areas of the protein on to the stationary phase. Therefore, for gradient elution the mobile phase changes from one with high ionic strength to one with low ionic strength. (b) Gradient optimisation for hydrophobic interaction chromatography (HIC). Column: Proteomix HIC Butyl‐NP5, 5 μm, NP 4.6 × 100 mm; mobile phase: A: 2 M ammonium sulphate, in 100 mM sodium phosphate pH 7.0; B: 100 mM sodium phosphate pH 7.0; flowrate: 0.5 ml/min. Detector: UV, 280 nm; column temperature: 30 °C; sample: NIST mAb 10 mg/ml (pI 9.18, in 12.5 mM histidine, pH 6.0); injection volume: 2 μl.

2 Graphs of mAU vs. minutes each displaying a curve with arrows pointing the peaks. One of the peaks found in the bottom graph is labeled IgG1.

Figure 7.7 (a) Affinity chromatography – interaction between the targeted protein and the immobilised ligand is highly selective, so much so that low efficiency arising from slow mass transfer of the protein on and off the ligand is usually not an issue. It is important that the immobilisation of the ligand is such that there are no non‐specific interactions with the bridging groups. (b) Specificity of Bio‐monolith Protein A to IgG1. Experimental conditions: column – Agilent Bio‐monolith Protein A; mobile phase: A (binding buffer) – sodium phosphate buffer, 20 mM, pH 8.0, B (eluting buffer) – citric acid, 0.1 M, pH 2.8; gradient: 0% B from 0 to 0.5 min (for washing), 100% B from 0.5–1.5‐ 2 min (for eluting), 0% B from 2.1–4 min (for equilibrating); sample: IgG1 (2.5 mg/ml) and E. coli lysate (10 mg/ml); injection: 5 μg; flowrate: 1.0 ml/min; detection: UV at 280 DAD from Agilent Bio‐inert 1260 LC system.

HIC is very similar to reversed‐phase chromatography (RPC, also commonly abbreviated as RPLC or RP‐HPLC). However, in RPC the mobile phase will contain a polar‐organic constituent, such as methanol or acetonitrile, that may promote sample denaturation, especially for proteins. RPC tends not to be suitable for protein purification, except in very special circumstances [79], but is a very versatile and commonly used application for protein analysis. RPC has a lower binding capacity than HIC. While HIC is commonly used for protein separation it has also been applied to lipids [94], carbohydrates [95] and nucleic acids [96]. Common HIC and RPC stationary phase chemistries (Table 7.6) can be used in either HIC or RPC but generally more hydrophobic chemistries are used in RPC with polar organic solvents in the mobile phase and less hydrophobic chemistries are used in HIC with highly or completely aqueous buffer containing mobile phases.

RPC is very much the most common mode of chromatography for small molecules [97]. Therefore, as far as proteins are concerned, RPC comes into its own in the elucidation of protein structure where it is used for peptide analysis of protein tryptic digests (Section 7.4).

7.2.4 Biological Function and/or Structure – Exploited by Affinity Chromatography

Affinity chromatography (Figure a and b) exploits the biological function or specific structure of an analyte for separation making it extremely specific and capable of purification to high purity from very complex sample mixtures [17]. It is this specificity that makes affinity chromatography very expensive to use. Affinity chromatography is probably best characterised by the purification of enzymes by immobilising substrates/ligands (Table 7.6) on to chromatography stationary phase matrices. The enzymes would bind to the immobilised substrate once introduced to the column [98,99]. The interaction between the stationary phase ligand and the analyte needs to be reversible so that the column can be reused and so the analytes can be eluted. Elution is usually carried out through the application of free‐ligand changes in pH or salt gradients. Affinity chromatography is now used as the primary capture step for purifying immunoglobulin molecules used for therapeutic applications, but is also used for separating nucleic acids, membrane‐bound proteins and even whole cells. The use of affinity methods requires a detailed knowledge of the biological function and to some extent structure of the analyte of interest. The stationary phases need to be relatively inert in terms of their binding characteristics but possess suitable chemistry to allow attachment of the ligand. In this way retention is exclusively by the affinity interaction and resolution is not lost under the influence of non‐specific interactions. Ligands are usually added along with a spacer of up to 10 units (usually hydrocarbon chains but can contain carbonyl or amine groups) in length to allow correct orientation of the ligand and stop steric hindrance during binding to closely immobilised ligands. The matrix is usually an agarose or dextran base. Unlike other forms of chromatographic separation, affinity chromatography can be carried out in both batch‐resin and packed‐column formats.

7.3 Engineering Proteins for Streamlined Chromatographic Separations

For the isolation of native proteins from biological tissue or clinical samples, the chromatography may be limited by the physicochemical properties of the protein of interest. However, with the advent of recombinant DNA technologies, proteins can now be engineered to be more easily purified via chromatographic methods. There are several options available when trying to engineer a protein for a more simplified purification. There are three main approaches for engineering proteins for more efficient purification.

  • The addition of a purification tag: Purification tags are small amino acid sequences (<20 residues) that can be used in conjunction with affinity chromatography for purification. Commonly used purification tags are:

    Immobilised metal affinity chromatography ( IMAC ) associated with the surface properties of a target protein. IMAC (also known as metal chelate chromatography) utilises immobilised divalent cations (Ni2+, Co2+, Zn2+, Cu2+) that interact via coordinate bonds with the imidazole groups of histidine, indole groups of tryptophan and to a lesser extent thiol groups of cysteines. Proteins harbouring these residues on their surface will be adsorbed to the stationary phase and can be selectively eluted by altering the pH of the mobile phase, the addition of a chelating agent (usually ethylenediaminetetraacetic acid [EDTA]) or a solution of a free functional group (imidazole, indole). The molecular biology techniques used to generate His‐tagged proteins are generated by molecular biology techniques (Section 7.2.3). Most IMAC resins have a cellulose base and incorporate a spacer arm between the metal ion and the resin bead. Proteins can be engineered via recombinant DNA technologies [100,101] to have these tags at either their N‐ or C‐terminal. The incorporation of 6× histidine residues is the most common form of tag used in IMAC separations, as cysteine incorporation tends to lead to protein aggregation and tryptophan incorporation can cause significant changes to the protein structure. Due to its specificity, IMAC can be used on very ‘dirty’ samples (crude cell extracts) and still achieve target protein purities of >95% from a single separation.

    Introduction of a FLAG‐Tag or S‐Tag on to the N‐terminal of a protein allows immunoaffinity chromatography to be used for protein purification. The FLAG tag (Asp‐Tyr‐Lys‐Asp‐Asp‐Asp‐Asp‐Lys) is an 8‐reside hydrophilic amino acid sequence that is recognised by an immobilised monoclonal antibody on the stationary phase. The binding on the FLAG tag to the stationary phase is Ca2+‐dependent. Elution is carried out via the introduction of EDTA to the column and subsequent chelation of the Ca2+. The FLAG tag can be removed through treatment of the eluate with enterokinase [102]. Similarly, the 15‐amino acid residue S‐tag (Lys‐Glu‐Thr‐Ala‐Ala‐Ala‐Lys‐Phe‐Glu‐Arg‐Gln‐His‐Met‐Asp‐Ser) derived from pancreatic ribonuclease A, can be used for protein purification, usually in batch preparations [103]. Removal of the S‐tag is via subtilisin treatment.

    Both of these small proprietary tag‐based purification systems are very useful for small preparations of recombinant proteins, but as the size of the preparation increases the financial costs may become prohibitive and the digestion product isolation can become more complex.

    Polyarginine C‐terminal tags have been used in conjunction with cation exchange chromatography for protein purification. The arginine tag is sequentially removed through the application of carboxypeptidase B [104].

  • The generation of a fusion protein: There are several systems (Chapter 6.2) available for the expression of fusion proteins that, like the small affinity tags discussed above, utilise recombinant DNA technologies. The fusion protein systems that have been characterised the most are glutathione S‐transferase (GST) and maltose binding protein (MBP). Both of these systems are supplied as proprietary kits [105].

    Both systems fuse the GST or MBP protein to the N‐ or C‐terminal of the protein of interest and then use immobilised ligands (Glutathione for GST fusions; amylose for MBP fusions) on agarose‐based stationary phases. Both systems can allow on‐column cleavage or elution via pH changes or the addition of free ligand and subsequent cleavage. Thrombin is used for GST cleavage and Factor Xa for MBP cleavage. If the digestion is carried out after the initial affinity capture step, then a follow‐on IEX separation is usually employed to separate the fusion protein from the protein of interest.

    The generation of a fusion protein not only allows for efficient purification of target proteins but also confers stability to otherwise unstable proteins [106] and can enhance solubility for aggregation‐prone protein preparations [107].

  • Manipulating the expression format of the protein: Recombinant DNA technologies have also allowed users to manipulate the expression format and titre of recombinant proteins in order to aid subsequent purifications. The use of strong induction promoters (the bacteriophage T7 promoter in E.coli [108] or the cytomegalovirus (CMV) promoter in mammalian cell cultures [109]) for expressing recombinant proteins has facilitated the generation of aggregated misfolded protein known as inclusion bodies [110]. These inclusion bodies can be efficiently isolated through cellular homogenisation, homogenate centrifugation and detergent washing to yield protein preparations that are >95% pure for the protein of interest. These washed inclusion bodies can then be solubilised and refolded [111] to generate soluble proteins that are easily captured through the application of IEX chromatography. This expression and purification scheme is applied to the generation of many bacterially expressed recombinant protein therapeutics that are generated on a commercial scale [112].

    Another application of recombinant DNA technologies, has allowed recombinant expressed proteins to be sorted and/or secreted to the periplasm (bacterial cells), cell membrane or growth media (bacterial, plant, insect and mammalian cells) for a more simplified purification strategy. As proteins are generated, specific sorting signals are built in to the amino acid sequence; these signals will ensure that a protein is sorted to the correct site of activity. These signals can be manipulated for engineering protein sorting [113,114]. Isolation of proteins from the periplasm of bacterial cells is carried out via an osmotic shock of the cells [115], while extracellular signals will ensure that expressed proteins are transported across the cell membrane into the growth media. As in the application of the strong promoter systems described above, sorting signals are much used in the generation of commercial‐scale protein therapeutics.

7.4 Example Chromatographic Separations of Biological Samples

7.4.1 On‐Column Protein Folding

  • Rationale: As described earlier (Section 7.3), the rapid expression of proteins in E.coli cell cultures can lead to the formation of inclusion bodies. Historically, these protein aggregates were seen as ‘lost’ products and efforts were made to try to express the protein in a more soluble format. However, the relatively high purity obtained through the generation and isolation of inclusion bodies has more recently made them more attractive for protein purification. There are many solubilisation and refolding protocols available for application to many proteins expressed as inclusions bodies. However, for some proteins the solubilisation and refolding procedures lead to a loss in yield and/or a reduction in product quality. On‐column refolding can in many circumstances circumvent these problems through stabilisation of the protein during the molecular shaping into its final conformation. In addition, on‐column refolding allows processing at much smaller volumes (refolding is usually carried out in volumes that are 20‐fold larger than the solubilisation and therefore necessitate large column loadings of dilute protein samples) and aids in the analysis and preparation of proteins generated via high throughput screening (HTS) technologies [116]. During method development activities combining the refolding and primary capture chromatography steps can simplify an overall purifications scheme. However, the on‐column refolding step has been employed as part of the primary capture chromatography step and after an initial chromatographic capture and crude purification.
  • Technical considerations: The application of on‐column protein refolding is dependent on the protein of interest, the available time, money and chromatographic hardware available. For inexperienced users it may be prudent to first define a test‐tube refolding protocol and then capture the column step for a refolding product and attempt to combine the two steps. Due to its high capacity and robust stationary phase chemistry, IEX is an ideal capture and separation mode for refold samples. The stationary phase will be exposed to chaotropes (e.g. 8 M urea or 5 M guanadine‐HCl), reducing agents (e.g. 10 mM dithiothreitol [DTT]), oxidising agents (e.g. 5 mM cystamine‐HCl) as well as extremes of pH, and as such will need to be properly cleaned, regenerated and stored after use. Both strong and weak exchangers can be applied and it is common to screen several exchangers for optimisation. Proteins should be loaded on to the column at concentrations of ≤10 mg/ml. The solubilisation sample should be carried out with ‘clean’ inclusion bodies that are devoid of extraction buffer components, e.g. detergents. Protein folding can be a spontaneous process both in vivo and in vitro in test‐tube preparations, but for immobilised protein it is advisable to slowly introduce the refolding environment through the application of a gradient. Elution is carried out using high salt buffers (0.5–1 M NaCl) or a change in the mobile phase pH.
  • Simplified method: A baseline AIEX protocol for on‐column refolding.

7.4.2 On‐Column Protein Refolding via Anion Exchange Chromatography

  1. Wash the inclusion body preparation to achieve a clean ‘white’ protein mass – this can be done with water or aqueous buffer/1 M urea mix.
  2. Store inclusion bodies as a 25–50% w/v slurry in the washing solution. This slurry can be stored at −80 °C prior to chromatographic processing.
  3. Prepare the solubilisation buffer (50 mM Tris‐HCl, 8 M urea, 10 mM DTT, pH 9.0) and then slowly add the inclusion body slurry to a final concentration of 10 mg/ml protein. Allow the solubilisation to mix for 30–60 minutes. Note that if a cation exchange chromatography (CIEX) column is used then the base buffer should be modified (e.g. sodium acetate).
  4. Equilibrate an anion exchange column with five column volumes of solubilisation buffer and then load the solubilisation mixture on to the column. Wash the column with five column volumes of solubilisation buffer.
  5. Refold the bound protein by applying the refold buffer (50 mM Tris‐HCl, pH 9.0) from 0 to 100% over 10 column volumes.
  6. Elute the column using a step elution with 50 mM Tris‐HCl, 0.5 M NaCl, pH 9.0.
  7. The column should be cleaned and stored after use.

For an example on‐column refolding of recombinant lysozyme (Figure 7.8a), showing the associated size exclusion separation of key elution samples from the on‐column refolding (Figure 7.8a), the diethylaminoethanol (DEAE) elution of the refolded lysozyme is resolved into conjoined two peaks. The peak 1 refolded sample shows a shift in retention time (16 minutes) compared to the solubilised load material (main peak max. at 7 minutes). The solubilisation sample resolves into two conjoined peaks (peak max. at 7 and 10 minutes, respectively). The DEAE elution peak 2 contains higher molecular weight material (lower retention time) with a similar retention time peak max. of 10 minutes to some of the solubilised material. The DEAE column cleaning step contains high molecular weight material similar to the solubilisation sample (peak max. at 7 minutes). The conclusions from this on‐column refolding experiment are that lysozyme can be captured post‐solubilisation and refolded using on‐column methods. The post peak material seen on the DEAE column contains higher molecular weight material that could be aggregates or misfolded protein. The cleaning step removes material that is likely to be non‐refolded protein.

Top: Graph displaying 3 overlapping curves for A280, %B, and cond. Bottom: Graph with 4 overlapping curves for A280_solubilisation, A280_elution peak 1, A280_elution peak 2, and A280_cleaning.

Figure 7.8 On‐column refolding. Recombinant lysozyme inclusion bodies (expressed in E.coli) were washed three time in purified water and then stored as a 56% w/v slurry at −80 °C. After thawing, 100 mg of slurry was added to solubilisation buffer (50 mM Tris‐HCl, 8 M urea, 10 mM DTT, pH 9.5) and stirred for 30 minutes for solubilisation. (a) The solubilisation was loaded on to a 1 ml DEAE FF column that had been pre‐equilibrated in solubilisation buffer. Then load washing was performed with solubilisation buffer. Refolding was carried out by introducing the refolding buffer (50 mM Tris‐HCl, pH 9.0) over 10 column volumes (0–100% B). The refolding protein was then eluted using 50 mM Tris–HCl, 0.5 M NaCl, pH 9.0 as a step elution for 10 column volumes. After elution, the column was cleaned with 0.5 M NaOH. All steps were carried out at 1 ml/min and the protein was monitored using A280. Elution fractions are highlighted as shaded regions. (b) Key fractions from the DEAE on‐column refold were diluted to 25 ml and separated using size exclusion chromatography (column: 5 ml of Bio‐Gel P‐6 gel filtration column) 0 at 1 ml per minute.

Associated analytical technologies: Assaying for protein structure can be a relatively complex undertaking. However, there are some techniques (circular dichroism, protein NMR or hydrogen‐deuterium exchange mass‐spectroscopy) that will provide detailed information on the folding dynamics and crucially the final 3D conformation of the protein. However, there are simpler analytical technologies that can be employed to monitor the changes in protein conformation, for example analytical ultracentrifugation and size exclusion chromatography.

7.4.3 Protein Structure Elucidation

Even after isolating an intact protein, it may still be difficult to characterise the purified product. However, such a ‘top‐down’ study of a large molecule can be complemented by a ‘bottom‐up’ approach to protein analysis. This could involve, for example, papain digestion of a monoclonal antibody, but this results in a few still relatively large fragments. On the other hand, deglycosylation of monoclonal antibodies allows glycan analysis by hydrophilic interaction liquid chromatography (HILIC) liquid chromatography–mass spectrometry (LC‐MS) to study the sugars responsible for molecular recognition, HILIC being a relatively recently popularised mode of LC involving a polar stationary phase and a mobile phase containing a high proportion of a polar organic solvent and a low proportion of aqueous buffer that is suitable for polar analytes and is characterised by good MS sensitivity [117]. This type of analysis is very valuable as it is post‐translational modifications such as glycosylation that endow proteins with many of their important properties. This is particularly the case in the study of monoclonal antibodies and ‘biosimilars’ used as biopharmaceutical drugs.

However, the most common form of the ‘bottom‐up’ approach is to carry out a tryptic digest of the protein and subsequently characterise the product peptides by LC (Figure 7.9) or, more frequently, LC‐MS or even 2D‐LC‐MS. This chromatography approach is now widely used and the chromatography used has been perfected to the point that the actual digestion is a significant variable. To this end, manufacturers are now introducing kits to carry out solid phase digestion to reduce the variability caused by time, temperature and autocatalytic digestion of the enzyme.

Graphs illustrating reproducibility of tryptic digestions using an immobilised enzyme kit system for 3 individual mAb digestions-UHPLC vanquish system, each displaying a fluctuating curve with an ascending line.

Figure 7.9 Reproducibility of tryptic digestions using an immobilised enzyme kit system. Three individual mAb Digestion–ns ‐ UHPLC Vanquish System, Acclaim C18 2 × 250 mm, 2 μm column.

Source: Courtesy of Thermo Fisher Scientific.

An even greater challenge is to determine low levels of biopharmaceutical drugs in biological fluids. This can be and is carried out on the intact protein but even here the bottom‐up approach can be applied. A signature product peptide may be found for a particular biopharmaceutical and levels of that signature peptide may be monitored as a surrogate for the biopharmaceutical.

7.5 Other Applications of Chromatography for Biological Sample Preparation and Analysis

While the content of this chapter has been focused mainly on the separation of proteins, advances in chromatographic hardware and column chemistry have allowed liquid and gas chromatography to be applied to many biologically relevant fields. This is not new. For many years catecholamines in biological fluids have been studied, e.g. by HPLC with electrochemical detection. Prostanoid levels have been monitored in the study of analgesic drugs and corticosteroids in the study of depression. Gas chromatography, after chemical derivatisation of analytes, can be used in the field of metabolomics and supercritical fluid chromatography has been used effectively with MS detection in the field of lipidomics. However, more recent illustrative examples include:

  • The purification of viral associated particles for vaccine production and characterisation [118].
  • Cell membrane biology and receptor characterisation [119].
  • The linkage of chromatography with mass spectroscopy (LC‐MS/GC‐MS) or NMR (LC‐NMR) has enabled the study of metabolic fluxes and perturbations in diseased cells [ 5,120] and engineered expression strains [121].
  • Clinical sample analysis has also benefitted from hyphenated techniques: lipid quantification [122], bile acid analysis [123] and prenatal screening [124].
  • The separation of gene expression products for pathogen identification and characterisation is also a relatively recent application of chromatography that has enabled a more efficient and cost‐effective approach to electrophoretic separation [125].

In summary, the application of chromatography to biological research has enabled biologists to gain a detailed insight into their sample composition and confidence in their data that was once only available to synthetic and analytical chemists.

That is not to say that chromatography in biology is a mature field. The pace of biomedical research shows no signs of abating. In the field of biopharmaceuticals the need for innovation is a constant and, going beyond large biological molecules as drugs, research into drug–antibody complexes and ‘new modalities’ such as oligonucleotides as drugs will throw up new challenges to the chromatographer.

References

  1. 1 Campíns‐Falcó, P., Sevillano‐Cabeza, A., Herráez‐Hernández, R. et al. (2012). Solid‐phase extraction and clean‐up procedures in pharmaceutical analysis. In: Encyclopedia of Analytical Chemistry, 1–17. Chichester, UK: Wiley.
  2. 2 Shabir, G.A., John Lough, W., Arain, S.A., and Bradshaw, T.K. (2007). Evaluation and application of best practice in analytical method validation. J. Liq. Chromatogr. Relat. Technol. 30: 311–333.
  3. 3 Hansen, S.H. and Pedersen‐Bjergaard, S. (2015). Analysis of small‐molecule drugs in biological fluids. In: Bioanalysis of Pharmaceuticals, 207–260. Wiley.
  4. 4 Samsonov, G.V., Pisarev, O.A., and Melenevsky, A.T. (1993). Chromatographic purification and superpurification of biologically active compounds using heteroreticular and composite ion exchange resins at low pressure. Pure Appl. Chem. 65: 2287–2290.
  5. 5 Pasikanti, K.K., Ho, P.C., and Chan, E.C.Y. (2008). Gas chromatography/mass spectrometry in metabolic profiling of biological fluids. J. Chromatogr. B 871: 202–211.
  6. 6 Latulippe, D.R. and Zydney, A.L. (2009). Size exclusion chromatography of plasmid DNA isoforms. J. Chromatogr. A 1216: 6295–6302.
  7. 7 Hong, P., Koza, S., and Bouvier, E.S.P. (2012). Size‐exclusion chromatography for the analysis of protein biotherapeutics and their aggregates. J. Liq. Chromatogr. Relat. Technol. 35: 2923–2950.
  8. 8 Ó'Fágáin, C., Cummins, P.M., and O'Connor, B.F. (2017). Gel‐filtration chromatography. In: Methods in Molecular Biology, 15–25. Springer.
  9. 9 Budelier, K. and Schorr, J. (2001). Purification of DNA by anion‐exchange chromatography. In: Current Protocols in Molecular Biology, Unit 2.1B. Hoboken, NJ, USA: Wiley.
  10. 10 Bolto, B., Dixon, D., and Eldridge, R. (2004). Ion exchange for the removal of natural organic matter. React. Funct. Polym. 60: 171–182.
  11. 11 Burness, A.T.H. and Pardoe, I.U. (1983). Chromatofocusing of sialoglycoproteins. J. Chromatogr. A 259: 423–432.
  12. 12 Mohammad, J. (2010). Chromatofocusing. Cold Spring Harb. Protoc. 2010: pdb.top67.
  13. 13 Canene‐Adams, K. (2013). Reverse‐phase HPLC analysis and purification of small molecules. Methods Enzymol. 533: 291–301.
  14. 14 Cummins, P.M. and O'Connor, B.F. (2011). Hydrophobic interaction chromatography. Methods Mol. Biol. 681: 431–437.
  15. 15 Chockalingam, P.S., Jurado, L.A., and Jarrett, H.W. (2001). DNA affinity chromatography. Mol. Biotechnol. 19: 189–199.
  16. 16 Urh, M., Simpson, D., and Zhao, K. (2009). Chapter 26: Affinity chromatography. In: Methods in Enzymology, 417–438. Elsevier.
  17. 17 Wiederschain, G.Y. (2007). Handbook of affinity chromatography. Biochemist 72: 793–793. Portland Press.
  18. 18 West, I. and Goldring, O. (1994). Lectin affinity chromatography. Mol. Biotechnol. 2: 147–155.
  19. 19 Norsko, J.K. (1990). Chemisorption on metal surfaces. Reports Prog. Phys. 53: 1253–1295.
  20. 20 Toyo'oka, T. (2009). Recent advances in separation and detection methods for thiol compounds in biological samples. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 877: 3318–3330.
  21. 21 Porath, J. (1992). Immobilized metal ion affinity chromatography. Protein Expr. Purif. 3: 263–281.
  22. 22 Tsai, C.F., Wang, Y.T., Lin, P.Y., and Chen, Y.J. (2011). Phosphoproteomics by highly selective IMAC protocol. NeuroMethods 57: 181–196.
  23. 23 Gilbert, M.T. (1987). Chiral chromatography. In: High Performance Liquid Chromatography, 291–312. Elsevier.
  24. 24 Lam, S. (1989). Chiral liquid chromatography. In: Chiral Liquid Chromatography (ed. W. Lough), 83–101. Blackie Publishing Group.
  25. 25 Lämmerhofer, M. (2012). Chiral separations and enantioselectivity. J. Chromatogr. A 1269: 1–2.
  26. 26 Asnin, L. (2012). Adsorption models in chiral chromatography. J. Chromatogr. A 1269: 3–25.
  27. 27 Fiori, M., Scintu, M.F., and Addis, M. (2013). Characterization of the lipid fraction in lamb meat: comparison of different lipid extraction methods. Food Anal. Methods 6: 1648–1656.
  28. 28 Perrone, P.A. and Brown, P.R. (1984). Ion‐pair chromatography of nucleotides. J. Chromatogr. A 317: 301–310.
  29. 29 Cecchi, T. and Passamonti, P. (2009). Retention mechanism for ion‐pair chromatography with chaotropic reagents. J. Chromatogr. A 1216: 1789–1797.
  30. 30 Dolan, J.W., Kirkland, J.J., and Snyder, L.R. (2010). Introduction to Modern Liquid Chromatography, 3e. Wiley.
  31. 31 Purnell, J.H. (1966). The correlation of separating power and efficiency of gas‐chromatographic columns. J. Chem. Soc. 1268–1274.
  32. 32 Miyabe, K. and Guiochon, G. (1999). Peak tailing and column radial heterogeneity in linear chromatography. J. Chromatogr. A 830: 263–274.
  33. 33 Miyabe, K., Matsumoto, Y., Niwa, Y. et al. (2009). An estimation of the column efficiency made by analyzing tailing peak profiles. J. Chromatogr. A 1216: 8319–8330.
  34. 34 Stoll, D.R. (2010). Recent progress in online, comprehensive two‐dimensional high‐performance liquid chromatography for non‐proteomic applications. Anal. Bioanal. Chem. 397: 979–986.
  35. 35 Anspach, F.B., Curbelo, D., Hartmann, R. et al. (1999). Expanded‐bed chromatography in primary protein purification. J. Chromatogr. A 865: 129–144.
  36. 36 Co, C.C., Ho, C.‐C., and Kumar, G. (2012). Motility‐based cell sorting by planar cell chromatography. Anal. Chem. 84: 10160–10164.
  37. 37 Moldoveanu, S. and David, V. (2015). Modern Sample Preparation for Chromatography, 1–453. Elsevier.
  38. 38 Kolars, J.C., Lown, K.S., Schmiedlin‐Ren, P. et al. (1994). CYP3A gene expression in human gut epithelium. Pharmacogenetics 4: 247–259.
  39. 39 Holland, N.T., Smith, M.T., Eskenazi, B., and Bastaki, M. (2003). Biological sample collection and processing for molecular epidemiological studies. Mutat. Res. 543: 217–234.
  40. 40 Note S (2001) Biological Sample Preparation 331.1. Molecules 1–12.
  41. 41 Stadtman, E.R. and Levine, R.L. (2006). Protein oxidation. Ann. NY Acad. Sci. 899: 191–208.
  42. 42 Laurence, J.S. and Middaugh, C.R. (2010). Fundamental structures and behaviors of proteins. In: Aggregation of Therapeutic Proteins, 1–61. Wiley.
  43. 43 Konermann, L. (2012). Protein Unfolding and Denaturants. In: eLS. Wiley.
  44. 44 Christie, W. and Han, X. (2012). Lipid Analysis: Isolation, Separation, Identification and Lipidomic, 2e. Pergamon.
  45. 45 Dell, A. and Reason, A.J. (1993). Carbohydrate analysis. Curr. Opin. Biotechnol. 4: 52–56.
  46. 46 Saulnier, L., Vigouroux, J., and Thibault, J.‐F. (1995). Isolation and partial characterization of feruloylated oligosaccharides from maize bran. Carbohydr. Res. 272: 241–253.
  47. 47 Stick, R.V. (2001). Carbohydrate‐based vaccines. In: Carbohydrates, 233–237. Elsevier.
  48. 48 Yu, C., Cohen, L.H., and Arbor, A. (2004). Tissue sample preparation — not the same old grind. Sample Prep. Perspect. 2–6.
  49. 49 Guthrie, J.W. (2012). General considerations when dealing with biological fluid samples. In: Comprehensive Sampling and Sample Preparation, 1–19. Elsevier.
  50. 50 Kralicek, A.V. and Ozawa, K. (2011). Advances in cell‐free protein synthesis for NMR sample preparation. In: Biomolecular NMR spectroscopy, 3–19.
  51. 51 Penquer, A. (2009). Review of the available uv detectors. EAS Publ. Ser. 37: 193–198.
  52. 52 Rutan, S. (1993). Diode array detection in HPLC. J. Chromatogr. A 657: 457.
  53. 53 Krattiger, B., Bruin, G.J., and Bruno, A. (1994). Refractive index detector. Anal. Chem. 66: 11A–15A.
  54. 54 Kohler, M., Haerdi, W., Christen, P., and Veuthey, J.L. (1997). The evaporative light scattering detector: some applications in pharmaceutical analysis. Trends Anal. Chem. 16: 475–484.
  55. 55 Mathews, B.T., Higginson, P.D., Lyons, R. et al. (2004). Improving quantitative measurements for the evaporative light scattering detector. Chromatographia 60: 625–633.
  56. 56 Geng, X., Shi, M., Ning, H. et al. (2018). A compact and low‐cost laser induced fluorescence detector with silicon based photodetector assembly for capillary flow systems. Talanta 182: 279–284.
  57. 57 Luci, G., Intorre, L., Ferruzzi, G. et al. (2018). Determination of ochratoxin a in tissues of wild boar (sus scrofa L.) by enzymatic digestion (ED) coupled to high‐performance liquid chromatography with a fluorescence detector (HPLC‐FLD). Mycotoxin. Res. 34 (1): 1–8.
  58. 58 McMinn, D.G. (1992). The flame ionization detector. Detect. Capill. Chromatogr. 23: 7–21.
  59. 59 Cohen, S., Gagnieu, M.‐C., Lefebvre, I., and Guitton, J. (2013). LC‐MS bioanalysis of nucleotides. In: Handbook of LC‐MS Bioanalysis, 559–572.
  60. 60 Yan, Z., Cheng, C., and Liu, S. (2012). Applications of mass spectrometry in analyses of steroid hormones. In: LC‐MS in Drug Bioanalysis, 251–286. Boston, MA: Springer.
  61. 61 Mohamed, M.A., Jaafar, J., Ismail, A.F. et al. (2017). Fourier transform infrared (FTIR) spectroscopy. In: Membrane Characterization, 3–29. Elsevier.
  62. 62 Subramanian, A. and Rodriguez‐Saona, L. (2009). Fourier transform infrared (FTIR) spectroscopy. In: Infrared Spectroscopy for Food Quality Analysis and Control, 145–178. Elsevier.
  63. 63 Silva Elipe, M.V. (2011). LC‐NMR and Other Hyphenated NMR Techniques, 1–220. Wiley.
  64. 64 Simeonov, E., Tsibranska, I., and Minchev, A. (1999). Solid–liquid extraction from plants — experimental kinetics and modelling. Chem. Eng. J. 73: 255–259.
  65. 65 Bucić‐Kojić, A., Planinić, M., Tomas, S. et al. (2007). Study of solid–liquid extraction kinetics of total polyphenols from grape seeds. J. Food Eng. 81: 236–242.
  66. 66 Majors, R.E. (2013). Novel sorbents for solid‐liquid extraction. LC GC Eur. 26: 685–691.
  67. 67 Golumbic, C. (1951). Liquid‐liquid extraction analysis. Anal. Chem. 23: 1210–1217.
  68. 68 Tedder, D.W. (2014). Liquid‐liquid extraction. In: Albr'ght's Chemical Engineering Handbook. CRC Press, 709–738.
  69. 69 Frank, K., Köhler, K., and Schuchmann, H.P. (2012). Stability of anthocyanins in high pressure homogenisation. Food Chem. 130: 716–719.
  70. 70 Ouzineb, K., Lord, C., Lesauze, N. et al. (2006). Homogenisation devices for the production of miniemulsions. Chem. Eng. Sci. 61: 2994–3000.
  71. 71 Shao, S., Guo, T., Gross, V. et al. (2016). Reproducible tissue homogenization and protein extraction for quantitative proteomics using micropestle‐assisted pressure‐cycling technology. J. Proteome Res. 15: 1821–1829.
  72. 72 Oldenburg, K., Pooler, D., Scudder, K. et al. (2005). High throughput sonication: evaluation for compound solubilization. Comb. Chem. High Throughput Screen. 8: 499–512.
  73. 73 Mahdi Jafari, S., He, Y., and Bhandari, B. (2006). Nano‐emulsion production by sonication and microfluidization – a comparison. Int. J. Food Prop. 9: 475–485.
  74. 74 Stepanskiy, L.G. (2012). Sonication‐induced unfolding proteins. J. Theor. Biol. 298: 77–81.
  75. 75 Campaniello, D., Bevilacqua, A., Sinigaglia, M., and Corbo, M.R. (2016). Using homogenization, sonication and thermo‐sonication to inactivate fungi. PeerJ 4: e2020.
  76. 76 Scheer, L. (2009). The use of syringe filters for analytical sample preparation: including HPLC and dissolution testing. Filtration 9: 308–309.
  77. 77 Fabry, T.L. (1987). Mechanism of erythrocyte aggregation and sedimentation. Blood 70: 1572–1576.
  78. 78 Pawliszyn, J. (2012). Theory of solid‐phase microextraction. In: Handbook of Solid Phase Microextraction, 13–59. Elsevier.
  79. 79 Mollerup, I., Jensen, S.W., Larsen, P. et al. (2002). Insulin, purification. Encycl. Bioprocess Technol. 992: 1219–1222.
  80. 80 Teagarden, D.L., Speaker, S.M., Maetin, S.W.H., and Österberg, T. (2010). Practical considerations for freeze‐drying in dual chamber package systems. In: Freeze Drying/Lyophilisation of Pharmaceutical and Biological Products, 494–526. CRC Press.
  81. 81 Burtis, C.A. (1990). Sample evaporation and its impact on the operating performance of an automated selective‐access analytical system. Clin. Chem. 36: 544–546.
  82. 82 Walker, G.M. and Beebe, D.J. (2002). Evaporation‐driven microfluidic sample concentration. In: 2nd Annual International IEEE‐EMBS Special Topic Conference on Microtechnologies in Medicine and Biol–gy ‐ Proceedings, 523–526.
  83. 83 Boutelle, M.G. and Fillenz, M. (1996). Clinical microdialysis: I role of on‐line measurement and quantitative microdialysis. In: Clinical Aspects of Microdialysis, 13–20.
  84. 84 Lange, E.C.M. (2013). Microdialysis in Drug Development, 13–33. New York, NY: Springer.
  85. 85 Baduel, C., Mueller, J.F., Tsai, H., and Gomez Ramos, M.J. (2015). Development of sample extraction and clean‐up strategies for target and non‐target analysis of environmental contaminants in biological matrices. J. Chromatogr. A 1426: 33–47.
  86. 86 Ng, L.L. (1983). Sample preparation by salts precipitation and quantitation by high‐performance liquid chromatography with uv detection of selected drugs in biological fluids. J. Chromatogr. A 257: 345–353.
  87. 87 Ellegren, H. and Låås, T. (1989). Size‐exclusion chromatography of DNA restriction fragments. Fragment length determinations and a comparison with the behaviour of proteins in size‐exclusion chromatography. J. Chromatogr. A 467: 217–226.
  88. 88 Vajda, J., Weber, D., Brekel, D. et al. (2016). Size distribution analysis of influenza virus particles using size exclusion chromatography. J. Chromatogr. A 1465: 117–125.
  89. 89 Ruysschaert, T., Marque, A., Duteyrat, J.L. et al. (2005). Liposome retention in size exclusion chromatography. BMC Biotechnol. 5: 11.
  90. 90 Benedikter, B.J., Bouwman, F.G., Vajen, T. et al. (2017). Ultrafiltration combined with size exclusion chromatography efficiently isolates extracellular vesicles from cell culture media for compositional and functional studies. Sci. Rep. 7: 15297.
  91. 91 Westman, E., Eriksson, S., Låås, T. et al. (1987). Separation of DNA restriction fragments by ion‐exchange chromatography on FPLC columns mono P and mono Q. Anal. Biochem. 166: 158–171.
  92. 92 Kennedy, R.M. (1990). Hydrophobic chromatography. Methods Enzymol. 182: 339–343.
  93. 93 Queiroz, J.A., Tomaz, C.T., and Cabral, J.M.S. (2008). Hydrophobic interaction chromatography of proteins. J. Chromatogr. A 1205: 46–59.
  94. 94 Fischer, W. (1996). Molecular analysis of lipid macroamphiphiles by hydrophobic interaction chromatography. J. Microbiol. Methods 7025: 129–144.
  95. 95 El Rassi, Z. (1995). Reversed‐phase and hydrophobic interaction chromatography of carbohydrates and glycoconjugates. J. Chromatogr. Libr. 58: 41–101.
  96. 96 Savard, J.M. and Schneider, J.W. (2007). Sequence‐specific purification of DNA oligomers in hydrophobic interaction chromatography using peptide nucleic acid amphiphiles: extended dynamic range. Biotechnol. Bioeng. 97: 367–376.
  97. 97 Lough, W. (2000). Reversed‐phase liquid chromatography. In: Encyclopaedia of Analytical Chemistry, 11442–11450. Wiley.
  98. 98 Cherkasov, I.A. (1972). Affinity chromatography of enzymes. Russ. Chem. Rev. 41: 891–903.
  99. 99 Farooqui, A.A. (1980). Purification of enzymes by heparin‐sepharose affinity chromatography. J. Chromatogr. A 184: 335–345.
  100. 100 Cheng, Y., Liu, D., Feng, Y., and Jing, G. (2003). An efficient fusion expression system for protein and peptide overexpression in Escherichia coli and NMR sample preparation. Protein Pept. Lett. 10: 175–181.
  101. 101 Loughran, S.T., Loughran, N.B., Ryan, B.J. et al. (2006). Modified his‐tag fusion vector for enhanced protein purification by immobilized metal affinity chromatography. Anal. Biochem. 355: 148–150.
  102. 102 Einhauer, A. and Jungbauer, A. (2001). The FLAGTM peptide, a versatile fusion tag for the purification of recombinant proteins. J. Biochem. Biophys. Methods 49: 455–465.
  103. 103 Raines, R.T., McCormick, M., Van Oosbree, T.R., and Mierendorf, R.C. (2000). The S.Tag fusion system for protein purification. Methods Enzymol. 326: 362–376.
  104. 104 Fuchs, S.M. and Raines, R.T. (2009). Polyarginine as a multifunctional fusion tag. Protein Sci. 14: 1538–1544.
  105. 105 GE Healthcare (2009). Glutathione S‐transferase (GST) gene fusion system. GST Gene Fusion Syst. 1–8.
  106. 106 Costa, S., Almeida, A., Castro, A., and Domingues, L. (2014). Fusion tags for protein solubility, purification and immunogenicity in Escherichia coli: the novel Fh8 system. Front. Microbiol. 5: 63.
  107. 107 Rabhi‐Essafi, I., Sadok, A., Khalaf, N., and Fathallah, D.M. (2007). A strategy for high‐level expression of soluble and functional human interferon alpha as a GST‐fusion protein in E. coli. Protein Eng. Des. Sel. 20: 201–209.
  108. 108 Tabor, S. (2001). Expression using the T7 RNA polymerase/promoter system. In: Current Protocols in Molecular Biology. Wiley, Chapter 16: Unit 16.2.
  109. 109 Moritz, B., Becker, P.B., and Göpfert, U. (2015). CMV promoter mutants with a reduced propensity to productivity loss in CHO cells. Sci. Rep. 5: 16952.
  110. 110 Ramón, A., Señorale‐Pose, M., and Marín, M. (2014). Inclusion bodies: not that bad…. Front. Microbiol. 5 (56): 1–6.
  111. 111 Singh, A., Upadhyay, V., and Panda, A.K. (2015). Solubilization and refolding of inclusion body proteins. In: Insoluble Proteins: Methods and Protocols, 283–291.
  112. 112 Panda, A.K. (2003). Bioprocessing of therapeutic proteins from the inclusion bodies of. Adv. Biochem. Eng. Biotechnol. 85: 43–93.
  113. 113 Haryadi, R., Ho, S., Kok, Y.J. et al. (2015). Optimization of heavy chain and light chain signal peptides for high level expression of therapeutic antibodies in CHO cells. PLoS One 10: e0116878.
  114. 114 Ng, D.T.W. and Sarkar, C.A. (2013). Engineering signal peptides for enhanced protein secretion from Lactococcus lactis. Appl. Environ. Microbiol. 79: 347–356.
  115. 115 Rathore, A.S., Bilbrey, R.E., and Steinmeyer, D.E. (2003). Optimization of an osmotic shock procedure for isolation of a protein product expressed in E. coli. Biotechnol. Prog. 19: 1541–1546.
  116. 116 Oganesyan, N., Kim, S.H., and Kim, R. (2005). On‐column protein refolding for crystallization. J. Struct. Funct. Genom. 6: 177–182.
  117. 117 McCalley, D.V. (2017). Understanding and manipulating the separation in hydrophilic interaction liquid chromatography. J. Chromatogr. A 1523: 49–71.
  118. 118 Cazares, L.H., Ward, M.D., Brueggemann, E.E. et al. (2016). Development of a liquid chromatography high resolution mass spectrometry method for the quantitation of viral envelope glycoprotein in Ebola virus‐like particle vaccine preparations. Clin. Proteomics 13: 18.
  119. 119 Caculitan, N.G., Kai, H., Liu, E.Y. et al. (2014). Size‐based chromatography of signaling clusters in a living cell membrane. Nano Lett. 14: 2293–2298.
  120. 120 Watson, G.W., Wickramasekara, S., Maier, C.S. et al. (2015). Assessment of global proteome in LNCaP cells by 2D‐RP/RP LC‐MS/MS following sulforaphane exposure. EuPA Open Proteom. 9: 34–40.
  121. 121 Albrecht, S., Kaisermayer, C., Reinhart, D. et al. (2018). Multiple reaction monitoring targeted LC‐MS analysis of potential cell death marker proteins for increased bioprocess control. Anal. Bioanal. Chem. 410: 3197–3207.
  122. 122 Blanchard, V., Ramin‐Mangata, S., Billon‐Crossouard, S. et al. (2018). Kinetics of plasma apolipoprotein E isoforms by LC‐MS/MS: a pilot study. J. Lipid Res. 59: 892–900.
  123. 123 Krautbauer, S. and Liebisch, G. (2018). LC‐MS/MS analysis of bile acids. In: Methods in Molecular Biology, vol. 1730, 103–110. Springer.
  124. 124 López Uriarte, G.A., Burciaga Flores, C.H., Torres de la Cruz, V.M. et al. (2018). Proteomic profile of serum of pregnant women carring a fetus with down syndrome using nano uplc Q‐tof ms/ms technology. J. Matern. Neonatal. Med. 31: 1483–1489.
  125. 125 Ueda, S., Yamaguchi, M., Eguchi, K., and Iwase, M. (2016). Identification of Cereulide‐producing Bacillus cereus by nucleic acid chromatography and reverse transcription real‐time PCR. Biocontrol Sci. 21: 45–50.

Further Reading

  1. Conductivity E, Tds S (2004) 760. Electrical Conductivity/Salinity Fact Sheet. Water Resour 0:2–6.
  2. Dong, M.W. (2006). Modern HPLC for Practising Scientists. Wiley.
  3. Gooding, K.M. and Regnier, F.E. (2002). HPLC of Biological Macro‐ Molecules, Revised and Expanded. CRC Press; Taylor and Francis Group.
  4. Grigore, M.‐N., Ivanescu, L., and Toma, C. (2014). Halophytes: An Integrative Anatomical Study. Springer International Publishing.
  5. Snyder, L.R. (1992). Chapter 1, Theory of chromatography. In: Journal of the Chromatography Library, A1–A68.
  6. Snyder, L.R., Joseph, J.K., and Dolan, J.W. (2010). Ionic samples: reversed‐phase, ion‐pair, and ion‐exchange chromatography. In: Introduction to Modern Liquid Chromatography, 303–360. Hoboken, NJ, USA: Wiley.
  7. Striegel, A.M., Yau, W.W., Kirkland, J.J., and Bly, D.D. (2009). Modern Size Exclusion Chromatography: Practice of Gel Permeation and Gel Filtration Chromatography, 2e. Wiley.
  8. Timms, J.F. and Cutillas, P.R. (2010). Overview of quantitative LC–MS techniques for proteomics and activitomics. Methods Mol. Biol. 658: 19–45.
  9. Walls, D. and Loughran, S. (2011). Protein Chromatography: Methods and Protocols. Springer.
..................Content has been hidden....................

You can't read the all page of ebook, please click here login for view all page.
Reset