Open access peer-reviewed chapter

Immunoassay Techniques Highlighting Biomarkers in Immunogenetic Diseases

Written By

Emilia Manole, Alexandra E. Bastian, Ionela D. Popescu, Carolina Constantin, Simona Mihai, Gisela F. Gaina, Elena Codrici and Monica T. Neagu

Submitted: 08 November 2017 Reviewed: 26 February 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.75951

From the Edited Volume

Immunogenetics

Edited by Nima Rezaei

Chapter metrics overview

1,862 Chapter Downloads

View Full Metrics

Abstract

Diagnosis of autoimmune diseases is crucial for the clinician and the patient alike. The immunoassay techniques most commonly used for this purpose are immunohistochemistry, ELISA, and Western blotting. For the detection of more specific biomarkers or the discovery of new ones for diagnostic purposes and as therapeutic targets, microarray techniques are increasingly used, for example, protein microarray, Luminex, and in recent years, surface plasmon resonance imaging. All of these technologies have undergone changes over time, making them easier to use. Similar technologies have been invented but responding to specific requirements for both diagnostic and research purposes. The goals are to study more analytes in the same sample, in a shorter time, and with increased accuracy. The reproducibility and reliability of the results are also a target pursued by manufacturers. In this chapter, we present these technologies and their utility in the diagnosis of immunogenetic diseases.

Keywords

  • immunoassay
  • protein biomarkers
  • autoimmune diseases
  • IHC
  • ELISA
  • WB
  • protein microarray
  • SPRi
  • Luminex

1. Introduction

An autoimmune pathology occurs when the immune system loses its ability to distinguish between its own cells and nonself cells, inducing the attack of self-tissue. This mechanism involves both the environmental factors and the genetic predisposition of the individual.

Proteomic technologies identify and separate different proteins of interest from biological samples, thus enabling their characterization as biomarkers, establishing their interactions, their role and the mechanisms in which they are involved, the identification of new diagnostic and therapeutic targets. The identification of protein biomarkers may be the basis for developing new methods of early diagnosis and treatment [1]. In general, an ideal biomarker should meet certain characteristics: be specific to a particular disease, be validated and confirmed as having specificity for that pathology, be able to early identify the disease, its testing to be easy and cheap as far as possible, reliable, and noninvasive [2, 3].

Although important advances have been made in deciphering immune function, the understanding of this function dysregulation and the specific autoimmune response remains limited. The domain is complex and includes, besides the disturbance of immune system functioning, gene alterations that regulate and control the self-tolerance. In this chapter, we will describe the techniques of highlighting the proteomic biomarkers involved in the pathogenesis of immunogenetic diseases.

In the case of immunogenetic diseases, one of the tissues that are first tested for specific biomarkers is blood, namely, the serum, which contains approximately 60–80 mg/mL proteins, besides amino acids, lipids, salts, and carbohydrates [4]. Applying proteomic immunoassay techniques for the diagnosis of immunogenetic diseases may also predict the course of disease, or result in a personalized treatment for patients [5, 6].

Proteomic biomarkers are particularly useful for providing the information on cellular signaling pathways, bringing early disease data, monitoring treatment response or adverse effects. They can be monitored from body fluids other than blood, such as: urine, saliva, cerebrospinal fluid and from different tissues (biopsies) [7].

The necessity to analyze very small amounts of proteins present in biological samples [8], as well as the increase in the number of proteins requiring simultaneous, reliable, reproducible, and significant investigations led to the modernization of the existing techniques and to the appearance of some new methods of biomarker investigation and analysis. Immunohistochemistry, ELISA, and Western blotting are of the old methods that changed, adapted, modernized over time, but remained “on barricades” for protein biomarker investigation, especially in autoimmune disorders. Immunoassay methodologies are the most commonly used tools in protein research, using the properties of antibodies to bind different protein domains and to mark them. Next, the methods abovementioned are the other high sensitivity technics for validating proteomic biomarkers such as protein microarray, surface plasmon resonance, and Luminex multiplex assays. In recent years, many multiplexed immunodetection techniques have been developed to simultaneously investigate multiple proteins (from several tens to several hundreds), in the same sample, and which are in very low amounts (Figure 1).

Figure 1.

The schematic representation of the immunoassay methods presented in this chapter, more or less in the order in which they appeared in time and how they evolved. These methods are based on the protein/antigen-antibody reaction that is shown on the left side—here is the indirect method: antigen → primary antibody → secondary antibody conjugated with a fluorochrome or an enzyme.

In many cases, the immunoassay techniques are used in conjunction for diagnostic, to confirm the presence of autoantibodies and then to characterize the expression of one or more specific biomarkers for a certain disease. More of this, these techniques can validate their mutual results.

Advertisement

2. Immunohistochemistry

The technique of immunohistochemistry (IHC) is a basic one, both in the anatomopathological diagnosis and in the research. It allows viewing of a protein of interest in a tissue section, specifying its location. This last aspect is very important and distinct IHC from other immunodetection techniques. The presence, reduction or the absence of the target protein allows a precise diagnosis or a personalized one. We do not intend to describe the technique itself, but we would like to mention it as the method of identifying immune antigens of interest, including immunogenetic diseases.

Based on the principle of the antigen-antibody reaction, this technique has undergone improvements over time. It started with a direct IHC method, the reaction antigen (target protein)-antibody, coupled with a fluorochrome. The first data on an attempt to use the direct IHC are from 1934 [9], but the use of fluorochrome for the first time was described in 1941 [10]. The introduction of an enzyme conjugated with an antibody and the visualization of the protein in light microscopy is due to Nakane and Pierce team [11]. The disadvantage of the IHC direct method is its low sensitivity.

Afterwards, an IHC indirect method was developed as follows: antigen-primary antibody, nonconjugated-secondary antibody (anti-primary antibody), conjugated with a fluorochrome or an enzyme, which convert a soluble substrate into an insoluble colored substrate [12, 13]. This method allowed the visual signal to be intensified.

The need to improve more the signal amplification has led to new changes. Thus the secondary antibody has been conjugated with other substances, such as biotin molecules, which in turn form complexes with streptavidin, forming a complex with an enzyme (e.g., horseradish peroxidase) [14]. More recently, an even more sensitive method was used in which a large number of secondary antibodies and enzymes are conjugated to a polymer chain (e.g., dextran) [15].

In the IHC technique, even an array-like reaction can be carried out on the same tissue section by targeting several proteins by using antibodies from different species (mouse, rabbit, goat, etc.), different enzymes coupled to the secondary antibody (e.g., horseradish peroxidase and alkaline phosphatase), different chromogens (e.g., 3,3′diaminobenzidine or 5-bromo-4-chloro-3-indolyl phosphate/nitro blue tetrazolium) or fluorochromes (e.g., FITC and rhodamine) with different colors.

Sometimes, especially when the protein of interest is low and the immunohistochemical signal is weak or with interruptions, a confirmation for protein expression by Western blotting is required. This confirmation is also required when we are not sure whether the antibody specifically binds to the protein of interest or if there is a nonspecific antibody labeling. The Western blot technique allows the identification of the protein as it is shown below.

Advertisement

3. Enzyme-linked immunosorbent assay (ELISA)

Old traditional ELISA technique was developed in 1971 by Engvall and Perlmann [16] and Van Weemen and Schuurs [17] and continues to be nowadays widely used as a routine diagnostic method allowing quantitation of a large variety of proteins [18]. The single-plex ELISA, the most utilized assay method performed in 96- or 384-well plates, has played a prominent role in the quantitative and qualitative identification of analytes.

Direct ELISA, the simplest type of ELISA, could accurately quantify a specific molecule with high sensitivity from a wide variety of samples, and it is faster [19]. But the signal is less amplified.

Indirect ELISA detection is a two-step ELISA which involves a primary antibody and a labeled secondary antibody [20]. This method presents a higher sensitivity and flexibility (different primary detection antibodies can be used with a single labeled secondary antibody). The disadvantage is the occurrence of nonspecific signals.

Beside direct and indirect detection models, two other ELISA methods appeared, to avoid false positive or false negative results, with a high specificity, suitable for complex samples, with more sensitivity and flexibility: sandwich ELISA (quantify antigens between the two layers of antibodies) [21] and competitive ELISA (based on a competitive binding process between the original antigen in the sample and the add-in antigen, the more antigen in the sample, the less labeled antigen is retained in the well and the weaker the signal) [22].

Another ELISA method is ELISpot assay, widely used to evaluate an immune response, for example, in allergies or in autoimmunity [23, 24]. This technique, performed on PVDF membranes, has advantages like specificity, sensitivity, and wide range of detection.

However, the use of ELISA for assessing multiple analytes might be time consuming due to the large number of workflows occurring simultaneously. Moreover, ELISA is designed as a solid-state immunoassay, and the use of a planar matrix can restrict immunoassay capacity, sensitivity, and detection quality [25].

Conventional single-target assays ELISA and Western blot are suitable for biomarker validation, but could be expensive, time consuming, and sample limiting. While most of the disease conditions may arise when only one single molecule is altered, more often it is the consequence of the interaction between several molecules within the inflammation milieu; therefore, studying the diseases necessitates a comprehensive perspective.

ELISA on a chip. In order to improve the method, in terms of using smaller quantities of samples, shortening the reaction time, avoiding sophisticated reading equipment, and reducing costs, a group of researchers tried to miniaturize the ELIZA platform [26]. They developed an ELIZA lab-on-a-chip system (ELIZA-LOC), which allows the use of only 5 μl of sample on a miniaturized 96-well plate combined with a CCD camera [27]. This system combines three functional elements: (i) reagent loading fluidics, (ii) assay and detection well plate, and (iii) reagent removal fluidics. The description of LOM technology (laminated object manufacturing) to obtain this system using polymer sheets was made by Rasooly et al. [28].

Besides miniaturization, another novelty is the washing step that is integrated directly in ELISA plate. The authors state that using this technology, there is no need for a specialized laboratory to perform the ELIZA test.

Advertisement

4. Western blot

The Western blot (WB), also known as immunoblot, is an analytical and quantitative technique for identifying specific proteins in many biological samples, liquid or tissue/cellular homogenates [29]. The WB technique brings concrete and useful information that cannot be offered by other immunoassay methods. If the target protein, present in the sample, is altered qualitatively or quantitatively, the band thickness is changed compared to a control being downregulated or overexpressed. The WB results can also guide us to a genetic investigation in case of partial deletion or duplication in the protein gene [30]. In addition, the WB method allows a quick comparison of target protein expression in many patients in medical diagnosis.

The WB technique was invented by Harry Towbin and co-workers in 1979. They used the method to identify bacterial or chicken ribosomal proteins separated on polyacrylamide gels containing urea. They called this method “electrophoretic blotting technique” [31]. The WB name was given 2 years later by Neal Burnette, which also brought some improvements to this method, including the use of SDS-PAGE gels [32]. The name “Western” was inspired by the earlier name of other blotting methods, “Southern”, named after the name of Edwin Southern, who published in 1975 a method for detecting specific DNA sequences [33], and “Northern” whose name was inspired by the name of the first blotting technique, “Southern”, a RNA detection technique, developed in 1977 by Alwine et al. [34].

Over time, the method has improved and has become easier to achieve, with nearly all materials commercially available: transfer devices, antibodies, pre-casting gels, digital imaging devices, and so on. However, in the most part, as methodology, the technique proposed by the Towbin team remains valid after 38 years. Burnette, Stark, and Towbin said after many years that they were surprised by the success and longevity of the method [35].

In summary, the Western blot method is a way to identify a target protein from a biological sample, a mixture of proteins, running it on polyacrylamide gel. The proteins in the sample are separated by SDS-PAGE gel electrophoresis, depending on their molecular weight. Because the gel is hard to handle, being fragile, the proteins are transferred to a membrane, usually nitrocellulose or PVDF (polyvinylidene fluoride), that maintains the gel pattern as a copy [7]. The electrical current causes the transfer. For visualization of the protein of interest, the membrane is probed by a specific primary antibody, it binds the specific epitope of the protein, and it is labeled by addition of a secondary antibody recognizing the primary antibody conjugated with a detection reagent (fluorophore, enzyme, and radioisotope). The visualization is done colorimetric, by chemiluminescence, on X-ray film, or directly in the membrane with the aid of an imaging system.

In order to be able to reuse a WB membrane that has already been exposed to primary and secondary antibodies, it is necessary to wash it. This operation is called stripping. Only membranes that have been treated with ECL (enhanced chemiluminescence kit) for protein visualization by chemiluminescence can be reused. This method is useful when we want to investigate more proteins on the same blot, for example, a protein of interest and a loading control protein. It saves biological samples, time, and substances. For stripping, special buffers are using that can efficiently remove antibodies but do not remove too much amount of the proteins on the membrane.

The WB system size may vary, with electrophoresis/transfer tank, gels, and membrane: mini, midi, and large, depending on the investigated protein size and the time needed for separation. However, the vast majority of investigators use now the mini system, sometimes the midi one, because of the existence of gradient gels and more sophisticated devices (see below). Transfer systems were developed by few companies to allow proteins the migration from gel to the membrane in different ways, using varying amounts of buffers: wet, semidry, or dry systems. New digital technologies offer a good and rapid bands visualization, avoiding underexposure or overexposure, as in the case of X-ray film developing. The images can be stored in a computer database and can be analyzed with software that measures the optical density of the bands.

4.1 Other methods based on the Western blotting technique

4.1.1 Multiplex Western blot

In the last few years, it has become a necessity to analyze multiple target proteins at the same time, in order to compare the expression of proteins involved in a specific pathology. First Multiplex WB experiment (multiplex Western blot (MWB)) was optimized by Anderson and Davison [36] to study different muscle proteins involved in muscular dystrophies. This method allowed a simultaneously screening of multiple proteins with a different size on a pair of blots, using a cocktail of monoclonal antibodies which permitted the identification of primary deficient and second deficient proteins in several muscle pathologies, knowing that the primary reduction of a protein causes the secondary reduction of another protein. The use of a MWB allowed establishing a biomarker profile for each patient, providing valuable information for diagnosis as well as for phenotype-genotype correlations. The MWB method proposed by Anderson used a biphasic polyacrylamide gel (with different concentrations) system electrophoresis, which separated the proteins with different molecular weights: molecular mass more than 200 kDa in the upper part of the gel, with 5.5–4% polyacrylamide gradient, while proteins with molecular mass under 150 kDa are separated in the lower phase, 7% polyacrylamide gel.

Introduction of this technique has revolutionized the medical diagnosis and opened new perspectives in biomedical research. Simultaneous analysis of several proteins involved in different pathologies by MWB reduced the cost and time for analysis. By this method, it could be determined and compared proteins in the same sample as well as a secondary reduction of other proteins in a specific disease [37, 38].

4.1.2 Capillary electrophoresis and capillary Western blotting

By this method, the molecules are separated by the size inside a capillary filled with an electrolyte. The advantage of the method is that the separating sieve matrix can be automatically pumped in and out because it contains rather unknown polymers than the typical cross-linked polymers for the gels. The big difference between the classical method and this one is that many samples can be run over and over again in an automated manner that saves a lot of time [35].

Capillary electrophoresis (CE) needs a smaller amount of sample than SDS-PAGE and offers a better resolution of a protein size. Proteins travel down the capillaries and are spaced according to the size. When the individual proteins reach the end of the capillary, they drop on a blotting membrane that moves along the capillary opening. It has been shown that classical protein standards such as carbonic anhydrase and lysozyme can be separated within an hour using only a few nanoliters of the sample [39].

O’Neill et al. [40] have been able to capture the resolved proteins on the capillary wall by photochemically activated molecules. This method allowed immune complexes to be formed after electrophoresis, in the capillary. Chemiluminescence reagents flowed through the capillary, and the image was taken with a CCD camera [40].

4.1.3 Microfluidic Western blotting

This technology reduces much more the amount of the sample required for WB and also the length of the capillaries from centimeters to microns, using microfluidic channels. He and Herr, in 2009, developed an automated immunoblotting method using a single streamlined microfluidic assay. A glass microfluidic chip, which has integrated a PAGE electrophoresis with subsequent in situ immunoblotting, allowed a rapid protein separation, directed electrophoretic transfer of resolved proteins to an in-line blotting membrane, and a high-efficiency identification of proteins of interest using antibody-functionalized membranes [41, 42]. The system requires only 0.01–0.5 μg of protein.

4.1.4 Dot blot

It is a method very similar to WB, but the proteins are not separated by gel electrophoresis. The samples are applied in small dots directly on the membrane and then spotted through circular templates. After membrane drying, the antibodies are applied. The visualization of target protein is made like at WB, chemiluminescent or colorimetric. Dot blot is used to test the specificity of some antibodies, to test the antibody concentration used for WB, or to evaluate the presence of a target protein in the sample before WB.

4.1.5 Far-Western blotting

It is used to detect a protein-protein interaction in vitro. Instead of the primary antibody for detecting the protein of interest, this method uses a nonantibody protein that binds to the protein of interest. Far-Western blotting detects proteins on the basis of the presence or the absence of binding sites for the protein probe. This method is important in characterization of protein interactions in biological processes such as signal transductions [43], receptor-ligand interactions, or screen libraries for interacting proteins.

Advertisement

5. Protein microarray

Protein microarray analysis has an increasingly use both for research purposes as well as for various biomedical applications, including the niches ones like evaluating markers of apoptosis activated by various therapies such as photodynamic therapy (PDT), assessment of epigenetic milieu, or transcriptional activity in treated cells [44]. Thus, protein microarray is a proteomic tool that can deliver high-throughput data for revealing new therapeutic targets [45].

Protein microarray history has spanned the last two decades, the basic principle being identical with ELISA, but there are several advantages such as spotting in terms of miniaturization, multiplexing, and large data obtained in an ELISA equivalent time. Briefly, biological samples of interest (e.g., serum, plasma, etc.) are incubated on a slide containing immobilized antibodies, proteins, or peptides. An antigen-antibody reaction occurs between an analyte from the tested sample and the corresponding antibody from slide followed by the detection step through various methods (e.g., fluorescence-based detection). The slide is further scanned, followed by image acquisition, data processing, and analysis. There are several classifications of this technique, but it could fall into two main categories: direct phase (e.g., antibody-, protein-, peptide array) and reverse or indirect phase where sample of interest is spotted on a slide and the corresponding antibody is further added.

Among all these variants, the antibody array type is preferred in tumor research domain or in biomarkers discovery/quantification due to technique’s high versatility and reproducibility [46]. The reverse phase array format could also be used for biomarker discovery because it is specificity but has the disadvantage of being more laborious.

It is worth to emphasize that protein microarray could be customized in terms of number and multiplicity of tested analytes one achieving new research and clinical benefits through this technology. Thus, although fundamental research purposes prevail when it comes to array platforms, there is also a recent increasing trend in clinical research, diagnostics, and even industry applications such as pharmacy or food. For instance, recent attempts are made in using array platform for autoimmune disease insights. Thus, novel antigen arrays have been developed in order to discover new autoantibody targets, providing analysis for hundreds of samples and of their reactivity pattern against thousands of antigens simultaneously [47].

Customizing an array in relation to clinical purpose confirms the flexibility of these platforms in assisting molecular management of the disease. A customized platform was designed in order to monitor severe acute respiratory syndrome (SARS) infection by screening hundreds of sera based on the reactivity against certain selected proteins from SARS coronavirus. Authors have reported that with this customized array, viral infection could be monitored for many months after infection [48]. This type of microarray platform has been further updated to a serological assay for the specific detection of IgM and IgG antibodies against the S1 receptor-binding subunit of the spike protein of emerging human coronavirus hCoV-EMC and SARS-CoV as antigens [49].

Protein microarray is a technology in continuous evolution offering multiple possibilities in updating other proteomic techniques. Therefore, the development of the “microwestern array” is a clear proof how traditional methods like Western blot can be linked to novel technology, thus significantly expanding the research technological arsenal [50].

Data generated by ELISA and WB require sometimes additional complementary proteomic methods to supplement and even support the scientific information. Such supportive task is often accomplished by protein microarrays providing important evidence on modulation of signaling networks and potential targets (or pathways); these factors or networks must first be identified, and array platforms allow exactly this development by exploring dozens of targets simultaneously within a single sample, providing lots of data which may be further investigated using traditional ELISA or WB techniques [51].

Advertisement

6. Luminex xMAP array

An important improvement in the biological assay field was made in the late 1990s when Luminex xMAP technology was launched. xMAP technology combines the principles of ELISA and flow cytometry, but goes beyond the limitations of solid-phase reaction kinetics and is suitable for high throughput, multiplex, and simultaneous detection of different biomarkers within a very small volume sample. Bringing together advanced fluidics, optics, and digital signal processing with proprietary microspheres, xMAP technology became one of the fastest growing multiplex technologies. Featuring a flexible open-architecture design, xMAP technology enables the configuration of various assays, quickly, cost effectively, and accurately, useful in clinical and research laboratories [52].

A key component of Luminex xMAP technology is represented by proprietary color-code polystyrene microspheres (beads) internally dyed with precise concentrations of two or three spectrally distinct fluorochromes. Through precise concentrations of these specific dyes, up to 500 distinctly bead sets can be developed, with a different spectral signature.

Based on fluorescent reporter signals, high-speed digital-signal processors identify each individual microsphere and quantify the result of every bioassay. The capability of adding multiple conjugated beads to each sample results in the ability to obtain multiple results from each sample [53].

There are different types of advanced detection platforms (as depicted in Table 1), and therefore, various biomarker panels could be analyzed. Accordingly, validation of novel biomarkers into multiplex immunoassay panels confers an attractive prospect of simultaneous measurement of multiple analytes in a single patient sample, enabling progres-sion monitoring and outcome prediction, even detecting major diseases in its earliest stages [54].

TechniqueAdvantagesDisadvantagesUse
IHCQualitative
Fast
Easy to detect
Relatively inexpensive
Medium specificity
Possible cross reactivity
Routine diagnstic tool
Shows the localisation of antigen
ELISAQuantitative
High sensitivity
Medium specificity
Higher throughput than Western blot
Automation potential
One protein/analyte at a time
False positive results
Labor intensive/time consuming/high reagent use
Costly setup for automation
To quantify a single protein
Confirmation of other screening method/validation
Western blotHigh specificity
High sensitivity
Quantitative
Automation potential
One or a small number of proteins/analytes at a time
Labor intensive/time consuming for the classical method. Resolved by the new equipments
Qualitative, quantitative, especially with the newest equipments
Difficult to transfer large or hydrophobic proteins—false negative results
Difficult to automate—for the classical method. Resolved by the new equipments
To identify the presence of a small number of proteins in the same sample (multiplex WB) or the presence of protein-protein interactions (Far-WB)
Confirmation of other screening method/antibodies validation
Bead-based array (e.g., Luminex® technology)High sensitivity
High throughput and speed
Multiplex and customizable panels of analytes
Open-architecture design
Low time, labor, and reagent use over traditional methods
Versatility
Flexibility
High cost for a specialized equipment and a validated antibody pairTo quantify (quantitative) multiple proteins/panels of analytes, in the same well from a small amount of sample
Clinical implementation—available IVD kits
Protein microarrayHigh sensitivity
Medium specificity
Highest throughput
Low time and reagent use
High cost for a specialized equipment and a validated antibody pairTo screen for changes across a large number of proteins, in the same well from a small amount of sample
SPRiVery high sensitivity
Label-free
Real time method
High throughput and speed
Multiplex and customizable panels of analytes
Open-architecture design
Low time, labor, and reagent use over traditional methods
Low time and reagent use
Biochip reusable
High cost for a specialized equipment
Suitable for liquid biological probes
To screen and quantify (quantitative) multiple protein interactions (and not only)/panels of analytes, from the same sample
Clinical implementation

Table 1.

Advantages and disadvantages of immunoassay methods presented in this chapter: IHC, ELISA, Western blot, bead-based array—Luminex technology, and chip-based array—protein microarray, SPRi.

Some of our studies illustrate significant dysregulation in circulating levels of cytokines and angiogenic factors in brain tumors, with over threefold upregulation of IL-6, IL-1 beta, TNF-alpha, and IL-10 and up to twofold upregulation of VEGF, FGF-2, IL-8, IL-2, and GM-CSF, with implications in tumor progression and aggressiveness, and also involved in disease-associated pain [55, 56].

Currently, Western blot is used to validate/confirm the identified biomarkers, and association between the xMAP technology and the Western blot was remarked in many studies. Interestingly, one of them emphasized the improvement in diagnostic sensitivity of HIV infection in early stages using xMAP technology, increasing the chances of an early accurate diagnosis. Thus, it was observed a superior sensitivity of Luminex xMAP compared with Western blot. Out of 87 confirmed HIV positive cases, Western blot confirmed 74.7% sensitivity, while Luminex xMAP identified 82.8% sensitivity (p < 0.05) [57]. Further advancements will be needed for a successful validation of current discoveries, and sustained efforts are necessary to expand the translation into clinical applications toward personalized medicine [58].

Advertisement

7. Surface plasmon resonance imaging, lab-on-a-chip

Since our goal is not describing surface plasmon resonance imaging (SPRi) methodology, we will not insist very much on the description of the technique. We will make a brief description of the principle on which SPRi is based.

The first SPR immunoassay was proposed by the team Liedberg, Nylander, and Lundström, in 1983 [59]. The SPR immunoassay method is label-free (unlike ELISA); no label molecule is required for analyte detection [60]. Moreover, the measurement is done in real time, which allows monitoring of the individual steps of this technology. SPRi is currently one of the most sensible platforms for studying a wide variety of interaction affinities [61, 62], involving nucleic acid sequences [63, 64, 65], peptides [66], proteins [67, 68], and carbohydrates [69]. It is possible to monitor hundreds of molecular interactions simultaneously.

The composition of a biochip consists essentially of a glass prism, coated with a thin gold film and a pre-functionalized surface chemistry. The sample to be analyzed is injected over the biochip, and the detection of a specific molecule can be performed by immobilizing a binding partner on the biochip. SPRi makes a nonlabeling and a real-time detection of biomarkers [70].

The SPRi platform allows the quantitative detection of multiple simultaneous multiplex interactions, and many studies are based on this application for screening a variety of analytes in different array types. The main advantage of using SPRi in immunodiagnostics is the possibility of monitoring the antigen-antibody reaction in real-time, estimating kinetics, how quickly it occurs and how durable it is. In addition, it does not require any labels.

In comparison to ELISA and Western Blot, SPRi has the advantage of investigating a large number of different analytes from the same sample (several hundred different spots can be placed on a biochip), and after washing the biochip, it is possible to immediately analyze another sample. SPRi takes less time than other methods. The disadvantage of SPRi would be that only liquid biological samples (blood, urine, cerebrospinal fluid, and cell culture medium) can be analyzed, and it does not analyze biological samples from different tissues/tissue lysates. SPRi is very effective when there are many samples and many different interactions to analyze, but for a small number of samples or to demonstrate only one type of interaction between two proteins, for example, WB is more efficient.

As a conclusion regarding the technologies presented in this chapter, we show Table 1 with the advantages and disadvantages of each method.

Advertisement

8. Immunoassay methods in immunogenetic disease diagnostic

Indirect immunofluorescence (IIF) for autoantibody analysis is one of the routine diagnostic methods. Different tissue sections or human tumor cell lines—HEp-2—are used as the source of antigen over which the serum of patients with specific autoantibodies is applied [71].

The antinuclear antibody (ANA) test is such a standard screening assay. The American College of Rheumatology declared HEp-2 IIF as the preferred method for ANA screening [72]. The large amount of ANAs can indicate an autoimmune disease, including systemic lupus erythematosus, Sjogren’s syndrome, scleroderma, rheumatoid arthritis, idiopathic inflammatory myopathy, and others.

One of the immunohistochemistry method applications in autoimmune disease diagnosis is the detection of the presence of MHC I and, more recently, of MHC II in skeletal muscle of patients with idiopathic inflammatory myopathies (IIMs). It is a group of autoimmune systemic diseases, of which the most common forms are dermatomyositis, polymyositis, and inclusion body myositis. The study of muscle biopsy makes the difference in diagnosis between subtypes, but also among other types of myopathies and IIMs. In addition to other pathological features, the presence of MHC I and MHC II in sarcolemma gives the certainty of diagnosis, as long as they are not present in normal muscles [73, 74, 75, 76]. Their overexpression in IIMs is induced by cytokines, including interferon and tumor necrosis factor alpha (TNF alpha) [77]. A study of 120 muscle biopsies from patients with different forms of IIMs showed a presence of MHC I in all biopsies and MHC II in 93% of them [76].

The MHC I expression appears early and precedes the lymphocyte infiltrate [78], persisting in late disease, and it is not attenuated by immunosuppressive treatment [79, 80, 81].

MHC II expression on antigen presenting cells activates T-helper cells and initiates an immune response without knowing the mechanism by which MHC II alleles mediate susceptibility to a given autoimmune disease [82].

From our experience in IIM cases where the IHC is not conclusive, a WB verification or validation is of great help in highlighting MHC I and II bands at their specific molecular weight.

From the more recent studies, we mention that the anti-signal recognition particle antibodies in the serum of IIM patients have diagnostic and prognostic value especially in the forms of immune-mediated necrotizing myopathy [83]. The authors draw attention to a mandatory IIF test along with the dot immunoassay to avoid false positive results from the latter method in pathologies not associated with IIM. The results sometimes depend on the nature of the antigen used in the technique and can be denatured.

ELISA is used as a diagnostic tool in autoimmune diseases, for evaluation of serum autoantibodies. Antinuclear antibodies (ANAs) directed against a variety of nuclear and cytoplasmic antigens are found with a high frequency in many systemic autoimmune disorders like systemic lupus erythematosus, scleroderma, Sjogren’s syndrome, myositis, etc. ANA-HEp-2 Screen ELISA is an immunoassay method for the qualitative combined detection of IgG antibodies against human serum HEp-2 cells. Each well is coated with Hep-2 cellular lysate. The test detects in a well plate total ANAs against double stranded DNA, histone, SS-A (Ro), SS-B (La), Sm, snRNP/Sm, Scl-70, PM-Scl, Jo-1, and centromeric antigens.

HEp-2 cells allow the recognition of over 30 nuclear and cytoplasmic patterns that are given upwards of 50 different autoantibodies [84, 85]. The specificity of the test is closely related to the quality of the antigens used [86]. It is one of the most common methods of diagnosis in organ-specific autoimmune diseases, such as Grave’s disease, primary biliary cirrhosis, insulin-dependent diabetes mellitus or systemic autoimmune disorders affecting different organs, such as systemic sclerosis, Sjogren’s syndrome, and mixed connective tissue disease rheumatoid arthritis [87, 88].

From recent research studies [89], we want to mention cortactin antibodies as new biomarkers in double seronegative myasthenia gravis (myasthenia gravis form dSNMG). ELISA tests validated by WB have demonstrated that the presence of cortactin autoantibodies is a biomarker to be taken into account, suggesting that the disease will be ocular or mild generalized and could be done routinely in the future.

Another work on rheumatoid arthritis shows that, apart from the autoantibody system that recognizes citrullinated proteins, the identification of another antibody system against carbamylated proteins has an important early diagnostic value, predicting a more severe course of disease [90]. The ELISA method used in this study could become routine for serum testing of patients with rheumatoid arthritis.

Western blot. To highlight the importance of WB technique in clinical diagnosis, we give some eloquent examples below. The WB method has been used in many studies, along with immunoprecipitation, ELISA, and flow cytometry, to demonstrate the quantitative or qualitative modification of proteins of interest in autoimmune diseases in order to find new biomarkers or therapeutic targets. WB has proven to be a good tool for serological tests.

Line-blot immunoassay is a Western blotting method that uses recombinant antigens immobilized on straight lines on a nylon strip. They are incubated with patient serum containing autoantibodies. They bind to the antigens present on the strip and are viewed colorimetrically. Interpretation of the results is done by comparing the color intensity of strips with the color of strips of a positive standard. There are some studies that have shown the utility of this method in the detection of autoantibodies present in serum but which could not be identified by IIF, for example, anti-SS-A/Ro in Sjogren’s syndrome [91, 92].

Haroon et al. have demonstrated using the WB method that there is an interaction between endoplasmic reticulum aminopeptidase 1 (ERAP1) with human leucocyte antigen (HLA)-B *27 in ankylosing spondylitis [93], and that the HLA-B27 molecules could alter the ERAP1 level. The functional interaction between ERAP1-peptide and HLA-B27 could thus be the missing link in the pathogenesis of ankylosing spondylitis.

Stagakis et al. studied whether anti-TNF therapy improves insulin resistance in rheumatoid arthritis [94]. Western blot was used to analyze the proteins p-Ser312 IRS-1 and p-AKT from peripheral blood mononuclear cell lysates. It has been established that anti-TNF alpha therapy has a positive effect, improving insulin sensitivity and reversing the defects in signaling insulin cascade in this disease.

Tsui et al. have conducted a study of the serum levels of noggin (NOG) and sclerostin (SOST) in patients with ankylosing spondylitis, more specifically, on the immune response to these two molecules [95]. The WB method was used to quantify the relative amounts of NOG/SOST-IgG immune complexes. An increased level of NOG/SOST-IgG immune complexes was found in patients with this pathology.

Rizzo et al. [96] showed that the dimeric form of the HLA-G molecule is associated with the response to methotrexate treatment in patients with early rheumatoid arthritis. HLA-G dimeric and monomeric forms have been highlighted by WB. The presence of dimeric form in plasma prior to methotrexate therapy could be a biomarker for the patient’s response to treatment.

Protein microarray. Antibody microarrays could provide a real-time vision of biological processes, such proteomic instrument being used in clinic to analyze serum and plasma in several pathologies including autoimmune disorders. One of the autoimmune diseases approached through protein array is systemic lupus erythematosus (SLE) where SLE clinical heterogeneity and the absence of robust biomarkers to evaluate the disease states and differentiate from other autoimmune conditions are yet to be resolved [97]. Thus, using an antibody-based leukocyte-capture microarray, mononuclear cells isolated from peripheral blood of 60 SLE patients were processed for obtaining proteomic patterns to distinguish SLE from healthy subjects. With this array platform, it was improved the conventional SLE diagnostics and disease states elucidation [98]. Moreover, an “in-house” antibody microarray comprising 135 human recombinant single-chain fragment variables aiming various immune proteins were used to examine systemic sclerosis (SSc) and SLE patients. This tailored array identified a significant number of differentially expressed proteins that delineate SLE from systemic sclerosis, thus surpassing disease classification through conventional clinical parameters, including, ANA, anti-DNA, SLEDAI-2 k, C1q, C3, C4, and CRP [99].

Another challenging field for protein array is related to rheumatoid arthritis (RA) as it could detect biomarkers specific for arthritis and not for autoimmune diseases in general [100]. Some research groups have started to develop different antigen arrays for differential diagnosis and even RA molecular classifying. Panels of proteins were detected, among these three proteins, namely, WIBG, GABARALP2, and ZNF706, were suggested as potential specific markers for RA early stages [101]. Hence, protein arrays bring valuable data to immune-disease background allowing exploration of numerous samples in parallel and thousands of targets.

Antibodies against ion channels, receptors, synaptic proteins, etc. confirm protein microarrays as a future potential tool in routine diagnosis [102]. Whatever commercially available or customized platforms, antibody arrays start to emerge in clinic by designing omics disease signatures helping the disease management.

The protein microarray was used in a study of pemphigus vulgaris, an autoimmune skin disease, to identify the entire set of antibodies, bringing extra data about the complex relationship between genetics and disease development [103]. The results were correlated with those obtained by the ELISA and proved to be compatible. The main targets for autoantibodies are desmoglein-3 and 1, but the study showed that there are autoantibodies that are not directed to desmoglein at a significant number of patients.

A study regarding ankylosing spondylitis using the protein microarray, confirmed by ELISA, to characterize anti-ankylosing spondylitis autoantibodies, showed that anti-protein phosphatase 1A (PPM1A) autoantibodies are present in the serum of the patients and that they could serve not only as biomarkers for diagnosis, disease severity, and response to anti-TNF therapy, but also as a therapeutic target [104].

Luminex xMAP technology has developed as an alternative to planar microarray methods. Bead-based immunoassays are one of them. The determinations by this method and by ELISA of anti-thyroid peroxidase and anti-thyroglobulin antibodies in autoimmune diseases have been shown to be compatibles [105].

There is a commercially available kit for ANA detection, which is low cost and saves time. Antigens corresponding to autoantibodies are linked to polystyrene microspheres labeled internally with different amounts of two different fluorochromes, resulting in 100 different color spectra. Each microbead carries an antigen specific for a single antibody [106].

Good results were obtained in assessing a number of antinuclear autoantibodies as: dsDNA, Sm and Sm/RNP (in systemic lupus erythematosus), SS-A/Ro and SS-B/La (in Sjogren’s syndrome), Jo-1 (myositis), ribosome (systemic lupus erythematosus), and centromere (systemic sclerosis) [107].

One of the problems with this technology could be the lack of a true quantitative calibration due to the difference in affinity of the antibodies for the antigen [108]. Some authors argue that it is also necessary to validate the results by other immunoassay methods [106], while others claim that the accuracy of the technique is similar to that observed by ELISA [109, 110].

There are studies in which the Luminex methodology is used for the analysis of serum biomarkers in various autoimmune diseases. Thus, in an article on ankylosing spondylitis, certain cytokines as hepatocyte growth factor (HGF), CXCL8, and matrix metalloproteinases (MMP-8 and MMP-9) identified from a large number of biomarkers by Luminex could be diagnostic targets, their serum levels being increased in this disease [111].

In other chapter regarding ankylosing spondylitis, Luminex bead-based technology was used for serum cytokines analysis, and the conclusion was that the utilization of TNF alpha inhibitors decreases the number of T cells producing proinflammatory cytokines [112].

Mou et al. showed, using Luminex technology in combination with PCR, that in ankylosing spondylitis patients from Southern China with HLA-B27 in their serum, HLA-B2704 subtype predominates. And the HLA-B2715 subtype may have a disease prognostic value, early onset being related to this subtype [113].

Surface plasmon resonance imaging. Despite its great sensitivity, this technology is relatively little used to determine the concentration of some analytes. Improving signal amplification methods is one of the research goals in this technique.

In some autoimmune diseases, such as rheumatoid arthritis (RA), psoriatic arthritis, systemic lupus erythematosus, or Sjogren’s syndrome, autoantibodies attack citrullinated proteins, and the presence of anti-citrullinated proteins, antibodies is a standard test in these cases. The use of SPRi for monitoring autoantibodies that bind to different citrullinated targets was first described by Lokate et al. SPRi has shown its ability to detect the interaction between citrullinated peptides and serum autoantibodies in RA patients in one step [114].

SPRi microarray technique was also used in a more recent research to identify autoantibodies against citrullinated protein (ACPA) profiles in patients with early onset rheumatoid arthritis. The authors made a comparative study using citrullinated and noncitrullinated peptides [115]. The study showed that SPRi is a suitable methodology for detecting ACPAs in the serum of patients with this pathology.

A subsequent study was also revealed by SPRi, the presence of citrullinated B-cell epitopes in fibrinogen [116].

A research team [117] showed the use of SPRi and gold particles to amplify the signal for the detection of inflammation biomarker TNF-alpha in serum. Also, the use of a specific buffer solution for sample dilution was utilized to reduce the nonspecific binding in real samples. Thus, a low limit of detection, as well as a good reproducibility and the longevity of chips, is a good motivation to use this immunoassay method to detect biomarkers that are in low concentrations in biological samples.

Buhl et al. reported in a research paper the use of SPRi technology for the anti-dsDNA detection in systemic lupus erythematosus [118].

Advertisement

9. Conclusions

Immunoassay methods have many advantages but some limitations too. Their importance in identifying different biomarkers for diagnosis or personalized therapy is essential. That is why they have diversified so much, in order to be able to answer all the challenges. Additionally, these methods and technologies have also specialized in an advanced degree, so that they can detect smaller amounts of molecules with as high a precision as possible in a shorter time. The antigen-antibody response gives them great sensitivity. The development of more advanced equipment leads to the automation of these methods and to a greater efficiency, with applicability in diagnosis and therapeutic monitoring, in discovery of new biomarkers and even in pharmacology.

In order to be used for diagnosis in different laboratories, these methods and kits should be standardized. The problems to be posed are: the clinical manifestation of the disease in different individuals, the source of the antigen, the specificity and sensitivity of the autoantibodies for different antigens, the reproducibility of the assay, and the precision and the accuracy of the method [91, 119, 120].

Some studies show a good correlation between IIF and ELISA methods [84, 121, 122], and others, on the contrary, show different results between these methods [71, 123].

Multiplex technologies are gaining more and more followers in recent years by allowing simultaneous analysis of a multitude of analysts, saving time and costs. However, there are studies showing that compared to the old methods, some false negative or false positive results are obtained [124, 125, 126, 127, 128]. Cross reactivities may also occur [129].

Assay kits produced by different manufacturers can show variable results also. More than this, the methodology used by each laboratory can lead to different results, even by using the same kit. International standardization is required. A collaboration between an international body and organizations responsible for quality of assessment of assays is desirable, so that a collaboration among clinicians, diagnostic laboratories, and manufacturers to be established.

Advertisement

Acknowledgments

All authors contributed equally to this work. Partially supported from Project ID: SMIS CSNR 1882/49159 (CAMED), PN-III-P1-1.2-PCCDI-2017-0341 (PATHDERM), PN-III-P1-1.2-PCCDI-2017-0782 (REGMED), PN 16.22.02.04/2016, PN 16.22.02.05/2016, PN 18.21.02.02/2018, and PN 18.21.01.06/2018.

References

  1. 1. Hanash SM. Biomedical applications of two-dimensional electrophoresis using immobilized pH gradients: Current status. Electrophoresis. 2000;21:1202-1209
  2. 2. National Institute of Aging Working group AB. Consensus report of the Working Group on: “Molecular and Biochemical Markers of Alzheimer’s disease”. Neurobiology of Aging. 1998;19(2):109-116
  3. 3. Gao J, Garulacan LA, Storm SM, Opiteck GJ, Dubaquie Y, Hefta SA, et al. Biomarker discovery in biological fluids. Methods. 2005;35(3):291-302
  4. 4. Burtis CA, Ashwood EA, editors. Tietz Fundamentals of Clinical Chemistry. 5th ed. 2001
  5. 5. Robinson WH, Steinman L, Utz PJ. Proteomics technologies for the study of autoimmune disease. Arthritis & Rheumatism. 2002;46(4):885-893
  6. 6. Hueber W, Robinson WH. Proteomic biomarkers for autoimmune disease. Proteomics. 2006;6(14):4100-4105
  7. 7. Kurien BT, Scofield RH. Western blotting. Methods. 2006;38(4):283-293
  8. 8. Coorssen JR, Blank PS, Albertorio F, Bezrukov L, Kolosova I, Backlund PS Jr, et al. Quantitative femto- to attomole immunodetection of regulated secretory vesicle proteins critical to exocytosis. Analytical Biochemistry. 2002;307(1):54-62
  9. 9. Marrack J. Nature of antibodies. Nature. 1934;133:292-293
  10. 10. Coons AH, Creech HJ, Jones RN. Immunological properties of an antibody containing a fluorescence group. Proceedings of the Society for Experimental Biology and Medicine. 1941;47(2):200-202
  11. 11. Nakane PK, Pierce GB Jr. Enzyme-labeled antibodies: Preparation and application for the localization of antigens. Journal of Histochemistry and Cytochemistry. 1966;14(12):929-931
  12. 12. Sternberger LA, Hardy PH Jr, Cuculis JJ, Meyer HG. The unlabeled antibody enzyme method of immunohistochemistry: Preparation and properties of soluble antigen-antibody complex (horseradish peroxidase-antihorseradish peroxidase) and its use in identification of spirochetes. Journal of Histochemistry and Cytochemistry. 1970;18(5):315-333
  13. 13. Mason DY, Sammons R. Alkaline phosphatase and peroxidase for double immunoenzymatic labelling of cellular constituents. Journal of Clinical Pathology. 1978;31(5):454-460
  14. 14. Hsu SM, Raine L. Protein A, avidin, and biotin in immunohistochemistry. Journal of Histochemistry and Cytochemistry. 1981;29(11):1349-1353
  15. 15. Ramos-Vara JA, Miller MA. Comparison of two polymer-based immunohistochemical detection systems: ENVISION+ and ImmPRESS. Journal of Microscopy. 2006;224(Pt 2):135-139
  16. 16. Engvall E, Perlmann P. Enzyme-linked immunosorbent assay (ELISA). Quantitative assay of immunoglobulin G. Immunochemistry. 1971;8(9):871-874
  17. 17. Van Weemen BK, Schuurs AH. Immunoassay using antigen-enzyme conjugates. FEBS Letters. 1971;15(3):232-236
  18. 18. Bilan R, Ametzazurra A, Brazhnik K, Escorza S, Fernandez D, Uribarri M, et al. Quantum-dot-based suspension microarray for multiplex detection of lung cancer markers: Preclinical validation and comparison with the Luminex xMAP system. Scientific Reports. 2017;7:44668
  19. 19. Engvall E. The ELISA, enzyme-linked immunosorbent assay. Clinical Chemistry. 2010;56:319-320
  20. 20. Lindström P, Wager O. IgG autoantibody to human serum albumin studied by the ELISA-technique. Scandinavian Journal of Immunology. 1978;7(5):419-425
  21. 21. Kato K, Hamaguchi Y, Okawa S, Ishikawa E, Kobayashi K. Use of rabbit antibody IgG bound onto plain and aminoalkylsilyl glass surface for the enzyme-linked sandwich immunoassay. Journal of Biochemistry. 1977;82(1):261-266
  22. 22. Yorde DE, Sasse EA, Wang TY, Hussa RO, Garancis JC. Competitive enzyme-liked immunoassay with use of soluble enzyme/antibody immune complexes for labeling. I. Measurement of human choriogonadotropin. Clinical Chemistry. 1976;22(8):1372-1377
  23. 23. Czerkinsky CC, Nilsson LA, Nygren H, Ouchterlony O, Tarkowski A. A solid-phase enzyme-linked immuno-spot (ELISPOT) assay for enumeration of specific antibody-secreting cells. Journal of Immunological Methods. 1983;65(1-2):109-121
  24. 24. Franci C, Ingles J, Castro R, Vidal J. Further studies on the ELISA-spot technique. Its application to particulate antigens and potential improvement in sensitivity. Journal of Immunological Methods. 1986;88(2):225-232
  25. 25. Liotta LA, Espina V, Mehta AI, Calvert V, Rosenblatt K, Geho D, et al. Protein microarrays: Meeting analytical challenges for clinical applications. Cancer Cell. 2003;3(4):317-325
  26. 26. Sapsford KE, Francis J, Sun S, Kostov Y, Rasooly A. Miniaturized 96-well ELISA chips for staphylococcal enterotoxin B detection using portable colorimetric detector. Analytical and Bioanalytical Chemistry. 2009;394(2):499-505
  27. 27. Sun S, Yang M, Kostov Y, Rasooly A. ELISA-LOC: Lab-on-a-chip for enzyme-linked immunodetection. Lab on a Chip. 2010;10(16):2093-2100
  28. 28. Rasooly A, Bruck HA, Kostov Y. An ELISA Lab-on-a-chip (ELISA-LOC). Methods in Molecular Biology. 2013;949:451-471
  29. 29. Ghosh R, Gilda JE, Gomes AV. The necessity of and strategies for improving confidence in the accuracy of western blots. Expert Review of Proteomics. 2014:1-12
  30. 30. Barresi R. From proteins to genes: Immunoanalysis in the diagnosis of muscular dystrophies. Skeletal Muscle. 2011;1(1):24
  31. 31. Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: Procedure and some applications. Proceedings of the National Academy of Sciences of the United States of America. 1979;76(9):4350-4354
  32. 32. Burnette WN. “Western blotting”: Electrophoretic transfer of proteins from sodium dodecyl sulfate-polyacrylamide gels to unmodified nitrocellulose and radiographic detection with antibody and radioiodinated protein A. Analytical Biochemistry. 1981;112(2):195-203
  33. 33. Southern EM. Detection of specific sequences among DNA fragments separated by gel electrophoresis. Journal of Molecular Biology. 1975;98(3):503-517
  34. 34. Alwine JC, Kemp DJ, Stark GR. Method for detection of specific RNAs in agarose gels by transfer to diazobenzyloxymethyl-paper and hybridization with DNA probes. Proceedings of the National Academy of Sciences of the United States of America. 1977;74(12):5350-5354
  35. 35. Mukhopadhyay R. Revamping the Western blot. ASBMB Today. 2012:14-16
  36. 36. Anderson LV, Davison K. Multiplex western blotting system for the analysis of muscular dystrophy proteins. The American Journal of Pathology. 1999;154(4):1017-1022
  37. 37. Gaina G, Manole E, Bordea C, Ionica E. Analysis of muscle Calpain-3 in LGMD 2A. Jurnal Studia Universitatis “Vasile Goldis” Arad. Seria Stiintele Vietii (Life Sciences Series). 2008;18:181-185
  38. 38. Gaina G, Manole E, Matanie C, Mihalcea A, Ionica E. Muscular dystrophies proteins evaluation by western blott and immunofluorescence. Romanian Biotechnological Letters. 2008;13(3):3729-3736
  39. 39. Anderson GJ, Cipolla CM, Kennedy RT. Western blotting using capillary electrophoresis. Analytical Chemistry. 2011;83:1350-1355
  40. 40. O’Neill RA, Bhamidipati A, Bi X, Deb-Basu D, Cahill L, Ferrante J, et al. Isoelectric focusing technology quantifies protein signaling in 25 cells. PNAS. 2006;103(44):16153-16158
  41. 41. He M, Herr AE. Microfluidic polyacrylamide gel electrophoresis with in situ immunoblotting for native protein analysis. Analytical Chemistry. 2009;81:8177-8184
  42. 42. He M, Herr AE. Polyacrylamide gel photopatterning enables automated protein immunoblotting in a two-dimensional microdevice. Journal of the American Chemical Society. 2010;132:2512-2513
  43. 43. Machida K, Mayer BJ. Detection of protein-protein interactions by far-western blotting. In: Kurien BT, Scofield RH, editors. Protein Blotting and Detection Methods in Molecular Biology (Methods and Protocols). Totowa, NJ: Humana Press; 2009
  44. 44. Demyanenko SV, Uzdensky AB, Sharifulina SA, Lapteva TO, Polyakova LP. PDT-induced epigenetic changes in the mouse cerebral cortex: A protein microarray study. Biochimica et Biophysica Acta. 2014;1840(1):262-270
  45. 45. Matei C, Tampa M, Caruntu C, Ion RM, Georgescu SR, Dumitrascu GR, et al. Protein microarray for complex apoptosis monitoring of dysplastic oral keratinocytes in experimental photodynamic therapy. Biological Research. 2014;47:33
  46. 46. Wilson JJ, Burgess R, Mao YQ, Luo S, Tang H, Jones VS, et al. Antibody arrays in biomarker discovery. Advances in Clinical Chemistry. 2015;69:255-324
  47. 47. Ayoglu B, Schwenk JM, Nilsson P. Antigen arrays for profiling autoantibody repertoires. Bioanalysis. 2016;8(10):1105-1126
  48. 48. Zhu H, Hu S, Jona G, Zhu X, Kreiswirth N, Willey BM, et al. Severe acute respiratory syndrome diagnostics using a coronavirus protein microarray. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(11):4011-4016
  49. 49. Reusken C, Mou H, Godeke GJ, van der Hoek L, Meyer B, Muller MA, et al. Specific serology for emerging human coronaviruses by protein microarray. Eurosurveillance: Bulletin Europeen sur les Maladies Transmissibles = European Communicable Disease Bulletin. 2013;18(14):20441
  50. 50. Mann M. Can proteomics retire the western blot? Journal of Proteome Research. 2008;7(8):3065
  51. 51. Bass JJ, Wilkinson DJ, Rankin D, Phillips BE, Szewczyk NJ, Smith K, et al. An overview of technical considerations for Western blotting applications to physiological research. Scandinavian Journal of Medicine and Science in Sports. 2017;27(1):4-25
  52. 52. http://info.luminexcorp.com/en-us/research/download-the-xmap-cookbook
  53. 53. http://core.phmtox.msu.edu/Scheduling/ItemDocs/65/LuminexUserManual.pdf
  54. 54. Tighe PJ, Ryder RR, Todd I, Fairclough LC. ELISA in the multiplex era: Potentials and pitfalls. Proteomics—Clinical Applications. 2015;9(3-4):406-422
  55. 55. Albulescu R, Codrici E, Popescu ID, Mihai S, Necula LG, Petrescu D, et al. Cytokine patterns in brain tumour progression. Mediators of Inflammation. 2013;2013:979748
  56. 56. Tanase C, Albulescu R, Codrici E, Popescu ID, Mihai S, Enciu AM, et al. Circulating biomarker panels for targeted therapy in brain tumors. Future Oncology. 2015;11(3):511-524
  57. 57. Kong W, Li Y, Cheng S, Yan C, An S, Dong Z, et al. Luminex xMAP combined with western blot improves HIV diagnostic sensitivity. Journal of Virological Methods. 2016;227:1-5
  58. 58. Krochmal M, Schanstra JP, Mischak H. Urinary peptidomics in kidney disease and drug research. Expert Opinion on Drug Discovery. 2017:1-10
  59. 59. Ingemar LBNCL. Surface plasmon resonance for gas detection and biosensing. Sensors and Actuators. 1983;4:299-304
  60. 60. Rich RL, Myszka DG. Higher-throughput, label-free, real-time molecular interaction analysis. Analytical Biochemistry. 2007;361(1):1-6
  61. 61. Paul S, Vadgama P, Ray AK. Surface plasmon resonance imaging for biosensing. IET Nanobiotechnology. 2009;3(3):71-80
  62. 62. Scarano S, Mascini M, Turner AP, Minunni M. Surface plasmon resonance imaging for affinity-based biosensors. Biosensors and Bioelectronics. 2010;25(5):957-966
  63. 63. Chen Y, Nguyen A, Niu L, Corn RM. Fabrication of DNA microarrays with poly(l-glutamic acid) monolayers on gold substrates for SPR imaging measurements. Langmuir. 2009;25(9):5054-5060
  64. 64. Fuchs J, Dell’Atti D, Buhot A, Calemczuk R, Mascini M, Livache T. Effects of formamide on the thermal stability of DNA duplexes on biochips. Analytical Biochemistry. 2010;397(1):132-134
  65. 65. Lee HJ, Wark AW, Li Y, Corn RM. Fabricating RNA microarrays with RNA-DNA surface ligation chemistry. Analytical Chemistry. 2005;77(23):7832-7837
  66. 66. Villiers MB, Cortes S, Brakha C, Marche P, Roget A, Livache T. Polypyrrole-peptide microarray for biomolecular interaction analysis by SPR imaging. Methods in Molecular Biology. 2009;570:317-328
  67. 67. Bellon S, Buchmann W, Gonnet F, Jarroux N, Anger-Leroy M, Guillonneau F, et al. Hyphenation of surface plasmon resonance imaging to matrix-assisted laser desorption ionization mass spectrometry by on-chip mass spectrometry and tandem mass spectrometry analysis. Analytical Chemistry. 2009;81(18):7695-7702
  68. 68. Lee HJ, Wark AW, Corn RM. Microarray methods for protein biomarker detection. The Analyst. 2008;133(8):975-983
  69. 69. Grant CF, Kanda V, Yu H, Bundle DR, McDermott MT. Optimization of immobilized bacterial disaccharides for surface plasmon resonance imaging measurements of antibody binding. Langmuir. 2008;24(24):14125-14132
  70. 70. Zeidan E, Li S, Zhou Z, Miller J, Sandros MG. Single-multiplex detection of organ injury biomarkers using SPRi based nano-immunosensor. Scientific Reports. 2016;6:36348
  71. 71. Hoffman IEA, Peene I, Veys EM, De Keyser F. Detection of specific antinuclear reactivities in patients with negative anti-nuclear antibody immunofluorescence screening tests. Clinical Chemistry. 2002;48(12):2171-2176
  72. 72. Meroni L, Schur PH. ANA screening: An old test with new recommendations. Annals of the Rheumatic Diseases. 2010;69(8):1420-1422
  73. 73. Jain A, Sharma MC, Sarkar C, Bhatia R, Singh S, Handa R. Major histocompatibility complex class I and II detection as a diagnostic tool in idiopathic inflammatory myopathies. Archives of Pathology and Laboratory Medicine. 2007;131(7):1070-1076
  74. 74. Sundaram C, Uppin MS, Meena AK. Major histocompatibility complex class I expression can be used as a diagnostic tool to differentiate idiopathic inflammatory myopathies from dystrophies. Neurology India. 2008;56(3):363-367
  75. 75. Van der Pas J, Hengstman GJ, ter Laak HJ, Borm GF, van Engelen BG. Diagnostic value of MHC class I staining in idiopathic inflammatory myopathies. Journal of Neurology, Neurosurgery, and Psychiatry. 2004;75(1):136-139
  76. 76. Das L, Blumbergs PC, Manavis J, Limaye VS. Major histocompatibility complex class I and II expression in idiopathic inflammatory myopathy. Applied Immunohistochemistry and Molecular Morphology. 2013;21(6):539-542
  77. 77. Hohlfeld R, Engel AG. The immunobiology of muscle. Immunology Today. 1994;15(6):269-274
  78. 78. Nagaraju K, Casciola-Rosen L, Lundberg I, Rawat R, Cutting S, Thapliyal R, et al. Activation of the endoplasmic reticulum stress response in autoimmune myositis: Potential role in muscle fibre damage and dysfunction. Arthritis & Rheumatism. 2005;52(6):1824-1835
  79. 79. Dalakas MC. Inflammatory muscle diseases: A critical review on pathogenesis and therapies. Current Opinion in Pharmacology. 2010;10(3):1-7
  80. 80. Dalakas MC, Hohlfeld R. Polymyositis and dermatomyositis. Lancet. 2003;362(9388):971-982
  81. 81. Civatte M, Schleinitz N, Krammer P, Fernandez C, Guis S, Veit V, et al. Class I MHC detection as a diagnostic tool in non informative muscle biopsies of patients suffering from dermatomyositis. Neuropathology and Applied Neurobiology. 2003;29(6):546-552
  82. 82. Daar AS, Fuggle SV, Fabre JW, Ting A, Morris PJ. The detailed distribution of MHC class II in normal human organs. Transplantation. 1984;38(3):293-298
  83. 83. Picard C, Vincent T, Lega JC, Hue S, Fortenfant F, Lakomy D, et al. Heterogeneous clinical spectrum of anti-SRP myositis and importance of the methods of detection of anti-SRP autoantibodies: A multicentric study. Immunologic Research. 2016;64(3):677-686
  84. 84. Orton SM, Peace-Brewer A, Schmitz JL, Freeman K, Miller WC, Folds JD. Practical evaluation of methods for detection and specificity of autoantibodies to extractable nuclear antigens. Clinical and Diagnostic Laboratory Immunology. 2004;11(2):297-301
  85. 85. González C, Guevara P, Alarcón I, Hernando M, Navajo JA, González-Buitrago JM. Antinuclear antibodies (ANA) screening by enzyme immunoassay with nuclear HEp-2 cell extract and recombinant antigens: Analytical and clinical evaluation. Clinical Biochemistry. 2002;35(6):463-469
  86. 86. Haass M, Lehmann HP. New aspects of autoantibody detection with a new combination of autoantigenic targets. Clinical and Applied Immunology Reviews. 2001;1(3-4):193-200
  87. 87. Jaskowski TD, Schroder C, Martins TB, Mouritsen CL, Litwin CM, Hill HR. Screening for antinuclear antibodies by enzyme immunoassay. American Journal of Clinical Pathology. 1996;105(4):468-473
  88. 88. Stinton LM, Fritzler MJ. A clinical approach to autoantibody testing in systemic autoimmune rheumatic disorders. Autoimmunity Reviews. 2007;7(1):77-84
  89. 89. Illa I, Cortes-Vicente E, Martinez MA, Gallardo E. Diagnostic utility of cortactin antibodies in myastenia gravis. Annals of the New York Academy of Sciences. 2018;1412(1):90-94
  90. 90. Shi J, Knevel R, Suwannalai P, van der Linden MP, Janssen GM, van Veelen PA, et al. Autoantibodies recognising carbamylated proteins are present in sera of patients with rheumatoid arthritis and predict joint damage. PNAS. 2011;108(42):17372-17377
  91. 91. Eissfeller P, Sticherling M, Scholz D, Hennig K, Lüttich T, Motz M, et al. Comparison of different test systems for simultaneous autoantibody detection in connective tissue diseases. Annals of the New York Academy of Sciences. 2005;1050:1-13
  92. 92. Gordon P, Khamashta MA, Rosenthal E, Simpson JM, Sharland G, Brucato A, et al. Anti-52 kDa Ro, anti-60 kDa Ro, and anti-La antibody profiles in neonathal lupus. The Journal of Rheumatology. 2004;31(12):2480-2487
  93. 93. Haroon N, Tsui FW, Uchanska-Ziegler B, Ziegler A, Inman RD. Endoplasmic reticulum aminopeptidase 1 (ERAP1) exhibits functionally signifi cant interaction with HLA-B27 and relates to subtype specifi city in ankylosing spondylitis. Annals of the Rheumatic Diseases. 2012;71(4):589-595
  94. 94. Stagakis I, Bertsias G, Karvounaris S, Kavousanaki M, Virla D, Raptopoulou A, et al. Anti-tumor necrosis factor therapy improves insulin resistance, beta cell function and insulin signaling in active rheumatoid arthritis patients with high insulin resistance. Arthritis Research and Therapy. 2012;14(3):R141
  95. 95. Tsui FWL, Tsui HW, Las Heras F, Pritzker KPH, Inman RD. Serum levels of novel noggin and sclerostin-immune complexes are elevated in ankylosing spondylitis. Annals of the Rheumatic Diseases. 2014;73(10):1873-1879
  96. 96. Rizzo R, Farina I, Bortolotti D, Galuppi E, Padovan M, Di Luca D, et al. The dimeric form of HLA-G molecule is associated with the response of early rheumatoid arthritis (ERA) patients to methotrexate. Clinical Rheumatology. 2017;36(3):701-705
  97. 97. Chen Z, Dodig-Crnković T, Schwenk JM, Tao S-c. Current applications of antibody microarrays. Clinical Proteomics. 2018;15:7
  98. 98. Lin MW, Ho JW, Harrison LC, dos Remedios CG, Adelstein S. An antibody-based leukocyte-capture microarray for the diagnosis of systemic lupus erythematosus. PLoS One. 2013;8(3):e58199
  99. 99. Carlsson A, Wuttge DM, Ingvarsson J, Bengtsson AA, Sturfelt G, Borrebaeck CA, et al. Serum protein profiling of systemic lupus erythematosus and systemic sclerosis using recombinant antibody microarrays. Molecular and Cellular Proteomics. 2011;10(5):M110.005033
  100. 100. Schumacher S, Muekusch S, Seitz H. Up-to-date applications of microarrays and their way to commercialization. Microarrays (Basel). 2015;4(2):196-213
  101. 101. Charpin C, Arnoux F, Martin M, Toussirot E, Lambert N, Balandraud N, et al. New autoantibodies in early rheumatoid arthritis. Arthritis Research and Therapy. 2013;15(4):R78
  102. 102. Abel L, Kutschki S, Turewicz M, Eisenacher M, Stoutjesdijk J, Meyer HE, et al. Autoimmune profiling with protein microarrays in clinical applications. Biochimica et Biophysica Acta. 2014;1844(5):9779-9787
  103. 103. Sajda T, Hazelton J, Patel M, Seiffert-Sinha K, Steinman L, Robinson W, et al. Multiplexed autoantigen microarrays identify HLA as a key driver of anti-desmoglein and -non-desmoglein reactivities in pemphigus. PNAS. 2016;113(7):1859-1864
  104. 104. Kim YG, Sohn DH, Zhao X, Sokolove J, Lindstrom TM, Yoo B, et al. Role of protein phosphatase magnesium-dependent 1A and anti-protein phosphatase magnesium-dependent 1A autoantibodiesin ankylosing spondylitis. Arthritis & Rheumatism. 2014;66(10):2793-2803
  105. 105. Gonzalez C, Garcia-Berrocal B, Talavan T, Cassas ML, Navajo JA, Gonzalez-Buitargo JM. Clinical evaluation of a microsphere bead-based flow cytometry assay for the simultaneous determination of anti thyroid peroxidase and anti thyroglobulin antibodies. Clinical Biochemistry. 2005;38(11):966-972
  106. 106. Gonzales-Buitrago JM. Multiplexed testing in the autoimmunity laboratory. Clinical Chemistry and Laboratory Medicine. 2006;44(10):1169-1174
  107. 107. Rouquete AM, Desgruelles C, Laroche P. Evaluation of the new multiplexed immunoassay, FIDIS, for simultaneous quantitative determination of antinuclear antibodies and comparison with conventional methods. American Journal of Clinical Pathology. 2003;120(5):676-681
  108. 108. Feng Y, Ke X, Ma R, Chen Y, Hu G, Liu F. Parallel detection of autoantibodies with microarray in rheumatoid diseases. Clinical Chemistry. 2004;50(2):416-422
  109. 109. Fritzler MJ. Advances and applications of multiplexed diagnostic technologies in autoimmune diseases. Lupus. 2006;15(7):422-427
  110. 110. Seideman J, Peritt D. A novel monoclonal antibody screening method using the Luminex-100 microsphere system. Journal of Immunological Methods. 2002;267(2):165-171
  111. 111. Mattey DL, Packham JC, Nixon NB, Coates L, Creamer P, Hailwood S, et al. Association of cytokine and matrix metalloproteinase profiles with disease activity and function in ankylosing spondylitis. Arthritis Research and Therapy. 2012;14(3):R127
  112. 112. Limon-Camacho L, Vargas-Rojas MI, Velasquez-Mellardo J, Casasola-Vargas J, Moctezuma JF, Burgos-Vargas R, et al. In vivo peripheral blood proinflammatory T cells in patients with ankylosing spondylitis. Journal of Rheumatology. 2012;39(4):830-835
  113. 113. Mou Y, Wu Z, Gu J, Liao Z, Lin Z, Wei Q, et al. HLA-B27 polymorphism in patients with juvenile and adult-onset ankylosing spondylitis in Southern China. Tissue Antigens. 2010;75(1):56-60
  114. 114. Lokate AMC, Beusink JB, Besselink GAJ, Pruijn GJM, Schasfoort RBM. Biomolecular interaction monitoring of autoantibodies by scanning surface plasmon resonance microarray imaging. Journal of the American Chemical Society. 2007;129(45):14013-14018
  115. 115. van Beers JBC, Willemze A, Jansen JJ, Engbers GHM, Salden M, Raats J, et al. ACPA fine-specificity profiles in early rheumatoid arthritis patients do not correlate with clinical features at baseline or with disease progression. Arthritis Research and Therapy. 2013;15(5):R140
  116. 116. van Beers JJ, Raijmakers R, Alexander LE, Stammen-Vogelzangs J, Lokate AM, Heck AJ, et al. Mapping of citrullinated fibrinogen B-cell epitopes in rheumatoid arthritis by imaging surface plasmon resonance. Arthritis Research and Therapy. 2010;12(6):R219
  117. 117. Martinez-Perdiguero J, Retolaza A, Bujanda L, Merino S. Surface plasmon resonance immunoassay for the detection of theTNFα biomarker in human serum. Talanta. 2014;119:492-497
  118. 118. Buhl A, Metzger JH, Heegaard NH, von Landenberg P, Flek M, Luppa PB. Novel biosensorbased analytic device for the detection of anti-double-stranded DNA antibodies. Clinical Chemistry. 2007;53(2):334-341
  119. 119. Vergani D, Alvarez F, Bianchi FB, Cancado ELR, Mackay IR, Manns MP, et al. Liver autoimmune serology: A consesus statement from the committee for autoimmune serology of the International Autoimmune Hepatitis Group. Journal of Hepatology. 2004;41(4):677-683
  120. 120. Törn C, Mueller P, Schlosser M, Bonifacio E, Bingley P. Diabetes Antibody Standardization Program: Evaluation of assays for autoantibodies to glutamic acid decarboxylase and islet antigen-2. Diabetologia. 2008;51(5):846-852
  121. 121. Hayashi N, Kawamoto T, Mukai M, Morinobu A, Koshiba M, Kondo S, et al. Detection of antinuclear antibodies by use of an enzyme immunoassay with nuclear HEp-2 cell extract and recombinant antigens: Comparison with immunofluorescence assay in 307 patients. Clinical Chemistry. 2001;47(9):1649-1659
  122. 122. Fenger M, Wiik A, Høter-Madsen M, Lykkegaard JJ, Rozenfeld T, Hansen MS, et al. Detection of antinuclear antibodies by solid-phase immunoassays and immunofluorescence analysis. Clinical Chemistry. 2004;50(11):2141-2147
  123. 123. Tozzoli R, Bizzaro N, Tonutti E, Pradella M, Manoni F, Vilalta D, et al. Immunoassay of anti-thyroid autoantibodies: High analytical variability in second generation methods. Clinical Chemistry and Laboratory Medicine. 2002;40(6):568-573
  124. 124. Fritzler MJ, Behmanesh F, Fritzler ML. Analysis of human sera that are polyreactive in an addressable laser bead immunoassay. Clinical Immunology. 2006;120(3):349-356
  125. 125. Nifli AP, Notas G, Mamoulaki M, Niniraki M, Ampartzaki V, Theodoropoulos PA, et al. Comparison of a multiplex, bead-based fluorescent assay and immunofluorescence methods for the detection of ANA and ANCA autoantibodies in human serum. Journal of Immunological Methods. 2006;311(1-2):189-197
  126. 126. Biagini RE, Parks CG, Smith JP, Sammons DL, Robertson SA. Analytical performance of the AtheNA MultiLyte ANAII assay in sera from lupus patients with multiple positive ANAs. Analytical and Bioanalytical Chemistry. 2007;388(3):613-618
  127. 127. Avaniss-Aghajani E, Berzon S, Sarkissian A. Clinical value of multiplex bead based immunoassay for detection of autoantibodies to nuclear antigens. Clinical and Vaccine Immunology. 2007;14(5):505-509
  128. 128. Salamunić I, Pauković-Sekulić B, Galetović A, Tandara L, Martinović-Kaliterna D. Comparative analysis of multiplex AtheNA Multi-Lyte ANA test system and conventional laboratory methods to detect autoantibodies. Biochemia Medica. 2008;18(1):88-98
  129. 129. Elshal MF, McCoy JP. Multiplex bead array assays: Performance evaluation and comparison of sensitivity to ELISA. Methods. 2006;38(4):317-323

Written By

Emilia Manole, Alexandra E. Bastian, Ionela D. Popescu, Carolina Constantin, Simona Mihai, Gisela F. Gaina, Elena Codrici and Monica T. Neagu

Submitted: 08 November 2017 Reviewed: 26 February 2018 Published: 05 November 2018