Abstract
Background
It is estimated that the average time between the diagnosis of Alzheimer’s disease (AD) and the patient’s death is 5-9 years. Therefore, both the initial phase of the disease and the preclinical state can be included in the critical period in disease diagnosis. Accordingly, huge progress has recently been observed in biomarker research to identify risk factors for dementia in older people with normal cognitive functions and mild cognitive impairments.
Methods
Electrochemical biosensors are excellent analytical tools that are used in the detection of AD biomarkers as they are easy to use, portable, and can do analysis in real time.
Results
This review presents the analytical techniques currently used to determine AD biomarkers in terms of their advantages and disadvantages; the most important clinical biomarkers of AD and their role in the disease. All recently used biorecognition molecules in electrochemical biosensor development, i.e., receptor protein, antibodies, aptamers and nucleic acids, are summarized for the first time. Novel electrochemical biosensors for AD biomarker detection, as ideal analytical platforms for point-of-care diagnostics, are also reviewed.
Conclusion
The article focuses on various strategies of biosensor chemical surface modifications to immobilize biorecognition molecules, enabling specific, quantitative AD biomarker detection in synthetic and clinical samples. In addition, this is the first review that presents innovative single-platform systems for simultaneous detection of multiple biomarkers and other important AD-associated biological species based on electrochemical techniques. The importance of these platforms in disease diagnosis is discussed.
Keywords: Alzheimer’s disease, biomarkers detection, early diagnosis, biosensors, surface modification, multiplex assays
1. INTRODUCTION
The morbidity of AD doubles every 5 years in people over 60 years and currently affects more than 44 million people worldwide [1-7]. It is predicted that this number will increase to 131.5 million people by 2050 [8], i.e., about 1 in 85 will be affected by AD if current trends persist without counteraction resulting from medical progress [9, 10]. The disease process can begin 20 years before the first signs of cognitive decline [11, 12]. Due to the great adaptation abilities of the human brain, the first important symptoms may appear only after the disappearance of about ¾ of the neurons. In this case, the brain exceeds the threshold of cognitive performance.
Despite medical progress and the development of scientific knowledge, AD remains an incurable disease, and its definitive diagnosis can only be made after the patient’s death based on the pathophysiological image of the brain [13]. To date, the molecular cause of AD-causing processes has not been clearly defined and remains a contentious issue [14, 15]. According to commonly accepted criteria, the occurrence of dementia is the condition for diagnosing AD [16]. The disease develops very slowly and the resulting changes are irreversible. In some cases, significant deterioration is observed within 2-3 years, in others, the course of dementia is slower and may last over 10 years. It is known that pathological abnormalities, such as amyloid plaques, form approximately 10-20 years before the appearance of cognitive dysfunction symptoms and considerable loss of nerve cells [17]. Early diagnosis of people without cognitive impairment (i.e., before the loss of neurons and synapses) would allow the use of new effective therapies in the future, giving hope for effective treatment and maintaining normal brain functions. The possibility of early detection of the disease, and thus the early use of anti-inflammatory drugs or antioxidants long before the onset of the first symptoms, can even prevent the occurrence of the disease [3].
Accordingly, great progress has recently been observed in biomarker research aimed at identifying dementia risk factors in people with normal cognitive functions [3, 5, 18]. Mass spectrometry (MS) [19], manganese-enhanced magnetic resonance imaging (MEMRI) [20], enzyme linked immunosorbent assay (ELISA) [21-23], flexible multi-analyte profiling (xMAP) [24], immunohistochemistry (IHC) [25-27], western-blot [28-30], fluorescence [31-33] and position emission tomography (PET) [34] are among techniques currently used to determine AD biomarkers. Despite the undoubted merits, the main disadvantages of these analytical methods are that they are time-consuming, relatively expensive, hardly available, require large sample volume and specialized equipment; and they are not yet adapted for point-of-care (POC) diagnostics (Table 1). The ease-of-use platforms with high sensitivity and specificity, and suitable for POC diagnostics are still critically needed.
Table 1.
Techniques for AD biomarkers detection.
| Technique | Advantages | Disadvantages | Refs. |
|---|---|---|---|
| IHC | The ability to simultaneously detect many antigens in hundreds of tissues. Routinely performed. Low technological requirements. Preservation of histological information. |
Expensive. Time-consuming. Variable antibody reactivity. Non-specific background signal. Variability dependent on fixation procedure, staining protocol, and antibody selection. |
[35-37] |
| ELISA | Cost-effective. Widely established. Absorbance is proportional to antigen concentration. Quantitative, very sensitive. |
Time-consuming. Inefficient. Insensitive to low level markers. Non-specific interactions. False positives/negatives possible, particularly with altered/mutated antigen. High amounts of protein lysate required. |
[38, 39] |
| MS – based technologies | No protein binding reagent required. Protein isoforms can be distinguished. Good combination of sensitivity and selectivity. Highly specific chemical information (accurate mass, characteristic fragment ions). |
Expensive. Time-consuming. Strict low-pressure requirements. Require specialized equipment. Hardly available. |
[38, 40-43] |
| MRI | High tissue contrast. Not or minimally invasive procedure. No radiation associated with imaging. No need for iodinated contrast. Superior soft tissue imaging with excellent spatial resolution. |
High cost. Limited availability. Claustrophobia due to the smaller patient bore. Contraindicated in patients with some metal implants and fragments. Requires specialized equipment. Hardly available. |
[44-46] |
| Western-blot | Separation of proteins according to molecular weight. Specific interaction of antibody and antigen can be directly visualized. Relatively simple and cost-effective method. |
Low- or medium-throughput. High amounts of protein lysate required. An imbalance in any step of the procedure may skew the entire process. |
[38, 47] |
| xMAP | Reduced cost and labour by multiplexing for complex projects. Smaller sample requirements for complex projects. Open architecture platform. |
Expensive. Cross reactivity of some antibodies, which has to be avoided. Requires the optimization during initial stages in the development of new assays. Hardly available. |
[48] |
| PET | Providing functional and biological information. May have diagnostic value detecting metastatic lesions that would have been missed on conventional imaging. |
High cost. Poor spatial resolution and lesion detectability. Shielding to avoid radiation exposure. Administration by intravenous injection of radiopharmaceutical compounds. |
[44, 49] |
| Fluorescence | Immune to light scattering. High specificity due to unique optical properties of molecules. It can measure analyte concentration in terms of fluorescence intensity and decay times. High sensitivity (especially with iodinated ligands). |
Susceptible to autofluorescence. Limitations associated with photostability and loss of recognition capability. |
[49, 50] |
The development of electrochemical biosensors is probably one of the most promising methods to solve some of the problems regarding sensitive, fast and cost-effective measurements [51]. Furthermore, their potential in microfabrication and portability can also be used to allow for their use in simple point-of-care devices and aid in drug-screening processes of effective therapeutic molecules for neurodegeneration associated with AD.
The other important issue related to biosensors is the proper immobilization of biorecognition molecules on the surface of electrodes, which affects the sensitivity and specificity of biomarker determination. The immobilization procedure must maintain the molecule responsible for biorecognition close to the transducer surface while retaining its biological activity in a reproducible manner. The immobilization layer should give the biological molecule enhanced stability [52-54]. There are different strategies of electrode surface functionalization for the immobilization of biorecognition molecules on a solid support. Nonetheless, the covalent binding between these molecules and the electrode surface is one of the most widely used. Just for examples, immobilization of antibodies on the glutaraldehyde (GA) layer [55] or on 3 cyanopropyltrimethoxysilane self-assembled monolayers [56] or also immobilization of thiolated antibodies on the gold surface [57] has been reported.
In recent years, the field of biosensors has been growing, and the application of nanotechnology has developed as one of the biggest opportunities to achieve higher sensitivity for biosensors [58]. At the present time, in the construction of electrochemical biosensors for biomedical applications associated with AD biomarkers detection, the use of different types of nanomaterials like porous magnetic microspheres [59], nanotubes [60], graphene oxide [61], indium tin oxide [56] and metallic nanoparticles [62] has been reported in this review.
The important challenge in electrochemical biosensors development concerns simultaneous multianalytes detection. This is extremely important in the context of detection of multiple AD biomarkers, which is fundamental for correct disease diagnosis and prognosis. Simultaneous detection of several clinically relevant biomarkers is essential for clinical applications and it is an effective solution in improving diagnostic value, while a single biomarker detection is usually not sufficient. Recently, the interest in electrochemical biosensors for simultaneous multiple biomarker detection, including AD biomarkers, has gained more attention than ever, and this issue will also be discussed in this review.
2. BIOMARKERS OF AD DIAGNOSIS
A biomarker has been defined as an objectively measurable change arising in biological environments such as human cells, tissues or body fluids. This change may describe the pathological condition or the body’s response to treatment when assessing the effectiveness of pharmacological therapy [63, 64]. Compounds that are “candidates” for AD biomarkers must reflect the basic neuropathological characteristics of this disease. Biomarker determination should be performed using a quick, safe and easy diagnostic test, which allows detecting the disease in the phase before the appearance of characteristic clinical symptoms [17].
The search for AD markers concerned the analysis of the cerebrospinal fluid since compounds that reflect all pathological conditions can be found in the fluid that washes the diseased tissue of the central nervous system. However, it is much easier to use blood as a source of biomarkers for diagnostic purposes, because
the access to the blood is very easy and the method of its collection is less invasive, which involves much less discomfort for the patient. Moreover, the detection of AD biomarkers in the blood is not an easy task. First of all, there is the so-called blood-brain barrier, hindering the transport of potential AD markers from the cerebrospinal fluid to the blood, which results in the possibility of too low blood concentrations [65]. Secondly, the blood component is a protein characterized by high abundance. For this reason, the determination of low-abundance proteins is much more difficult, especially that the concentration of proteins in the blood is higher than in the cerebrospinal fluid [66].
At present, three cerebrospinal fluid (CSF) biomarkers for AD diagnosis have been established and published worldwide: amyloid β peptides (Aβ1-42 and Aβ1-40), total tau protein and phosphorylated tau protein [67]. S100B protein, apolipoprotein E and glycated albumin are also major etiological AD factors [68-72], which are listed as the important biomarkers of this disease. These biomarkers with a significant role in the clinical practice of AD are presented below:
(I) Amyloid-β (Aβ) peptides consisting of about 40 amino acids are components of another large protein called Aβ-APP peptide precursor protein. β-amyloid is
formed from the precursor protein APP by non-amyloidogenic and amyloidogenic proteolysis (Fig. 1) [73].
Fig. (1).
A series of endoproteolytic cleavage of the APP protein leading to the formation of peptides by non-amyloidogenic and amyloidogenic pathways. In the first of them, the process takes place using α- and γ-secretase enzymes, while in the second one, β- and γ-secretase are involved. Non-amyloidogenic secretion products are soluble α-APPs peptide and non-toxic P3 and P7 peptides. Cleavage of APP with α-secretase prevents β-amyloid formation. The amyloidogenic hydrolysis products are soluble β-APPs protein and P11 peptide. The latter is further hydrolyzed by γ-secretase to release a 39-43 amino acid β-amyloid peptide because γ-secretase can cleave P11 peptide at slightly different locations. A form of β-amyloid 42 (Aβ1-42) is also produced, but in a smaller amount [74]. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Each of the forms of soluble oligomers resulting from APP transformations has a different mechanism of neurotoxic action. The oligomeric forms display a neurotoxic action: di-, tri-, tetramers and higher forms have been distinguished. Aβ peptide dimers are involved in the initiation of the proinflammatory reaction by activating glial cells. These cells secrete agents with potential neurotoxic effects, i.e., free radicals or cytokinins. Thus, dimers alone do not act directly adversely on nerve cells [75]. Medium soluble and globular forms have the ability to binding to synapses and destroy the connections between them. Their long-term neurotoxic effect is manifested by the death of nerve cells in a specific region of the hippocampus-C1 [76]. Protofibrillar forms affected the electrophysiological properties of membranes, including action potential and membrane depolarization [77]. The cause of cell death is the toxic effect of aggregated forms of Aβ [78]. Histopathological examination reveals outside neurons the senile plaques containing fibrillar Aβ [79, 80]. Elevated levels of the β-amyloid peptide can be detected even many years before the symptoms of the disease are recorded [72, 81]. From the point of view of early diagnosis, it is important to observe the dynamics of plasma changes in the level of β-amyloid peptide composed of 40 and 42 amino acids, which may enable an assessment of Alzheimer’s disease risk. The physiological concentration of Aβ present in human plasma as well as in the cerebrospinal fluid ranges from 0.1 to 0.5 nM [82]. An increase in total Aβ concentration is observed in the first asymptomatic preclinical phase [83]. As the disease progresses and amyloid deposits gradually form in the brain, Aβ, and in particular Aβ1-42 levels, often decrease to normal. This information indicates that fundamental biochemical events relevant to AD can be monitored in blood.
(II) Another important AD biomarker is tau protein. The aggregation of tau into neurofibrillary tangles within brain tissues is associated with AD pathology. This protein is responsible for the binding and stabilization of microtubules in neuronal axons [74], a process that is inhibited when tau becomes phosphorylated. AD-related pathology proceeds in precisely designate stages, based on the conception that neurofibrillary tangles develop from an accumulation of abnormal tau. Abnormal phosphorylation is a decisive step causing the formation of tau filaments (soluble and insoluble) [84]. During the neurodegeneration process, tau undergoes hyperphosphorylation, which results in conformational changes and its dissociation from microtubules, as well as self-association in glycates [85]. Complementary animal models indicate that tau hyperphosphorylation alone can cause neurodegeneration, leading researchers to conclude that hyperphosphorylated tau is toxic to neurons and plays an important role in AD neuropathology [86]. High levels of tau in the CSF of AD patients can reflect the intensity of neuronal damage and brain degeneration [87]. In AD, total tau (t-tau) or phosphorylated tau (p-tau) concentrations are increased compared to cognitively normal individuals [88].
(III) The brain in Alzheimer’s disease is characterized by high levels of S100 family proteins, especially S100B protein – a protein that binds calcium ions [89, 90]. In the brain, S100B protein is secreted primarily in glial cells, such as astrocytes and oligodendrocytes [91, 92]. Its elevated level was found in the serum and cerebrospinal fluid of patients with AD [93, 94]. It is believed that its higher concentration plays a role in the pathogenesis of neurodegenerative processes [69, 89]. S100B induces both neurotrophic and potential neurotoxic effects, depending on its concentration in the body [95]. Nanomolar concentrations of S100B stimulate the growth of neuritis [92], while micromolar concentrations of this protein are detrimental to these structures [96]. High levels of extracellular S100B protein are detected after serious brain injuries or in the case of neurodegenerative diseases such as AD [69, 97, 98]. Controlling the S100B protein level may be useful in evaluating the severity of dementia progressing with disease development [68].
(IV) Another important AD biomarker is glycated albumin. Albumin is a protein found in the highest concentration in both cerebrospinal fluid and plasma. Albumin present in plasma is mainly produced by the liver, and a small portion of it enters the brain [99]. The process of albumin glycation results in the formation of toxic aggregates [100] and significantly inhibits the slowing of Aβ1-42 aggregation [101]. It can be speculated that glycation reduces the effect of aggregation inhibition by Aβ [101]. A similar mechanism has been proposed for prions that bind Aβ monomers and oligomers, depending on their conformational state [102, 103]. This reduction in the inhibitory effect of albumin on Aβ fibrillation is particularly important in the brain parenchyma, where Aβ aggregation plays a key role in the onset of AD. Albumin-related glycine modification affects cells on both sides of the “blood-brain barrier” and weakens the inhibition of Aβ filament formation, usually associated with native albumin [104]. The accumulation of pathological amounts of proteins, modified as a result of the glycation process in the patient’s brain, causes that these modifications may contribute to AD etiology. As already mentioned, the causes and pathological mechanisms of AD are not fully understood and are still under discussion [101]. However, higher levels of glycated albumin were found in the cerebrospinal fluid and plasma of Alzheimer’s patients compared to the control groups [101, 105, 106].
(V) Apolipoprotein E (ApoE) is a 299 amino acid protein encoded by the APOE gene [107]. ApoE lipoproteins bind to several cell surface receptors to deliver lipids, as well as to hydrophobic Aβ peptide, which is believed to initiate toxic events leading to synaptic dysfunction and neurodegeneration in AD [108]. The apoE protein is supposed to influence AD pathogenesis through a variety of actions. It influences the innate immune system, the effects of the blood-brain barrier, for the accumulation of Aβ and synaptic function [109]. APOE4 alleles are correlated with cholinergic dysfunction and increased amyloid burden [110]. Several studies have demonstrated the important involvement of apoE in AD. This was first suggested by Strittmatter and Roses [10], who showed that, of the three polymorphic forms of APOE, carriers of APOE4 are more liable to develop AD. Additionally, the cognitive changes in the APOE4 carriers were demonstrated to occur several years earlier, with a dose-dependent effect [86]. However, the exact influence of APOE on AD and dementia pathophysiology is unclear. ApoE isoforms differentially regulate Aβ aggregation and clearance in the brain, and have distinct functions in brain lipid transport, glucose metabolism, neuronal signaling, neuroinflammation and mitochondrial function [108].
3. SENSORS
According to the IUPAC definition, a chemical sensor is a device that converts chemical information about the concentration of a particular sample component to an analytically useful signal. It consists of two parts: an analytically active layer in which the intermolecular (receptor-analyte) recognition process takes place and transducer. The chemical or physicochemical signal, generated as a result of the recognition process, is converted in the transducer section into an analytical signal. The transducer is, therefore, part of the sensor responsible for converting chemical information into an analytically useful signal [111].
The sensor should generate a repeatable analytical signal in a short time and have two characteristics: selectivity and sensitivity. The first feature determines the ability to accurately measure a particular size by omitting the influence of other parameters and the analytically active part is responsible for it. The second one determines the sensor’s predisposition to measure the smallest values of the sought-after quantity. The analytically active and transducer parts of the sensor correspond to this parameter [112].
Depending on the type of analytically active material, sensors are divided into chemical sensors and biosensors. In chemical sensors, analytically active parts are synthetic molecules (receptors) that selectively recognize analyte molecules [113]. In biosensors, the biological material is an analytically active material, most often involving antibodies, isolated enzymes, natural receptors, microorganisms, tissues, organelles, DNA, RNA or whole cells [113-115].
Biosensors are usually classified according to the type of signal transmitted, type of transducer, and are divided into electrochemical, electric, acoustic, optical and thermal/calorimetric sensors [110, 113]. Electrochemical transducers have been frequently used in biosensors for the detection of AD biomarkers due to the advantages of cost-effectiveness, easy production, and user friendliness [110]. Therefore, this review concerns biosensors based on these types of transducers (Fig. 2).
Fig. (2).
Scheme of a biosensor detection strategy. Interaction between biorecognition molecule and specific biomarkers generating bio-recognition signal. Transducers convert the biological recognition event into a measurable signal. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Biosensors are also often classified according to the reaction type that generates the analytical signal. Analyzing this parameter, we can distinguish between catalytic biosensors (e.g., enzymatic) and affinity biosensors (receptor protein reactions – analyte, antibody – antigen, hybridization reactions) [116].
The development of biosensors began in 1962, from the first mentions of the use of biosensors in the publication of Clark and Lyons [117]. The first amperometric biosensor described there was an enzyme electrode designed for glucose determination [118]. Since then, intensive research has been underway, resulting in the development of increasingly advanced, more sensitive and reliable biosensors [119].
3.1. Intermolecular Recognition
The operation of chemical sensors is based on intermolecular recognition processes occurring at the interface between the analytically active layer and the analyzed solution. The domain dealing with these issues is supramolecular chemistry, defined by one of its founders – J. M. Lehn – as chemistry of molecular aggregates and intermolecular bonds [120].
In the studies of supramolecular systems of the “guest-host” complexes, the main areas of interest include: determination of the complex structure, type of interactions and stoichiometry of supramolecular components, as well as constant stability of supramolecular systems and kinetic parameters [121].
Most often, the “host” (receptor) is a large molecule having specific “guests” (analytics) binding sites in the structure. The role of “guest” can be played by cations, anions or more complex molecules. Selective formation of the “guest-host” complex (receptor-analyte) requires mutual complementarity of its constituting molecules, i.e., achieving the appropriate geometric, spherical, energetic and electronic state. The term intermolecular recognition formulated by J. Rebek seems to be the most accurate: effective intermolecular recognition requires surfaces of complementary sizes, shapes and functions [121]. During complex formation, the analyte molecules fit into the structure of the “host” molecule as the “key to the lock”. This concept was used for the first time over a hundred years ago (in 1894) by E. Fischer and it has been functioning until now.
4. BIORECOGNITION MOLECULES
Synthetic and biological recognition molecules are at the heart of most of current bioreceptor assays. In principle, any biomolecules that have the capability of recognizing a target analyte can be used as a bioreceptor. Enzymes were the first recognition molecules combined in biosensor designs with broad range sensing applications. Nevertheless, other bioreceptors elements such as protein affinity systems and antibodies were introduced very shortly in the design of biosensors. Owing to the introduction of bioengineering techniques, and the complications to obtain recognition element against small size molecules, many novel biosensor recognition elements have been developed and synthesized [122]. Among the various types of biorecognition elements: receptor proteins, antibodies, aptamers and nucleic acids have been recently integrated into the biosensor designs as a popular choice for the detection of AD biomarkers, as discussed below.
4.1. Receptor Proteins
Receptor proteins are embedded in the cell membrane and span across the membrane, so that part of them is protruding out of the cell and the other part is located inside the cell. Receptor proteins are responsible for opening and closing of membrane channels for the transport of specific metabolites. These proteins are molecules characterized by a specific affinity for hormones, antibodies and other biologically active elements.
In the first years of using receptors in analytically active layers of biosensors [115], their low acquisition efficiency, relative instability, laborious isolation and long-term purification of proteins from cell membranes were quite a hindrance. Despite this, receptors as recognition elements showed high affinity and binding specificity for a specific analyte. Over time, due to the development of cell recombination techniques and gene expression systems, it is now possible to obtain receptor proteins in huge quantities from the point of view of biosensors. Therefore, the receptors are successfully used as analytically active elements in the biosensitive layer. In the literature, there are several successful applications of histidine-tagged receptor proteins that act as biosensor recognition elements [123-127].
Mikuła et al. used RAGE receptor domains to determine the presence of S100B protein in the buffer and in human plasma as well as in the presence of other potential markers for early diagnosis of Alzheimer’s disease (Aβ1-40). RAGE receptor domains were fused with poly-histidine tags. They are polypeptide elements that are introduced into the protein sequence by genetic engineering. Such a polyhistidine label consists of 2 to 10 histidines. The tag containing 6 histidines, often designated with His6-tag or 6xHis-tag abbreviations, was patented by Roche [128]. The presence of human plasma and other AD biomarkers (Aβ1-40) has no influence on biosensor performance. The detection limit was in the pM range and indicated that the biosensor was suitable for the determination of S100B protein in physiological samples [123, 128].
Histidine-tagged domains of RAGE receptor immobilized in biosensor monolayer were also used to determine Aβ16–23 and Aβ1–40 peptides [124-126]. These biosensors displayed good analytical parameters such as selectivity, sensitivity, detection limit in nM range and no effect of the human plasma matrix on the analytical signal. Considering the above arguments, the proposed biosensors could be included in the measurement tools suitable for cost-effective theranostics.
4.2. Antibodies
Antibodies (Abs), which belong to the most exquisitely designed molecules in nature, play an important role in a number of sensor devices due to their excellent target specificity and affinity. Many applications of Abs have been reported in the area of immunosensor development [129]. However, there is a huge gap between a vast number of priority analytes and a limited number of available immunosensors. Therefore, Abs engineering is a powerful tool for modifying antibodies properties. An article of Janata from 1975 is considered as the first development of an immunosensor [130]. However, it actually presents a biosensor with a receptor that is not an antibody, concanavalin A. Currently, immunosensors are the subject of not only many experimental articles, but also a number of reviews [131-134].
Guillozet-Bongaarts et al. introduced the tau-C3 monoclonal antibody specific to tau cleaved at aspartic acid 421 by caspase [135]. John Hardy et al. described transgenic AD mice in which Aβ was significantly reduced and amyloid plaques were removed after injection of anti-Aβ antibody [136]. Wang et al. used antibodies immobilized on the surface of a gold electrode in optimal orientation by protein G interaction as an analytically active element of an electrochemical biosensor for the detection of tau protein. The application of protein G for antibody immobilization ensured that Fab antibody binding domains were oriented away from the biosensor surface and free to react with target antigens. It increased the loading capacity of the biosensor and its sensitivity to antigens [137]. The latter biosensor was based on a microelectrode array with four gold microband electrodes and could detect the full-length of tau protein. It displayed a significantly lower quantification limit (0.03 pM) than the critical cut-off value (4.3 pM) of CSF tau protein, and this is important because recently described biosensors have a detection limit about five orders of magnitude higher than the CSF tau cut-off value, which differentiates AD cases from controls [138].
4.3. Aptamers
Aptamers are small single-stranded DNA or RNA nucleotides or peptides that exhibit high binding affinity and specificity for their targets such as metal ions and amino acids, proteins, antibodies, whole cells, bacteria or viruses [139-144]. They were first reported in 1990 by Ellington and Szostak [145], who presented RNA molecules that bind to a small organic dye. Since then, short strands of DNA or RNA that assume specific three-dimensional conformations and which are selected to target various molecules have been defined as nucleic acid aptamers. For in vitro selection of RNA or DNA aptamers, molecules from large populations of random sequences generated in a process known as SELEX (systematic evolution of ligands by exponential enrichment) are used. It is a combinatorial chemical technique involving the screening of specific ligands by repeated splitting and amplification rounds from a large nucleic acid library containing over a thousand different candidates [146].
Aptamers have a significant advantage in biomarker discovery over other recognition molecules due to their ability to distinguish between different modified forms and isoforms of the same protein. Moreover, it is important that aptamers affinity can be adapted by optimizing their recognition sequence or/and manipulating binding reaction conditions, making aptamers ideal molecular recognition tools [147].
Cleavage of the amyloid precursor protein (APP) by BACE-1 (β-site APP cleaving enzyme-1) results in the production of amyloid-β (Aβ). Thus, this enzyme should be a valuable target for the interference of Aβ production and the treatment of AD. Liang et al. selected A1-aptamer DNA with BACE1 binding properties of good affinity and specificity, and thus a potential of decreasing Aβ40 and Aβ42 production [148]. In turn, Rentmeister et al. selected RNA aptamers against the short cytoplasmic tail (B1-CT) of this APP cleaving enzyme from the β-site. They can specifically bind to B1-CT without affecting other essential biological activities, so that they can potentially be used to prevent or slow the onset of AD [149]. Zhou et al. described an electrochemical biosensor that uses Aβ oligomer antibodies and a nanocomposite of gold nanoparticles with aptamer and thionine as a recognition element for determining Aβ oligomers. Compared with the known Aβ detection method, the aptamer-based electrochemical assay was characterized by high sensitivity due to signal amplification by high thionine load on AuNPs, and high specificity resulting from highly specific recognition of Aβ oligomers with DNA aptamer antibodies. Due to these advantages, the biosensor was successfully applied to determine Aβ oligomers in artificial cerebrospinal fluid samples [62]. A sensitive inkjet-printed electrochemical aptasensor has been successfully fabricated for the detection of lysozyme. Lysozyme also plays an important role as a biomarker in various disease diagnosis such as AD. Aptamers were immobilized on the working electrode in such a way that the printed ink contained the dispersion of the CNT aptamer complex (dispersed CNT-aptamer complex). This method allows to control aptamer density as well as high-resolution patternability. The high affinity between single-stranded DNA and carbon nanotube (CNT) was used to immobilize aptamers. The biosensor was characterized by a 90 ng/mL detection limit and a reasonable shelf-life of approximately 21 days at room temperature [150].
4.4. Nucleic Acids
Nucleic acid based electrochemical biosensors have applications in diagnostics of AD. These biosensors primarily use deoxyribonucleic acid (DNA) as an oligonucleotide probe. The fundamental principle behind nucleic acid-based biosensors depends, among others, on sequence complementarity as per Chargaff's rules of base pairing (for DNA, A = T, G ≡ C), with the exception of aptamers [151]. Additionally, the complementary strand has to be antiparallel, which is a consequence of the double helix model described by Watson and Crick [154]. The principle of aptamer based detection is more akin to antigen-antibody or receptor-ligand interactions [152, 153].
Nucleic acid based biosensors are developed by immobilization of nucleic acids (DNA, RNA, oligonucleotides) to a solid support by adsorption, covalent bonding or ionic interaction [8]. The immobilization process also aids in probe orientation and ready accessibility to target element. Hybridization based biosensors rely on the duplex formation between nucleic acids. Hybridization normally takes place between a known DNA sequence, i.e., probe and an unknown counterpart i.e., target DNA, but DNA–RNA and RNA–RNA hybridizations can also occur [6]. The single-strand nucleic acid can bind through electrostatic interactions, hydrophobic interactions, or their complementary shapes [151].
DNA electrochemical biosensors have been developed for the detection of apoE genotypes in PCR-amplified DNA extracted from human blood [155-157]. An electrochemical DNA biosensor using a disposable electrochemical printed chip for the detection of apoE 4 from unpurified PCR amplicons has been reported by Ahmed et al. [155]. Marrazza et al. demonstrated a DNA electrochemical biosensor for apoE genotyping coupled with PCR. This method can discriminate the six genotypes of apoE [157]. Another sensitive DNA electrochemical biosensor for detection of the apoE 4 gene has been developed by Lu et al. The biotinylated oligonucleotide probe with a sequence complementary to apoE 4 gene at codon 112 was immobilized on the Au electrode via Au-S bond. To eliminate nonspecific adsorption of the conjugates, the electrode was blocked with 6-mercaptohexanol (MCH). The immobilized biotinylated oligonucleotide probe captures complementary apoE 4 gene [158].
5. ELECTROCHEMICAL BIOSENSORS FOR AD BIOMARKERS DETECTION
5.1. Singleplex Assays
Detection of AD biomarkers has been coupled with electrochemical transducers due to their high sensitivity, specificity, ease of use and fast response to the analyte of interest. Transducers can be classified by the mode of electrochemical transduction [159]. Typically, in (bio-) electrochemistry, the test reaction either generates a measurable current (amperometric), measurable potential or charge accumulation (potentiometric), or measurably alters the conductive properties of a medium (conductometric) between electrodes [160]. Cyclic voltammetry (CV), square wave voltammetry (SWV), differential pulse voltammetry (DPV) and electrochemical impedance spectroscopy (EIS) are among the most common electroanalytical techniques used for the detection of AD biomarkers [110]. Marrazza et al. developed new procedures for the detection of apolipoprotein E polymorphism in human blood based on coupling DNA electrochemical or piezoelectric sensors with polymerase chain reaction (PCR). The electrochemical sensor was obtained by immobilizing single-stranded oligonucleotides onto graphite screen-printed electrodes by adsorption at controlled potential [157]. Another ultrasensitive sandwich-type electrochemical immunosensor for the quantitative detection of APOE4 was designed based on fractal gold (FracAu) nanostructures and enzyme amplification. FracAu nanostructures were directly electrodeposited by hydrogen tetra-chloroaurate (HAuCl4) on a polyelectrolyte modified indium tin oxide (ITO) electrode. The detection performance of the modified interface was investigated using cyclic voltammetry (CV). After functionalization with HRP-labeled APOE4 antibody, human APOE4 could be quantitatively detected based on the current response [161]. Mikuła et al. presented an electrochemical biosensor consists a system of thiol derivative of pentetic acid (DPTA) complex with Cu(II) created on gold electrode surface for immobilization His-tagged domains of RAGE (Fig. 3). Domains of RAGE has been applied as an analytical active element for the determination of the glycated albumin. The analytical signals of a biosensor are generated based on the change in the electrochemical properties of the Cu(II) redox centers upon binding glycated albumin by His-tagged domains of RAGE. The recognition process was observed using the Osteryoung square – wave voltammetry (OSWV). The presence of 70 pM serum human albumin as well as 10 nM Aβ1-40 and S100B protein has a slight influence on the biosensor responses [162].
Fig. (3).
Schematic representation of glycated albumin detection. The immobilization of His6–RAGE domains consists of: (i) formation of a mixed layer of N-acetylcysteamine (NAC) and the thiol derivative of pentetic acid (DPTA); (ii) complexation of Cu(II) by DPTA; (iii) oriented immobilization of His6–RAGE domains via coordination bonds between Cu(II) sites from the DPTA–Cu(II) complex and imidazole nitrogen atoms of a histidine tag. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Another development in the field is a novel sandwich-type biosensor, which was capable of electrochemical detection of α-1 antitrypsin (AAT, a recognized biomarker for Alzheimer’s disease). The biosensor was composed of 3, 4, 9, 10-perylene tetracarboxylic acid/carbon nanotubes (PTCA-CNTs) as a sensing platform and alkaline phosphatase-labeled AAT antibody-functionalized silver nanoparticles as a signal enhancer. The biosensor exhibited desirable performance for AAT determination with wide linearity in the range from 0.05 to 20.0 pM and a low detection limit of 0.01 pM. Finally, the developed sensor was successfully applied to the analysis of AAT concentration in serum samples [60]. Another novel 384-multiwell microelectrode array (MMEA) based on a measurement system for sensitive label-free real-time monitoring of neurodegenerative processes using impedance spectroscopy was reported recently by Jahnke and colleagues. The MMEA system, in combination with the SH-SY5Y cell-based tauopathy model, introduced a novel 5-fold tau mutation, which eliminated the need for artificial tauopathy induction, and in consequence, allowed to quantitatively monitor the efficacy of potential novel therapeutics like SRN-003-556. The designed tauopathy screening system could be a useful tool to identify and develop novel therapeutics in the field of tau-related neurodegenerative diseases [163]. Dai et al. described a single-use, in vitro biosensor for the detection of t-tau protein in phosphate-buffer saline (PBS) and undiluted human serum. This biosensor consisted of three electrodes: working, counter, and reference electrodes fabricated on a PET sheet. Both working and counter electrodes were composed of a thin 10-nm-thick gold film. Measurements of t-tau protein in both 0.1 M PBS and undiluted human serum in the concentration range of 1000 pg/mL to 100,000 pg/mL showed excellent results and good linearity of calibration curves [164]. Another development in the field is a biosensor for the detection of tau protein intended for electrochemical observing of misfolding proteins. The biosensor monitored tau-tau binding and misfolding in the early stages of tau oligomerization (Fig. 4). The binding event between immobilized tau (tau-Au) acting as a recognition element and the tau protein solution was detected by the electrochemical impedance spectroscopy method. After binding of tau to tau-Au, the charge transfer resistance (Rct) decreased as a result of the creation of the tau-tau-Au interface. A linear relationship between tau solution concentration and Rct was noticed from 0.2 to 1.0 µM. Both electrochemical data and surface analysis indicated electrostatic and conformational changes induced by tau-tau binding. The designed electrochemical platform was highly selective for tau protein compared to bovine serum albumin and allowed the fast sample analysis [138]. Another development in the field is a sensitive and selective electrochemical platform designed by using gold nanoparticles (AuNPs) modified with Aβ1–16-heme
Fig. (4).
Illustration of the tau-based biosensor and tau-tau-Au interface. The tau-Au surface (a) was exposed to the protein solution under conditions forming the tau-tau-Au surface (b). Adapted with permission from Ref [138]. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
for the detection of total Aβ peptides. The gold nanoparticles modified with Aβ1–16-heme were captured by a monoclonal antibody specific for the common Aβ N-terminus. The antibody was immobilized onto the gold electode surface. Anchored AuNPs modified with Aβ1–16-heme demonstrated electrocatalytic O2 reduction. The incubation of the monoclonal antibody immobilized on the electrode surface with native Aβ decreased the amount of AuNPs modified with Aβ1–16-heme linked onto the electrode, causing a decrease in the reduction current of O2 to H2O2. Voltammetric responses were found to be proportional to the Aβ concentrations from 0.02 to 1.50 nM, and a detection limit of 10 pM was obtained [9]. Derkus et al. reported a novel immunosensor for the simultaneous quantification of myelin basic protein and tau protein in the cerebrospinal fluid and human serum. The immunosensor was developed based on the screen printed carbon electrode modified with graphene oxide and amine functionalized 1st generation trimethylolpropane tris[poly(propyleneglycol)] (pPG) dendrimers. A novel carrier system graphene oxide/pPG nanocomposite structure was used for the immobilization of antibodies of anti-myelin basic protein and anti-tau protein. The detection limits for immunosensor presented (0.30 nM for myelin basic protein and 0.15 nM for tau proteins) indicated its suitability for the levels required for analysis in a neuroclinic [61]. Another four-electrode electrochemical biosensor for the detection of tau protein was reported by Wang et al. The biosensor was based on the antibodies used as analytically active molecules immobilized on a self-assembled monolayer and protein G deposited on a gold microband electrodes surface. The assay performed by using electrochemical impedance spectroscopy was fast, very sensitive and displayed a linear response with increasing tau concentrations. The presence of human serum and bovine serum albumin has no influence on the biosensor performance. The detection limit for the full-length 2N4R tau protein was in the pM range (0.03 pM). Taking into account the above parameters, this technology could be adapted for the detection of different biomarkers to allow a multiple assay to identify AD progression in medical samples analysis [137]. Another sensitive approach utilizes the antibody-modified carbon fiber microelectrodes for in vivo analysis of Aβ concentration. Using this method, the Aβ concentrations in the interstitial fluid of the hippocampus of transgenic and wild- type mice with human Aβ were detected. The Aβ contained a tyrosine residue that can be oxidized at approximately 0.65 V (vs. Ag/AgCl) on a carbon surface. These authors used the square wave voltammetry to measure tyrosine oxidation. The antibody was immobilized on the electrode surface to achieve selectivity because there is a lot of other proteins that contain tyrosine. The authors also monitored Aβ half-life in vivo. They administered a γ-secretase inhibitor that inhibits Aβ generation and demonstrated that the Aβ levels decreased in a concentration-dependent manner after inhibitor treatment [165].
Another platform designated for electrochemical immunosensor employs indium tin oxide disposable sheets were modified using 3 cyanopropyltrimethoxysilane self-assembled monolayers as elements for precise immobilized anti-CRP antibody via covalent interactions without the need for any cross-linking agent. This sensitive approach allows the electrochemical detection of C-reactive protein (CRP) by analysis of charge transfer resistance changes. The proposed immunosensor was sensitive, with a detection limit of 0.455 fg mL−1. Detection of CRP in human serum samples was measured by fabricated biosensor to determine the feasibility of the biosensing system for medical purposes [56]. Another miniaturized platform designated for ultrasensitive biosensor employs monoclonal amyloid-beta antibodies (mAb) as a biorecognition elements immobilized on a disc-shaped platinum/iridium (Pt/Ir) microelectrode surface. The novel approach in the modification strategy of microelectrode relied on electropolymerization by conducting free amine-containing aromatic polymer (poly (ortho-phenylenediamine) (PoPD)), followed by cross-linking with glutaraldehyde (GA) for subsequent covalent coupling of mAβ on the microelectrode surface, GA created a stable ‘click compound’ on the microelectrode surface by covalently linking to the amine groups of hydroxylysine or lysine in the antibodies (Fig. 5). This approach improved the impedimetric detection performance of Aβ1-40 in terms of charge transfer resistance (about 400- fold difference) compared to the adsorption-based immobilization method. The real diagnostic applicability of this biosensing platform was evaluated using brain tissue lysate samples. Diagnostic performance of immunosensor was proven to be more effective than conventional ELISA in terms of lowest detection limit 4.81 pg mL-1, sample volume consumption and assay simplicity [55].
Fig. (5).
Schematic representation of the reaction mechanisms occurring during the preparation of immunosensor and subsequent detection of Aβ1-40. Adapted with permission from Ref [55]. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Another sensitive approach utilizes the genomic DNA samples wherein enzyme-assisted electrochemical detection of apoE4 gene enables signal amplification by using ferrocene (Fc)-capped gold nanoparticles modified with streptavidin. Benefitting from amplified signal, the apoE4 gene in genomic DNAs was detected at less than 0.1 pM level. Biotinylated oligonucleotide
probes capture complementary apoE 4 gene immobilized on the gold electrodes surface. This is followed by hybridization with apoE 4 gene at codon 112 or apoE 2/3 gene with a single base mismatch relative to apoE 4 gene, enzymatic cleavage by restriction enzyme HhaI and then attachment of ferrocene (Fc) – capped gold nanoparticle modified with streptavidin (Fig. 6). Cleavage only occurs at the complementary apoE 4 duplex, therefore this sequence can be discriminated against other apoE sequences [158].
Fig. (6).
Schematic of electrochemical detection of apoE 4 gene. Adapted with permission from Ref. [158]. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Another extremely sensitive electrochemical apatasensor method for the quantitation of amyloid beta
(Aβ) was reported recently. This approach revealed immobilization of a specific RNA aptamer on the gold nanostructure (synthesized by electrodeposition using polyethylene glycol) (Fig. 7). Binding of Aβ peptide to specific RNA aptamer was detected by ferro/ferricyanide redox marker. The applicability of the developed aptasensor was tested in the real samples (human blood serum and artificial cerebrospinal fluid) for the demonstration of its viability [166].
Fig. (7).
Fabrication steps of the aptasensor and Aβ determination: I– electrodeposition of fern leaves-like gold nanostructure (FLGN), II– aptamer immobilization, III– 6-mercaptohexanol (MCH) immobilization, IV– Aβ incubation. Adapted with permission from Ref. [166]. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Another platform designated for peptide-based biosensor employs irregular shaped microporous gold nanostructure electrodeposited on a polycrystalline gold surface using sodium alendronate. Aβ1–42-binding peptide was immobilized on the gold nanostructure by using a specific peptide sequence that creates a strong bond with the surface of a microporous gold nanostructure through the thiol group of its cysteine residue. In selecting this peptide sequence, a high specificity to capture Aβ1–42 was considered. Ferro/ferricyanide redox probe was employed as a redox marker to electrochemically follow the binding of Aβ1–42 by the peptide. The applicability of biosensors for the quantitation of Aβ1–42 was tested in spiked serum samples and artificial cerebrospinal fluid. The presented electrochemical biosensor is free of interferences and able to detect Aβ1–42 with a detection limit of 0.2 pg mL−1 [167]. It is critically important to diagnose AD at the early stages of its progression, which allows successful treatment and recovery of patients. Therefore, it is essential to develop simple and sensitive diagnostic methods that can detect AD biomarkers at very low concentrations in biological fluids. Some of the recently reported biosensors are able to determine AD biomarkers with remarkable sensitivity (the values of detection limit at pM) (Table 2).
Table 2.
Specifications of electrochemical biosensors for the detection of single AD biomarkers.
| AD Biomarker | Biorecognition Molecules | Techniques | Linear Range of Detection | Detection Limit | Refs. |
|---|---|---|---|---|---|
| Aβ oligomers | Protein-binding peptide Cellular prion protein (residues 95-110) Cellular prion protein |
SWV EIS EIS |
0.5 – 20 nM 0.5 – 100 pM 0.1 pM – 10 nM |
0.048 nM 0.5 pM 0.1 pM |
[168] [169] [170] |
| Aβ 1-40 | mAβab | EIS | 1−104 pg/ml | 4.81 pg/ml | [55] |
| Aβ 1-42 | Specific RNA aptamer Aβ1–42-binding peptide Anti-Aβ1-42 |
DPV DPV CV |
0.002–1.28 ng/ml 3–7000 pg /ml 0.5 – 500 ng/ml |
0.4 pg/ml 0.2 pg/ml 0.1 ng/ml |
[166] [167] [171] |
| Total Aβ | Anti mAβ antibody | EIS | 2.65 nM–2.04µM | 0.57 nM | [172] |
| S100B protein | His6-RAGE domain Monoclonal anti-S100B His6-RAGE domain |
SWV DPV SWV |
1 – 20 pM 0.1 – 100 pg/ml 1 – 20 pM |
0.52 pM 0.1 pg/ml 0.9 pM |
[128] [173] [123] |
| Glycated albumin | His6-RAGE domain | SWV | 1– 20 pM | 2.3 pM | [162] |
| APOE4 | HRP-labelled APOE4 antibody | CV | 1 – 10 ng/ml | 0.3 ng/ml | [161] |
| Tau-441 | Oriented antibodies 39E10 Tau protein Anti-tau antibody |
CV, EIS CV, EIS EIS, DPV |
0.01 pM-10nM 0.2 – 1.0 µM 0.5 – 15.1 nM |
0.03 pM 0.2 µM 0.15 nM |
[137] [138] [61] |
5.2. Multiplex Assays
It is imperative to deviate from the detection of single AD biomarker and develop a diagnosis methodology of multiplex biomarker detection to reflect the complexity of AD pathogenesis. Simultaneous detection of two or more markers achieves higher sensitivity than one marker to distinguish Alzheimer's disease patients and others, indicating that each marker may be complementary and not redundant with the other for an accurate diagnosis. Measurement of different analytes in a single sample from individual patients in parallel appears to considerably improve the accuracy of AD diagnosis [174].
Simultaneous detection of a series of clinically relevant protein biomarkers is indispensable for clinical applications. However, the levels of different biomarkers may cover an expansive range; a biosensor for such markers requires not only high sensitivity but also a broad detection range [175]. For instance, the cut-off levels of Aβ1-42, t-tau and p-tau181 are 530 pg/mL (117.4 pM), 350 pg/mL (7.6 pM) and 80 pg/mL (1.7 pM) [176]. Hence, although many biosensors for specific biomarkers have been reported, the development of multiplex detection of AD biomarkers remains strongly limited.
Simultaneous sensing of multiple specific biomarkers can be achieved through either multi- label or multi-electrode approaches. In multi-label systems, a single electrode is used for the detection of various biomarkers. On the other hand, multiple sensing areas are used to detect multiple biomarkers in multi-electrode systems [177]. The designing of a biosensor that is capable of simultaneous determination of two or more analytes in a single measurement is a great challenge. The following examples of electrochemical biosensor for multiple AD biomarker detection will be presented herein.
In the first paper, the novel ratiometric electrochemical biosensor for the dual determination of copper ions (Cu2+) and Aβ1-42 based on a 2,2′-azinobis-(3-ethylbenzothiazoline-6-sulphonate) (ABTS) and poly(diallyldimethylammonium chloride) (PDDA)-bi functionalized single-walled carbon nanotubes (CNTs) composite with the detection limits of 0.04 μM for Cu2+ and 0.5 ng mL-1 for Aβ1-42, respectively, was presented by Yu et al. [178]. Neurokinin B (NKB) immobilized on the ABTS-PDDA/CNTs modified electrode surface was used as specific recognition of Cu2+ element by forming a [CuII(NKB)2] complex with Cu2+. The modified electrode also generated the electrochemical signal toward the Aβ1–42 monomer, when a certain amount of the monomer was added to Cu2+-contained PBS buffer, due to the release of copper ions from the complex through Aβ binding to Cu2+. Consequently, the designed electrochemical approach was capable of monitoring two important biological species: copper ions, which are directly involved in Aβ aggregation and Aβ1-42; by one single biosensor in plasma and hippocampus of normal and AD rats [178].
Recently, the simultaneous determination of four AD biomarkers on one microchip has been presented by Song et al. [179]. Sensitive and selective detection of multiple biomarkers including Tau, ApoE4, Amyloid-β and miRNA-101 on mini-pillar-based biosensor that confines the reagent in open-channel microreactors for simultaneously sensing multiple biomarkers is achieved. Such a mini-pillar sensor mainly consists of a mini-pillar array with an electrode array for anchoring the droplets to electrochemical signal acquisition and circuit integration unit for aggregation of multiple signals (Fig. 8).
Fig. (8).
The open-channel mini-pillar sensor for multiple electrochemical detection. Reprinted with permission from Ref [179]. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
The mechanism of proteinaceous biomarkers, including Tau, ApoE 4 and Amyloid-β sensing, is based on electrode resistance change with the electrochemical signals fluctuations (Fig. 9a). After immobilization of antibody and block protein on gold nanodendrites working electrode, the initial signal is recorded. The antibody-antigen specific recognition causes the increases of surface resistance and signal decrease. The
Fig. (9).
Electrochemical array toward multiple and simultaneous AD biomarkers detection. a) The mechanism of electrochemical detection of proteinaceous biomarkers. b) The mechanism of electrochemical detection of miRNA-101. Reprinted with permission from Ref [179]. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
gold nanodendrites are obtained by electrodeposition onto the working electrode and caused enhanced sensitivity via improving probe-binding capacity and response signal.
The detection of miRNA-101 is based on altering the positioning of the reporter (Fc) relative to the electrode surface (thereby producing a target-dependent change in current) during the combining of miRNA-101 to redox-reporter-modified hairpin DNA probe immobilized on working electrode by Au–S bond. All target biomarkers concentrations are determined by using the signal fluctuations (Fig. 9b) [179].
CONCLUSION AND FUTURE DIRECTIONS
With the increasing aging population, the number of people with AD is set to rise. The need is now greater than ever to develop technologies for the rapid, sensitive, reliable and cost-effective methods for AD biomarkers detection based on new analytical technologies. In this review, we have presented a snapshot of the recent developments in this field and an overview of the pace at which electrochemical biosensors have sought to address this gap. Moving forward, there are definitely challenges that must be addressed, for the field to continue its growth momentum.
The important strategy of development is the capability for integration of analytical technologies on a single platform. The development of electrochemical biosensors that allow for simultaneous detection of multiple biomarkers of an AD can achieve higher detection sensitivity while reducing false positives. This can be achieved by the simultaneous use of biorecognition molecules with different specificities equipped with appropriate redox active labels, so that many AD biomarkers could be detected in parallel. However, there are still problems arising from mutual interference or overlapping of the electrochemical signal. This challenge can be achieved by finding appropriate redox active labels which differ in electrochemical behavior when attached to biorecognition molecules on the same electrode platform.
Another challenge is to improve the sensitivity and specificity of biosensors and make them affordable and accessible at the point of care. In recent years, nanomaterials have been widely used in biosensing as one of the candidates for further sensitivity actions in the development of highly sensitive devices for early diagnosis and point-of-care applications. New advances in nanomaterials, nanofabrication technologies and biomimetic surfaces can all be further explored for developing biosensors that continue to push the boundaries of AD biomarkers detection.
There is a need for transition from development stages towards commercialization of biosensors for point-of-care diagnostics of AD. This is especially important for biosensors where detection limits and the sensitivity achieved are several orders of magnitude better than physiologically and diagnostically relevant levels. By driving the development of fabrication techniques of biosensors, through product development, other practical aspects, including costs and portability, could also be examined.
ACKNOWLEDGEMENTS
The author thanks the Institute of Animal Reproduction and Food Research, as well as Polish Academy of Sciences in Olsztyn, Poland for their financial support.
LIST OF ABBREVIATIONS
- mAb
Monoclonal Antibody
- mAβab
Monoclonal amyloid-beta antibodies
- HRP
Horse Reddish Peroxidase
- AuNPs
Gold Nanoparticles
- SAM
Self Assembled Monolayer
- DPTA
Thiol Derivative of Pentetic Acid
- His6-RAGE
His-tagged domains of Receptor for Advanced Glycation End products
- Aβ
Amyloid beta
- MCH
6-mercaptohexanol
- Fc
Ferrocene
- MS
Mass Spectrometry
- MRI
Magnetic Resonance Imaging
Funding Statement
This work has been supported by the Institute of Animal Reproduction and Food Research and Polish Academy of Sciences in Olsztyn, Poland.
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
This work has been supported by the Institute of Animal Reproduction and Food Research and Polish Academy of Sciences in Olsztyn, Poland.
CONFLICT OF INTEREST
The author declares no conflict of interest, financial or otherwise.
REFERENCES
- 1.Zarowitz B.J., Stefanacci R., Hollenack K., O’Shea T., Gruber J., Tangalos E.G. The application of evidence-based principles of care in older persons (issue 5): Alzheimer’s disease. J. Am. Med. Dir. Assoc. 2007;8(3):183–193. doi: 10.1016/j.jamda.2006.08.008. [DOI] [PubMed] [Google Scholar]
- 2.Leszek J., Małyszczak K., Janicka B., Kiejna A., Wiak A. Total tau in cerebrospinal fluid differentiates Alzheimer’s disease from vascular dementia. Med. Sci. Monit. 2003;9(11):CR484–CR488. [PubMed] [Google Scholar]
- 3.Lu H., Zhu X-C., Jiang T., Yu J-T., Tan L. Body fluid biomarkers in Alzheimer’s disease. Ann. Transl. Med. 2015;3(5):70. doi: 10.3978/j.issn.2305-5839.2015.02.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Andreasen N., Blennow K. CSF biomarkers for mild cognitive impairment and early Alzheimer’s disease. Clin. Neurol. Neurosurg. 2005;107(3):165–173. doi: 10.1016/j.clineuro.2004.10.011. [DOI] [PubMed] [Google Scholar]
- 5.Dmitrzak-Węglarz M., Hauser J. Proteomic analysis in quest for biologic markers of psychiatric diseases. Psychiatria. 2020;3:118–127. Available at: https://journals.viamedica.pl/psychiatria/article/download/29193/23958 (Accessed date: 5 June 2020). [Google Scholar]
- 6.Schilling M.A. Unraveling Alzheimer’s: making sense of the relationship between diabetes and Alzheimer’s disease1. J. Alzheimers Dis. 2016;51(4):961–977. doi: 10.3233/JAD-150980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Srikanth V., Maczurek A., Phan T., Steele M., Westcott B., Juskiw D., Münch G. Advanced glycation endproducts and their receptor RAGE in Alzheimer’s disease. Neurobiol. Aging. 2011;32(5):763–777. doi: 10.1016/j.neurobiolaging.2009.04.016. [DOI] [PubMed] [Google Scholar]
- 8.Davtyan H., Zagorski K., Rajapaksha H., Hovakimyan A., Davtyan A., Petrushina I., Kazarian K., Cribbs D.H., Petrovsky N., Agadjanyan M.G., Ghochikyan A. Alzheimer’s disease Advax(CpG)- adjuvanted MultiTEP-based dual and single vaccines induce high-titer antibodies against various forms of tau and Aβ pathological molecules. Sci. Rep. 2016;6:28912. doi: 10.1038/srep28912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Liu L., Zhao F., Ma F., Zhang L., Yang S., Xia N. Electrochemical detection of β-amyloid peptides on electrode covered with N-terminus-specific antibody based on electrocatalytic O2 reduction by Aβ(1-16)-heme-modified gold nanoparticles. Biosens. Bioelectron. 2013;49:231–235. doi: 10.1016/j.bios.2013.05.028. [DOI] [PubMed] [Google Scholar]
- 10.Strittmatter W.J., Roses A.D. Apolipoprotein E and Alzheimer’s disease. Annu. Rev. Neurosci. 1996;19(1):53–77. doi: 10.1146/annurev.ne.19.030196.000413. [DOI] [PubMed] [Google Scholar]
- 11.Kalaria R.N., Maestre G.E., Arizaga R., Friedland R.P., Galasko D., Hall K., Luchsinger J.A., Ogunniyi A., Perry E.K., Potocnik F., Prince M., Stewart R., Wimo A., Zhang Z.X., Antuono P. World Federation of Neurology Dementia Research Group. Alzheimer’s disease and vascular dementia in developing countries: prevalence, management, and risk factors. Lancet Neurol. 2008;7(9):812–826. doi: 10.1016/S1474-4422(08)70169-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gandy S. Perspective: prevention is better than cure. Nature. 2011;475(7355):S15. doi: 10.1038/475S15a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dickson D.W. The pathogenesis of senile plaques. J. Neuropathol. Exp. Neurol. 1997;56(4):321–339. doi: 10.1097/00005072-199704000-00001. [DOI] [PubMed] [Google Scholar]
- 14.Leshchyns’ka I., Liew H.T., Shepherd C., Halliday G.M., Stevens C.H., Ke Y.D., Ittner L.M., Sytnyk V. Aβ-dependent reduction of NCAM2-mediated synaptic adhesion contributes to synapse loss in Alzheimer’s disease. Nat. Commun. 2015;6:8836. doi: 10.1038/ncomms9836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Xue Y., Zhang Z., Wen C., Liu H., Wang S., Li J., Zhuge Q., Chen W., Ye Q. Characterization of Alzheimer’s disease using ultra-high b-values apparent diffusion coefficient and diffusion kurtosis imaging. Aging Dis. 2019;10(5):1026–1036. doi: 10.14336/AD.2018.1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Blennow K. Cerebrospinal fluid protein biomarkers for Alzheimer’s disease. NeuroRx. 2004;1(2):213–225. doi: 10.1602/neurorx.1.2.213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Fagan A.M., Roe C.M., Xiong C., Mintun M.A., Morris J.C., Holtzman D.M. Cerebrospinal fluid tau/β-amyloid(42) ratio as a prediction of cognitive decline in nondemented older adults. Arch. Neurol. 2007;64(3):343–349. doi: 10.1001/archneur.64.3.noc60123. [DOI] [PubMed] [Google Scholar]
- 18.Coupé P., Manjón J.V., Lanuza E., Catheline G. Lifespan changes of the human brain in Alzheimer’s disease. Sci. Rep. 2019;9(1):3998. doi: 10.1038/s41598-019-39809-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Wang R., Sweeney D., Gandy S.E., Sisodia S.S. The profile of soluble amyloid beta protein in cultured cell media. Detection and quantification of amyloid beta protein and variants by immunoprecipitation-mass spectrometry. J. Biol. Chem. 1996;271(50):31894–31902. doi: 10.1074/jbc.271.50.31894. [DOI] [PubMed] [Google Scholar]
- 20.McAvoy T., Lassman M.E., Spellman D.S., Ke Z., Howell B.J., Wong O., Zhu L., Tanen M., Struyk A., Laterza O.F. Quantification of tau in cerebrospinal fluid by immunoaffinity enrichment and tandem mass spectrometry. Clin. Chem. 2014;60(4):683–689. doi: 10.1373/clinchem.2013.216515. [DOI] [PubMed] [Google Scholar]
- 21.Yang T., Hong S., O’Malley T., Sperling R.A., Walsh D.M., Selkoe D.J. New ELISAs with high specificity for soluble oligomers of amyloid β-protein detect natural Aβ oligomers in human brain but not CSF. Alzheimers Dement. 2013;9(2):99–112. doi: 10.1016/j.jalz.2012.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wang L.S., Leung Y.Y., Chang S.K., Leight S., Knapik-Czajka M., Baek Y., Shawa L.M., Lee V.M.Y., Trojanowski J.Q., Clark C.M. Comparison of xMAP and ELISA assays for detecting CSF biomarkers of AD. J Alzheimers. 2012;31:439–445. doi: 10.3233/JAD-2012-120082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Veerabhadrappa B., Delaby C., Hirtz C., Vialaret J., Alcolea D., Lleó A., Fortea J., Santosh M.S., Choubey S., Lehmann S. Detection of amyloid beta peptides in body fluids for the diagnosis of alzheimer’s disease: Where do we stand? Crit. Rev. Clin. Lab. Sci. 2020;57(2):99–113. doi: 10.1080/10408363.2019.1678011. [DOI] [PubMed] [Google Scholar]
- 24.Kang M.K., Lee J., Nguyen A.H., Sim S.J. Label-free detection of ApoE4-mediated β-amyloid aggregation on single nanoparticle uncovering Alzheimer’s disease. Biosens. Bioelectron. 2015;72:197–204. doi: 10.1016/j.bios.2015.05.017. [DOI] [PubMed] [Google Scholar]
- 25.Shankar G.M., Leissring M.A., Adame A., Sun X., Spooner E., Masliah E., Selkoe D.J., Lemere C.A., Walsh D.M. Biochemical and immunohistochemical analysis of an Alzheimer’s disease mouse model reveals the presence of multiple cerebral Abeta assembly forms throughout life. Neurobiol. Dis. 2009;36(2):293–302. doi: 10.1016/j.nbd.2009.07.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.den Haan J., Morrema T.H.J., Verbraak F.D., de Boer J.F., Scheltens P., Rozemuller A.J., Bergen A.A.B., Bouwman F.H., Hoozemans J.J. Amyloid-beta and phosphorylated tau in post-mortem Alzheimer’s disease retinas. Acta Neuropathol. Commun. 2018;6(1):147. doi: 10.1186/s40478-018-0650-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhang H., Zhu X., Pascual G., Wadia J.S., Keogh E., Hoozemans J.J., Siregar B., Inganäs H., Stoop E.J.M., Goudsmit J., Apetri A., Koudstaal W., Wilson I.A. Structural basis for recognition of a unique epitope by a human anti-tau antibody. Structure. 2018;26(12):1626–1634.e4. doi: 10.1016/j.str.2018.08.012. [DOI] [PubMed] [Google Scholar]
- 28.Sengupta U., Portelius E., Hansson O., Farmer K., Castillo-Carranza D., Woltjer R., Zetterberg H., Galasko D., Blennow K., Kayed R. Tau oligomers in cerebrospinal fluid in Alzheimer’s disease. Ann. Clin. Transl. Neurol. 2017;4(4):226–235. doi: 10.1002/acn3.382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Shi L., Baird A.L., Westwood S., Hye A., Dobson R., Thambisetty M., Lovestone S. A decade of blood biomarkers for Alzheimer’s disease research: an evolving field, improving study designs, and the challenge of replication. J. Alzheimers Dis. 2018;62(3):1181–1198. doi: 10.3233/JAD-170531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Houck A.L., Hernández F., Ávila J. A simple model to study tau pathology. J. Exp. Neurosci. 2016;10:31–38. doi: 10.4137/JEN.S25100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Roohk H.V., Zaidi A.R. A review of glycated albumin as an intermediate glycation index for controlling diabetes. J. Diabetes Sci. Technol. 2008;2(6):1114–1121. doi: 10.1177/193229680800200620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Carrico Z.M., Le G., Malinow R. A fluorescence assay for detecting amyloid-β using the cytomegalovirus enhancer/promoter. J. Biol. Methods. 2017;4(3):77. doi: 10.14440/jbm.2017.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lv G., Shen Y., Zheng W., Yang J., Li C., Lin J. Fluorescence detection and dissociation of amyloid-β species for the treatment of Alzheimer’s disease. Adv. Ther. 2019;2(9):1900054. doi: 10.1002/adtp.201900054. [DOI] [Google Scholar]
- 34.Law W.P., Wang W.Y.S., Moore P.T., Mollee P.N., Ng A.C.T. Cardiac amyloid imaging with 18F-Florbetaben PET: a pilot study. J. Nucl. Med. 2016;57(11):1733–1739. doi: 10.2967/jnumed.115.169870. [DOI] [PubMed] [Google Scholar]
- 35.McGuire J.N., Overgaard J., Pociot F. Mass spectrometry is only one piece of the puzzle in clinical proteomics. Brief. Funct. Genomics Proteomics. 2008;7(1):74–83. doi: 10.1093/bfgp/eln005. [DOI] [PubMed] [Google Scholar]
- 36.Gremel G., Grannas K., Sutton L.A., Pontén F., Zieba A. In situ protein detection for companion diagnostics. Front. Oncol. 2013;3:271. doi: 10.3389/fonc.2013.00271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Dunstan R.W., Wharton K.A., Jr, Quigley C., Lowe A. The use of immunohistochemistry for biomarker assessment-can it compete with other technologies? Toxicol. Pathol. 2011;39(6):988–1002. doi: 10.1177/0192623311419163. [DOI] [PubMed] [Google Scholar]
- 38.Boellner S., Becker K.F. Reverse phase protein arrays-quantitative assessment of multiple biomarkers in biopsies for clinical use. Microarrays (Basel) 2015;4(2):98–114. doi: 10.3390/microarrays4020098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Nimse S.B., Sonawane M.D., Song K.S., Kim T. Biomarker detection technologies and future directions. Analyst (Lond.) 2016;141(3):740–755. doi: 10.1039/C5AN01790D. [DOI] [PubMed] [Google Scholar]
- 40.Fliser D., Novak J., Thongboonkerd V., Argilés A., Jankowski V., Girolami M.A., Jankowski J., Mischak H. Advances in urinary proteome analysis and biomarker discovery. J. Am. Soc. Nephrol. 2007;18(4):1057–1071. doi: 10.1681/ASN.2006090956. [DOI] [PubMed] [Google Scholar]
- 41.Villiers L., Loots D. Using metabolomics for elucidating the mechanisms related to tuberculosis treatment failure. Curr. Metabolomics. 2014;1(4):306–317. doi: 10.2174/2213235X113016660006. [DOI] [Google Scholar]
- 42.Gale P.J., Vestal M.L. The development of time-of-flight mass spectrometry. In: Gross M.L., Caprioli R.M., editors. The Encyclopedia of Mass Spectrometry. Vol. 9. Amsterdam: Elsevier Science B.V; 2016. pp. 34–42. [DOI] [Google Scholar]
- 43.Rockwood A.L., Kushnir M.M., Clarke N.J. Mass spectrometry. In: Rifai N., Horvath A.R., Wittwer C.T., Hoofnagle A., editors. Principles and Applications of Clinical Mass Spectrometry. Amsterdam: Elsevier Science B.V; 2018. pp. 33–65. [DOI] [Google Scholar]
- 44.Lecchi M., Fossati P., Elisei F., Orecchia R., Lucignani G. Current concepts on imaging in radiotherapy. Eur. J. Nucl. Med. Mol. Imaging. 2008;35(4):821–837. doi: 10.1007/s00259-007-0631-y. [DOI] [PubMed] [Google Scholar]
- 45.Magni-Manzoni S., Malattia C., Lanni S., Ravelli A. Advances and challenges in imaging in juvenile idiopathic arthritis. Nat. Rev. Rheumatol. 2012;8(6):329–336. doi: 10.1038/nrrheum.2012.30. [DOI] [PubMed] [Google Scholar]
- 46.Dibble E.H., Karantanis D., Mercier G., Peller P.J., Kachnic L.A., Subramaniam R.M. PET/CT of cancer patients: part 1, pancreatic neoplasms. AJR Am. J. Roentgenol. 2012;199(5):952–967. doi: 10.2214/AJR.11.8182. [DOI] [PubMed] [Google Scholar]
- 47.Villafañez F., Gottifredi V., Soria G. Development and optimization of a miniaturized western blot-based screening platform to identify regulators of post-translational modifications. High Throughput. 2019;8(2):15. doi: 10.3390/ht8020015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Vrzalová J. Multiplex xMAP immunoanalysis and examples of its application. PhD Thesis, Charles University in Prague Faculty of Medicine in Pilsen. 2020 Available at: https://dspace.cuni.cz/bitstream/handle/20.500.11956/ 22085/ RPTX_2008_ 2_11160_ GR999101_ 277740_0_74168.pdf?sequence=1&isAllowed=y (Accessed date: 10 June 2020).
- 49.Villena Gonzales W., Mobashsher A.T., Abbosh A. The progress of glucose monitoring-a review of invasive to minimally and non-invasive techniques, devices and sensors. Sensors (Basel) 2019;19(4):800. doi: 10.3390/s19040800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Cottet M., Faklaris O., Zwier J.M., Trinquet E., Pin J.P., Durroux T. Original fluorescent ligand-based assays open new perspectives in G-protein coupled receptor drug screening. Pharmaceuticals. 2011;4(1):202–214. doi: 10.3390/ph4010202. [DOI] [Google Scholar]
- 51.Qureshi A., Gurbuz Y., Niazi J.H. Biosensors for cardiac biomarkers detection: a review. Sens. Actuators B Chem. 2012;171:62–76. doi: 10.1016/j.snb.2012.05.077. [DOI] [Google Scholar]
- 52.Park M., Tsai S.L., Chen W. Microbial biosensors: engineered microorganisms as the sensing machinery. Sensors (Basel) 2013;13(5):5777–5795. doi: 10.3390/s130505777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhou Y., Chiu C.W., Liang H. Interfacial structures and properties of organic materials for biosensors: an overview. Sensors (Basel) 2012;12(11):15036–15062. doi: 10.3390/s121115036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Putzbach W., Ronkainen N.J. Immobilization techniques in the fabrication of nanomaterial-based electrochemical biosensors: a review. Sensors (Basel) 2013;13(4):4811–4840. doi: 10.3390/s130404811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zakaria N., Ramli M.Z., Ramasamy K., Meng L.S., Yean C.Y., Banga Singh K.K., Zain Z.M., Low K.F. An impedimetric micro-immunosensing assay to detect Alzheimer’s disease biomarker: Aβ40. Anal. Biochem. 2018;555:12–21. doi: 10.1016/j.ab.2018.05.031. [DOI] [PubMed] [Google Scholar]
- 56.Sonuç Karaboğa M.N., Sezgintürk M.K. A novel silanization agent based single used biosensing system: Detection of C-reactive protein as a potential Alzheimer’s disease blood biomarker. J. Pharm. Biomed. Anal. 2018;154:227–235. doi: 10.1016/j.jpba.2018.03.016. [DOI] [PubMed] [Google Scholar]
- 57.Wang X., Mei Z., Wang Y., Tang L. Comparison of four methods for the biofunctionalization of gold nanorods by the introduction of sulfhydryl groups to antibodies. Beilstein J. Nanotechnol. 2017;8:372–380. doi: 10.3762/bjnano.8.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Topkaya S.N., Azimzadeh M., Ozsoz M. Electrochemical biosensors for cancer biomarkers detection: recent advances and challenges. Electroanalysis. 2016;28(7):1402–1419. doi: 10.1002/elan.201501174. [DOI] [Google Scholar]
- 59.de la Escosura-Muñiz A., Plichta Z., Horák D., Merkoçi A. Alzheimer’s disease biomarkers detection in human samples by efficient capturing through porous magnetic microspheres and labelling with electrocatalytic gold nanoparticles. Biosens. Bioelectron. 2015;67:162–169. doi: 10.1016/j.bios.2014.07.086. [DOI] [PubMed] [Google Scholar]
- 60.Zhu G., Lee H.J. Electrochemical sandwich-type biosensors for α-1 antitrypsin with carbon nanotubes and alkaline phosphatase labeled antibody-silver nanoparticles. Biosens. Bioelectron. 2017;89(Pt 2):959–963. doi: 10.1016/j.bios.2016.09.080. [DOI] [PubMed] [Google Scholar]
- 61.Derkus B., Acar Bozkurt P., Tulu M., Emregul K.C., Yucesan C., Emregul E. Simultaneous quantification of Myelin Basic Protein and Tau proteins in cerebrospinal fluid and serum of Multiple Sclerosis patients using nanoimmunosensor. Biosens. Bioelectron. 2017;89(Pt 2):781–788. doi: 10.1016/j.bios.2016.10.019. [DOI] [PubMed] [Google Scholar]
- 62.Zhou Y., Li C., Li X., Zhu X., Ye B., Xu M. A sensitive aptasensor for the detection of β-amyloid oligomers based on metal-organic frameworks as electrochemical signal probes. Anal. Methods. 2018;10(36):4430–4437. doi: 10.1039/C8AY00736E. [DOI] [Google Scholar]
- 63.Fuellen G., Jansen L., Cohen A.A., Luyten W., Gogol M., Simm A., Saul N., Cirulli F., Berry A., Antal P., Köhling R., Wouters B., Möller S. Health and aging: unifying concepts, scores, biomarkers and pathways. Aging Dis. 2019;10(4):883–900. doi: 10.14336/AD.2018.1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mayeux R. Biomarkers: potential uses and limitations. NeuroRx. 2004;1(2):182–188. doi: 10.1602/neurorx.1.2.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Henry M.S., Passmore A.P., Todd S., McGuinness B., Craig D., Johnston J.A. The development of effective biomarkers for Alzheimer’s disease: a review. Int. J. Geriatr. Psychiatry. 2013;28(4):331–340. doi: 10.1002/gps.3829. [DOI] [PubMed] [Google Scholar]
- 66.Zürbig P., Jahn H. Use of proteomic methods in the analysis of human body fluids in Alzheimer research. Electrophoresis. 2012;33(24):3617–3630. doi: 10.1002/elps.201200360. [DOI] [PubMed] [Google Scholar]
- 67.Sharma N., Singh A.N. Exploring biomarkers for Alzheimer’s disease. J. Clin. Diagn. Res. 2016;10(7):KE01–KE06. doi: 10.7860/JCDR/2016/18828.8166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Chaves M.L., Camozzato A.L., Ferreira E.D., Piazenski I., Kochhann R., Dall’Igna O., Mazzini G.S., Souza D.O., Portela L.V. Serum levels of S100B and NSE proteins in Alzheimer’s disease patients. J. Neuroinflammation. 2010;7:6. doi: 10.1186/1742-2094-7-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Mrak R.E., Griffinbc W.S. The role of activated astrocytes and of the neurotrophic cytokine S100B in the pathogenesis of Alzheimer’s disease. Neurobiol. Aging. 2001;22(6):915–922. doi: 10.1016/S0197-4580(01)00293-7. [DOI] [PubMed] [Google Scholar]
- 70.Peskind E.R., Griffin W.S.T., Akama K.T., Raskind M.A., Van Eldik L.J. Cerebrospinal fluid S100B is elevated in the earlier stages of Alzheimer’s disease. Neurochem. Int. 2001;39(5-6):409–413. doi: 10.1016/S0197-0186(01)00048-1. [DOI] [PubMed] [Google Scholar]
- 71.Petzold A., Jenkins R., Watt H.C., Green A.J.E., Thompson E.J., Keir G., Fox N.C., Rossor M.N. Cerebrospinal fluid S100B correlates with brain atrophy in Alzheimer’s disease. Neurosci. Lett. 2003;336(3):167–170. doi: 10.1016/S0304-3940(02)01257-0. [DOI] [PubMed] [Google Scholar]
- 72.Shuvaev V.V., Laffont I., Serot J.M., Fujii J., Taniguchi N., Siest G. Increased protein glycation in cerebrospinal fluid of Alzheimer’s disease. Neurobiol. Aging. 2001;22(3):397–402. doi: 10.1016/S0197-4580(00)00253-0. [DOI] [PubMed] [Google Scholar]
- 73.Nunan J., Small D.H. Regulation of APP cleavage by alpha-, beta- and gamma-secretases. FEBS Lett. 2000;483(1):6–10. doi: 10.1016/S0014-5793(00)02076-7. [DOI] [PubMed] [Google Scholar]
- 74.Zetterberg H., Blennow K., Hanse E. Amyloid β and APP as biomarkers for Alzheimer’s disease. Exp. Gerontol. 2010;45(1):23–29. doi: 10.1016/j.exger.2009.08.002. [DOI] [PubMed] [Google Scholar]
- 75.Roher A.E., Chaney M.O., Kuo Y.M., Webster S.D., Stine W.B., Haverkamp L.J., Woods A.S., Cotter R.J., Tuohy J.M., Krafft G.A., Bonnell B.S., Emmerling M.R. Morphology and toxicity of Abeta-(1-42) dimer derived from neuritic and vascular amyloid deposits of Alzheimer’s disease. J. Biol. Chem. 1996;271(34):20631–20635. doi: 10.1074/jbc.271.34.20631. [DOI] [PubMed] [Google Scholar]
- 76.Kim H.J., Chae S.C., Lee D.K., Chromy B., Lee S.C., Park Y.C., Klein W.L., Krafft G.A., Hong S.T. Selective neuronal degeneration induced by soluble oligomeric amyloid beta protein. FASEB J. 2003;17(1):118–120. doi: 10.1096/fj.01-0987fje. [DOI] [PubMed] [Google Scholar]
- 77.Hartley D.M., Walsh D.M., Ye C.P., Diehl T., Vasquez S., Vassilev P.M., Teplow D.B., Selkoe D.J. Protofibrillar intermediates of amyloid beta-protein induce acute electrophysiological changes and progressive neurotoxicity in cortical neurons. J. Neurosci. 1999;19(20):8876–8884. doi: 10.1523/JNEUROSCI.19-20-08876.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Mattson M.P. Pathways towards and away from Alzheimer’s disease. Nature. 2004;430(7000):631–639. doi: 10.1038/nature02621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Luca A., Calandra C., Luca M. Molecular bases of Alzheimer’s disease and neurodegeneration: the role of neuroglia. Aging Dis. 2018;9(6):1134–1152. doi: 10.14336/AD.2018.0201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Terry R.D. Neuropathological changes in Alzheimer disease. Prog. Brain Res. 1994;101:383–390. doi: 10.1016/S0079-6123(08)61964-0. [DOI] [PubMed] [Google Scholar]
- 81.Mayeux R., Honig L.S., Tang M.X., Manly J., Stern Y., Schupf N., Mehta P.D. Plasma A[β]40 and A[β]42 and Alzheimer’s disease: relation to age, mortality, and risk. Neurology. 2003;61(9):1185–1190. doi: 10.1212/01.WNL.0000091890.32140.8F. [DOI] [PubMed] [Google Scholar]
- 82.Stanyon H.F., Viles J.H. Human serum albumin can regulate amyloid-β peptide fiber growth in the brain interstitium: implications for Alzheimer disease. J. Biol. Chem. 2012;287(33):28163–28168. doi: 10.1074/jbc.C112.360800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Graff-Radford N.R., Crook J.E., Lucas J., Boeve B.F., Knopman D.S., Ivnik R.J., Smith G.E., Younkin L.H., Petersen R.C., Younkin S.G. Association of low plasma Abeta42/Abeta40 ratios with increased imminent risk for mild cognitive impairment and Alzheimer disease. Arch. Neurol. 2007;64(3):354–362. doi: 10.1001/archneur.64.3.354. [DOI] [PubMed] [Google Scholar]
- 84.Šimić G., Babić Leko M., Wray S., Harrington C., Delalle I., Jovanov-Milošević N., Bažadona D., Buée L., de Silva R., Di Giovanni G., Wischik C., Hof P.R. Tau protein hyperphosphorylation and aggregation in Alzheimer’s disease and other tauopathies, and possible neuroprotective strategies. Biomolecules. 2016;6(1):6. doi: 10.3390/biom6010006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Pérez-Ruiz E., Decrop D., Ven K., Tripodi L., Leirs K., Rosseels J., van de Wouwer M., Geukens N., De Vos A., Vanmechelen E., Winderickx J., Lammertyn J., Spasic D. Digital ELISA for the quantification of attomolar concentrations of Alzheimer’s disease biomarker protein Tau in biological samples. Anal. Chim. Acta. 2018;1015:74–81. doi: 10.1016/j.aca.2018.02.011. [DOI] [PubMed] [Google Scholar]
- 86.Safieh M., Korczyn A.D., Michaelson D.M. ApoE4: an emerging therapeutic target for Alzheimer’s disease. BMC Med. 2019;17(1):64. doi: 10.1186/s12916-019-1299-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liscic R.M., Yang Y.H. Biomarkers in Alzheimer’s disease. Transl. Neurosci. Clin. 2016;2(1):1–2. doi: 10.18679/CN11-6030_R.2016.002. [DOI] [Google Scholar]
- 88.Reitz C., Mayeux R. Alzheimer disease: epidemiology, diagnostic criteria, risk factors and biomarkers. Biochem. Pharmacol. 2014;88(4):640–651. doi: 10.1016/j.bcp.2013.12.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ramzy T., Wafai H., Harvy M., Morsy S., Ashour M., Morsy F. Serum levels of S100B and oxidative stress parameters in Alzheimer’s disease (AD) in experimental animals. J. Appl. Sci. Res. 2020;7:1411–1418. Available at: http://www.aensiweb.com/old/jasr/jasr/2011/1411-1418.pdf (Accessed date: 5 June 2020). [Google Scholar]
- 90.Ostendorp T. Structure and function of the metal- binding protein s100b and its interaction with the receptor for advanced glycation end products. PhD Thesis, University of Konstanz: Germany. 2020 Available at: http://kops.uni-konstanz.de/bitstream/handle/123456789/8123/Ostendorp_Diss.pdf?sequence=1&isAllowed=y (Accessed date: 5 June 2020).
- 91.Donato R. S100: a multigenic family of calcium-modulated proteins of the EF-hand type with intracellular and extracellular functional roles. Int. J. Biochem. Cell Biol. 2001;33(7):637–668. doi: 10.1016/S1357-2725(01)00046-2. [DOI] [PubMed] [Google Scholar]
- 92.Huttunen H.J., Kuja-Panula J., Sorci G., Agneletti A.L., Donato R., Rauvala H. Coregulation of neurite outgrowth and cell survival by amphoterin and S100 proteins through receptor for advanced glycation end products (RAGE) activation. J. Biol. Chem. 2000;275(51):40096–40105. doi: 10.1074/jbc.M006993200. [DOI] [PubMed] [Google Scholar]
- 93.Leclerc E., Fritz G., Weibel M., Heizmann C.W., Galichet A. S100B and S100A6 differentially modulate cell survival by interacting with distinct RAGE (receptor for advanced glycation end products) immunoglobulin domains. J. Biol. Chem. 2007;282(43):31317–31331. doi: 10.1074/jbc.M703951200. [DOI] [PubMed] [Google Scholar]
- 94.Bianchi R., Kastrisianaki E., Giambanco I., Donato R. S100B protein stimulates microglia migration via RAGE-dependent up-regulation of chemokine expression and release. . J. Biol. Chem. 2011;286(9):7214–7226. doi: 10.1074/jbc.M110.169342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Heizmann C.W., Ackermann G.E., Galichet A. Pathologies involving the S100 proteins and RAGE. Subcell. Biochem. 2007;45:93–138. doi: 10.1007/978-1-4020-6191-2_5. [DOI] [PubMed] [Google Scholar]
- 96.Rothermundt M., Peters M., Prehn J.H.M., Arolt V. S100B in brain damage and neurodegeneration. Microsc. Res. Tech. 2003;60(6):614–632. doi: 10.1002/jemt.10303. [DOI] [PubMed] [Google Scholar]
- 97.Griffin W.S., Yeralan O., Sheng J.G., Boop F.A., Mrak R.E., Rovnaghi C.R., Burnett B.A., Feoktistova A., Van Eldik L.J. Overexpression of the neurotrophic cytokine S100 beta in human temporal lobe epilepsy. J. Neurochem. 1995;65(1):228–233. doi: 10.1046/j.1471-4159.1995.65010228.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Yan S.S., Wu Z.Y., Zhang H.P., Furtado G., Chen X., Yan S.F., Schmidt A.M., Brown C., Stern A., LaFaille J., Chess L., Stern D.M., Jiang H. Suppression of experimental autoimmune encephalomyelitis by selective blockade of encephalitogenic T-cell infiltration of the central nervous system. Nat. Med. 2003;9(3):287–293. doi: 10.1038/nm831. [DOI] [PubMed] [Google Scholar]
- 99.Prajapati K.D., Sharma S.S., Roy N. Current perspectives on potential role of albumin in neuroprotection. Rev. Neurosci. 2011;22(3):355–363. doi: 10.1515/rns.2011.028. [DOI] [PubMed] [Google Scholar]
- 100.Bouma B., Kroon-Batenburg L.M.J., Wu Y.P., Brünjes B., Posthuma G., Kranenburg O., de Groot P.G., Voest E.E., Gebbink M.F.B. Glycation induces formation of amyloid cross-β structure in albumin. J. Biol. Chem. 2003;278(43):41810–41819. doi: 10.1074/jbc.M303925200. [DOI] [PubMed] [Google Scholar]
- 101.Ramos-Fernández E., Tajes M., Palomer E., Ill-Raga G., Bosch-Morató M., Guivernau B., Román-Dégano I., Eraso-Pichot A., Alcolea D., Fortea J., Nuñez L., Paez A., Alameda F., Fernández-Busquets X., Lleó A., Elosúa R., Boada M., Valverde M.A., Muñoz F.J. Posttranslational nitro-glycative modifications of albumin in Alzheimer’s disease: implications in cytotoxicity and amyloid-β peptide aggregation. J. Alzheimers Dis. 2014;40(3):643–657. doi: 10.3233/JAD-130914. [DOI] [PubMed] [Google Scholar]
- 102.Rubel A.A., Ryzhova T.A., Antonets K.S., Chernoff Y.O., Galkin A. Identification of PrP sequences essential for the interaction between the PrP polymers and Aβ peptide in a yeast-based assay. Prion. 2013;7(6):469–476. doi: 10.4161/pri.26867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Younan N.D., Sarell C.J., Davies P., Brown D.R., Viles J.H. The cellular prion protein traps Alzheimer’s Aβ in an oligomeric form and disassembles amyloid fibers. FASEB J. 2013;27(5):1847–1858. doi: 10.1096/fj.12-222588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Rózga M., Kłoniecki M., Jabłonowska A., Dadlez M., Bal W. The binding constant for amyloid Abeta40 peptide interaction with human serum albumin. Biochem. Biophys. Res. Commun. 2007;364(3):714–718. doi: 10.1016/j.bbrc.2007.10.080. [DOI] [PubMed] [Google Scholar]
- 105.Kikuchi S., Shinpo K., Takeuchi M., Yamagishi S., Makita Z., Sasaki N., Tashiro K. Glycation-a sweet tempter for neuronal death. Brain Res. Brain Res. Rev. 2003;41(2-3):306–323. doi: 10.1016/S0165-0173(02)00273-4. [DOI] [PubMed] [Google Scholar]
- 106.Costa M., Horrillo R., Ortiz A.M., Pérez A., Mestre A., Ruiz A., Boada M., Grancha S. Increased Albumin Oxidation in Cerebrospinal Fluid and Plasma from Alzheimer’s Disease Patients. J. Alzheimers Dis. 2018;63(4):1395–1404. doi: 10.3233/JAD-180243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Verghese P.B., Castellano J.M., Holtzman D.M. Apolipoprotein E in Alzheimer’s disease and other neurological disorders. Lancet Neurol. 2011;10(3):241–252. doi: 10.1016/S1474-4422(10)70325-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Liu C.C., Liu C.C., Kanekiyo T., Xu H., Bu G. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat. Rev. Neurol. 2013;9(2):106–118. doi: 10.1038/nrneurol.2012.263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Huynh T.V., Davis A.A., Ulrich J.D., Holtzman D.M. Apolipoprotein E and Alzheimer’s disease: the influence of apolipoprotein E on amyloid-β and other amyloidogenic proteins. J. Lipid Res. 2017;58(5):824–836. doi: 10.1194/jlr.R075481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Shui B., Tao D., Florea A., Cheng J., Zhao Q., Gu Y., Li W., Jaffrezic-Renault N., Mei Y., Guo Z. Biosensors for Alzheimer’s disease biomarker detection: A review. Biochimie. 2018;147:13–24. doi: 10.1016/j.biochi.2017.12.015. [DOI] [PubMed] [Google Scholar]
- 111.Janata J., Josowicz M., DeVaney D.M. Chemical sensors. Anal. Chem. 1994;66(12):207R–228R. doi: 10.1021/ac00084a010. [DOI] [PubMed] [Google Scholar]
- 112.Mehrotra P. Biosensors and their applications - A review. J. Oral Biol. Craniofac. Res. 2016;6(2):153–159. doi: 10.1016/j.jobcr.2015.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Radecki J., Radecka H., Cieśla J., Tudek B. Chemical sensors and biosensors in genetically modified food control. BioTechnologia. 2020;3:67–78. Available at: https://rcin.org.pl/Content/87403/POZN271_113871_biotechnolofia-2006-no3-radecki.pdf (Accessed date: 2 June 2020). [Google Scholar]
- 114.Puzan B., Sankiewicz A., Gorodkiewicz E. Biosensors SPRI as a diagnostic tool in the future. Chemik. 2020;68:528–535. Available at: https://www.researchgate.net/publication/286060331_Biosensors_SPRI_as_a_diagnostic_tool_in_the_future (Accessed date: 2 June 2020). [Google Scholar]
- 115.Chambers J.P., Arulanandam B.P., Matta L.L., Weis A., Valdes J.J. Biosensor recognition elements. Curr. Issues Mol. Biol. 2008;10(1-2):1–12. [PubMed] [Google Scholar]
- 116.Subrahmanyam S., Piletsky S.A., Turner A.P.F. Application of natural receptors in sensors and assays. Anal. Chem. 2002;74(16):3942–3951. doi: 10.1021/ac025673+. [DOI] [PubMed] [Google Scholar]
- 117.Clark L.C., Jr, Lyons C. Electrode systems for continuous monitoring in cardiovascular surgery. Ann. N. Y. Acad. Sci. 1962;102:29–45. doi: 10.1111/j.1749-6632.1962.tb13623.x. [DOI] [PubMed] [Google Scholar]
- 118.Mohanty S.P., Kougianos E. Biosensors: a tutorial review. IEEE Potentials. 2006;25(2):35–40. doi: 10.1109/MP.2006.1649009. [DOI] [Google Scholar]
- 119.Kissinger P.T. Biosensors-a perspective. Biosens. Bioelectron. 2005;20(12):2512–2516. doi: 10.1016/j.bios.2004.10.004. [DOI] [PubMed] [Google Scholar]
- 120.Schroeder G. Selected aspects of supramolecular chemistry; BETAGRAF P.U.H.: Poznań. 2020 Available at: https://chemia.ug.edu.pl/sites/default/files/_nodes/strona- chemia/16862/files/betagraf_2009.pdf (Accessed date: 2 June 2020).
- 121.Schroeder G. Supramolecular materials; BETAGRAF P.U.H.: Poznań. 2020 Available at: http://supra. home. amu. edu.pl/ files/monographs/materialy_supramolekularne.pdf (Accessed date: 2 June 2020).
- 122.Bazin I., Tria S.A., Hayat A., Marty J.L. New biorecognition molecules in biosensors for the detection of toxins. Biosens. Bioelectron. 2017;87:285–298. doi: 10.1016/j.bios.2016.06.083. [DOI] [PubMed] [Google Scholar]
- 123.Kurzątkowska K., Jankowska A., Wysłouch-Cieszyńska A., Zhukova L., Puchalska M., Dehaen W., Radecka H., Radecki J. Voltammetric detection of the S100B protein using His-tagged RAGE domain immobilized onto a gold electrode modified with a dipyrromethene-Cu(II) complex and different diluents. J. Electroanal. Chem. (Lausanne Switz.) 2016;767:76–83. doi: 10.1016/j.jelechem.2016.02.012. [DOI] [Google Scholar]
- 124.Jargiło A., Grabowska I., Radecka H., Sulima M., Marszałek I., Wysłouch-Cieszyńska A., Dehaen W., Radecki J. Redox active dipyrromethene Cu(II) monolayer for oriented immobilization of his-tagged RAGE domains - the base of electrochemical biosensor for determination of Aβ 16-23′. Electroanalysis. 2013;25(5):1185–1193. doi: 10.1002/elan.201200537. [DOI] [Google Scholar]
- 125.Mikuła E., Sulima M., Marszałek I., Wysłouch-Cieszyńska A., Verwilst P., Dehaen W., Radecki J., Radecka H. Oriented immobilization of His-tagged protein on a redox active thiol derivative of DPTA-Cu(II) layer deposited on a gold electrode-the base of electrochemical biosensors. Sensors (Basel) 2013;13(9):11586–11602. doi: 10.3390/s130911586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Zborowska M., Sulima M., Marszałek I., Wysłouch- Cieszyńska A., Radecka H., Radecki J. Nitrilotriacetic acid-copper(II) monolayer deposited on a gold electrode for the immobilization of histidine tagged V domain of receptor for advanced glycation end products-the basis of amyloid-beta peptide sensing. Anal. Lett. 2014;47(8):1375–1391. doi: 10.1080/00032719.2013.867501. [DOI] [Google Scholar]
- 127.Hochuli E., Bannwarth W., Döbeli H., Gentz R., Stüber D. Genetic approach to facilitate purification of recombinant proteins with a novel metal chelate adsorbent. Nat. Biotechnol. 1988;6:1321–1325. doi: 10.1038/nbt1188-1321. [DOI] [Google Scholar]
- 128.Mikuła E., Wysłouch-Cieszyńska A., Zhukova L., Puchalska M., Verwilst P., Dehaen W., Radecki J., Radecka H. Voltammetric detection of S100B protein using His-tagged receptor domains for advanced glycation end products (RAGE) immobilized onto a gold electrode surface. Sensors (Basel) 2014;14(6):10650–10663. doi: 10.3390/s140610650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Bontidean I., Berggren C., Johansson G., Csöregi E., Mattiasson B., Lloyd J.R., Jakeman K.J., Brown N.L. Detection of heavy metal ions at femtomolar levels using protein-based biosensors. Anal. Chem. 1998;70(19):4162–4169. doi: 10.1021/ac9803636. [DOI] [PubMed] [Google Scholar]
- 130.Janata J. An immunoelectrode. J. Am. Chem. Soc. 1975;97(10):2914–2916. doi: 10.1021/ja00843a058. [DOI] [Google Scholar]
- 131.Dzantiev B., Zherdev A. Antibody-based biosensors. In: Nikolelis D.P., Varzakas T., Erdem A., Nikoleli G.P., editors. Portable Biosensing of Food Toxicants and Environmental Pollutants. Boca Raton: CRC Press Taylor & Francis Group; 2013. pp. 161–196. [DOI] [Google Scholar]
- 132.Ricci F., Volpe G., Micheli L., Palleschi G. A review on novel developments and applications of immunosensors in food analysis. Anal. Chim. Acta. 2007;605(2):111–129. doi: 10.1016/j.aca.2007.10.046. [DOI] [PubMed] [Google Scholar]
- 133.Skottrup P.D., Nicolaisen M., Justesen A.F. Towards on-site pathogen detection using antibody-based sensors. Biosens. Bioelectron. 2008;24(3):339–348. doi: 10.1016/j.bios.2008.06.045. [DOI] [PubMed] [Google Scholar]
- 134.Van Dorst B., Mehta J., Bekaert K., Rouah-Martin E., De Coen W., Dubruel P., Blust R., Robbens J. Recent advances in recognition elements of food and environmental biosensors: a review. Biosens. Bioelectron. 2010;26(4):1178–1194. doi: 10.1016/j.bios.2010.07.033. [DOI] [PubMed] [Google Scholar]
- 135.Guillozet-Bongaarts A.L., Garcia-Sierra F., Reynolds M.R., Horowitz P.M., Fu Y., Wang T., Cahill M.E., Bigio E.H., Berry R.W., Binder L.I. Tau truncation during neurofibrillary tangle evolution in Alzheimer’s disease. Neurobiol. Aging. 2005;26(7):1015–1022. doi: 10.1016/j.neurobiolaging.2004.09.019. [DOI] [PubMed] [Google Scholar]
- 136.Hardy J., Selkoe D.J. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–356. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
- 137.Wang S.X., Acha D., Shah A.J., Hills F., Roitt I., Demosthenous A., Bayford R.H. Detection of the tau protein in human serum by a sensitive four-electrode electrochemical biosensor. Biosens. Bioelectron. 2017;92:482–488. doi: 10.1016/j.bios.2016.10.077. [DOI] [PubMed] [Google Scholar]
- 138.Esteves-Villanueva J.O., Trzeciakiewicz H., Martic S. A protein-based electrochemical biosensor for detection of tau protein, a neurodegenerative disease biomarker. Analyst (Lond.) 2014;139(11):2823–2831. doi: 10.1039/C4AN00204K. [DOI] [PubMed] [Google Scholar]
- 139.Ciesiolka J., Gorski J., Yarus M. Selection of an RNA domain that binds Zn2+. RNA. 1995;1(5):538–550. [PMC free article] [PubMed] [Google Scholar]
- 140.Fang X., Tan W. Aptamers generated from cell-SELEX for molecular medicine: a chemical biology approach. Acc. Chem. Res. 2010;43(1):48–57. doi: 10.1021/ar900101s. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Mascini M., Palchetti I., Tombelli S. Nucleic acid and peptide aptamers: fundamentals and bioanalytical aspects. Angew. Chem. Int. Ed. Engl. 2012;51(6):1316–1332. doi: 10.1002/anie.201006630. [DOI] [PubMed] [Google Scholar]
- 142.Hong K.L., Sooter L.J. Single-stranded DNA aptamers against pathogens and toxins: identification and biosensing applications. BioMed Res. Int. 2015;2015:419318. doi: 10.1155/2015/419318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Mallikaratchy P. Evolution of complex target SELEX to identify aptamers against mammalian cell-surface antigens. Molecules. 2017;22(2):E215. doi: 10.3390/molecules22020215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Ruscito A., DeRosa M.C. Small-molecule binding aptamers: selection strategies, characterization, and applications. Front Chem. 2016;4:14. doi: 10.3389/fchem.2016.00014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Ellington A.D., Szostak J.W. In vitro selection of RNA molecules that bind specific ligands. . Nature. 1990;346(6287):818–822. doi: 10.1038/346818a0. [DOI] [PubMed] [Google Scholar]
- 146.Li Y. A quarter century of in vitro selection. J. Mol. Evol. 2015;81(5-6):137–139. doi: 10.1007/s00239-015-9723-7. [DOI] [PubMed] [Google Scholar]
- 147.Chen K., Fu T., Sun W., Huang Q., Zhang P., Zhao Z., Zhang X., Tan W. DNA-supramolecule conjugates in theranostics. Theranostics. 2019;9(11):3262–3279. doi: 10.7150/thno.31885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Liang H., Shi Y., Kou Z., Peng Y., Chen W., Li X., Li S., Wang Y., Wang F., Zhang X. Inhibition of BACE1 activity by a DNA aptamer in an Alzheimer’s disease cell model. PLoS One. 2015;10(10):e0140733. doi: 10.1371/journal.pone.0140733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Rentmeister A., Bill A., Wahle T., Walter J., Famulok M. RNA aptamers selectively modulate protein recruitment to the cytoplasmic domain of beta-secretase BACE1. in vitro. RNA. 2006;12(9):1650–1660. doi: 10.1261/rna.126306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Khan N.I., Maddaus A.G., Song E., Khan N.I., Maddaus A.G., Song E. A low-cost inkjet-printed aptamer-based electrochemical biosensor for the selective detection of lysozyme. Biosensors (Basel) 2018;8(1):7. doi: 10.3390/bios8010007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Bora U., Sett A., Singh D. Nucleic acid based biosensors for clinical applications. Biosens. J. 2013;2:1–8. doi: 10.4172/2090-4967.1000104. [DOI] [Google Scholar]
- 152.Macgregor R.B., Poon G.M.K. The DNA double helix fifty years on. Comput. Biol. Chem. 2003;27(4-5):461–467. doi: 10.1016/j.compbiolchem.2003.08.001. [DOI] [PubMed] [Google Scholar]
- 153.Szabat M., Pedzinski T., Czapik T., Kierzek E., Kierzek R. Structural aspects of the antiparallel and parallel duplexes formed by DNA, 2′-O-methyl RNA and RNA oligonucleotides. PLoS One. 2015;10(11):e0143354. doi: 10.1371/journal.pone.0143354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Pray L. Discovery of DNA structure and function: Watson and Crick. Nature Education. 2020;1(1):100. Available at: https://www.fcav.unesp.br/Home/departamentos/tecnologia/marcostuliooliveira/discovery-of-dna-double-helix_-watson-and-crick-_-learn-science-at-scitable.pdf (Accessed date: 2 June 2020). [Google Scholar]
- 155.Ahmed M.U., Idegami K., Chikae M., Kerman K., Chaumpluk P., Yamamura S., Tamiya E. Electrochemical DNA biosensor using a disposable electrochemical printed (DEP) chip for the detection of SNPs from unpurified PCR amplicons. Analyst (Lond.) 2007;132(5):431–438. doi: 10.1039/b615242b. [DOI] [PubMed] [Google Scholar]
- 156.Guo K., Li X., Kraatz H.B. Exploiting the interactions of PNA-DNA films with Ni2+ ions: detection of nucleobase mismatches and electrochemical genotyping of the single-nucleotide mismatch in apoE 4 related to Alzheimer’s disease. Biosens. Bioelectron. 2011;27(1):187–191. doi: 10.1016/j.bios.2011.06.013. [DOI] [PubMed] [Google Scholar]
- 157.Marrazza G., Tombelli S., Mascini M., Manzoni A. Detection of human apolipoprotein E genotypes by DNA biosensors coupled with PCR. Clin. Chim. Acta. 2001;307(1-2):241–248. doi: 10.1016/S0009-8981(01)00454-5. [DOI] [PubMed] [Google Scholar]
- 158.Lu H., Wu L., Wang J., Wang Z., Yi X., Wang J., Wang N. Voltammetric determination of the Alzheimer’s disease-related ApoE 4 gene from unamplified genomic DNA extracts by ferrocene-capped gold nanoparticles. Mikrochim. Acta. 2018;185(12):549. doi: 10.1007/s00604-018-3087-9. [DOI] [PubMed] [Google Scholar]
- 159.Thévenot D.R., Toth K., Durst R.A., Wilson G.S. Electrochemical biosensors: recommended definitions and classification. Biosens. Bioelectron. 2001;16(1-2):121–131. doi: 10.1016/s0956-5663(01)00115-4. [DOI] [PubMed] [Google Scholar]
- 160.Grieshaber D., MacKenzie R., Vörös J., Reimhult E. Electrochemical biosensors - sensor principles and architectures. Sensors (Basel) 2008;8(3):1400–1458. doi: 10.3390/s80314000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Liu Y., Xu L.P., Wang S., Yang W., Wen Y., Zhang X. An ultrasensitive electrochemical immunosensor for apolipoprotein E4 based on fractal nanostructures and enzyme amplification. Biosens. Bioelectron. 2015;71:396–400. doi: 10.1016/j.bios.2015.04.068. [DOI] [PubMed] [Google Scholar]
- 162.Mikuła E., Wysłouch-Cieszyńska A., Zhukova L., Verwilst P., Dehaen W., Radecki J., Radecka H. Electrochemical biosensor for the detection of glycated albumin. Curr. Alzheimer Res. 2017;14(3):345–351. doi: 10.2174/1567205013666161108110542. [DOI] [PubMed] [Google Scholar]
- 163.Jahnke H.G., Krinke D., Seidel D., Lilienthal K., Schmidt S., Azendorf R., Fischer M., Mack T., Striggow F., Althaus H., Schober A., Robitzki A.A. A novel 384- multiwell microelectrode array for the impedimetric monitoring of Tau protein induced neurodegenerative processes. Biosens. Bioelectron. 2017;88:78–84. doi: 10.1016/j.bios.2016.07.074. [DOI] [PubMed] [Google Scholar]
- 164.Dai Y., Molazemhosseini A., Liu C.C.A.A. Single-Use, In vitro biosensor for the detection of T-Tau protein, A biomarker of neuro-degenerative disorders, in PBS and human serum using differential pulse voltammetry (DPV). . Biosensors (Basel) 2017;7(1):1–11. doi: 10.3390/bios7010010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Hassan Q., Kerman K. Electrochemical approaches for the detection of amyloid-β, tau, and α-synuclein. Curr. Opin. Electrochem. 2018;14:89–95. doi: 10.1016/j.coelec.2018.12.009. [DOI] [Google Scholar]
- 166.Negahdary M., Heli H. An ultrasensitive electrochemical aptasensor for early diagnosis of Alzheimer’s disease, using a fern leaves-like gold nanostructure. Talanta. 2019;198:510–517. doi: 10.1016/j.talanta.2019.01.109. [DOI] [PubMed] [Google Scholar]
- 167.Negahdary M., Heli H. An electrochemical peptide-based biosensor for the Alzheimer biomarker amyloid-β(1-42) using a microporous gold nanostructure. Mikrochim. Acta. 2019;186(12):766. doi: 10.1007/s00604-019-3903-x. [DOI] [PubMed] [Google Scholar]
- 168.Li H., Xie H., Cao Y., Ding X., Yin Y., Li G. A general way to assay protein by coupling peptide with signal reporter via supermolecule formation. . Anal. Chem. 2013;85(2):1047–1052. doi: 10.1021/ac302906c. [DOI] [PubMed] [Google Scholar]
- 169.Rushworth J.V., Ahmed A., Griffiths H.H., Pollock N.M., Hooper N.M., Millner P.A. A label-free electrical impedimetric biosensor for the specific detection of Alzheimer’s amyloid-beta oligomers. Biosens. Bioelectron. 2014;56:83–90. doi: 10.1016/j.bios.2013.12.036. [DOI] [PubMed] [Google Scholar]
- 170.Sun L., Zhong Y., Gui J., Wang X., Zhuang X., Weng J. A hydrogel biosensor for high selective and sensitive detection of amyloid-beta oligomers. Int. J. Nanomedicine. 2018;13:843–856. doi: 10.2147/IJN.S152163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Rama E.C., González-García M.B., Costa-García A. Competitive electrochemical immunosensor for amyloid- beta 1-42 detection based on gold nanostructurated screen-printed carbon electrodes. Sens. Actuators B Chem. 2014;201:567–571. doi: 10.1016/j.snb.2014.05.044. [DOI] [Google Scholar]
- 172.Lien T.T.N., Takamura Y., Tamiya E., Vestergaard M.C. Modified screen printed electrode for development of a highly sensitive label-free impedimetric immunosensor to detect amyloid beta peptides. Anal. Chim. Acta. 2015;892:69–76. doi: 10.1016/j.aca.2015.08.036. [DOI] [PubMed] [Google Scholar]
- 173.Liu Y., Wang H., Chen J., Liu C., Li W., Kong J., Yang P., Liu B. A sensitive microchip-based immunosensor for electrochemical detection of low-level biomarker s100b. Electroanalysis. 2013;25(4):1050–1055. doi: 10.1002/elan.201200525. [DOI] [Google Scholar]
- 174.Kruse N., Schlossmacher M.G., Schulz-Schaeffer W.J., Vanmechelen E., Vanderstichele H., El-Agnaf O.M., Mollenhauer B. A first tetraplex assay for the simultaneous quantification of total α-Synuclein, Tau, β-Amyloid42 and DJ-1 in human cerebrospinal fluid. PLoS One. 2016;11(4):e0153564. doi: 10.1371/journal.pone.0153564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Chang H., Kang H., Ko E., Jun B-H., Lee H-Y., Lee Y-S., Jeong D.H. PSA detection with femtomolar sensitivity and a broad dynamic range using sers nanoprobes and an area-scanning method. ACS Sens. 2016;1(6):645–649. doi: 10.1021/acssensors.6b00053. [DOI] [Google Scholar]
- 176.Kern S., Zetterberg H., Kern J., Zettergren A., Waern M., Höglund K., Andreasson U., Wetterberg H., Börjesson-Hanson A., Blennow K., Skoog I. Prevalence of preclinical Alzheimer disease: Comparison of current classification systems. Neurology. 2018;90(19):e1682–e1691. doi: 10.1212/WNL.0000000000005476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Ceylan O., Mishra G.K., Yazici M., Qureshi A., Niazi J.H., Gurbuz Y. A hand-held point-of-care biosensor device for detection of multiple cancer and cardiac disease biomarkers using interdigitated capacitive arrays. IEEE Trans. Biomed. Circuits Syst. 2018;12(6):1440–1449. doi: 10.1109/TBCAS.2018.2870297. [DOI] [PubMed] [Google Scholar]
- 178.Yu Y., Wang P., Zhu X., Peng Q., Zhou Y., Yin T., Liang Y., Yin X. Combined determination of copper ions and β-amyloid peptide by a single ratiometric electrochemical biosensor. Analyst (Lond.) 2017;143(1):323–331. doi: 10.1039/C7AN01683B. [DOI] [PubMed] [Google Scholar]
- 179.Song Y., Xu T., Zhu Q., Zhang X. Integrated individually electrochemical array for simultaneously detecting multiple Alzheimer’s biomarkers. Biosens. Bioelectron. 2020;162:112253. doi: 10.1016/j.bios.2020.112253. [DOI] [PubMed] [Google Scholar]









