Electrochemical Aptasensors for Detection of Neurodegenerative Diseases Biomarkers: Comparison
Please note this is a comparison between Version 3 by Edyta Mikuła and Version 2 by Sirius Huang.

Neurodegenerative diseases (NDs) are becoming a major global health problem. They constitute an incurable diverse group of disorders characterized by the progressive degeneration of the structure and function of the central or peripheral nervous system. There is an enormous unmet demand worldwide for methods for the early detection of ND biomarkers. Among the various approaches reported so far, biosensors are powerful analytical implements that have been applied to detect biomarkers of NDs. Electrochemical aptasensors have been at the forefront of this development not only thanks to their low cost and simple design but also due to advances in nanomaterials modifying the surface of the transducers involved.

  • aptasensor
  • aptamer
  • neurodegenerative disease biomarkers
  • electrochemical detection
  • Amyloid β peptide
  • tau protein
  • α-synuclein
  • human cellular prion protein

1. Introduction

Each year, tens of millions of people around the world are diagnosed with NDs including Alzheimer’s disease (AD), Parkinson’s disease (PD) or prion diseases. These are known as “protein misfolding disorders” and are caused by proteins that are prone to aggregation. In NDs, there is a slow, progressive loss of the structure or dysfunction of neurons in the central nervous system, leading to deficits in specific brain functions. They are considered incurable due to the lack of a known way to reverse the progressive degeneration of neurons. Most NDs begin years before symptoms are present, and the first symptoms appear when a significant number of neurons are damaged or the damage affects a specific part of the central nervous system. For many of these illnesses, diagnosis and treatment are limited. Early diagnosis of age-related disorders is an increasingly pressing health challenge worldwide and is crucial in the management of the disease and determines the patient’s living conditions [1][2]. The detection of NDs can be accomplished by determining the concentration of specific biomarkers including α-synuclein (α-syn), amyloid β peptide (Aβ), tau protein or human cellular prion protein (PrPC) in body fluids. Therefore, non-invasive, simple, fast and real-time methods of detecting the biomarkers of NDs are particularly sought after. To fulfill these requirements, electrochemical biosensors offer potential diagnostic and theranostic applications. Biosensors are analytical devices that convert the result of a biological reaction into an analytical signal. Each biosensor consists of two basic components: an analytically active layer (the receptor part) and a transducer. The recognition elements include molecules constituting biological material, e.g., nucleic acids (DNA or RNA aptamers), proteins, enzymes, antibodies or whole cells, immobilized on a suitable carrier. In the analytically active layer, the process of intermolecular recognition (receptor–analyte) takes place. During this process, a physicochemical signal is generated, and a transducer converts it into an analytically useful signal. The most commonly used transducers are optical, piezoelectric or electrochemical systems [3]. In the last few years, special emphasis has been placed on aptamers as the molecular recognition elements in electrochemical biosensors (aptasensors), which has been confirmed by the rapid increase in the number of scientific papers in this field [4][5][6][7]. According to Web of Science, publications concerning electrochemical aptamer-based biosensors represent about 28% of the total number of publications on electrochemical biosensors [3]. Aptamers are short, single- or double-stranded DNA or RNA nucleotides or peptides [8]. They have emerged as good alternatives to antibodies in the design of electrochemical biosensors. They are distinguished by a low immunogenicity, longer shelf life, easy storage and the possibility of transport at ambient temperature. The advantages of aptamer manufacturing processes include their lower cost, batch-to-batch variability, simple upscaling and purification. These processes do not require experiments with animals or cells. Additionally, they can be selected under non-physiological conditions [9][10]]. They also exhibit a high binding affinity and specificity for a broad range of targets (such as proteins, amino acids, peptides, drugs, small metal ions and small organic molecules, as well as bacteria and viruses and even whole cells) due to their specific three-dimensional structures [11][12][13][14][15]. Moreover, aptamers display a specificity for some targets (e.g., small molecules or ions) that cannot be recognized by antibodies. They are also able to distinguish forms and isoforms of the same protein, which is an important advantage for biomarker detection. Furthermore, it is possible to manipulate the binding reaction conditions and optimize the aptamers’ recognition sequences to fine-tune their affinities [16]. Aptamers are also good candidates for conjugation, labeling or functionalization, which is often achieved in the step of aptamer synthesis [17]20[18][19]. With each new aptamer–protein couple, a systematic analysis of their binding process, conditions and structure of the created complexes, as well as their interfacial behavior, is necessary [19][20]. In this framework, a suitable method for aptamer immobilization on a solid surface is critical for the successful elaboration of the biosensor. The selection of the aptamer deposition strategy strongly relies on the modification of the aptamer and the physicochemical features of the electrode surface [8][20]. The electrode material used for the deposition of aptamers should allow the precise control of the packing density and spatial orientation of the aptamers in the deposited layer via their controlled immobilization on the surface. This bind should be strong and stable, providing the conformational flexibility required for the process of the specific recognition of the analyte. The aptamer immobilization procedure should ensure the formation of a stable, reproducible receptor layer and the sensitive detection of the analyte, and it should limit the non-specific adsorption of matrix components. An important issue to be addressed in the development of protein aptasensors is the occurrence of false-positive signals due to the non-specific adsorption of proteins found in biological fluids [21]. The most frequently described methods of assembly of aptamers on solid surfaces are physisorption, chemisorption and affinity conjugation (avidin–biotin). Physical absorption based on electrostatic interactions is the simplest method for depositing of aptamers on solid surfaces. However, the low stability of the electrostatically deposited layer caused by the rapid desorption of the aptamers from the surface is problematic in this method. The solution may be additional stabilization by covalent bonding [21]. The second very common method of attaching aptamers to solid surfaces is chemisorption. Aptamers are covalently deposited on the surfaces of the solid electrodes depending on the type of substrate and its functionalization. However, they do not spontaneously form covalent bonds with the electrode surfaces. Therefore, it is necessary to functionalize either the aptamer or the electrode surface [22]. Possibly the most popular approach is the chemisorption of a thiol-modified aptamer on a gold surface [23][24]. However, this method is unstable over time. Generally, aptamers are co-immobilized on the gold electrode surface with alkanethiols [25][26], dithiols [27][28] or anti-biofouling agents [29][30] to fill unmodified areas. One of the most effective approaches to aptamer immobilization is based on the strong affinity of biotin to avidin [31], streptavidin [32] and neutravidin [33]. Compared to other aptamer immobilization methods, the high affinity of biotin to streptavidin increases the potential number of aptamers that can be deposited on the surface of the detection platform. This is because each streptavidin molecule can bind to two biotinylated aptamers. This implies a reduction in non-specific adsorption and an improvement in the signal-to-noise ratio (S/N) of the biosensor [34]. The appropriate preparation of the detection platform ensures the low sensitivity and high selectivity of the constructed aptasensor. Therefore, it is important to implement a signal amplification strategy through the use of functional nanomaterials in the detection layer [35][36]. A wide range of nanomaterials, including silica nanoparticles [37] and precious metals [38][39], carbon nanomaterials [40][41], nanocomposites and nanohybrids [42][43], polymers [44][45] and metal oxides [46], can be used in the construction of electrochemical aptasensors for biomedical applications. Advances in electrochemical aptasensors have resulted not only from the use of nanomaterials but also from the interdisciplinary combination of nanotechnology, engineering, nanomaterial chemistry and related fields [3][47][48][49][50][51][52]. Recently, there is a huge demand for new portable analytical devices with better selectivity and specificity for health-care applications, mainly for addressing the point-of-care (POC) detection of biomarkers for chronic and emergent diseases such as cancer, neurodegenerative disorders, cardiovascular diseases or chronic respiratory diseases [3]. This represents the main reason for the development of innovative techniques for the modification and immobilization of aptamers as biorecognition molecules with unique properties in the field of electrochemical aptasensors [53][54]. Nevertheless, their numerous assets and significant advances have not yet resulted in their entry to the market of electrochemical aptasensors [55][56][57]. Electrochemical aptasensors are widely used in the detection of ND biomarkers due to their numerous advantages such as their speed of analysis, low cost, easy operation and high miniaturization potential.

2. Electrochemical Aptasensors for the Detection of Neurodegenerative Diseases Biomarkers

A measurable change arising in biological surroundings such as human tissues or body fluids is defined as a biomarker. This change should arise from the pathological condition or response of the body to treatment when assessing the effectiveness of pharmacological therapy [58][59]. Thus, biomarkers are compounds that need to reflect the basic neuropathological features of a disease, and their determination should be conducted by means of a safe, easy and quick diagnostic test [60].
There has recently been great progress in the determination of biomarkers aimed at detecting these diseases in the phase preceding the appearance of distinctive clinical symptoms [61][62]. Enzyme-linked immunosorbent assay (ELISA) [63][64][65], magnetic resonance imaging (MRI) [66], manganese-enhanced magnetic resonance imaging (MEMRI) [67], mass spectrometry (MS) [68][69], flexible multi-analyte profiling (xMAP) [70], positron emission tomography (PET) [71][72], surface plasmon resonance [73], Western blotting [74][75], scanning tunneling electron microscopy [76], immunohistochemistry [77][78][79] and fluorescence [80][81][82] are some of the major techniques used for the detection of biomarkers of NDs. Despite their numerous advantages, these methods are relatively expensive, time-consuming and difficult to access, and they often require specialized equipment, qualified personnel and large volumes of samples. Moreover, they are not yet suitable for POC diagnostics.
The development of easy-of-use electrochemical aptamer-based platforms with a high sensitivity and specificity is probably one of the most promising methods to solve some of the problems regarding fast and cost-effective measurements appropriate for POC diagnostics due to their potential in terms of their portability and novel microfabrication technologies [83][84][85][86].
Recently developed sensing platforms for electrochemical detection of biomarkers of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and prion diseases are summarized in Table 1.
Table 1. Specifications of electrochemical aptasensors for the detection of ND biomarkers.
Biomarker Biorecognition Element Techniques Linear Range of Detection LOD Ref.
α-syn

oligomer
DNA Apt DPV 6 × 10−11–1.5 × 10−7 M 10−11 M [87]
SH-DNA Apt EIS 10−12 M [88]
DNA Apt-MB DPV 10−15–10−9 M 6.4 × 10−16 M [89]
NH2-DNA Apt EIS 10−19–10−14 M 7 × 10−20 M (buffer)

9 × 10−20 M (plasma)
[90]
Total

α-syn
DNA Apt-AuNPs conjugate Vm 10−11–10−6 M 10−11 M [91]
RNA Apt DPV 0.002–1.28 ng/mL 0.4 pg/mL [92]
AβOs Ab-DNA Apt sandwich DPV 10−10 M [93]
DNA Apt-AuNFs DPV 10−9–2 × 10−6 M 4.5 × 10−10 M [94]
DNA Apt EIS 10−10–5 × 10−7 M 3 × 10−11 M [95]
Apt-Poly T-CuNPs DPV 10−11–2.2 × 10−9 M 3.5 × 10−12 M [96]
DNA Apt1 Apt2 LSV 10−12–10−8 M 4.3 × 10−13 M [97]
SH–stem-loop DNA Apt-Fc ACV 10−12–2 × 10−7 M 3 × 10−13 M [98]
NH2-DNA Apt DPV 4.43 × 10−14–4.43 × 10−6 M 10−14 M [99]
DNA Apt–Fc ACV 10−13–1.5 × 10−6 M 2 × 10−15 M [100]
THAS DPV 10−15–10−11 M 5 × 10−16 M [101]
SH-DNA Apt DPV 5 × 10−16–5 × 10−13 M 2.5 × 10−16 M [102]
SiO2Ag- DNA Apt bioconjugate DPV 5 pg/mL–10 ng/mL 1.22 pg/mL [103]
SH-DNA Apt DPV 0.5–10 pg/mL

160 fg/mL (buffer)

900 fg/mL (serum)
[104]
DNA Apt–SnS2 NSs EIS 10−4–103 ng/mL

238.9 fg/mL (PB)

56.9 fg/mL (HS)
[105]
AβOs(40) DNA Apt SWV 0.100–1.00 µM 9.3 × 10−11 M [106]
Tau-381 protein NH2-DNA Apt DPV 1.0–10−10 M 7 × 10−13 M [107]
Anti-tau Ab + tau-381 DNA Apt DPV 0.5–10−10 M 4.2 × 10−13 M [108]
NH2-DNA Apt–MWCNTs EIS 10−15–1 × 10−9 M 10−15 M [109]
SH-DNA Apt–VG@Au DPV 0.1 pg/mL–1–1 ng/mL 0.034 pg/mL [110]
PrPC Biot DNA Apt with dT15 spacer DPV 10−12–10−9 M 8 × 10−13 M [111]
Biot DNA Apt with dT15 spacer CV 10−12–10−5 M 5 × 10−13 M [112]
MB DNA Apt and FcA SWV 2 × 10−13–10−5 M 1.6 × 10−13 M [113]
DNA1 Apt and DNA2 Apt SWV 2 × 10−14–2.8 × 10−13 M 7.6 × 10−15 M [114]
Abbreviations: Apt—aptamer, EIS—electrochemical impedance spectroscopy, MB—methylene blue, DPV—differential pulse voltammetry, Vm—voltammetry, LSV—linear sweep voltammetry, AuNFs—gold nanoflowers, Ab—antibody, ACV—alternative current voltammetry, Biot—biotinylated, THAS—triple-helix aptamer switch, Poly T-CuNPs—poly(thymine) copper nanoparticles, MWCNTs—multiwalled carbon nanotubes, DNA Apt–SnS2 NSs—DNA aptamer onto vertical aligned tin disulphide nanosheets, PB—physiological buffer, HS—human serum samples, VG@Au—vertical graphene modified with nanogold, CV—cyclic voltammetry, FcA—ferrocenecarboxylic acid.

References

  1. Gao, H.-M.; Hong, J.-S. Why neurodegenerative diseases are progressive: Uncontrolled inflammation drives disease progression. Trends Immunol. 2008, 29, 357–365.
  2. Amor, S.; Puentes, F.; Baker, D.; van der Valk, P. Inflammation in neurodegenerative diseases. Immunol. 2010, 129, 154–169.
  3. Villalonga, A.; Pérez-Calabuig, A.M.; Villalonga, R. Electrochemical biosensors based on nucleic acid aptamers. Anal. Bioanal. Chem. 2020, 412, 55–72.
  4. Stanciu, L.A.; Wei, Q.; Barui, A.K.; Noor, M. Recent advances in aptamer-based biosensors for global health applications. Annu. Rev. Biomed. Eng. 2021, 23, 433–459.
  5. Yoo, H.; Jo, H.; Oh, S.S. Detection and beyond: Challenges and advances in aptamer-based biosensors. Mater. Adv. 2020, 1, 2663.
  6. Khan, N.I.; Song, E. Lab-on-a-chip systems for aptamer-based biosensing. Micromachines 2020, 11, 220.
  7. Rahman, M.M. Progress in electrochemical biosensing of SARS-CoV-2 virus for COVID-19 management. Chemosensors 2022, 10, 287.
  8. Mascini, M.; Palchetti, I.; Tombelli, S. Nucleic acid and peptide aptamers: Fundamentals and bioanalytical aspects. Angew. Chem. Int. Ed. Engl. 2012, 51, 1316–1332.
  9. Zhang, Y.; Lai, B.S.; Juhas, M. Recent advances in aptamer discovery and applications. Molecules 2019, 24, 941.
  10. Dunn, M.R.; Jimenez, R.; Chaput, J.C. Analysis of aptamer discovery and technology. Nat. Rev. Chem. 2017, 1, 0076.
  11. Chen, C.; Zhou, S.; Cai, Y.Q.; Tang, F.Q. Nucleic acid aptamer application in diagnosis and therapy of colorectal cancer based on cell-selex technology. NPJ Precis. Oncol. 2017, 1, 37.
  12. Zhou, J.; Rossi, J. Aptamers as targeted therapeutics: Current potential and challenges. Nat. Rev. Drug Discovery 2016, 16, 181–202.
  13. Feng, S.L.; Chen, C.T.; Wang, W.; Que, L. An aptamer nanopore-enabled microsensor for detection of theophylline. Biosens. Bioelectron. 2018, 105, 36–41.
  14. Umrao, S.; Jain, V.; Anusha; Chakraborty, B.; Roy, R. Protein-induced fluorescence enhancement as aptamer sensing mechanism for thrombin detection. Sens. Actuators B 2018, 267, 294–301.
  15. Saraf, N.; Woods, E.R.; Peppler, M.; Seal, S. Highly selective aptamer based organic electrochemical biosensor with pico-level detection. Biosens. Bioelectron. 2018, 117, 40–46.
  16. Wang, H.Q.; Wu, Z.; Tang, L.J.; Yu, R.Q.; Jiang, J.H. Fluorescence protection assay: A novel homogeneous assay platform toward development of aptamer sensors for protein detection. Nucleic Acids Res. 2011, 39, e122.
  17. Elskens, J.P.; Elskens, J.M.; Madder, A. Chemical modification of aptamers for increased binding affinity in diagnostic applications: Current status and future prospects. Int. J. Mol. Sci. 2020, 21, 4522.
  18. Nerantzaki, M.; Loth, C.; Lutz, J.-F. Chemical conjugation of nucleic acid aptamers and synthetic polymers. Polym. Chem. 2021, 12, 3498–3509.
  19. Ozturk, M.; Nilsen-Hamilton, M.; Ilgu, M. Aptamer applications in neuroscience. Pharmaceuticals 2021, 14, 1260.
  20. Kong, H.Y.; Byun, J. Nucleic acid aptamers: New methods for selection, stabilization, and application in biomedical science. Biomol. Ther. 2013, 21, 423–434.
  21. Arshavsky-Graham, S.; Heuer, C.; Jiang, X.; Segal, E. Aptasensors versus immunosensors—Which will prevail? Eng. Life Sci. 2022, 22, 319–333.
  22. Pividori, M.I.; Merkoci, A.; Alegret, S. Electrochemical genosensor design: Immobilisation of oligonucleotides onto transducer surfaces and detection methods. Biosens. Bioelectron. 2000, 15, 291–303.
  23. Li, Y.; Lu, Y. Functional Nucleic Acids for Analytical Applications, 1st ed.; Springer: New York, NY, USA, 2009.
  24. Oberhaus, F.V.; Frense, D.; Beckmann, D. Immobilization techniques for aptamers on gold electrodes for the electrochemical detection of proteins: A review. Biosensors 2020, 10, 45.
  25. Nan, M.-N.; Bi, Y.; Xue, H.-L.; Long, H.-T.; Xue, S.-L.; Pu, L.-M.; Prusky, D. Modification performance and electrochemical characteristics of different groups of modified aptamers applied for label-free electrochemical impedimetric sensors. Food Chem. 2021, 337, 127761.
  26. Zamay, G.S.; Zamay, T.N.; Kolovskii, V.A.; Shabanov, A.V.; Glazyrin, Y.E.; Veprintsev, D.V.; Krat, A.V.; Zamay, S.S.; Kolovskaya, O.S.; Gargaun, A.; et al. Electrochemical aptasensor for lung cancer-related protein detection in crude blood plasma samples. Sci. Rep. 2016, 6, 34350.
  27. Mazaafrianto, D.N.; Ishida, A.; Maeki, M.; Tani, H.; Tokeshi, M. Label-free electrochemical sensor for Ochratoxin A using a microfabricated electrode with immobilized aptamer. ACS Omega 2018, 3, 16823–16830.
  28. Ding, S.; Mosher, C.; Lee, X.Y.; Das, S.R.; Cargill, A.A.; Tang, X.; Chen, B.; McLamore, E.S.; Gomes, C.; Hostetter, J.M.; et al. Rapid and label-free detection of interferon gamma via an electrochemical aptasensor comprised of a ternary surface monolayer on a gold interdigitated electrode array. ACS Sens. 2017, 2, 210–217.
  29. Su, Z.; Xu, H.; Xu, X.; Zhang, Y.; Ma, Y.; Li, C.; Xie, Q. Effective covalent immobilization of Quinone and aptamer onto a gold electrode via thiol addition for sensitive and selective protein biosensing. Talanta 2017, 164, 244–248.
  30. Cui, M.; Wang, Y.; Jiao, M.; Jayachandran, S.; Wu, Y.; Fan, X.; Luoet, X. Mixed self-assembled aptamer and newly designed zwitterionic peptide as antifouling biosensing interface for electrochemical detection of alpha-fetoprotein. ACS Sens. 2017, 2, 490–494.
  31. Liu, N.; Song, J.; Lu, Y.; Davis, J.J.; Gao, F.; Luo, X. Electrochemical aptasensor for ultralow fouling cancer cell quantification in complex biological media based on designed branched peptides. Anal. Chem. 2019, 91, 8334–8340.
  32. Jain, A.; Cheng, K. The principles and applications of avidin-based nanoparticles in drug delivery and diagnosis. J. Control. Release 2017, 245, 27–40.
  33. Pandey, A.K.; Rajput, Y.S.; Sharma, R.; Singh, D. Immobilized aptamer on gold electrode senses trace amount of aflatoxin M1. Appl. Nanosci. 2017, 7, 893–903.
  34. Karapetis, S.; Nikolelis, D.; Hianik, T. Label-free and redox markers-based electrochemical aptasensors for aflatoxin M1 detection. Sensors 2018, 18, 4218.
  35. Tahiri-Alaoui, A.; Frigotto, L.; Manville, N.; Ibrahim, J.; Romby, P.; James, W. High affinity nucleic acid aptamers for streptavidin incorporated into bi-specific capture ligands. Nucleic Acids Res. 2002, 30, e45.
  36. Gan, X.; Zhao, H. Understanding signal amplification strategies of nanostructured electrochemical sensors for environmental pollutants. Curr. Opin. Electrochem. 2019, 17, 56–64.
  37. Liu, J.; Morris, M.D.; Macazo, F.C.; Schoukroun-Barnes, L.R.; White, R.J. The current and future role of aptamers in electroanalysis. J. Electrochem. Soc. 2014, 161, H301–H313.
  38. Walcarius, A. Silica-based electrochemical sensors and biosensors: Recent trends. Curr. Opin. Electrochem. 2018, 10, 88–97.
  39. Negahdary, M. Electrochemical aptasensors based on the gold nanostructures. Talanta 2020, 216, 120999.
  40. Pan, M.; Yang, J.; Liu, K.; Yin, Z.; Ma, T.; Liu, S.; Xu, L.; Wang, S. Noble metal nanostructured materials for chemical and biosensing systems. Nanomaterials 2020, 10, 209.
  41. Wang, Z.; Yu, J.; Gui, R.; Jin, H.; Xia, Y. Carbon nanomaterials-based electrochemical aptasensors. Biosens. Bioelectron. 2016, 79, 136–149.
  42. Amiri, M.; Nekoueian, K.; Saberi, R.S. Graphene-family materials in electrochemical aptasensors. Anal. Bioanal. Chem. 2021, 413, 673–699.
  43. Idris, A.O.; Mamba, B.; Feleni, U. Poly (propylene imine) dendrimer: A potential nanomaterial for electrochemical application. Mater. Chem. Phys. 2020, 244, 122641.
  44. Amouzadeh Tabrizi, M.; Acedo, P. An electrochemical impedance spectroscopy-based aptasensor for the determination of SARS-CoV-2-RBD using a carbon nanofiber–gold nanocomposite modified screen-printed electrode. Biosensors 2022, 12, 142.
  45. Gui, R.; Jin, H.; Guo, H.; Wang, Z. Recent advances and future prospects in molecularly imprinted polymers-based electrochemical biosensors. Biosens. Bioelectron. 2018, 100, 56–70.
  46. Kaya, S.I.; Cetinkaya, A.; Ozkan, S.A. Latest advances in determination of bisphenols with nanomaterials, molecularly imprinted polymers and aptamer based electrochemical sensors. Crit. Rev. Anal. Chem. 2022, 52, 1223–1243.
  47. George, J.M.; Antony, A.; Mathew, B. Metal oxide nanoparticles in electrochemical sensing and biosensing: A review. Microchim. Acta 2018, 185, 358.
  48. Ma, M.; Lu, X.; Guo, Y.; Wang, L.; Liang, X. Combination of metal-organic frameworks (MOFs) and covalent organic frameworks (COFs): Recent advances in synthesis and analytical applications of MOF/COF composites. TrAC Trends Anal. Chem. 2022, 157, 116741.
  49. Ye, Q.; Zhang, Z.; Liu, J.; Wang, X. Screen-printed electrode-based biosensors modified with functional nucleic acid probes and their applications in this pandemic age: A review. Anal. Methods 2022, 14, 2961–2975.
  50. Onaş, A.M.; Dascălu, C.; Raicopol, M.D.; Pilan, L. critical design factors for electrochemical aptasensors based on target-induced conformational changes: The case of small-molecule targets. Biosensors 2022, 12, 816.
  51. Villalonga, A.; Mayol, B.; Villalonga, R.; Vilela, D. Electrochemical aptasensors for clinical diagnosis. A review of the last five years. Sens. Actuators B 2022, 369, 132318.
  52. Kurup, C.P.; Mohd-Naim, N.F.; Ahmed, M.U. Recent trends in nanomaterial-based signal amplification in electrochemical aptasensors. Crit. Rev. Biotechnol. 2022, 42, 794–812.
  53. Yousef, H.; Liu, Y.; Zheng, L. Nanomaterial-based label-free electrochemical aptasensors for the detection of thrombin. Biosensors 2022, 12, 253.
  54. Song, S.; Wang, L.; Li, J.; Fan, C.; Zhao, J. Aptamer-based biosensors. Trends Anal. Chem. 2008, 27, 108–117.
  55. Plaxco, K.W.; Soh, H.T. Switch-based biosensors: A new approach towards real-time, in vivo molecular detection. Trends. Biotechnol. 2011, 29, 1–5.
  56. Kaur, H.; Bruno, J.G.; Kumar, A.; Sharma, T.K. Aptamers in the therapeutics and diagnostics pipelines. Theranostics 2018, 8, 4016.
  57. Majdinasab, M.; Marty, J.L. Recent advances in electrochemical aptasensors for detection of biomarkers. Pharmaceuticals 2022, 15, 995.
  58. Ghoorchian, A.; Moradi, M.; Aghajani, S.; Afkhami, A.; Madrakian, T.; Thomas, S.; Nguyen, T.A.; Ahmadi, M. Miniaturized bioelectrochemical devices. In Micro and Nano Technologies, Micro- and Nanotechnology Enabled Applications for Portable Miniaturized Analytical Systems, 1st ed.; Thomas, S., Ahmadi, M., Nguyen, T.A., Afkhami, A., Madrakian, T., Eds.; Elsevier: Amsterdam, The Netherlands, 2021; Chapter 5; pp. 89–108.
  59. Fuellen, G.; Jansen, L.; Cohen, A.A.; Luyten, W.; Gogol, M.; Simm, A.; Saul, N.; Cirulli, F.; Berry, A.; Antal, P.; et al. Health and aging: Unifying concepts, scores, biomarkers and pathways. Aging Dis. 2019, 10, 883.
  60. Mayeux, R. Biomarkers: Potential uses and limitations. NeuroRx 2004, 1, 182–188.
  61. Fagan, A.M.; Roe, C.M.; Xiong, C.; Mintun, M.A.; Morris, J.C.; Holtzman, D.M. Cerebrospinal fluid tau/β-Amyloid42 ratio as a prediction of cognitive decline in nondemented older adults. Arch. Neurol. 2007, 64, 343.
  62. Counts, S.E.; Ikonomovic, M.D.; Mercado, N.; Vega, I.E.; Mufson, E.J. Biomarkers for the early detection and progression of Alzheimer’s disease. Neurotherapeutics 2017, 14, 35–53.
  63. Coupé, P.; Manjón, J.V.; Lanuza, E.; Catheline, G. Lifespan changes of the human brain in Alzheimer’s disease. Sci. Rep. 2019, 9, 3998.
  64. 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, 99–113.
  65. Shinohara, M.; Hirokawa, J.; Shimodaira, A.; Tashiro, Y.; Suzuki, K.; Gheni, G.; Fukumori, A.; Matsubara, T.; Morishima, M.; Saito, Y.; et al. ELISA evaluation of tau accumulation in the brains of patients with Alzheimer disease. J. Neuropathol. Exp. Neurol. 2021, 80, 652–662.
  66. Yang, C.-M.; Chang, J.-Y.; Chen, M.-Y.; Lai, C.-S. A systematic study and potential limitations of proton-ELISA platform for α-synuclein antigen detection. Chemosensors 2022, 10, 5.
  67. Noor, M.B.T.; Zenia, N.Z.; Kaiser, M.S.; Mahmud, M.; Al Mamun, S. Detecting Neurodegenerative Disease from MRI: A Brief Review on a Deep Learning Perspective. In International Conference on Brain Informatics; Liang, P., Goel, V., Shan, C., Eds.; Lecture Notes in Computer Science; Springer: Cham, Switzerland, 2019; pp. 115–125.
  68. Jun, Y.; Qinqing, L. Manganese-enhanced magnetic resonance imaging: Application in central nervous system diseases. Front. Neurol. 2020, 11, 143.
  69. Agrawal, I.; Tripathi, P.; Biswas, S. Mass spectrometry based protein biomarkers and drug target discovery and clinical diagnosis in age-related progressing neurodegenerative diseases. Drug Metab. Rev. 2022, 54, 22–36.
  70. Azevedo, R.; Jacquemin, C.; Villain, N.; Fenaille, F.; Lamari, F.; Becher, F. Mass spectrometry for neurobiomarker discovery: The relevance of post-translational modifications. Cells 2022, 11, 1279.
  71. 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.
  72. Wang, Y.T.; Edison, P. Tau imaging in neurodegenerative diseases using Positron Emission Tomography. Curr. Neurol. Neurosci. Rep. 2019, 19, 45.
  73. Ricci, M.; Cimini, A.; Camedda, R.; Chiaravalloti, A.; Schillaci, O. Tau biomarkers in dementia: Positron Emission Tomography radiopharmaceuticals in tauopathy assessment and future perspective. Int. J. Mol. Sci. 2021, 22, 13002.
  74. Rezabakhsh, A.; Rahbarghazi, R.; Fathi, F. Surface plasmon resonance biosensors for detection of Alzheimer’s biomarkers; an effective step in early and accurate diagnosis. Biosen. Bioelectron. 2020, 167, 112511.
  75. 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, 226–235.
  76. 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. Alzheimer’s Dis. 2018, 62, 1181–1198.
  77. Kang, D.-Y.; Lee, J.-H.; Oh, B.-K.; Choi, J.-W. Ultra-sensitive immunosensor for β-amyloid (1-42) using scanning tunneling microscopy-based electrical detection. Biosens. Bioelectron. 2009, 24, 1431–1436.
  78. 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 Aβ assembly forms throughout life. Neurobiol. Dis. 2009, 36, 293–302.
  79. 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, 147.
  80. Zhang, H.; Zhu, X.; Pascual, G.; Wadia, J.S.; Keogh, E.; Hoozemans, J.J.; Siregar, B.; Inganäs, H.; Stoop, E.J.M.; Goudsmit, J.; et al. Structural basis for recognition of a unique epitope by a human anti-tau antibody. Structure 2018, 26, 1626–1634.
  81. Zhou, J.; Jangili, P.; Son, S.; Ji, M.S.; Won, M.; Kim, J.S. Fluorescent diagnostic probes in neurodegenerative diseases. Adv. Mater. 2020, 32, 2001945.
  82. Lv, G.; Shen, Y.; Zheng, W.; Yang, J.; Li, C.; Lin, J. Fluorescence detection and dissociation of amyloid-βspecies for the treatment of Alzheimer’s disease. Adv. Ther. 2019, 2, 1900054.
  83. Liu, B.; Shen, H.; Hao, Y.; Zhu, X.; Li, S.; Huang, Y.; Qu, P.; Xu, M. Lanthanide functionalized metal-organic coordination polymer: Toward novel turn-on fluorescent sensing of amyloid β-peptide. Anal. Chem. 2018, 90, 12449–12455.
  84. Mir, M.; Palma-Florez, S.; Lagunas, A.; López-Martínez, M.J.; Samitier, J. Biosensors integration in blood–brain barrier-on-a-chip: Emerging platform for monitoring neurodegenerative diseases. ACS Sensors 2022, 7, 1237–1247.
  85. Negahdary, M.; Angnes, L. Electrochemical aptamer-based nanobiosensors for diagnosing Alzheimer’s disease: A review. Biomater. Adv. 2022, 135, 112689.
  86. Aggarwal, N.; Choudhury, S.; Chibh, S.; Panda, J.J. Aptamer-nanoconjugates as emerging theranostic systems in neurodegenerative disorders. Colloid Interface Sci. Commun. 2022, 46, 100554.
  87. Margiana, R.; Hammid, A.T.; Ahmad, I.; Alsaikhan, F.; Jalil, A.T.; Tursunbaev, F.; Umar, F.; Romero Parra, R.M.; Mustafa, Y.F. Current progress in aptasensor for ultra-low level monitoring of Parkinson’s disease biomarkers. Crit. Rev. Anal. Chem. 2022, 1–16.
  88. Taghdisi, S.M.; Danesh, N.M.; Nameghi, M.A.; Ramezani, M.; Alibolandi, M.; Hassanzadeh-Khayat, M.; Emrani, A.S.; Abnous, K. A novel electrochemical aptasensor based on nontarget-induced high accumulation of methylene blue on the surface of electrode for sensing of α-synuclein oligomer. Biosens. Bioelectron. 2019, 123, 14–18.
  89. Sun, K.; Xia, N.; Zhao, L.; Liu, K.; Hou, W.; Li, L. Aptasensors for the selective detection of alpha-synuclein oligomer by colorimetry, surface plasmon resonance and electrochemical impedance spectroscopy. Sens. Actuators B 2017, 245, 87–94.
  90. Jang, S.J.; Lee, C.-S.; Kim, T.H. α-Synuclein oligomer detection with aptamer switch on reduced graphene oxide electrode. Nanomaterials 2020, 10, 832.
  91. Tao, D.; Wang, J.; Song, S.; Cai, K.; Jiang, M.; Cheng, J.; Hu, L.; Jafrezic-Renault, N.; Guo, Z.; Pan, H. Polythionine and gold nanostar-based impedimetric aptasensor for label-free detection of α-synuclein oligomers. J. Appl. Electrochem. 2021, 51, 1523–1533.
  92. You, X.; Gopinath, S.C.B.; Lakshmipriya, T.; Li, D. High-affinity detection of Alpha-Synuclein by aptamer-gold conjugates on an amine-modified dielectric surface. J. Anal. Methods Chem. 2019, 2019, 6526850.
  93. 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.
  94. Zhou, Y.; Zhang, H.; Liu, L.; Li, C.; Chang, Z.; Zhu, X.; Ye, B.; Xu, M. Fabrication of an antibody-aptamer sandwich assay for electrochemical evaluation of levels of β-amyloid oligomers. Sci. Rep. 2016, 6, 35186.
  95. Zhou, Y.; Li, C.; Li, X.; Zhu, X.; Ye, B.; Xu, M. A sensitive aptasensor for detection of β-amyloid oligomers based on metal-organic frameworks as electrochemical signal probes. Anal. Methods 2018, 10, 4430–4437.
  96. Zhang, Y.; Figueroa-Miranda, G.; Lyu, Z.; Zafiu, C.; Willbold, D.; Offenhäusser, A.; Mayer, D. Monitoring amyloid-β proteins aggregation based on label-free aptasensor. Sens. Actuators B 2019, 288, 535–542.
  97. Zhou, Y.; Lv, Y.; Dong, H.; Liu, L.; Mao, G.; Zhang, Y.; Xu, M. Ultrasensitive assay of amyloid-beta oligomers using Au-vertical graphene/carbon cloth electrode based on poly(thymine)-templated copper nanoparticles as probes. Sens. Actuators B 2021, 331, 129429.
  98. Liao, X.; Ge, K.; Cai, Z.; Qiu, S.; Wu, S.; Li, Q.; Liu, Z.; Gao, F.; Tang, Q. Hybridization chain reaction triggered poly adenine to absorb silver nanoparticles for label-free electrochemical detection of Alzheimer’s disease biomarkers amyloid β-peptide oligomers. Anal. Chim. Acta 2022, 1192, 339391.
  99. Zhang, Y.; Figueroa-Miranda, G.; Wu, C.; Willbold, D.; Offenhäusser, A.; Mayer, D. Electrochemical dual-aptamer biosensors based on nanostructured multielectrode arrays for the detection of neuronal biomarkers. Nanoscale 2020, 12, 16501–16513.
  100. Tao, D.; Xie, C.; Fu, S.; Rong, S.; Song, S.; Ye, H.; Jaffrezic-Renault, N.; Guo, Z. Thionine-functionalized three-dimensional carbon nanomaterial based aptasensor for analysis of Aβ oligomers in serum. Anal. Chim. Acta 2021, 1183, 338990.
  101. Zhang, Y.; Figueroa-Miranda, G.; Zafiu, C.; Willbold, D.; Offenhäusser, A.; Mayer, D. Amperometric aptasensor for Amyloid β oligomer detection by optimized stem-loop structures with an adjustable detection range. ACS Sens. 2019, 4, 3042–3050.
  102. Wang, X.; Gu, X.; Li, L.; Yu, B.; Lv, L.; Chen, Q.; Xu, M. An excellent electrochemical aptasensor for amyloid-β oligomers based on a triple-helix aptamer switch via target-triggered signal transduction DNA displacement events. Anal. Bioanal. Chem. 2021, 413, 3707–3716.
  103. Wang, X.; Li, L.; Gu, X.; Yu, B.; Jiang, M. Switchable electrochemical aptasensor for amyloid-β oligomers detection based on triple helix switch coupling with labeled signaling displaced-probe. Microchim. Acta 2021, 188, 49.
  104. You, M.; Yang, S.; An, Y.; Zhang, F.; He, P. A novel electrochemical biosensor with molecularly imprinted polymers and aptamer-based sandwich assay for determining amyloid-β oligomer. J. Electroanal. Chem. 2020, 862, 114017.
  105. Liu, Y.; Xu, Q.; Zhang, Y.; Ren, B.; Huang, L.; Cai, H.; Xu, T.; Liu, Q.; Zhang, X. An electrochemical aptasensor based on AuPt alloy nanoparticles for ultrasensitive detection of amyloid-β oligomers. Talanta 2021, 231, 122360.
  106. Tri Murti, B.; Huang, Y.-J.; Darumas Putri, A.; Lee, C.-P.; Hsieh, C.-M.; Wei, S.-M.; Tsai, M.-L.; Peng, C.-W.; Yang, P.-K. Free-standing vertically aligned tin disulfide nanosheets for ultrasensitive aptasensor design toward Alzheimer’s diagnosis applications. Chem. Eng. J. 2023, 452, 139394.
  107. Deng, C.; Liu, H.; Si, S.; Zhu, X.; Tu, Q.; Jin, Y.; Xiang, J. An electrochemical aptasensor for amyloid-β oligomer based on double-stranded DNA as “conductive spring”. Microchim. Acta 2020, 187, 239.
  108. Tao, D.; Shui, B.; Gu, Y.; Cheng, J.; Zhang, W.; Jaffrezic-Renault, N.; Song, S.; Guo, Z. Development of a label-free electrochemical aptasensor for the detection of Tau381 and its preliminary application in AD and non-AD patients’ sera. Biosensors 2019, 9, 84.
  109. Shui, B.; Tao, D.; Cheng, J.; Mei, Y.; Jaffrezic-Renault, N.; Guo, Z. Novel electrochemical aptamer-antibody sandwich assay for the detection of tau-381 in human serum. Analyst 2018, 143, 3549–3554.
  110. Yin, M.; Xu, D.; Yu, J.; Huang, S.; Gopinath, S.C.B.; Kang, P. Impedance spectroscopy for identifying tau protein to monitor anesthesia-based issues. Biotechnol. Appl. Biochem. 2022, 69, 1805–1811.
  111. Liu, Y.; Liu, X.; Li, M.; Liu, Q.; Xu, T. Portable vertical electrochemical aptasensing platform for point-of-care testing of Tau protein in the blood. Biosensors 2022, 12, 564.
  112. Miodek, A.; Castillo, G.; Hianik, T.; Korri-Youssoufi, H. Electrochemical aptasensor of cellular prion protein based on modified polypyrrole with redox dendrimers. Biosens. Bioelectron. 2014, 56, 104–111.
  113. Miodek, A.; Castillo, G.; Hianik, T.; Korri-Youssoufi, H. Electrochemical aptasensor of human cellular prion based on multiwalled carbon nanotubes modified with dendrimers: A platform for connecting redox markers and aptamers. Anal Chem. 2013, 85, 7704–7712.
  114. Yu, P.; Zhang, X.; Zhou, J.; Xiong, E.; Li, X.; Chen, J. Smart protein biogate as a mediator to regulate competitive host-guest interaction for sensitive ratiometric electrochemical assay of prion. Sci. Rep. 2015, 5, 16015.
  115. Yu, P.; Zhang, X.; Xiong, E.; Zhou, J.; Li, X.; Chen, J. A label-free and cascaded dual-signaling amplified electrochemical aptasensing platform for sensitive prion assay. Biosens. Bioelectron. 2016, 85, 471–478.
More
Video Production Service