1
|
Schuler M, Gerwert G, Mann M, Woitzik N, Langenhoff L, Hubert D, Duman D, Höveler A, Galkowski S, Simon J, Denz R, Weber S, Kwon EH, Wanka R, Kötting C, Güldenhaupt J, Beyer L, Tönges L, Mollenhauer B, Gerwert K. Alpha-synuclein misfolding as fluid biomarker for Parkinson's disease measured with the iRS platform. EMBO Mol Med 2025:10.1038/s44321-025-00229-z. [PMID: 40281305 DOI: 10.1038/s44321-025-00229-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Misfolding and aggregation of alpha-synuclein (αSyn) play a key role in the pathophysiology of Parkinson's disease (PD). Despite considerable advances in diagnostics, an early and differential diagnosis of PD still represents a major challenge. We innovated the immuno-infrared sensor (iRS) platform for measuring αSyn misfolding. We analyzed cerebrospinal fluid (CSF) from two cohorts comprising PD cases, atypical Parkinsonian disorders, and disease controls. We obtained an AUC of 0.90 (n = 134, 95% CI 0.85-0.96) for separating PD/MSA from controls by determination of the αSyn misfolding by iRS. Using two thresholds divided individuals as unaffected/affected by misfolding with an intermediate area in between. Comparing the affected/unaffected cases, controls versus PD/MSA cases were classified with 97% sensitivity and 92% specificity. The spectral data revealed misfolding from an α-helical/random-coil αSyn in controls to β-sheet enriched αSyn in PD and MSA cases. Moreover, a first subgroup analysis implied the potential for patient stratification in clinically overlapping cases. The iRS, directly measuring all αSyn conformers, is complementary to the αSyn seed-amplification assays (SAAs), which however only amplify seeding competent conformers.
Collapse
Affiliation(s)
- Martin Schuler
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Grischa Gerwert
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Marvin Mann
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Nathalie Woitzik
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Lennart Langenhoff
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Diana Hubert
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Deniz Duman
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Adrian Höveler
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Sandy Galkowski
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Jonas Simon
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Robin Denz
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr-University Bochum, Bochum, Germany
| | - Sandrina Weber
- University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
| | - Eun-Hae Kwon
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Robin Wanka
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Carsten Kötting
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Jörn Güldenhaupt
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Léon Beyer
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Lars Tönges
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Brit Mollenhauer
- University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
- Paracelsus-Elena-Klinik, Klinikstraße 16, 34128, Kassel, Germany
- Scientific employee with an honorary contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Klaus Gerwert
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany.
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany.
| |
Collapse
|
2
|
Lista S, Mapstone M, Caraci F, Emanuele E, López-Ortiz S, Martín-Hernández J, Triaca V, Imbimbo C, Gabelle A, Mielke MM, Nisticò R, Santos-Lozano A, Imbimbo BP. A critical appraisal of blood-based biomarkers for Alzheimer's disease. Ageing Res Rev 2024; 96:102290. [PMID: 38580173 DOI: 10.1016/j.arr.2024.102290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/18/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024]
Abstract
Biomarkers that predict the clinical onset of Alzheimer's disease (AD) enable the identification of individuals in the early, preclinical stages of the disease. Detecting AD at this point may allow for more effective therapeutic interventions and optimized enrollment for clinical trials of novel drugs. The current biological diagnosis of AD is based on the AT(N) classification system with the measurement of brain deposition of amyloid-β (Aβ) ("A"), tau pathology ("T"), and neurodegeneration ("N"). Diagnostic cut-offs for Aβ1-42, the Aβ1-42/Aβ1-40 ratio, tau and hyperphosphorylated-tau concentrations in cerebrospinal fluid have been defined and may support AD clinical diagnosis. Blood-based biomarkers of the AT(N) categories have been described in the AD continuum. Cross-sectional and longitudinal studies have shown that the combination of blood biomarkers tracking neuroaxonal injury (neurofilament light chain) and neuroinflammatory pathways (glial fibrillary acidic protein) enhance sensitivity and specificity of AD clinical diagnosis and improve the prediction of AD onset. However, no international accepted cut-offs have been identified for these blood biomarkers. A kit for blood Aβ1-42/Aβ1-40 is commercially available in the U.S.; however, it does not provide a diagnosis, but simply estimates the risk of developing AD. Although blood-based AD biomarkers have a great potential in the diagnostic work-up of AD, they are not ready for the routine clinical use.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Mark Mapstone
- Department of Neurology, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA.
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania 95125, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, Troina 94018, Italy.
| | | | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Rome 00015, Italy.
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy.
| | - Audrey Gabelle
- Memory Resources and Research Center, Montpellier University of Excellence i-site, Montpellier 34295, France.
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA.
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", Rome 00133, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome 00143, Italy.
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain; Physical Activity and Health Research Group (PaHerg), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid 28041, Spain.
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma 43122, Italy.
| |
Collapse
|
3
|
Kavungal D, Magalhães P, Kumar ST, Kolla R, Lashuel HA, Altug H. Artificial intelligence-coupled plasmonic infrared sensor for detection of structural protein biomarkers in neurodegenerative diseases. SCIENCE ADVANCES 2023; 9:eadg9644. [PMID: 37436975 PMCID: PMC10337894 DOI: 10.1126/sciadv.adg9644] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
Diagnosis of neurodegenerative disorders (NDDs) including Parkinson's disease and Alzheimer's disease is challenging owing to the lack of tools to detect preclinical biomarkers. The misfolding of proteins into oligomeric and fibrillar aggregates plays an important role in the development and progression of NDDs, thus underscoring the need for structural biomarker-based diagnostics. We developed an immunoassay-coupled nanoplasmonic infrared metasurface sensor that detects proteins linked to NDDs, such as alpha-synuclein, with specificity and differentiates the distinct structural species using their unique absorption signatures. We augmented the sensor with an artificial neural network enabling unprecedented quantitative prediction of oligomeric and fibrillar protein aggregates in their mixture. The microfluidic integrated sensor can retrieve time-resolved absorbance fingerprints in the presence of a complex biomatrix and is capable of multiplexing for the simultaneous monitoring of multiple pathology-associated biomarkers. Thus, our sensor is a promising candidate for the clinical diagnosis of NDDs, disease monitoring, and evaluation of novel therapies.
Collapse
Affiliation(s)
- Deepthy Kavungal
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Pedro Magalhães
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Senthil T. Kumar
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Rajasekhar Kolla
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Hilal A. Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Institute of Bioengineering, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Hatice Altug
- Bionanophotonic Systems Laboratory, Institute of Bioengineering, School of Engineering, EPFL, Lausanne, Switzerland
| |
Collapse
|
4
|
Beyer L, Stocker H, Rujescu D, Holleczek B, Stockmann J, Nabers A, Brenner H, Gerwert K. Amyloid-beta misfolding and GFAP predict risk of clinical Alzheimer's disease diagnosis within 17 years. Alzheimers Dement 2023; 19:1020-1028. [PMID: 35852967 DOI: 10.1002/alz.12745] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/29/2022] [Accepted: 06/10/2022] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Blood-based biomarkers for Alzheimer's disease (AD) are urgently needed. Here, four plasma biomarkers were measured at baseline in a community-based cohort followed over 17 years, and the association with clinical AD risk was determined. METHODS Amyloid beta (Aβ) misfolding status as a structure-based biomarker as well as phosphorylated tau 181 (P-tau181), glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) concentration levels were determined at baseline in heparin plasma from 68 participants who were diagnosed with AD and 240 controls without dementia diagnosis throughout follow-up. RESULTS Aβ misfolding exhibited high disease prediction accuracy of AD diagnosis within 17 years. Among the concentration markers, GFAP showed the best performance, followed by NfL and P-tau181. The combination of Aβ misfolding and GFAP increased the accuracy. DISCUSSION Aβ misfolding and GFAP showed a strong ability to predict clinical AD risk and may be important early AD risk markers. Aβ misfolding illustrated its potential as a prescreening tool for AD risk stratification in older adults.
Collapse
Affiliation(s)
- Léon Beyer
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Hannah Stocker
- Network Aging Research, Heidelberg University, Heidelberg, Germany
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Dan Rujescu
- Department of Psychiatry, Medical University of Vienna, Vienna, Austria
| | | | - Julia Stockmann
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Nabers
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Hermann Brenner
- Network Aging Research, Heidelberg University, Heidelberg, Germany
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Klaus Gerwert
- Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
5
|
Nazari MT, Schnorr C, Rigueto CVT, Alessandretti I, Melara F, da Silva NF, Crestani L, Ferrari V, Vieillard J, Dotto GL, Silva LFO, Piccin JS. A review of the main methods for composite adsorbents characterization. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:88488-88506. [PMID: 36334205 DOI: 10.1007/s11356-022-23883-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
Adsorption is a promising technology for removing several contaminants from aqueous matrices. In the last years, researchers worldwide have been working on developing composite adsorbents to overcome some limitations and drawbacks of conventional adsorbent materials, which depend on various factors, including the characteristics of the adsorbents. Therefore, it is essential to characterize the composite adsorbents to describe their properties and structure and elucidate the mechanisms, behavior, and phenomenons during the adsorption process. In this sense, this work aimed to review the main methods used for composite adsorbent characterization, providing valuable information on the importance of these techniques in developing new adsorbents. In this paper, we reviewed the following methods: X-Ray diffraction (XRD); spectroscopy; scanning electron microscopy (SEM); N2 adsorption/desorption isotherms (BET and BJH methods); thermogravimetry (TGA); point of zero charge (pHPZC); elemental analysis; proximate analysis; swelling and water retention capacities; desorption and reuse.
Collapse
Affiliation(s)
- Mateus T Nazari
- Graduate Program in Civil and Environmental Engineering (PPGEng), University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | - Carlos Schnorr
- Universidad De La Costa, Calle 58 # 55-66, 080002, Barranquilla, Atlántico, Colombia
| | - Cesar V T Rigueto
- Graduate Program in Food Science and Technology (PPGCTA), Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Ingridy Alessandretti
- Graduate Program in Food Science and Technology (PPGCTA), University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | - Flávia Melara
- Graduate Program in Civil and Environmental Engineering (PPGEng), University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| | - Nathália F da Silva
- Research Group On Adsorptive and Catalytic Process Engineering (ENGEPAC), Federal University of Santa Maria, Av. Roraima, 1000-7, Santa Maria, RS, 97105-900, Brazil
| | - Larissa Crestani
- Research Group On Adsorptive and Catalytic Process Engineering (ENGEPAC), Federal University of Santa Maria, Av. Roraima, 1000-7, Santa Maria, RS, 97105-900, Brazil
| | - Valdecir Ferrari
- Graduate Program in Metallurgical, Materials, and Mining Engineering (PPG3M), Federal University of Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Julien Vieillard
- CNRS, INSA Rouen, UNIROUEN, COBRA (UMR 6014 and FR 3038), Normandie University, Evreux, France
| | - Guilherme L Dotto
- Research Group On Adsorptive and Catalytic Process Engineering (ENGEPAC), Federal University of Santa Maria, Av. Roraima, 1000-7, Santa Maria, RS, 97105-900, Brazil.
| | - Luis F O Silva
- Universidad De La Costa, Calle 58 # 55-66, 080002, Barranquilla, Atlántico, Colombia
| | - Jeferson S Piccin
- Graduate Program in Civil and Environmental Engineering (PPGEng), University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
- Graduate Program in Food Science and Technology (PPGCTA), University of Passo Fundo (UPF), Passo Fundo, RS, Brazil
| |
Collapse
|
6
|
ATR-FTIR Biosensors for Antibody Detection and Analysis. Int J Mol Sci 2022; 23:ijms231911895. [PMID: 36233197 PMCID: PMC9570191 DOI: 10.3390/ijms231911895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
Quality control of drug products is of paramount importance in the pharmaceutical world. It ensures product safety, efficiency, and consistency. In the case of complex biomolecules such as therapeutic proteins, small variations in bioprocess parameters can induce substantial variations in terms of structure, impacting the drug product quality. Conditions for obtaining highly reproducible grafting of 11-mercaptoundecanoic acid were determined. On that basis, we developed an easy-to-use, cost effective, and timesaving biosensor based on ATR-FTIR spectroscopy able to detect immunoglobulins during their production. A germanium crystal, used as an internal reflection element (IRE) for FTIR spectroscopy, was covalently coated with immunoglobulin-binding proteins. This thereby functionalized surface could bind only immunoglobulins present in complex media such as culture media or biopharmaceutical products. The potential subsequent analysis of their structure by ATR-FTIR spectroscopy makes this biosensor a powerful tool to monitor the production of biotherapeutics and assess important critical quality attributes (CQAs) such as high-order structure and aggregation level.
Collapse
|
7
|
Applications of Single-Molecule Vibrational Spectroscopic Techniques for the Structural Investigation of Amyloid Oligomers. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196448. [PMID: 36234985 PMCID: PMC9573641 DOI: 10.3390/molecules27196448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022]
Abstract
Amyloid oligomeric species, formed during misfolding processes, are believed to play a major role in neurodegenerative and metabolic diseases. Deepening the knowledge about the structure of amyloid intermediates and their aggregation pathways is essential in understanding the underlying mechanisms of misfolding and cytotoxicity. However, structural investigations are challenging due to the low abundance and heterogeneity of those metastable intermediate species. Single-molecule techniques have the potential to overcome these difficulties. This review aims to report some of the recent advances and applications of vibrational spectroscopic techniques for the structural analysis of amyloid oligomers, with special focus on single-molecule studies.
Collapse
|
8
|
Aitekenov S, Sultangaziyev A, Abdirova P, Yussupova L, Gaipov A, Utegulov Z, Bukasov R. Raman, Infrared and Brillouin Spectroscopies of Biofluids for Medical Diagnostics and for Detection of Biomarkers. Crit Rev Anal Chem 2022; 53:1561-1590. [PMID: 35157535 DOI: 10.1080/10408347.2022.2036941] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
This review surveys Infrared, Raman/SERS and Brillouin spectroscopies for medical diagnostics and detection of biomarkers in biofluids, that include urine, blood, saliva and other biofluids. These optical sensing techniques are non-contact, noninvasive and relatively rapid, accurate, label-free and affordable. However, those techniques still have to overcome some challenges to be widely adopted in routine clinical diagnostics. This review summarizes and provides insights on recent advancements in research within the field of vibrational spectroscopy for medical diagnostics and its use in detection of many health conditions such as kidney injury, cancers, cardiovascular and infectious diseases. The six comprehensive tables in the review and four tables in supplementary information summarize a few dozen experimental papers in terms of such analytical parameters as limit of detection, range, diagnostic sensitivity and specificity, and other figures of merits. Critical comparison between SERS and FTIR methods of analysis reveals that on average the reported sensitivity for biomarkers in biofluids for SERS vs FTIR is about 103 to 105 times higher, since LOD SERS are lower than LOD FTIR by about this factor. High sensitivity gives SERS an edge in detection of many biomarkers present in biofluids at low concentration (nM and sub nM), which can be particularly advantageous for example in early diagnostics of cancer or viral infections.HighlightsRaman, Infrared spectroscopies use low volume of biofluidic samples, little sample preparation, fast time of analysis and relatively inexpensive instrumentation.Applications of SERS may be a bit more complicated than applications of FTIR (e.g., limited shelf life for nanoparticles and substrates, etc.), but this can be generously compensated by much higher (by several order of magnitude) sensitivity in comparison to FTIR.High sensitivity makes SERS a noninvasive analytical method of choice for detection, quantification and diagnostics of many health conditions, metabolites, and drugs, particularly in diagnostics of cancer, including diagnostics of its early stages.FTIR, particularly ATR-FTIR can be a method of choice for efficient sensing of many biomarkers, present in urine, blood and other biofluids at sufficiently high concentrations (mM and even a few µM)Brillouin scattering spectroscopy detecting visco-elastic properties of probed liquid medium, may also find application in clinical analysis of some biofluids, such as cerebrospinal fluid and urine.
Collapse
Affiliation(s)
- Sultan Aitekenov
- Department of Chemistry, School of Sciences and Humanities (SSH), Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Alisher Sultangaziyev
- Department of Chemistry, School of Sciences and Humanities (SSH), Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Perizat Abdirova
- Department of Chemistry, School of Sciences and Humanities (SSH), Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Lyailya Yussupova
- Department of Chemistry, School of Sciences and Humanities (SSH), Nazarbayev University, Nur-Sultan, Kazakhstan
| | | | - Zhandos Utegulov
- Department of Physics, School of Sciences and Humanities (SSH), Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Rostislav Bukasov
- Department of Chemistry, School of Sciences and Humanities (SSH), Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
9
|
Stocker H, Nabers A, Perna L, Möllers T, Rujescu D, Hartmann AM, Holleczek B, Schöttker B, Stockmann J, Gerwert K, Brenner H. Genetic predisposition, Aβ misfolding in blood plasma, and Alzheimer's disease. Transl Psychiatry 2021; 11:261. [PMID: 33934115 PMCID: PMC8088439 DOI: 10.1038/s41398-021-01380-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease is highly heritable and characterized by amyloid plaques and tau tangles in the brain. The aim of this study was to investigate the association between genetic predisposition, Aβ misfolding in blood plasma, a unique marker of Alzheimer associated neuropathological changes, and Alzheimer's disease occurrence within 14 years. Within a German community-based cohort, two polygenic risk scores (clinical Alzheimer's disease and Aβ42 based) were calculated, APOE genotype was determined, and Aβ misfolding in blood plasma was measured by immuno-infrared sensor in 59 participants diagnosed with Alzheimer's disease during 14 years of follow-up and 581 participants without dementia diagnosis. Associations between each genetic marker and Aβ misfolding were assessed through logistic regression and the ability of each genetic marker and Aβ misfolding to predict Alzheimer's disease was determined. The Alzheimer's disease polygenic risk score and APOE ε4 presence were associated to Aβ misfolding (odds ratio, 95% confidence interval: per standard deviation increase of score: 1.25, 1.03-1.51; APOE ε4 presence: 1.61, 1.04-2.49). No association was evident for the Aβ polygenic risk score. All genetic markers were predictive of Alzheimer's disease diagnosis albeit much less so than Aβ misfolding (areas under the curve: Aβ polygenic risk score: 0.55; AD polygenic risk score: 0.59; APOE ε4: 0.63; Aβ misfolding: 0.84). Clinical Alzheimer's genetic risk was associated to early pathological changes (Aβ misfolding) measured in blood, however, predicted Alzheimer's disease less accurately than Aβ misfolding itself. Genetic predisposition may provide information regarding disease initiation, while Aβ misfolding could be important in clinical risk prediction.
Collapse
Affiliation(s)
- Hannah Stocker
- Network Aging Research, Heidelberg University, Heidelberg, Germany.
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany.
- Medical Faculty, Heidelberg University, Heidelberg, Germany.
| | - Andreas Nabers
- Department of Biophysics, Competence Center for Biospectroscopy, Ruhr-University Bochum, Bochum, Germany
- Faculty of Biology and Biotechnology, Department of Biophysics, Ruhr University Bochum, Bochum, Germany
| | - Laura Perna
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Tobias Möllers
- Network Aging Research, Heidelberg University, Heidelberg, Germany
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Dan Rujescu
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Halle, Halle, Germany
| | - Annette M Hartmann
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Halle, Halle, Germany
| | | | - Ben Schöttker
- Network Aging Research, Heidelberg University, Heidelberg, Germany
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Julia Stockmann
- Department of Biophysics, Competence Center for Biospectroscopy, Ruhr-University Bochum, Bochum, Germany
- Faculty of Biology and Biotechnology, Department of Biophysics, Ruhr University Bochum, Bochum, Germany
| | - Klaus Gerwert
- Department of Biophysics, Competence Center for Biospectroscopy, Ruhr-University Bochum, Bochum, Germany
- Faculty of Biology and Biotechnology, Department of Biophysics, Ruhr University Bochum, Bochum, Germany
| | - Hermann Brenner
- Network Aging Research, Heidelberg University, Heidelberg, Germany
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
10
|
Mayerhöfer TG, Pahlow S, Popp J. Recent technological and scientific developments concerning the use of infrared spectroscopy for point-of-care applications. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 251:119411. [PMID: 33450450 DOI: 10.1016/j.saa.2020.119411] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 06/12/2023]
Abstract
In this contribution we review selected point-of-care applications of infrared spectroscopy and the technological innovations they are based on. After a short introduction summarizing the general idea behind point-of-care applications we introduce the reader to important infrared spectroscopy sensing principles on a very basic level. We discuss the role of optical components like quantum cascade lasers, supercontinuum sources, waveguides and how they are potentially going to revolutionize point-of-care applications. First, we focus on the technological solutions of some principal problems like increasing the pathlength in a transmission cell to enhance the sensitivity for solutes in aqueous solutions and discuss indirect methods which circumvent the problem of low transmittance. In the second part we show how the technological progress of the last decades enabled scientific progress leading to selected concrete and outstanding point-of-care solutions and applications based on infrared spectroscopy. These include the detection and quantification of malaria parasitemia, early recognition of Alzheimer's disease long before the onset of clinical symptoms and a non-invasive method for testing the blood glucose content. The selected examples demonstrate and showcase that infrared spectroscopy is on the way to become an indispensable technique for point-of-care applications.
Collapse
Affiliation(s)
- Thomas G Mayerhöfer
- Leibniz Institute of Photonic Technology (IPHT), Albert-Einstein-Str. 9, D-07745 Jena, Germany; Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Jena, D-07743, Helmholtzweg 4, Germany
| | - Susanne Pahlow
- Leibniz Institute of Photonic Technology (IPHT), Albert-Einstein-Str. 9, D-07745 Jena, Germany; Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Jena, D-07743, Helmholtzweg 4, Germany
| | - Jürgen Popp
- Leibniz Institute of Photonic Technology (IPHT), Albert-Einstein-Str. 9, D-07745 Jena, Germany; Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University, Jena, D-07743, Helmholtzweg 4, Germany.
| |
Collapse
|
11
|
Stockmann J, Verberk IMW, Timmesfeld N, Denz R, Budde B, Lange-Leifhelm J, Scheltens P, van der Flier WM, Nabers A, Teunissen CE, Gerwert K. Amyloid-β misfolding as a plasma biomarker indicates risk for future clinical Alzheimer's disease in individuals with subjective cognitive decline. Alzheimers Res Ther 2020; 12:169. [PMID: 33357241 PMCID: PMC7761044 DOI: 10.1186/s13195-020-00738-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND We evaluated Aβ misfolding in combination with Aβ42/40 ratio as a prognostic tool for future clinical progression to mild cognitive impairment (MCI) or dementia due to Alzheimer's disease (AD) in individuals with subjective cognitive decline (SCD). METHODS Baseline plasma samples (n = 203) from SCD subjects in the SCIENCe project and Amsterdam Dementia Cohort (age 61 ± 9 years; 57% male, mean follow-up time 2.7 years) were analyzed using immuno-infrared-sensor technology. Within 6 years of follow-up, 22 (11%) individuals progressed to MCI or dementia due to AD. Sensor readout values > 1646 cm- 1 reflected normal Aβ folding; readouts at ≤ 1646 cm- 1 reflected low and at < 1644 cm- 1 high misfolding. We used Cox proportional hazard models to quantify Aβ misfolding as a prognostic biomarker for progression to MCI and dementia due to AD. The accuracy of the predicted development of MCI/AD was determined by time-dependent receiver operating characteristic (t-ROC) curve analyses that take individual follow-up and conversion times into account. Statistical models were adjusted for age, sex, and APOEε4 status. Additionally, plasma Aβ42/40 data measured by SIMOA were statistically analyzed and compared. RESULTS All 22 patients who converted to MCI or AD-dementia within 6 years exhibited Aβ misfolding at baseline. Cox analyses revealed a hazard ratio (HR) of 19 (95% confidence interval [CI] 2.2-157.8) for future conversion of SCD subjects with high misfolding and of 11 (95% CI 1.0-110.1) for those with low misfolding. T-ROC curve analyses yielded an area under the curve (AUC) of 0.94 (95% CI 0.86-1.00; 6-year follow-up) for Aβ misfolding in an age, sex, and APOEε4 model. A similar model with plasma Aβ42/40 ratio yielded an AUC of 0.92 (95% CI, 0.82-1.00). The AUC increased to 0.99 (95% CI, 0.99-1.00) after inclusion of both Aβ misfolding and the Aβ42/40 ratio. CONCLUSIONS A panel of structure- and concentration-based plasma amyloid biomarkers may predict conversion to clinical MCI and dementia due to AD in cognitively unimpaired subjects. These plasma biomarkers provide a noninvasive and cost-effective alternative for screening early AD pathological changes. Follow-up studies and external validation in larger cohorts are in progress for further validation of our findings.
Collapse
Affiliation(s)
- Julia Stockmann
- Competence Center for Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr University Bochum, Faculty of Biology and Biotechnology, Bochum, Germany
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Nina Timmesfeld
- Ruhr University Bochum, Department of Medical Informatics, Biometry and Epidemiology, Bochum, Germany
| | - Robin Denz
- Ruhr University Bochum, Department of Medical Informatics, Biometry and Epidemiology, Bochum, Germany
| | - Brian Budde
- Competence Center for Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr University Bochum, Faculty of Biology and Biotechnology, Bochum, Germany
| | - Julia Lange-Leifhelm
- Competence Center for Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr University Bochum, Faculty of Biology and Biotechnology, Bochum, Germany
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Andreas Nabers
- Competence Center for Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
- Department of Biophysics, Ruhr University Bochum, Faculty of Biology and Biotechnology, Bochum, Germany
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Klaus Gerwert
- Competence Center for Biospectroscopy, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany.
- Department of Biophysics, Ruhr University Bochum, Faculty of Biology and Biotechnology, Bochum, Germany.
| |
Collapse
|
12
|
Beyer L, Günther R, Koch JC, Klebe S, Hagenacker T, Lingor P, Biesalski AS, Hermann A, Nabers A, Gold R, Tönges L, Gerwert K. TDP-43 as structure-based biomarker in amyotrophic lateral sclerosis. Ann Clin Transl Neurol 2020; 8:271-277. [PMID: 33263951 PMCID: PMC7818221 DOI: 10.1002/acn3.51256] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/04/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022] Open
Abstract
Pathologic alterations of Transactivation response DNA‐binding protein 43 kilo Dalton (TDP‐43) are a major hallmark of amyotrophic lateral sclerosis (ALS). In this pilot study, we analyzed the secondary structure distribution of TDP‐43 in cerebrospinal fluid of ALS patients (n = 36) compared to Parkinson´s disease patients (PD; n = 30) and further controls (Ctrl; n = 24) using the immuno‐infrared sensor technology. ALS patients could be discriminated from PD and Ctrl with a sensitivity/specificity of 89 %/77 % and 89 %/83 %, respectively. Our findings demonstrate that TDP‐43 misfolding measured by the immuno‐infrared sensor technology has the potential to serve as a biomarker candidate for ALS.
Collapse
Affiliation(s)
- Léon Beyer
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, Bochum, Germany.,Faculty of Biology and Biotechnology, Department of Biophysics, Ruhr University Bochum, Bochum, Germany
| | - René Günther
- Department of Neurology, Technische Universität Dresden, Dresden, Germany.,German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Jan Christoph Koch
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany
| | - Stephan Klebe
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Tim Hagenacker
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Paul Lingor
- Department of Neurology, University Medicine Göttingen, Göttingen, Germany.,School of Medicine, Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Munich, Germany
| | - Anne-Sophie Biesalski
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section "Albrecht-Kossel", Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Andreas Nabers
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, Bochum, Germany.,Faculty of Biology and Biotechnology, Department of Biophysics, Ruhr University Bochum, Bochum, Germany
| | - Ralf Gold
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, Bochum, Germany.,Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Lars Tönges
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, Bochum, Germany.,Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Klaus Gerwert
- Center for Protein Diagnostics (ProDi), Ruhr University Bochum, Bochum, Germany.,Faculty of Biology and Biotechnology, Department of Biophysics, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
13
|
Agbaria AH, Beck G, Lapidot I, Rich DH, Kapelushnik J, Mordechai S, Salman A, Huleihel M. Diagnosis of inaccessible infections using infrared microscopy of white blood cells and machine learning algorithms. Analyst 2020; 145:6955-6967. [PMID: 32852502 DOI: 10.1039/d0an00752h] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Physicians diagnose subjectively the etiology of inaccessible infections where sampling is not feasible (such as, pneumonia, sinusitis, cholecystitis, peritonitis), as bacterial or viral. The diagnosis is based on their experience with some medical markers like blood counts and medical symptoms since it is harder to obtain swabs and reliable laboratory results for most cases. In this study, infrared spectroscopy with machine learning algorithms was used for the rapid and objective diagnosis of the etiology of inaccessible infections and enables an assessment of the error for the subjective diagnosis of the etiology of these infections by physicians. Our approach allows for diagnoses of the etiology of both accessible and inaccessible infections as based on an analysis of the innate immune system response through infrared spectroscopy measurements of white blood cell (WBC) samples. In the present study, we examined 343 individuals involving 113 controls, 89 inaccessible bacterial infections, 54 accessible bacterial infections, 60 inaccessible viral infections, and 27 accessible viral infections. Using our approach, the results show that it is possible to differentiate between controls and infections (combined bacterial and viral) with 95% accuracy, and enabling the diagnosis of the etiology of accessible infections as bacterial or viral with >94% sensitivity and > 90% specificity within one hour after the collection of the blood sample with error rate <6%. Based on our approach, the error rate of the physicians' subjective diagnosis of the etiology of inaccessible infections was found to be >23%.
Collapse
Affiliation(s)
- Adam H Agbaria
- Department of Physics, Ben-Gurion University, Beer-Sheva 84105, Israel
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Blond P, Bevernaegie R, Troian-Gautier L, Lagrost C, Hubert J, Reniers F, Raussens V, Jabin I. Ready-to-Use Germanium Surfaces for the Development of FTIR-Based Biosensors for Proteins. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:12068-12076. [PMID: 33007158 DOI: 10.1021/acs.langmuir.0c02681] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Germanium is particularly suitable for the design of FTIR-based biosensors for proteins. The grafting of stable and thin organic layers on germanium surfaces remains, however, challenging. To tackle this problem, we developed a calix[4]arene-tetradiazonium salt decorated with four oligo(ethylene glycol) chains and a terminal reactive carboxyl group. This versatile molecular platform was covalently grafted on germanium surfaces to yield robust ready-to-use surfaces for biosensing applications. The grafted calixarene monolayer prevents nonspecific adsorption of proteins while allowing bioconjugation with biomolecules such as bovine serum albumin (BSA) or biotin. It is shown that the native form of the investigated proteins was maintained upon immobilization. As a proof of concept, the resulting calix[4]arene-based germanium biosensors were used through a combination of ATR-FTIR spectroscopy and fluorescence microscopy for the selective detection of streptavidin from a complex medium. This study opens real possibilities for the development of sensitive and selective FTIR-based biosensors devoted to the detection of proteins.
Collapse
Affiliation(s)
- Pascale Blond
- Laboratoire de Chimie Organique, Université libre de Bruxelles (ULB), Avenue F. D. Roosevelt 50, CP160/06, B-1050 Brussels, Belgium
- Laboratory for the Structure and Function of Biological Membranes, Centre for Structural Biology and Bioinformatics, Université libre de Bruxelles (ULB), Boulevard du Triomphe, CP206/02, B-1050 Brussels, Belgium
| | - Robin Bevernaegie
- Laboratoire de Chimie Organique, Université libre de Bruxelles (ULB), Avenue F. D. Roosevelt 50, CP160/06, B-1050 Brussels, Belgium
| | - Ludovic Troian-Gautier
- Laboratoire de Chimie Organique, Université libre de Bruxelles (ULB), Avenue F. D. Roosevelt 50, CP160/06, B-1050 Brussels, Belgium
| | | | - Julie Hubert
- Chemistry of Surfaces, Interfaces and Nanomaterials, Université libre de Bruxelles (ULB), Boulevard du Triomphe, CP 255, B-1050 Brussels, Belgium
| | - François Reniers
- Chemistry of Surfaces, Interfaces and Nanomaterials, Université libre de Bruxelles (ULB), Boulevard du Triomphe, CP 255, B-1050 Brussels, Belgium
| | - Vincent Raussens
- Laboratory for the Structure and Function of Biological Membranes, Centre for Structural Biology and Bioinformatics, Université libre de Bruxelles (ULB), Boulevard du Triomphe, CP206/02, B-1050 Brussels, Belgium
| | - Ivan Jabin
- Laboratoire de Chimie Organique, Université libre de Bruxelles (ULB), Avenue F. D. Roosevelt 50, CP160/06, B-1050 Brussels, Belgium
| |
Collapse
|
15
|
The Effect of (-)-Epigallocatechin-3-Gallate on the Amyloid-β Secondary Structure. Biophys J 2020; 119:349-359. [PMID: 32579965 DOI: 10.1016/j.bpj.2020.05.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/20/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023] Open
Abstract
Amyloid-β (Aβ) is a macromolecular structure of great interest because its misfolding and aggregation, along with changes in the secondary structure, have been correlated with its toxicity in various neurodegenerative diseases. Small drug-like molecules can modulate the amyloid secondary structure and therefore have raised significant interest in applications to active and passive therapies targeting amyloids. In this study, we investigate the interactions of epigallocatechin-3-gallate (EGCG), found in green tea, with Aβ polypeptides, using a combination of in vitro immuno-infrared sensor measurements, docking, molecular dynamics simulations, and ab initio calculations. We find that the interactions of EGCG are dominated by only a few residues in the fibrils, including hydrophobic π-π interactions with aromatic rings of side chains and hydrophilic interactions with the backbone of Aβ, as confirmed by extended (1-μs-long) molecular dynamics simulations. Immuno-infrared sensor data are consistent with degradation of Aβ fibril induced by EGCG and inhibition of Aβ fibril and oligomer formation, as manifested by the recovery of the amide-I band of monomeric Aβ, which is red-shifted by 26 cm-1 when compared to the amide-I band of the fibrillar form. The shift is rationalized by computations of the infrared spectra of Aβ42 model structures, suggesting that the conformational change involves interchain hydrogen bonds in the amyloid fibrils that are broken upon binding of EGCG.
Collapse
|
16
|
Tiiman A, Jelić V, Jarvet J, Järemo P, Bogdanović N, Rigler R, Terenius L, Gräslund A, Vukojević V. Amyloidogenic Nanoplaques in Blood Serum of Patients with Alzheimer's Disease Revealed by Time-Resolved Thioflavin T Fluorescence Intensity Fluctuation Analysis. J Alzheimers Dis 2020; 68:571-582. [PMID: 30814355 PMCID: PMC6484272 DOI: 10.3233/jad-181144] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Biomarkers are central to current research on molecular mechanisms underlying Alzheimer's disease (AD). Their further development is of paramount importance for understanding pathophysiological processes that eventually lead to disease onset. Biomarkers are also crucial for early disease detection, before clinical manifestation, and for development of new disease modifying therapies. OBJECTIVE The overall aim of this work is to develop a minimally invasive method for fast, ultra-sensitive and cost-effective detection of structurally modified peptide/protein self-assemblies in the peripheral blood and in other biological fluids. Specifically, we focus here on using this method to detect structured amyloidogenic oligomeric aggregates in the blood serum of apparently healthy individuals and patients in early AD stage, and measure their concentration and size. METHODS Time-resolved detection of Thioflavin T (ThT) fluorescence intensity fluctuations in a sub-femtoliter observation volume element was used to identify in blood serum ThT-active structured amyloidogenic oligomeric aggregates, hereafter called nanoplaques, and measure with single-particle sensitivity their concentration and size. RESULTS The concentration and size of structured amyloidogenic nanoplaques are significantly higher in the blood serum of individuals diagnosed with AD than in control subjects. CONCLUSION A new method with the ultimate, single-particle sensitivity was successfully developed. The proposed approach neither relies on the use of immune-based probes, nor on the use of radiotracers, signal-amplification or protein separation techniques, and provides a minimally invasive test for fast and cost-effective early determination of structurally modified peptides/proteins in the peripheral blood, as shown here, but also in other biological fluids.
Collapse
Affiliation(s)
- Ann Tiiman
- Department of Clinical Neuroscience (CNS), Center for Molecular Medicine CMM L8:01, Karolinska Institutet, Stockholm, Sweden
| | - Vesna Jelić
- Department of Neurobiology, Care Sciences and Society (NVS), Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, Stockholm, Sweden.,The National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Petter Järemo
- Department of Internal Medicine, The Vrinnevi Hospital, Norrköping, Sweden
| | - Nenad Bogdanović
- Department of Neurobiology, Care Sciences and Society (NVS), Division of Clinical Geriatrics, Karolinska Institutet, Huddinge, Sweden.,Department of Geriatric Medicine, University of Oslo, Oslo, Norway
| | - Rudolf Rigler
- Department of Medical Biochemistry and Biophysics (MBB), Karolinska Institutet, Stockholm, Sweden
| | - Lars Terenius
- Department of Clinical Neuroscience (CNS), Center for Molecular Medicine CMM L8:01, Karolinska Institutet, Stockholm, Sweden
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Arrhenius Laboratories, Stockholm University, Stockholm, Sweden
| | - Vladana Vukojević
- Department of Clinical Neuroscience (CNS), Center for Molecular Medicine CMM L8:01, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Ganesan M, Paranthaman S. Studies on the structure and conformational flexibility of secondary structures in amyloid beta — A quantum chemical study. JOURNAL OF THEORETICAL & COMPUTATIONAL CHEMISTRY 2020. [DOI: 10.1142/s0219633620500145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Density functional theory (DFT) calculations are performed to study the conformational flexibility of secondary structures in amyloid beta (A[Formula: see text]) polypeptide. In DFT, M06-2X/6-31[Formula: see text]G(d, p) method is used to optimize the secondary structures of 2LFM and 2BEG in gas phase and in solution phase. Our calculations show that the secondary structures are energetically more stable in solution phase than in gas phase. This is due to the presence of strong solvent interaction with the secondary structures considered in this study. Among the backbone [Formula: see text] and [Formula: see text] dihedral angles, [Formula: see text] varies significantly in sheet structure. This is due to the absence of intermolecular hydrogen bond (H-bond) interactions in sheets considered in this study. Our calculations show that the conformational transition of helix/coil to sheet or vice-versa is due to the floppiness of the amino acid residues. This is observed from the Ramachandran map of the studied secondary structures. Further, it is noted that the intramolecular H-bond interactions play a significant role in the conformational transition of secondary structures of A[Formula: see text].
Collapse
Affiliation(s)
- Mahendiraprabu Ganesan
- Department of Physics and International Research Centre, Kalasalingam Academy of Research and Education (Deemed to be University), Krishnankoil 626126, India
| | - Selvarengan Paranthaman
- Department of Physics and International Research Centre, Kalasalingam Academy of Research and Education (Deemed to be University), Krishnankoil 626126, India
| |
Collapse
|
18
|
Baumgartner B, Freitag S, Lendl B. 3D Printing for Low-Cost and Versatile Attenuated Total Reflection Infrared Spectroscopy. Anal Chem 2020; 92:4736-4741. [DOI: 10.1021/acs.analchem.9b04043] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Bettina Baumgartner
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, Getreidemarkt 9/E164, 1060 Wien, Austria
| | - Stephan Freitag
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, Getreidemarkt 9/E164, 1060 Wien, Austria
| | - Bernhard Lendl
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, Getreidemarkt 9/E164, 1060 Wien, Austria
| |
Collapse
|
19
|
Agbaria AH, Rosen GB, Lapidot I, Rich DH, Mordechai S, Kapelushnik J, Huleihel M, Salman A. Rapid diagnosis of infection etiology in febrile pediatric oncology patients using infrared spectroscopy of leukocytes. JOURNAL OF BIOPHOTONICS 2020; 13:e201900215. [PMID: 31566906 DOI: 10.1002/jbio.201900215] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/27/2019] [Accepted: 09/15/2019] [Indexed: 06/10/2023]
Abstract
Rapid diagnosis of the etiology of infection is highly important for an effective treatment of the infected patients. Bacterial and viral infections are serious diseases that can cause death in many cases. The human immune system deals with many viral and bacterial infections that cause no symptoms and pass quietly without treatment. However, oncology patients undergoing chemotherapy have a very weak immune system caused by leukopenia, and even minor pathogen infection threatens their lives. For this reason, physicians tend to prescribe immediately several types of antibiotics for febrile pediatric oncology patients (FPOPs). Uncontrolled use of antibiotics is one of the major contributors to the development of resistant bacteria. Therefore, for oncology patients, a rapid and objective diagnosis of the etiology of the infection is extremely critical. Current identification methods are time-consuming (>24 h). In this study, the potential of midinfrared spectroscopy in tandem with machine learning algorithms is evaluated for rapid and objective diagnosis of the etiology of infections in FPOPs using simple peripheral blood samples. Our results show that infrared spectroscopy enables the diagnosis of the etiology of infection as bacterial or viral within 70 minutes after the collection of the blood sample with 93% sensitivity and 88% specificity.
Collapse
Affiliation(s)
- Adam H Agbaria
- Department of Physics, Ben-Gurion University, Beer-Sheva, Israel
| | - Guy Beck Rosen
- Department of Hematology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Itshak Lapidot
- Department of Electrical and Electronics Engineering, ACLP-Afeka Center for Language Processing, Afeka Tel-Aviv Academic College of Engineering, Tel-Aviv, Israel
| | - Daniel H Rich
- Department of Physics, Ben-Gurion University, Beer-Sheva, Israel
| | - Shaul Mordechai
- Department of Physics, Ben-Gurion University, Beer-Sheva, Israel
| | - Joseph Kapelushnik
- Department of Hematology, Soroka University Medical Center, Beer-Sheva, Israel
| | - Mahmoud Huleihel
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ahmad Salman
- Department of Physics, SCE-Sami Shamoon College of Engineering, Beer-Sheva, Israel
| |
Collapse
|
20
|
Prediction of Alzheimer's disease diagnosis within 14 years through Aβ misfolding in blood plasma compared to
APOE4
status, and other risk factors. Alzheimers Dement 2020; 16:283-291. [DOI: 10.1016/j.jalz.2019.08.189] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
21
|
Lue LF, Kuo YM, Sabbagh M. Advance in Plasma AD Core Biomarker Development: Current Findings from Immunomagnetic Reduction-Based SQUID Technology. Neurol Ther 2019; 8:95-111. [PMID: 31833027 PMCID: PMC6908530 DOI: 10.1007/s40120-019-00167-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Indexed: 11/28/2022] Open
Abstract
New super-sensitive biomarker assay platforms for measuring Alzheimer's disease (AD) core pathological markers in plasma have recently been developed and tested. Research findings from these technologies offer promising evidence for identifying the earliest stages of AD and correlating them with brain pathological progression. Here, we review findings using immunomagnetic reduction, one of these ultrasensitive technologies. The principles, technology and assays developed, along with selected published findings will be discussed. The major findings from this technology were significant increases of amyloid beta (Aβ) 42 and total tau (t-tau) levels in subjects clinically diagnosed with early AD when compared with cognitively normal control (NC) subjects. The composite marker of the product of Aβ42 and t-tau discriminated subjects with early AD from NC subjects with high accuracy. The potential of this technology for the purpose of early or preclinical disease stage detection has yet to be explored in subjects who have also been assessed with brain imaging and cerebrospinal fluid AD core biomarker measurements.
Collapse
Affiliation(s)
- Lih-Fen Lue
- Civin Neuropathology Laboratory, Banner Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ, 85351, USA.
- School of Life Sciences, Arizona State University, 427 East Tyler Mall, Tempe, AZ, 85281, USA.
| | - Yu-Min Kuo
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University Medical School, 1 Dasyue Road, Tainan, Taiwan
| | - Marwan Sabbagh
- Cleveland Clinic Lou Ruvo Center for Brain Health, 888 W Bonneville Ave, Las Vegas, NV, 89106, USA
| |
Collapse
|
22
|
Nabers A, Hafermann H, Wiltfang J, Gerwert K. Aβ and tau structure-based biomarkers for a blood- and CSF-based two-step recruitment strategy to identify patients with dementia due to Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2019; 11:257-263. [PMID: 30911600 PMCID: PMC6416642 DOI: 10.1016/j.dadm.2019.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) diagnosis requires invasive CSF analysis or expensive brain imaging. Therefore, a minimal-invasive reliable and cost-effective blood test is requested to power large clinical AD trials at reduced screening failure. METHODS We applied an immuno-infrared sensor to measure the amyloid-β (Aβ) and tau secondary structure distribution in plasma and CSF as structure-based biomarkers for AD (61 disease controls, 39 AD cases). RESULTS Within a first diagnostic screening step, the structure-based Aβ blood biomarker supports AD identification with a sensitivity of 90%. In a second diagnostic validation step, the combined use of the structure-based CSF biomarkers Aβ and tau excluded false-positive cases which offers an overall specificity of 97%. DISCUSSION The primary Aβ-based blood biomarker funnels individuals with suspected AD for subsequent validation of the diagnosis by structure-based combined analysis of the CSF biomarkers Aβ and tau. Our novel two-step recruitment strategy substantiates the diagnosis of AD with a likelihood of 29.
Collapse
Affiliation(s)
- Andreas Nabers
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Henning Hafermann
- LVR-Hospital Essen, Department of Psychiatry and Psychotherapy, Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August-University Goettingen, Göttingen, Germany
- German Center for Neurodegenrative Diseases (DZNE), Göttingen, Germany
- iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | - Klaus Gerwert
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
23
|
Veerabhadrappa B, Delaby C, Hirtz C, Vialaret J, Alcolea D, Lleó A, Fortea J, Santosh MS, 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 2019; 57:99-113. [PMID: 31661652 DOI: 10.1080/10408363.2019.1678011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease characterized by progressive decline of cognitive abilities. Amyloid beta peptides (Aβ), Tau proteins and the phosphorylated form of the Tau protein, p-Tau, are the core pathological biomarkers of the disease, and their detection for the diagnosis of patients is progressively being implemented. However, to date, their quantification is mostly performed on cerebrospinal fluid (CSF), the collection of which requires an invasive lumbar puncture. Early diagnosis has been shown to be important for disease-modifying treatment, which is currently in development, to limit the progression of the disease. Nevertheless, the diagnosis is often delayed to the point where the disease has already progressed, and the tools currently available do not allow for a systematic follow-up of patients. Thus, the search for a molecular signature of AD in a body fluid such as blood or saliva that can be collected in a minimally invasive way offers hope. A number of methods have been developed for the quantification of core biomarkers, especially in easily accessible fluids such as the blood, that improve their accuracy, specificity and sensitivity. This review summarizes and compares these approaches, focusing in particular on their use for Aβ detection, the earliest biomarker to be modified in the course of AD. The review also discusses biomarker quantification in CSF, blood and saliva and their clinical applications.
Collapse
Affiliation(s)
- Bhavana Veerabhadrappa
- Center for Incubation Innovation Research and Consultancy (CIIRC), Jyothy Institute of Technology, Bengaluru, India
| | - Constance Delaby
- INSERM U1183, Laboratoire de Biochimie-Protéomique Clinique, CHU de Montpellier, Université de Montpellier, Montpellier, France.,Sant Pau Memory Unit, Department of Neurology, Institut D'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Christophe Hirtz
- INSERM U1183, Laboratoire de Biochimie-Protéomique Clinique, CHU de Montpellier, Université de Montpellier, Montpellier, France
| | - Jérôme Vialaret
- INSERM U1183, Laboratoire de Biochimie-Protéomique Clinique, CHU de Montpellier, Université de Montpellier, Montpellier, France
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Institut D'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Institut D'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Institut D'Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mysore Sridhar Santosh
- Center for Incubation Innovation Research and Consultancy (CIIRC), Jyothy Institute of Technology, Bengaluru, India
| | | | - Sylvain Lehmann
- INSERM U1183, Laboratoire de Biochimie-Protéomique Clinique, CHU de Montpellier, Université de Montpellier, Montpellier, France
| |
Collapse
|
24
|
Blat A, Wiercigroch E, Smeda M, Wislocka A, Chlopicki S, Malek K. Fourier transform infrared spectroscopic signature of blood plasma in the progression of breast cancer with simultaneous metastasis to lungs. JOURNAL OF BIOPHOTONICS 2019; 12:e201900067. [PMID: 31265171 DOI: 10.1002/jbio.201900067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 06/09/2023]
Abstract
Despite advanced diagnostic techniques used for detecting cancer, this disease still remains a leading cause of death in the developed world. What is more, the greatest danger for patients is not related with growing of tumor but rather with metastasis of cancer cells to the distant organs. In this study, Fourier transform infrared (FTIR) spectroscopy was used to track chemical changes in blood plasma to find spectral markers of metastatic breast cancer during the disease progression. Plasma samples were taken 1-5 weeks after orthotropic inoculation of 4T1 metastatic breast cancer cells to mice. The earliest changes detected by FTIR spectroscopy in plasma were correlated with unsaturation of phospholipids and secondary structures of proteins that appeared 2 and 3 weeks, respectively, after 4T1 cells inoculation (micrometastatic phase). Significant alternations in the content and structure of lipids and carbohydrates were identified in plasma at the later stages (macrometastatic phase). When large primary tumors in breast and macrometastases in lung were developed, all bands in FTIR spectra significantly differed from those at earlier phases of the cancer progression. In conclusion, we showed that each phase of the breast cancer progression and its pulmonary metastasis can be characterized by a specific panel of spectral markers.
Collapse
Affiliation(s)
- Aneta Blat
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Ewelina Wiercigroch
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Marta Smeda
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland
| | - Adrianna Wislocka
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland
- Chair of Pharmacology, Jagiellonian University Medical College, Grzegorzecka 16, 31-531 Krakow, Poland
| | - Kamilla Malek
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland
| |
Collapse
|
25
|
Kowalczuk D, Pitucha M. Application of FTIR Method for the Assessment of Immobilization of Active Substances in the Matrix of Biomedical Materials. MATERIALS 2019; 12:ma12182972. [PMID: 31540255 PMCID: PMC6766236 DOI: 10.3390/ma12182972] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/08/2019] [Accepted: 09/11/2019] [Indexed: 12/02/2022]
Abstract
Background: The purpose of the study was to demonstrate the usefulness of the Fourier transform infrared spectroscopy (FTIR) method for the evaluation of the modification process of biomaterials with the participation of active substances. Methods: Modified catheter samples were prepared by activating the matrix with an acid, iodine, or bromine, and then immobilizing the active molecules. To carry out the modification process, the Fourier transform infrared-attenuated total reflectance (FTIR-ATR) method was used. Results: FTIR analysis indicated the presence of the immobilized substances in the catheter matrix and site-specific reactions. Conclusion: We surmise that the infrared spectroscopic technique is an ideal tool for the assessment of the drug immobilization and the changes occurring in the course of the modification process.
Collapse
Affiliation(s)
- Dorota Kowalczuk
- Chair and Department of Medicinal Chemistry, Faculty of Pharmacy with Division of Medical Analytics, the Medical University of Lublin, 20-090 Lublin, Poland.
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Faculty of Pharmacy with Division of Medical Analytics, the Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
26
|
Budde B, Schartner J, Tönges L, Kötting C, Nabers A, Gerwert K. Reversible Immuno-Infrared Sensor for the Detection of Alzheimer's Disease Related Biomarkers. ACS Sens 2019; 4:1851-1856. [PMID: 31241315 DOI: 10.1021/acssensors.9b00631] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The development of biosensors for medical purposes is a growing field. An immuno-infrared biosensor for the preclinical detection of Alzheimer's disease (AD) in body fluids was developed. The key element of this sensor is an ATR crystal with chemically modified surface to catch the biomarker out of the body fluid. So far, the immuno-infrared sensor can be used only once and requires time-consuming steps of sensor exchange, sensor cleaning, and novel surface functionalization. Here, we developed an immuno-infrared sensor providing a reusable surface and showcase its performance by the detection of the AD biomarker proteins Aβ and Tau in human cerebrospinal fluid (CSF). The sensor surface is covalently coated with the immunoglobulin binding proteins Protein A or Protein G. These were employed for noncovalent immobilization of antibodies and the subsequent immobilization and analysis of their antigens. The reversible antibody immobilization can be repeated several times with the same or different antibodies. Further, the more specific binding of the antibody via its Fc region instead of the conventional NHS coupling leads to a 3-4-fold higher antigen binding capacity of the antibody. Thus, the throughput, sensitivity, and automation capacity of the immuno-infrared biosensor are significantly increased as compared to former immuno-infrared assays. This immuno-sensor can be used with any antibody that binds to Protein A or Protein G.
Collapse
Affiliation(s)
- Brian Budde
- Department of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, D-44801 Bochum, Germany
- Center for Protein Diagnostics (ProDi), Gesundheitscampus 4, 44801 Bochum, Germany
| | - Jonas Schartner
- Department of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, D-44801 Bochum, Germany
- Center for Protein Diagnostics (ProDi), Gesundheitscampus 4, 44801 Bochum, Germany
| | - Lars Tönges
- Department of Neurology, Ruhr-Universität Bochum, St. Josef-Hospital, Gudrunstrasse 56, D-44791 Bochum, Germany
- Center for Protein Diagnostics (ProDi), Gesundheitscampus 4, 44801 Bochum, Germany
| | - Carsten Kötting
- Department of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, D-44801 Bochum, Germany
- Center for Protein Diagnostics (ProDi), Gesundheitscampus 4, 44801 Bochum, Germany
| | - Andreas Nabers
- Department of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, D-44801 Bochum, Germany
- Center for Protein Diagnostics (ProDi), Gesundheitscampus 4, 44801 Bochum, Germany
| | - Klaus Gerwert
- Department of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, D-44801 Bochum, Germany
- Center for Protein Diagnostics (ProDi), Gesundheitscampus 4, 44801 Bochum, Germany
| |
Collapse
|
27
|
Ostrander JS, Lomont JP, Rich KL, Saraswat V, Feingold BR, Petti MK, Birdsall ER, Arnold MS, Zanni MT. Monolayer Sensitivity Enables a 2D IR Spectroscopic Immuno-biosensor for Studying Protein Structures: Application to Amyloid Polymorphs. J Phys Chem Lett 2019; 10:3836-3842. [PMID: 31246039 PMCID: PMC6823637 DOI: 10.1021/acs.jpclett.9b01267] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Immunosensors use antibodies to detect and quantify biomarkers of disease, though the sensors often lack structural information. We create a surface-sensitive two-dimensional infrared (2D IR) spectroscopic immunosensor for studying protein structures. We tether antibodies to a plasmonic surface, flow over a solution of amyloid proteins, and measure the 2D IR spectra. The 2D IR spectra provide a global assessment of antigen structure, and isotopically labeled proteins give residue-specific structural information. We report the 2D IR spectra of fibrils and monomers using a polyclonal antibody that targets human islet amyloid polypeptide (hIAPP). We observe two fibrillar polymorphs differing in their structure at the G24 residue, which supports the hypothesis that hIAPP polymorphs form from a common oligomeric intermediate. This work provides insight into the structure of hIAPP, establishes a new method for studying protein structures using 2D IR spectroscopy, and creates a spectroscopic immunoassay applicable for studying a wide range of biomarkers.
Collapse
Affiliation(s)
- Joshua S. Ostrander
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Justin P. Lomont
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Kacie L. Rich
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Vivek Saraswat
- Department of Materials Science and Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Benjamin R. Feingold
- Department of Materials Science and Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Megan K. Petti
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Erin R. Birdsall
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Michael S. Arnold
- Department of Materials Science and Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Martin T. Zanni
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
28
|
Ghidoni R, Squitti R, Siotto M, Benussi L. Innovative Biomarkers for Alzheimer's Disease: Focus on the Hidden Disease Biomarkers. J Alzheimers Dis 2019; 62:1507-1518. [PMID: 29504534 DOI: 10.3233/jad-170953] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The criteria for the clinical diagnosis of AD include the analysis of biomarkers of the underlying brain disease pathology; a set of cerebrospinal fluid (CSF) tests, amyloid-β1-42 (Aβ42), total-tau (t-tau), and phosphorylated tau (p-tau), are available and their performance in a clinical setting has been assessed in several studies. Thus, in dementia research, great advances have been made in the discovery of putative biomarkers; however, disappointingly, few of them have been translated into clinically applicable assays. To find biomarkers able to reliably detect AD pathology already at prodromal stages and in blood is even more important. Recent technical breakthroughs have provided ultrasensitive methods that allow the detection of brain-specific proteins in blood. In the present review, we will focus on the usefulness of ultrasensitive technologies for biomarker discovery and trace elements detection; moreover, we will review studies on circulating nano-compartments, a promising novel source of material for molecular diagnostics.
Collapse
Affiliation(s)
- Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Rosanna Squitti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
29
|
Hrubešová K, Fousková M, Habartová L, Fišar Z, Jirák R, Raboch J, Setnička V. Search for biomarkers of Alzheimer's disease: Recent insights, current challenges and future prospects. Clin Biochem 2019; 72:39-51. [PMID: 30953619 DOI: 10.1016/j.clinbiochem.2019.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/03/2019] [Indexed: 12/12/2022]
Abstract
Due to the trend of prolonged lifespan leading to higher incidence of age-related diseases, the demand for reliable biomarkers of dementia rises. In this review, we present novel biomarkers of high potential, especially those found in blood, urine or saliva, which could lead to a more comfortable patient experience and better time- and cost-effectivity, compared to the currently used diagnostic methods. We focus on biomarkers that might allow for the detection of Alzheimer's disease before its clinical manifestations. Such biomarkers might be helpful for better understanding the etiology of the disease and identifying its risk factors. Moreover, it could be a base for developing new treatment or at least help to prolong the presymptomatic stage in patients suffering from Alzheimer's disease. As potential candidates, we present, for instance, neurofilament light in both cerebrospinal fluid and blood plasma or amyloid β in plasma. Above all, we provide an overview of different approaches to the diagnostics, analyzing patient's biofluids as a whole using molecular spectroscopy. Infrared and Raman spectroscopy and especially chiroptical methods provide information not only on the chemical composition, but also on molecular structure. Therefore, these techniques are promising for the diagnostics of Alzheimer's disease, as the accumulation of amyloid β in abnormal conformation is one of the hallmarks of this disease.
Collapse
Affiliation(s)
- Kateřina Hrubešová
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Markéta Fousková
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Lucie Habartová
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Roman Jirák
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Jiří Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Vladimír Setnička
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic.
| |
Collapse
|
30
|
Habartová L, Hrubešová K, Syslová K, Vondroušová J, Fišar Z, Jirák R, Raboch J, Setnička V. Blood-based molecular signature of Alzheimer's disease via spectroscopy and metabolomics. Clin Biochem 2019; 72:58-63. [PMID: 30954438 DOI: 10.1016/j.clinbiochem.2019.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 01/02/2023]
Abstract
OBJECTIVES With over 35 million cases worldwide, Alzheimer's disease (AD) represents the main cause of dementia. The differentiation of AD from other types of dementia is challenging and its early diagnosis is complicated. The established biomarkers are not only based on the invasive collection of cerebrospinal fluid, but also lack sufficient sensitivity and specificity. Therefore, much current effort is aimed at the identification of new biomarkers of AD in peripheral blood. DESIGN AND METHODS We focused on blood-based analyses using chiroptical spectroscopy (Raman optical activity, electronic circular dichroism) supplemented with conventional vibrational spectroscopy (infrared, Raman) and metabolomics (high-performance liquid chromatography with a high-resolution mass detection). RESULTS This unique approach enabled us to identify the spectral pattern of AD and variations in metabolite levels. Subsequent linear discriminant analysis of the spectral data resulted in differentiation between the AD patients and control subjects. CONCLUSIONS It may be stated that this less invasive approach has strong potential for the identification of disease-related changes within essential plasmatic biomolecules and metabolites.
Collapse
Affiliation(s)
- Lucie Habartová
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic.
| | - Kateřina Hrubešová
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Kamila Syslová
- Department of Organic Technology, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Jana Vondroušová
- Department of Organic Technology, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine of the Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Roman Jirák
- Department of Psychiatry, First Faculty of Medicine of the Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Jiří Raboch
- Department of Psychiatry, First Faculty of Medicine of the Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Vladimír Setnička
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| |
Collapse
|
31
|
Nabers A, Perna L, Lange J, Mons U, Schartner J, Güldenhaupt J, Saum KU, Janelidze S, Holleczek B, Rujescu D, Hansson O, Gerwert K, Brenner H. Amyloid blood biomarker detects Alzheimer's disease. EMBO Mol Med 2019; 10:emmm.201708763. [PMID: 29626112 PMCID: PMC5938617 DOI: 10.15252/emmm.201708763] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is currently incurable, but there is general agreement that a minimally invasive blood biomarker for screening in preclinical stages would be crucial for future therapy. Diagnostic tools for detection of AD are either invasive like cerebrospinal fluid (CSF) biomarkers or expensive such as positron emission tomography (PET) scanning. Here, we determine the secondary structure change of amyloid‐β (Aβ) in human blood. This change used as blood amyloid biomarker indicates prodromal AD and correlates with CSF AD biomarkers and amyloid PET imaging in the cross‐sectional BioFINDER cohort. In a further population‐based longitudinal cohort (ESTHER), the blood biomarker detected AD several years before clinical diagnosis in baseline samples with a positive likelihood ratio of 7.9; that is, those who were diagnosed with AD over the years were 7.9 times more likely to test positive. This assay may open avenues for blood screening of early AD stages as a funnel for further more invasive and expensive tests.
Collapse
Affiliation(s)
- Andreas Nabers
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Laura Perna
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julia Lange
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Ute Mons
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jonas Schartner
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Jörn Güldenhaupt
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Kai-Uwe Saum
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | - Dan Rujescu
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Halle, Halle, Germany
| | - Oskar Hansson
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Klaus Gerwert
- Department of Biophysics, Ruhr-University Bochum, Bochum, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Network Aging Research (NAR), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
32
|
Fallah MA, Hauser K. Immobilization approaches can affect protein dynamics: a surface-enhanced infrared spectroscopic study on lipid–protein interactions. Biomater Sci 2019; 7:3204-3212. [DOI: 10.1039/c9bm00140a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Near-field detection of SEIRA reveals that surface immobilization alters conformational properties of α-synuclein.
Collapse
Affiliation(s)
| | - Karin Hauser
- Department of Chemistry
- University of Konstanz
- 78457 Konstanz
- Germany
| |
Collapse
|
33
|
Mordechai S, Shufan E, Porat Katz BS, Salman A. Early diagnosis of Alzheimer's disease using infrared spectroscopy of isolated blood samples followed by multivariate analyses. Analyst 2018; 142:1276-1284. [PMID: 27827489 DOI: 10.1039/c6an01580h] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, particularly in the elderly. The disease is characterized by cognitive decline that typically starts with insidious memory loss and progresses relentlessly to produce global impairment of all higher cortical functions. Due to better living conditions and health facilities in developed countries, which result in higher overall life spans, these countries report upward trends of AD among their populations. There are, however, no specific diagnostic tests for AD and clinical diagnosis is especially difficult in the earliest stages of the disease. Early diagnosis of AD is frequently subjective and is determined by physicians (generally neurologists, geriatricians, and psychiatrists) depending on their experience. Diagnosing AD requires both medical history and mental status testing. Having trouble with memory does not mean you have AD. AD has no current cure, but treatments for symptoms are available and research continues. In this study, we investigated the potential of infrared microscopy to differentiate between AD patients and controls, using Fourier transform infrared (FTIR) spectroscopy of isolated blood components. FTIR is known as a quick, safe, and minimally invasive method to investigate biological samples. For this goal, we measured infrared spectra from white blood cells (WBCs) and plasma taken from AD patients and controls, with the consent of the patients or their guardians. Applying multivariate analysis, principal component analysis (PCA) followed by linear discriminant analysis (LDA), it was possible to differentiate among the different types of mild, moderate, and severe AD, and the controls, with 85% accuracy when using the WBC spectra and about 77% when using the plasma spectra. When only the moderate and severe stages were included, an 83% accuracy was obtained using the WBC spectra and about 89% when using the plasma spectra.
Collapse
Affiliation(s)
- S Mordechai
- Department of Physics, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | | | | | | |
Collapse
|
34
|
Haas J, Schwartz M, Rengstl U, Jetter M, Michler P, Mizaikoff B. Chem/bio sensing with non-classical light and integrated photonics. Analyst 2018; 143:593-605. [PMID: 29260151 DOI: 10.1039/c7an01011g] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Modern quantum technology currently experiences extensive advances in applicability in communications, cryptography, computing, metrology and lithography. Harnessing this technology platform for chem/bio sensing scenarios is an appealing opportunity enabling ultra-sensitive detection schemes. This is further facilliated by the progress in fabrication, miniaturization and integration of visible and infrared quantum photonics. Especially, the combination of efficient single-photon sources together with waveguiding/sensing structures, serving as active optical transducer, as well as advanced detector materials is promising integrated quantum photonic chem/bio sensors. Besides the intrinsic molecular selectivity and non-destructive character of visible and infrared light based sensing schemes, chem/bio sensors taking advantage of non-classical light sources promise sensitivities beyond the standard quantum limit. In the present review, recent achievements towards on-chip chem/bio quantum photonic sensing platforms based on N00N states are discussed along with appropriate recognition chemistries, facilitating the detection of relevant (bio)analytes at ultra-trace concentration levels. After evaluating recent developments in this field, a perspective for a potentially promising sensor testbed is discussed for reaching integrated quantum sensing with two fiber-coupled GaAs chips together with semiconductor quantum dots serving as single-photon sources.
Collapse
Affiliation(s)
- J Haas
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| | | | | | | | | | | |
Collapse
|
35
|
Etezadi D, Warner JB, Lashuel HA, Altug H. Real-Time In Situ Secondary Structure Analysis of Protein Monolayer with Mid-Infrared Plasmonic Nanoantennas. ACS Sens 2018; 3:1109-1117. [PMID: 29845861 PMCID: PMC6133232 DOI: 10.1021/acssensors.8b00115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Dynamic detection
of protein conformational changes at physiological
conditions on a minute amount of samples is immensely important for
understanding the structural determinants of protein function in health
and disease and to develop assays and diagnostics for protein misfolding
and protein aggregation diseases. Herein, we experimentally demonstrate
the capabilities of a mid-infrared plasmonic biosensor for real-time
and in situ protein secondary structure analysis in aqueous environment
at nanoscale. We present label-free ultrasensitive dynamic monitoring
of β-sheet to disordered conformational transitions in a monolayer
of the disease-related α-synuclein protein under varying stimulus
conditions. Our experiments show that the extracted secondary structure
signals from plasmonically enhanced amide I signatures in the protein
monolayer can be reliably and reproducibly acquired with second derivative
analysis for dynamic monitoring. Furthermore, by using a polymer layer
we show that our nanoplasmonic approach of extracting the frequency
components of vibrational signatures matches with the results attained
from gold-standard infrared transmission measurements. By facilitating
conformational analysis on small quantities of immobilized proteins
in response to external stimuli such as drugs, our plasmonic biosensor
could be used to introduce platforms for screening small molecule
modulators of protein misfolding and aggregation.
Collapse
|
36
|
Agbaria AH, Beck Rosen G, Lapidot I, Rich DH, Huleihel M, Mordechai S, Salman A, Kapelushnik J. Differential Diagnosis of the Etiologies of Bacterial and Viral Infections Using Infrared Microscopy of Peripheral Human Blood Samples and Multivariate Analysis. Anal Chem 2018; 90:7888-7895. [PMID: 29869874 DOI: 10.1021/acs.analchem.8b00017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human viral and bacterial infections are responsible for a variety of diseases that are still the main causes of death and economic burden for society across the globe. Despite the different responses of the immune system to these infections, some of them have similar symptoms, such as fever, sneezing, inflammation, vomiting, diarrhea, and fatigue. Thus, physicians usually encounter difficulties in distinguishing between viral and bacterial infections on the basis of these symptoms. Rapid identification of the etiology of infection is highly important for effective treatment and can save lives in some cases. The current methods used for the identification of the nature of the infection are mainly based on growing the infective agent in culture, which is a time-consuming (over 24 h) and usually expensive process. The main objective of this study was to evaluate the potential of the mid-infrared spectroscopic method for rapid and reliable identification of bacterial and viral infections based on simple peripheral blood samples. For this purpose, white blood cells (WBCs) and plasma were isolated from the peripheral blood samples of patients with confirmed viral or bacterial infections. The obtained spectra were analyzed by multivariate analysis: principle component analysis (PCA) followed by linear discriminant analysis (LDA), to identify the infectious agent type as bacterial or viral in a time span of about 1 h after the collection of the blood sample. Our preliminary results showed that it is possible to determine the infectious agent with high success rates of 82% for sensitivity and 80% for specificity, based on the WBC data.
Collapse
Affiliation(s)
- Adam H Agbaria
- Department of Physics , Ben-Gurion University , Beer-Sheva 84105 , Israel
| | - Guy Beck Rosen
- Department of Pediatric Hematology/Oncology , Soroka University Medical Center , Beer-Sheva 84105 , Israel
| | - Itshak Lapidot
- Department of Electrical and Electronics Engineering, ACLP-Afeka Center for Language Processing , Afeka Tel-Aviv Academic College of Engineering , Tel-Aviv 69107 , Israel
| | - Daniel H Rich
- Department of Physics , Ben-Gurion University , Beer-Sheva 84105 , Israel
| | - Mahmoud Huleihel
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences , Ben-Gurion University of the Negev , Beer-Sheva 84105 , Israel
| | - Shaul Mordechai
- Department of Physics , Ben-Gurion University , Beer-Sheva 84105 , Israel
| | - Ahmad Salman
- Department of Physics , SCE-Sami Shamoon College of Engineering , Beer-Sheva 84100 , Israel
| | - Joseph Kapelushnik
- Department of Pediatric Hematology/Oncology , Soroka University Medical Center , Beer-Sheva 84105 , Israel
| |
Collapse
|
37
|
Blond P, Mattiuzzi A, Valkenier H, Troian-Gautier L, Bergamini JF, Doneux T, Goormaghtigh E, Raussens V, Jabin I. Grafting of Oligo(ethylene glycol)-Functionalized Calix[4]arene-Tetradiazonium Salts for Antifouling Germanium and Gold Surfaces. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:6021-6027. [PMID: 29724105 DOI: 10.1021/acs.langmuir.8b00464] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Biosensors that can determine protein concentration and structure are highly desired for biomedical applications. For the development of such biosensors, the use of Fourier transform infrared (FTIR) spectroscopy with the attenuated internal total reflection (ATR) configuration is particularly attractive, but it requires appropriate surface functionalization of the ATR optical element. Indeed, the surface has to specifically interact with a target protein in close contact with the optical element and must display antifouling properties to prevent nonspecific adsorption of other proteins. Here, we report robust monolayers of calix[4]arenes bearing oligo(ethylene glycol) (oEG) chains, which were grafted on germanium and gold surfaces via their tetradiazonium salts. The formation of monolayers of oEGylated calix[4]arenes was confirmed by AFM, IR, and contact angle measurements. The antifouling properties of these modified surfaces were studied by ATR-FTIR spectroscopy and fluorescence microscopy, and the nonspecific absorption of bovine serum albumin was found to be reduced by 85% compared to that of unmodified germanium. In other words, the organic coating by oEGylated calix[4]arenes provides remarkable antifouling properties, opening the way for the design of germanium- or gold-based biosensors.
Collapse
Affiliation(s)
- Pascale Blond
- Laboratoire de Chimie Organique , Université Libre de Bruxelles (ULB) , avenue F. D. Roosevelt 50 , CP160/06, B-1050 Brussels , Belgium
- Laboratory for the Structure and Function of Biological Membranes, Center for Structural Biology and Bioinformatics , Université Libre de Bruxelles (ULB) , avenue F. D. Roosevelt 50 , CP206/02, B-1050 Brussels , Belgium
| | - Alice Mattiuzzi
- Laboratoire de Chimie Organique , Université Libre de Bruxelles (ULB) , avenue F. D. Roosevelt 50 , CP160/06, B-1050 Brussels , Belgium
- X4C , Rue Chêne Bonnet 128 , 6110 Montigny-le-Tilleul , Belgium
| | - Hennie Valkenier
- Engineering of Molecular NanoSystems , Ecole Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB) , avenue F. D. Roosevelt 50 , CP165/64, B-1050 Brussels , Belgium
| | - Ludovic Troian-Gautier
- Laboratoire de Chimie Organique , Université Libre de Bruxelles (ULB) , avenue F. D. Roosevelt 50 , CP160/06, B-1050 Brussels , Belgium
| | - Jean-François Bergamini
- Institut des Sciences Chimiques de Rennes (Equipe MaCSE), CNRS, UMR 6226 , Université de Rennes 1 , Campus de Beaulieu, Bat 10C, 35042 Cedex Rennes , France
| | - Thomas Doneux
- Chimie Analytique et Chimie des Interfaces , Université Libre de Bruxelles (ULB) , Campus de la Plaine, boulevard du Triomphe , CP255, B-1050 Brussels , Belgium
| | - Erik Goormaghtigh
- Laboratory for the Structure and Function of Biological Membranes, Center for Structural Biology and Bioinformatics , Université Libre de Bruxelles (ULB) , avenue F. D. Roosevelt 50 , CP206/02, B-1050 Brussels , Belgium
| | - Vincent Raussens
- Laboratory for the Structure and Function of Biological Membranes, Center for Structural Biology and Bioinformatics , Université Libre de Bruxelles (ULB) , avenue F. D. Roosevelt 50 , CP206/02, B-1050 Brussels , Belgium
| | - Ivan Jabin
- Laboratoire de Chimie Organique , Université Libre de Bruxelles (ULB) , avenue F. D. Roosevelt 50 , CP160/06, B-1050 Brussels , Belgium
| |
Collapse
|
38
|
Staniszewska-Slezak E, Wiercigroch E, Fedorowicz A, Buczek E, Mateuszuk L, Baranska M, Chlopicki S, Malek K. A possible Fourier transform infrared-based plasma fingerprint of angiotensin-converting enzyme inhibitor-induced reversal of endothelial dysfunction in diabetic mice. JOURNAL OF BIOPHOTONICS 2018; 11:e201700044. [PMID: 28700133 DOI: 10.1002/jbio.201700044] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/10/2017] [Accepted: 06/06/2017] [Indexed: 06/07/2023]
Abstract
Angiotensin-converting enzyme inhibitors (ACE-I) display vasoprotective activity and represent the cornerstone in the treatment of cardiovascular diseases. In this study, we tested whether Fourier transform infrared (FTIR)-based analysis of blood plasma is sensitive to detect vasoprotective effects of treatment with perindopril including reversal of endothelial dysfunction in diabetes. For this purpose, plasma samples were collected from untreated db/db mice, db/db mice treated with 2 or 10 mg/kg perindopril and db+ mice. The effect of perindopril on endothelial function was examined in ex vivo aortic rings; 10 mg/kg but not 2 mg/kg of perindopril reversed endothelial dysfunction. In plasma of db/db mice, the balance between conformations of plasma proteins was noted, and treatment with perindopril at a high dose but not at a low dose reversed this effect. This was revealed by amide II/amide I ratio attributed to increased β-sheet formation. Spectral markers at 3010, 1520/1238 cm-1 , representative for unsaturation degree of lipids and phosphorylation of tyrosine, respectively, were also affected by perindopril treatment. In conclusion, although metabolic abnormalities associated with type 2 diabetes mellitus such as hypertriglyceridemia and hyperglycemia strongly affected spectral FTIR profile of diabetic plasma, we identified FTIR features that seem to be associated with the vasoprotective activity of ACE-I.
Collapse
Affiliation(s)
- Emilia Staniszewska-Slezak
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Ewelina Wiercigroch
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Andrzej Fedorowicz
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Chair of Pharmacology, Jagiellonian University, Krakow, Poland
| | - Elżbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Lukasz Mateuszuk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Malgorzata Baranska
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Chair of Pharmacology, Jagiellonian University, Krakow, Poland
| | - Kamilla Malek
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| |
Collapse
|
39
|
Schartner J, Güldenhaupt J, Katharina Gaßmeyer S, Rosga K, Kourist R, Gerwert K, Kötting C. Highly stable protein immobilizationviamaleimido-thiol chemistry to monitor enzymatic activity. Analyst 2018; 143:2276-2284. [DOI: 10.1039/c8an00301g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Combining a novel protein immobilisation method with multivariate curve resolution enables the direct observation of biocatalysis by ATR-FTIR spectroscopy.
Collapse
Affiliation(s)
- Jonas Schartner
- Department of Biophysics
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| | - Jörn Güldenhaupt
- Department of Biophysics
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| | | | - Katharina Rosga
- Department of Biophysics
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| | - Robert Kourist
- Junior Research Group for Microbial Biotechnology
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| | - Klaus Gerwert
- Department of Biophysics
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| | - Carsten Kötting
- Department of Biophysics
- Ruhr-Universität Bochum
- 44801 Bochum
- Germany
| |
Collapse
|
40
|
Lomont JP, Rich KL, Maj M, Ho JJ, Ostrander JS, Zanni MT. Spectroscopic Signature for Stable β-Amyloid Fibrils versus β-Sheet-Rich Oligomers. J Phys Chem B 2017; 122:144-153. [PMID: 29220175 DOI: 10.1021/acs.jpcb.7b10765] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We use two-dimensional IR (2D IR) spectroscopy to explore fibril formation for the two predominant isoforms of the β-amyloid (Aβ1-40 and Aβ1-42) protein associated with Alzheimer's disease. Two-dimensional IR spectra resolve a transition at 1610 cm-1 in Aβ fibrils that does not appear in other Aβ aggregates, even those with predominantly β-sheet-structure-like oligomers. This transition is not resolved in linear IR spectroscopy because it lies under the broad band centered at 1625 cm-1, which is the traditional infrared signature for amyloid fibrils. The feature is prominent in 2D IR spectra because 2D lineshapes are narrower and scale nonlinearly with transition dipole strengths. Transmission electron microscopy measurements demonstrate that the 1610 cm-1 band is a positive identification of amyloid fibrils. Sodium dodecyl sulfate micelles that solubilize and disaggregate preaggregated Aβ samples deplete the 1625 cm-1 band but do not affect the 1610 cm-1 band, demonstrating that the 1610 cm-1 band is due to very stable fibrils. We demonstrate that the 1610 cm-1 transition arises from amide I modes by mutating out the only side-chain residue that could give rise to this transition, and we explore the potential structural origins of the transition by simulating 2D IR spectra based on Aβ crystal structures. It was not previously possible to distinguish stable Aβ fibrils from the less stable β-sheet-rich oligomers with infrared light. This 2D IR signature will be useful for Alzheimer's research on Aβ aggregation, fibril formation, and toxicity.
Collapse
Affiliation(s)
- Justin P Lomont
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Kacie L Rich
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Michał Maj
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Jia-Jung Ho
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Joshua S Ostrander
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| | - Martin T Zanni
- Department of Chemistry, University of Wisconsin-Madison , Madison, Wisconsin 53706, United States
| |
Collapse
|
41
|
Schartner J, Nabers A, Budde B, Lange J, Hoeck N, Wiltfang J, Kötting C, Gerwert K. An ATR-FTIR Sensor Unraveling the Drug Intervention of Methylene Blue, Congo Red, and Berberine on Human Tau and Aβ. ACS Med Chem Lett 2017; 8:710-714. [PMID: 28740603 DOI: 10.1021/acsmedchemlett.7b00079] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/09/2017] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease affects millions of human beings worldwide. The disease progression is characterized by the formation of plaques and neurofibrillary tangles in the brain, which are based on aggregation processes of the Aβ peptide and tau protein. Today there is no cure and even no in vitro assay available for the identification of drug candidates, which provides direct information concerning the protein secondary structure label-free. Therefore, we developed an attenuated total reflection Fourier transform infrared spectroscopy (ATR-FTIR) sensor, which uses surface bound antibodies to immobilize a desired target protein. The secondary structure of the protein can be evaluated based on the secondary structure sensitive frequency of the amide I band. Direct information about the effect of a drug candidate on the secondary structure distribution of the total target protein fraction within the respective body fluid can be detected by a frequency shift of the amide I band. Thereby, the extent of the amide I shift is indicative for the compound efficiency. The functionality of this approach was demonstrated by the quantification of the effect of the drug candidate methylene blue on the pathogenic misfolded tau protein as extracted from cerebrospinal fluid (CSF). Methylene blue induces a shift from pathogenic folded β-sheet dominated to the healthy monomeric state. A similar effect was observed for congo red on pathogenic Aβ isoforms from CSF. In addition, the effect of berberine on synthetic Aβ1-42 is studied. Berberine seems to decelerate the aggregation process of synthetic Aβ1-42 peptides.
Collapse
Affiliation(s)
- Jonas Schartner
- Department
of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| | - Andreas Nabers
- Department
of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| | - Brian Budde
- Department
of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| | - Julia Lange
- Department
of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| | - Nina Hoeck
- Department
of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| | - Jens Wiltfang
- Department
of Psychiatry and Psychotherapy, Georg-August-University Göttingen, University Medical
Center, 37099 Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), 37099 Göttingen, Germany
- iBiMED,
Medical Sciences Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Carsten Kötting
- Department
of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| | - Klaus Gerwert
- Department
of Biophysics, Ruhr-Universität Bochum, Universitätsstrasse 150, 44801 Bochum, Germany
| |
Collapse
|
42
|
Hoarau M, Badieyan S, Marsh ENG. Immobilized enzymes: understanding enzyme – surface interactions at the molecular level. Org Biomol Chem 2017; 15:9539-9551. [DOI: 10.1039/c7ob01880k] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Interactions between immobilized enzymes and supporting surfaces are complex and context-dependent and can significantly alter enzyme structure, stability and activity.
Collapse
Affiliation(s)
- Marie Hoarau
- Department of Chemistry
- University of Michigan
- Ann Arbor
- USA
| | | | - E. Neil G. Marsh
- Department of Chemistry
- University of Michigan
- Ann Arbor
- USA
- Department of Biological Chemistry
| |
Collapse
|
43
|
Haas J, Mizaikoff B. Advances in Mid-Infrared Spectroscopy for Chemical Analysis. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2016; 9:45-68. [PMID: 27070183 DOI: 10.1146/annurev-anchem-071015-041507] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Infrared spectroscopy in the 3-20 μm spectral window has evolved from a routine laboratory technique into a state-of-the-art spectroscopy and sensing tool by benefitting from recent progress in increasingly sophisticated spectra acquisition techniques and advanced materials for generating, guiding, and detecting mid-infrared (MIR) radiation. Today, MIR spectroscopy provides molecular information with trace to ultratrace sensitivity, fast data acquisition rates, and high spectral resolution catering to demanding applications in bioanalytics, for example, and to improved routine analysis. In addition to advances in miniaturized device technology without sacrificing analytical performance, selected innovative applications for MIR spectroscopy ranging from process analysis to biotechnology and medical diagnostics are highlighted in this review.
Collapse
Affiliation(s)
- Julian Haas
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, 89069 Ulm, Germany;
| | - Boris Mizaikoff
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, 89069 Ulm, Germany;
| |
Collapse
|
44
|
Nabers A, Ollesch J, Schartner J, Kötting C, Genius J, Hafermann H, Klafki H, Gerwert K, Wiltfang J. Amyloid-β-Secondary Structure Distribution in Cerebrospinal Fluid and Blood Measured by an Immuno-Infrared-Sensor: A Biomarker Candidate for Alzheimer's Disease. Anal Chem 2016; 88:2755-62. [PMID: 26828829 DOI: 10.1021/acs.analchem.5b04286] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The misfolding of the Amyloid-beta (Aβ) peptide into β-sheet enriched conformations was proposed as an early event in Alzheimer's Disease (AD). Here, the Aβ peptide secondary structure distribution in cerebrospinal fluid (CSF) and blood plasma of 141 patients was measured with an immuno-infrared-sensor. The sensor detected the amide I band, which reflects the overall secondary structure distribution of all Aβ peptides extracted from the body fluid. We observed a significant downshift of the amide I band frequency of Aβ peptides in Dementia Alzheimer type (DAT) patients, which indicated an overall shift to β-sheet. The secondary structure distribution of all Aβ peptides provides a better marker for DAT detection than a single Aβ misfold or the concentration of a specific oligomer. The discrimination between DAT and disease control patients according to the amide I frequency was in excellent agreement with the clinical diagnosis (accuracy 90% for CSF and 84% for blood). The amide I band maximum above or below the decisive marker frequency appears as a novel spectral biomarker candidate of AD. Additionally, a preliminary proof-of-concept study indicated an amide I band shift below the marker band already in patients with mild cognitive impairment due to AD. The presented immuno-IR-sensor method represents a promising, simple, robust, and label-free diagnostic tool for CSF and blood analysis.
Collapse
Affiliation(s)
- Andreas Nabers
- Department of Biophysics, Ruhr-University Bochum , 44780 Bochum, Germany
| | - Julian Ollesch
- Department of Biophysics, Ruhr-University Bochum , 44780 Bochum, Germany
| | - Jonas Schartner
- Department of Biophysics, Ruhr-University Bochum , 44780 Bochum, Germany
| | - Carsten Kötting
- Department of Biophysics, Ruhr-University Bochum , 44780 Bochum, Germany
| | - Just Genius
- Clinics for Psychiatry and Psychotherapy, LVR-Clinical Center Essen , 45147 Essen, Germany
| | - Henning Hafermann
- Clinics for Psychiatry and Psychotherapy, LVR-Clinical Center Essen , 45147 Essen, Germany
| | - Hans Klafki
- Department of Psychiatry and Psychotherapy, Georg-August-University Göttingen, University Medical Center , 37099 Göttingen, Germany
| | - Klaus Gerwert
- Department of Biophysics, Ruhr-University Bochum , 44780 Bochum, Germany
| | - Jens Wiltfang
- Clinics for Psychiatry and Psychotherapy, LVR-Clinical Center Essen , 45147 Essen, Germany.,Department of Psychiatry and Psychotherapy, Georg-August-University Göttingen, University Medical Center , 37099 Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE) , 37099 Göttingen, Germany
| |
Collapse
|