Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Alzheimers Dement ; 2022 Jul 19.
Article in English | MEDLINE | ID: mdl-35852967

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 (Aß) 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: Aß 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 Aß misfolding and GFAP increased the accuracy. DISCUSSION: Aß misfolding and GFAP showed a strong ability to predict clinical AD risk and may be important early AD risk markers. Aß misfolding illustrated its potential as a prescreening tool for AD risk stratification in older adults.

2.
Alzheimers Res Ther ; 13(1): 145, 2021 08 30.
Article in English | MEDLINE | ID: mdl-34461995

ABSTRACT

BACKGROUND: To understand the potential for early intervention and prevention measures in Alzheimer's disease, the association between risk factors and early pathological change needs to be assessed. Hence, the aim of this study was to determine whether risk factors of Alzheimer's clinical syndrome (clinical AD), such as body mass index (BMI), are associated with Aß misfolding in blood, a strong risk marker for AD among older adults. METHODS: Information on risk factors and blood samples were collected at baseline in the ESTHER study, a population-based cohort study of older adults (age 50-75 years) in Germany. Aß misfolding in blood plasma was analyzed using an immuno-infrared-sensor in a total of 872 participants in a nested case-control design among incident dementia cases and matched controls. Associations between risk factors and Aß misfolding were assessed by multiple logistic regression. For comparison, the association between the risk factors and AD incidence during 17 years of follow-up was investigated in parallel among 5987 cohort participants. RESULTS: An inverse association with Aß misfolding was seen for BMI at age 50 based on reported weight history (aOR 0.64, 95% CI 0.43-0.96, p = 0.03). Similar but not statistically significant associations were seen for BMI at baseline (i.e., mean age 68) and at age 40. No statistically significant associations with Aß misfolding were found for other risk factors, such as diabetes, smoking, and physical activity. On the other hand, low physical activity was associated with a significantly reduced risk of developing clinical AD compared to physical inactivity. CONCLUSIONS: Our results support that AD pathology may be detectable and associated with reduced weight even in middle adulthood, many years before clinical diagnosis of AD. Physical activity might reduce the risk of onset of AD symptoms.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Adult , Aged , Alzheimer Disease/epidemiology , Body Mass Index , Cohort Studies , Humans , Middle Aged , Plasma
3.
Transl Psychiatry ; 11(1): 261, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33934115

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, Aß 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 Aß42 based) were calculated, APOE genotype was determined, and Aß 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 Aß misfolding were assessed through logistic regression and the ability of each genetic marker and Aß misfolding to predict Alzheimer's disease was determined. The Alzheimer's disease polygenic risk score and APOE ε4 presence were associated to Aß 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 Aß polygenic risk score. All genetic markers were predictive of Alzheimer's disease diagnosis albeit much less so than Aß misfolding (areas under the curve: Aß polygenic risk score: 0.55; AD polygenic risk score: 0.59; APOE ε4: 0.63; Aß misfolding: 0.84). Clinical Alzheimer's genetic risk was associated to early pathological changes (Aß misfolding) measured in blood, however, predicted Alzheimer's disease less accurately than Aß misfolding itself. Genetic predisposition may provide information regarding disease initiation, while Aß misfolding could be important in clinical risk prediction.


Subject(s)
Alzheimer Disease , Alzheimer Disease/genetics , Amyloid beta-Peptides , Genetic Predisposition to Disease , Humans , Plaque, Amyloid , Plasma
4.
Alzheimers Dement (Amst) ; 13(1): e12151, 2021.
Article in English | MEDLINE | ID: mdl-33614887

ABSTRACT

INTRODUCTION: Determining potential risk factors of amyloid beta (Aß) misfolding in blood, a risk marker for clinical Alzheimer's disease (AD), could have important implications for its utility in future research and clinical settings. METHODS: Participants aged 50 to 75 years attending a general health examination were recruited for a prospective community-based cohort study in Saarland, Germany, in 2000 to 2002. For these analyses, participants with available Aß misfolding measurements and clinical AD information at 17-year follow-up were included (n = 444). RESULTS: Age did not show any association with Aß misfolding in plasma; however, a strong association of both age and Aß misfolding with the incidence of clinical AD was evident. Education and cardiovascular diseases were likewise not associated with Aß misfolding. DISCUSSION: Structural measurement of Aß misfolding in blood plasma is an age-independent risk marker of clinical AD among older adults, supporting that risk of clinical AD is already largely determined before older adulthood.

6.
Ann Clin Transl Neurol ; 8(1): 271-277, 2021 01.
Article in English | MEDLINE | ID: mdl-33263951

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.


Subject(s)
Amyotrophic Lateral Sclerosis/cerebrospinal fluid , Amyotrophic Lateral Sclerosis/diagnosis , Biomarkers/cerebrospinal fluid , DNA-Binding Proteins/cerebrospinal fluid , Aged , DNA-Binding Proteins/chemistry , Female , Humans , Male , Middle Aged , Pilot Projects , Protein Structure, Secondary , Sensitivity and Specificity , Spectrophotometry, Infrared/methods
7.
Acta Neuropathol Commun ; 8(1): 222, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33308303

ABSTRACT

The neuropathology of Alzheimer's disease (AD) is characterized by hyperphosphorylated tau neurofibrillary tangles (NFTs) and amyloid-beta (Aß) plaques. Aß plaques are hypothesized to follow a development sequence starting with diffuse plaques, which evolve into more compact plaques and finally mature into the classic cored plaque type. A better molecular understanding of Aß pathology is crucial, as the role of Aß plaques in AD pathogenesis is under debate. Here, we studied the deposition and fibrillation of Aß in different plaque types with label-free infrared and Raman imaging. Fourier-transform infrared (FTIR) and Raman imaging was performed on native snap-frozen brain tissue sections from AD cases and non-demented control cases. Subsequently, the scanned tissue was stained against Aß and annotated for the different plaque types by an AD neuropathology expert. In total, 160 plaques (68 diffuse, 32 compact, and 60 classic cored plaques) were imaged with FTIR and the results of selected plaques were verified with Raman imaging. In diffuse plaques, we detect evidence of short antiparallel ß-sheets, suggesting the presence of Aß oligomers. Aß fibrillation significantly increases alongside the proposed plaque development sequence. In classic cored plaques, we spatially resolve cores containing predominantly large parallel ß-sheets, indicating Aß fibrils. Combining label-free vibrational imaging and immunohistochemistry on brain tissue samples of AD and non-demented cases provides novel insight into the spatial distribution of the Aß conformations in different plaque types. This way, we reconstruct the development process of Aß plaques in human brain tissue, provide insight into Aß fibrillation in the brain, and support the plaque development hypothesis.


Subject(s)
Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/diagnostic imaging , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Disease Progression , Female , Humans , Male , Plaque, Amyloid/classification , Plaque, Amyloid/metabolism , Spectroscopy, Fourier Transform Infrared , Spectrum Analysis, Raman
8.
Alzheimers Res Ther ; 12(1): 169, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33357241

ABSTRACT

BACKGROUND: We evaluated Aß misfolding in combination with Aß42/40 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 Aß folding; readouts at ≤ 1646 cm- 1 reflected low and at < 1644 cm- 1 high misfolding. We used Cox proportional hazard models to quantify Aß 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 Aß42/40 data measured by SIMOA were statistically analyzed and compared. RESULTS: All 22 patients who converted to MCI or AD-dementia within 6 years exhibited Aß 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 Aß misfolding in an age, sex, and APOEε4 model. A similar model with plasma Aß42/40 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 Aß misfolding and the Aß42/40 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.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Aged , Alzheimer Disease/diagnosis , Amyloid beta-Peptides , Biomarkers , Cognitive Dysfunction/diagnosis , Disease Progression , Female , Humans , Male , Middle Aged , Peptide Fragments , ROC Curve
9.
Biophys J ; 119(2): 349-359, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32579965

ABSTRACT

Amyloid-ß (Aß) 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 Aß 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 Aß, as confirmed by extended (1-µs-long) molecular dynamics simulations. Immuno-infrared sensor data are consistent with degradation of Aß fibril induced by EGCG and inhibition of Aß fibril and oligomer formation, as manifested by the recovery of the amide-I band of monomeric Aß, 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 Aß42 model structures, suggesting that the conformational change involves interchain hydrogen bonds in the amyloid fibrils that are broken upon binding of EGCG.


Subject(s)
Amyloid beta-Peptides , Catechin , Amyloid , Catechin/analogs & derivatives , Peptide Fragments , Protein Structure, Secondary
10.
Alzheimers Dement ; 16(2): 283-291, 2020 02.
Article in English | MEDLINE | ID: mdl-31611055

ABSTRACT

INTRODUCTION: Alzheimer's disease (AD) has a long prodromal stage and identifying high-risk individuals is critical. We aimed to investigate the ability of Aß misfolding in blood plasma, APOE4 status, and dementia risk factors to predict diagnosis of AD. METHODS: Within a community-based cohort, Aß misfolding in plasma measured by immuno-infrared sensor and APOE genotype were determined at baseline in 770 participants followed over 14 years. Associations between Aß misfolding, APOE4, and other predictors with clinical AD, vascular dementia, and mixed dementia diagnoses were assessed. RESULTS: Aß misfolding was associated with a 23-fold increased odds of clinical AD diagnosis within 14 years. No association was observed with vascular dementia/mixed dementia diagnoses. APOE4-positive participants had a 2.4-fold increased odds of clinical AD diagnosis within 14 years. DISCUSSION: Aß misfolding in blood plasma was a strong, specific risk prediction marker for clinical AD even many years before diagnosis in a community-based setting.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/blood , Apolipoprotein E4 , Biomarkers/blood , Aged , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Apolipoprotein E4/blood , Apolipoprotein E4/genetics , Case-Control Studies , Dementia, Vascular , Female , Genotype , Humans , Male , Risk Factors , Time Factors
11.
ACS Sens ; 4(7): 1851-1856, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31241315

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 Aß 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.


Subject(s)
Alzheimer Disease/diagnosis , Amyloid beta-Peptides/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/immunology , Antibodies, Immobilized/immunology , Antibodies, Monoclonal/immunology , Bacterial Proteins/chemistry , Biomarkers/cerebrospinal fluid , Germanium/chemistry , Humans , Immunoglobulin G/immunology , Peptide Fragments/immunology , Reproducibility of Results , Spectroscopy, Fourier Transform Infrared/instrumentation , Spectroscopy, Fourier Transform Infrared/methods , Staphylococcal Protein A/chemistry , tau Proteins/immunology
12.
Alzheimers Dement (Amst) ; 11: 257-263, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30911600

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-ß (Aß) 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 Aß 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 Aß and tau excluded false-positive cases which offers an overall specificity of 97%. DISCUSSION: The primary Aß-based blood biomarker funnels individuals with suspected AD for subsequent validation of the diagnosis by structure-based combined analysis of the CSF biomarkers Aß and tau. Our novel two-step recruitment strategy substantiates the diagnosis of AD with a likelihood of 29.

13.
EMBO Mol Med ; 10(5)2018 05.
Article in English | MEDLINE | ID: mdl-29626112

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-ß (Aß) 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.


Subject(s)
Alzheimer Disease/blood , Amyloid beta-Peptides/blood , Amyloid/blood , Biomarkers/blood , Aged , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/chemistry , Biomarkers/cerebrospinal fluid , Cohort Studies , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Positron-Emission Tomography/methods , Protein Structure, Secondary , ROC Curve
14.
ACS Med Chem Lett ; 8(7): 710-714, 2017 Jul 13.
Article in English | MEDLINE | ID: mdl-28740603

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 Aß 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 ß-sheet dominated to the healthy monomeric state. A similar effect was observed for congo red on pathogenic Aß isoforms from CSF. In addition, the effect of berberine on synthetic Aß1-42 is studied. Berberine seems to decelerate the aggregation process of synthetic Aß1-42 peptides.

15.
Anal Chem ; 88(5): 2755-62, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26828829

ABSTRACT

The misfolding of the Amyloid-beta (Aß) peptide into ß-sheet enriched conformations was proposed as an early event in Alzheimer's Disease (AD). Here, the Aß 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 Aß peptides extracted from the body fluid. We observed a significant downshift of the amide I band frequency of Aß peptides in Dementia Alzheimer type (DAT) patients, which indicated an overall shift to ß-sheet. The secondary structure distribution of all Aß peptides provides a better marker for DAT detection than a single Aß 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.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/chemistry , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Aged , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Female , Humans , Male , Middle Aged , Prospective Studies , Protein Structure, Secondary
16.
J Biophotonics ; 9(3): 224-34, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25808829

ABSTRACT

The secondary structure change of the Abeta peptide to beta-sheet was proposed as an early event in Alzheimer's disease. The transition may be used for diagnostics of this disease in an early state. We present an Attenuated Total Reflection (ATR) sensor modified with a specific antibody to extract minute amounts of Abeta peptide out of a complex fluid. Thereby, the Abeta peptide secondary structure was determined in its physiological aqueous environment by FTIR-difference-spectroscopy. The presented results open the door for label-free Alzheimer diagnostics in cerebrospinal fluid or blood. It can be extended to further neurodegenerative diseases. An immunologic ATR-FTIR sensor for Abeta peptide secondary structure analysis in complex fluids is presented.


Subject(s)
Amyloid beta-Peptides/chemistry , Spectroscopy, Fourier Transform Infrared/instrumentation , Water/chemistry , Animals , Biomimetics , Chick Embryo , Models, Molecular , Protein Structure, Secondary
17.
Anal Chem ; 87(14): 7467-75, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26102158

ABSTRACT

Protein immobilization studied by attenuated total reflection Fourier transform infrared (ATR-FT-IR) difference spectroscopy is an emerging field enabling the study of proteins at atomic detail. Gold or glass surfaces are frequently used for protein immobilization. Here, we present an alternative method for protein immobilization on germanium. Because of its high refractive index and broad spectral window germanium is the best material for ATR-FT-IR spectroscopy of thin layers. So far, this technique was mainly used for protein monolayers, which lead to a limited signal-to-noise ratio. Further, undesired protein-protein interactions can occur in a dense layer. Here, the germanium surface was functionalized with thiols and stepwise a dextran brush was generated. Each step was monitored by ATR-FT-IR spectroscopy. We compared a 70 kDa dextran with a 500 kDa dextran regarding the binding properties. All surfaces were characterized by atomic force microscopy, revealing thicknesses between 40 and 110 nm. To analyze the capability of our system we utilized N-Ras on mono-NTA (nitrilotriacetic acid) functionalized dextran, and the amount of immobilized Ras corresponded to several monolayers. The protein stability and loading capacity was further improved by means of tris-NTA for immobilization. Small-molecule-induced changes were revealed with an over 3 times higher signal-to-noise ratio compared to monolayers. This improvement may allow the observation of very small and so far hidden changes in proteins upon stimulus. Furthermore, we immobilized green fluorescent protein (GFP) and mCherry simultaneously enabling an analysis of the surface by fluorescence microscopy. The absence of a Förster resonance energy transfer (FRET) signal demonstrated a large protein-protein distance, indicating an even distribution of the protein within the dextran.


Subject(s)
Chemical Fractionation , Dextrans/isolation & purification , Germanium/chemistry , Green Fluorescent Proteins/chemistry , Immobilized Proteins/chemistry , Luminescent Proteins/chemistry , Dextrans/chemistry , Germanium/isolation & purification , Microscopy, Atomic Force , Models, Molecular , Molecular Structure , Particle Size , Spectroscopy, Fourier Transform Infrared , Surface Properties , Red Fluorescent Protein
18.
Chembiochem ; 15(17): 2529-34, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25256748

ABSTRACT

Protein immobilization on solid surfaces has become a powerful tool for the investigation of protein function. Physiologically relevant molecular reaction mechanisms and interactions of proteins can be revealed with excellent signal-to-noise ratio by vibrational spectroscopy (ATR-FTIR) on germanium crystals. Protein immobilization by thiol chemistry is well-established on gold surfaces, for example, for surface plasmon resonance. Here, we combine features of both approaches: a germanium surface functionalized with different thiols to allow specific immobilization of various histidine-tagged proteins with over 99% specific binding. In addition to FTIR, the surfaces were characterized by XPS and fluorescence microscopy. Secondary-structure analysis and stimulus-induced difference spectroscopy confirmed protein activity at the atomic level, for example, physiological cation channel formation of Channelrhodopsin 2.


Subject(s)
Germanium/chemistry , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Sulfhydryl Compounds/chemistry , Histidine/chemistry , Histidine/genetics , Histidine/metabolism , Models, Molecular , Molecular Structure , Rhodopsin/chemistry , Rhodopsin/metabolism , Spectroscopy, Fourier Transform Infrared
SELECTION OF CITATIONS
SEARCH DETAIL
...