Synonym |
Biotinylated Aβ-Protein (1-40) |
Species |
Human |
Protein Accession |
P05067 |
Purity |
≥ 95% |
Endotoxin Level |
< 1 EU/μg |
Biological Activity |
Not specified |
Expression System |
Escherichia coli |
Fusion Tag |
Not specified |
Predicted Molecular Mass |
4417 Da |
Formulation |
Supplied as a lyophilized powder |
Reconstitution |
Reconstitute in PBS or deionized water |
Storage & Stability |
Store at -20°C upon arrival. Avoid repeated freeze-thaw cycles. |
FAQ
What is Biotinyl-Amyloid β-Protein (1-40) and how does it work in research applications?
Biotinyl-Amyloid β-Protein (1-40) is a modified form of the amyloid beta (Aβ) peptide, which is
primarily associated with Alzheimer's disease research. The peptide consists of the first 40 amino acids
of the amyloid beta sequence and is biotinylated, meaning that a biotin molecule is covalently attached
to it. This modification facilitates the detection, capture, and isolation of the peptide when used in
various experimental settings. Biotin is a small, naturally occurring molecule that binds strongly to
streptavidin and avidin, proteins commonly used in research for pulling down biotinylated molecules.
This strong binding can be used in a variety of assays, including Western blotting, immunoassays, and
pull-down experiments. In research, the Biotinyl-Amyloid β-Protein (1-40) is often utilized to
understand the aggregation properties of amyloid beta peptides which form the core of the amyloid
plaques found in the brains of Alzheimer's patients. Studying these properties can provide insights into
the pathological process of fibrillogenesis where soluble Aβ peptides aggregate and form insoluble
plaques. This process is a key focus of Alzheimer's research because disrupting or preventing Aβ
aggregation is a potential therapeutic strategy for treating or preventing the disease. The
biotinylation of the peptide adds a convenient handle for studying these interactions. Researchers can
immobilize the peptide on surfaces coated with avidin or streptavidin, allowing for the real-time
analysis of oligomerization and aggregation using techniques like atomic force microscopy or surface
plasmon resonance. Moreover, biotinylated Aβ (1-40) is advantageous in high-throughput screening assays
for potential therapeutic candidates that can inhibit Aβ aggregation. In these assays, biotinylated
peptides allow for the efficient and reproducible capture of peptides for subsequent analysis. By
tagging amyloid beta with biotin, it becomes easier for researchers to perform detailed analyses of
amyloidogenic pathways and potential inhibitors, thereby providing a strategic tool for Alzheimer's
disease research that may lead to the development of effective therapeutic agents.
Why is
biotinylation used for Amyloid β-Protein (1-40) and what are its benefits in experimental protocols?
Biotinylation of Amyloid β-Protein (1-40) is a strategic modification employed to facilitate
multiple experimental procedures, enhancing both the efficiency and sensitivity of various assays. The
primary advantage of using biotinylation is the ability to leverage the strong affinity between biotin
and avidin or streptavidin proteins. This interaction is one of the strongest non-covalent bonds in
nature, which makes it a particularly reliable and robust method for molecular capture and detection.
Biotinylated molecules can be easily detected or affinity-captured using avidin- or streptavidin-coated
surfaces or beads, thereby simplifying the separation and purification procedures necessary in many
biochemical assays. In terms of practical applications, biotinylation allows for enhanced versatility in
numerous experimental protocols. For instance, availing the strong biotin-avidin interaction enables
researchers to immobilize biotinylated Aβ peptides onto solid phases for use in enzyme-linked
immunosorbent assays (ELISAs) or other binding assays. These applications are crucial for determining
the kinetics of peptide aggregation, binding specificity with other molecular entities, or screening of
drug candidates that might inhibit amyloid aggregation or toxicity. Furthermore, biotinylated peptides
can be used in pull-down experiments to identify peptide interactions with other proteins or small
molecules in complex biological mixtures. This application is particularly relevant in understanding
protein-protein interactions or even peptide-DNA/RNA interactions within the scope of neurodegenerative
research. Another essential benefit of biotinylation is its minimal impact on the physiological
properties of the molecule to which it is attached. The small size of the biotin moiety does not
significantly alter the structural or functional properties of the peptide, allowing the amyloid β
peptide to retain its biological activity and aggregation propensity. This retention of natural
properties is vital when studying the pathophysiological processes involved in amyloid aggregation and
Alzheimer's disease, as the biotinylated peptides provide a faithful representation of the non-modified
peptides within experimental settings. Additionally, biotinylation extends the functional application of
the peptides for diagnostic purposes. Biotinylated Aβ (1-40) can be used to generate diagnostic tools or
biosensors for the early detection of Alzheimer's disease markers in biological samples, providing both
research and clinical diagnostic opportunities. In summary, incorporating biotinylation into amyloid β
peptide research provides substantial experimental flexibility and robustness, making it a powerful tool
in advancing understanding and detection methodologies for amyloidogenic processes and potential
therapeutic interventions.
How is Biotinyl-Amyloid β-Protein (1-40) used in Alzheimer's disease
research, and what discoveries have been made using it?
Biotinyl-Amyloid β-Protein (1-40) serves as
a pivotal tool in Alzheimer's disease research, contributing to our understanding of the molecular
mechanisms underlying amyloid fibril formation and accumulation. Alzheimer's disease is characterized by
the accumulation of amyloid plaques in the brain, primarily composed of amyloid β (Aβ) peptides. Among
these, the Aβ (1-40) variant is one of the more prevalent forms and is integral to studies focused on
understanding the formation and propagation of amyloid plaques. Using Biotinyl-Amyloid β-Protein (1-40)
has advanced research by allowing scientists to explore several key aspects of amyloid behavior. Due to
the biotinylation, these peptides are particularly useful in binding assays, where they can be
immobilized to study their interaction with other molecules, such as antibodies, small inhibitory
molecules, or even other proteins. This has enabled researchers to characterize potential inhibitors of
Aβ aggregation, providing a pathway toward developing therapeutic agents that may halt or slow down
plaque formation in the Alzheimer's affected brain. Several important discoveries have emerged from
studies using this biotinylated peptide. One such discovery involves the identification of small
molecules and peptides that can inhibit Aβ oligomerization to prevent plaque formation. By screening
libraries of compounds with biotinylated Aβ, researchers can more readily identify those that bind
directly to Aβ and modify its aggregation pathway. These inhibitors are critical in the development of
drugs aimed at modulating Aβ aggregation, providing a targeted approach to ameliorate one of the
neuropathological hallmarks of Alzheimer's disease. Additionally, biotinylated Aβ has been instrumental
in elucidating the structure-activity relationships (SAR) within amyloid fibrils. Techniques such as
cryo-electron microscopy and nuclear magnetic resonance (NMR) spectroscopy, used alongside biotinylated
Aβ, have led to greater insights into the structural dynamics of Aβ fibrils. Understanding these
structures at high resolution allows for the rational design of molecules that can interfere directly
with fibril formation or destabilize existing fibrils. Furthermore, using Biotinyl-Amyloid β-Protein
(1-40) in conjunction with animal models has helped in understanding the in vivo dynamics of Aβ
aggregation, plaque formation, and the accompanying neuroinflammatory processes. Researchers can track
how the peptide distributes and propagates through the model system, offering insights into how amyloid
deposition contributes to cognitive decline. Overall, biotinylated Aβ peptides remain a versatile and
essential resource in Alzheimer's research, driving discoveries that enhance our understanding of the
disease pathology and aiding in the pursuit of effective therapeutic interventions.
What
challenges are commonly associated with using Biotinyl-Amyloid β-Protein (1-40) in experiments, and how
can they be addressed?
While Biotinyl-Amyloid β-Protein (1-40) offers many advantages for research,
using it comes with its own set of challenges that can potentially impact experimental outcomes. One of
the main challenges is ensuring the integrity and stability of the peptide during experiments. Amyloid β
peptides are inherently prone to aggregation, which is exacerbated by their biochemical properties and
experimental conditions, such as temperature, pH, and peptide concentration. Researchers must carefully
control these factors to prevent premature aggregation, which can lead to inconsistent or misleading
results. Another significant challenge involves preventing non-specific binding, a common issue
encountered due to the biotin-streptavidin interaction's high affinity and ubiquity. Non-specific
interactions could result in elevated background noise in assays or unspecific signal detection. To
mitigate this, researchers often employ blocking strategies using biotin-free buffers or incorporating
excess free biotin before adding streptavidin to block potential non-specific sites. Calibration of
these reagents is essential to balance out specific versus non-specific interactions effectively.
Another problem is the potential alteration of biotinylation on the peptide's physicochemical
properties. While biotin is generally inert, its incorporation might, in some cases, influence the
peptide's solubility or folding, thereby impacting its physiological behavior. Peptide scientists
usually conduct calibration experiments—deploying a non-biotinylated form of the peptide in parallel—to
establish baselines for comparison and ensure that the biotin's presence does not dramatically skew
data. Furthermore, the biotinylation process needs to be optimal to ensure uniform labeling, especially
when scaled for high-throughput analyses. Utilizing commercial preparations with verified degrees of
biotinylation helps address issues related to variability in peptide labeling. Another relevant area of
concern includes handling the storage and handling of the peptide. Given the peptide's tendency to
aggregate under suboptimal storage conditions, adhering to recommended storage guidelines is crucial.
Peptides should be stored in lyophilized form at low temperatures and reconstituted with the correct
solvents just prior to use. Use of antioxidants or buffering agents may also support peptide stability.
Lastly, data interpretation from experiments involving biotinylated peptides requires careful
consideration because the presence of biotin might impact readouts, depending on the assay design.
Thoroughly validating experimental setups and confirming findings with orthogonal approaches are
recommended to mitigate misinterpretations. Addressing these challenges involves maintaining rigorous
experimental protocols, optimizing buffer systems, and implementing comprehensive controls and
validations to ensure that the experimental artifacts do not overshadow actual scientific discoveries.
Researchers are encouraged to adapt protocols that account for these nuances, ensuring the reliable use
of Biotinyl-Amyloid β-Protein (1-40) in advancing Alzheimer's research.
How does the use of
Biotinyl-Amyloid β-Protein (1-40) facilitate drug discovery in the context of neurodegenerative
diseases?
In the realm of neurodegenerative disease research, particularly Alzheimer's disease,
Biotinyl-Amyloid β-Protein (1-40) plays a critical role in facilitating the drug discovery process.
Biotinylated proteins like Aβ (1-40) are instrumental in high-throughput screening platforms designed to
identify small molecules or biological agents that can interfere with Aβ aggregation or toxicity, one of
the main pathological features of Alzheimer's. Using biotinylated peptides provides a streamlined
approach to evaluating the efficacy of numerous compounds in inhibiting amyloid plaque formation or
disrupting existing amyloid structures, both potential therapeutic strategies to diminish
neurodegeneration. The biotin group allows the amyloid peptide to be immobilized on various surfaces
without altering its biological activity. This immobilization is particularly advantageous in drug
screening assays, where multiple drug candidates can be assessed for their interaction with the amyloid
peptide and their ability to block pathological interactions or conformational changes associated with
disease progression. The peptides' immobilization facilitates techniques such as enzyme-linked
immunosorbent assays (ELISAs) or fluorescence polarization, which measure binding interactions and
thereby help to identify compounds with therapeutic potential. Once potential inhibitors are identified
through high-throughput screens, the biotinylated peptides further aid in characterizing these drugs'
mechanisms of action. For example, researchers can perform binding kinetic studies to determine how
tightly and specifically a drug candidate interacts with the Aβ (1-40) aggregates, utilizing surface
plasmon resonance (SPR) or biolayer interferometry (BLI). These methods rely on the consistent and
robust immobilization of biotinylated peptides to accurately track interaction dynamics. Additionally,
researchers might employ competitive binding assays, facilitated by the biotin tag, to detail how new
drugs displace conventional biotin-binding interactions, offering insights into competitive inhibition
properties. Furthermore, the versatility of biotinylated peptides enables their use in structural
biology, aiding in the elucidation of the molecular structures of peptide-drug complexes via techniques
such as X-ray crystallography or NMR. Understanding these structures can inform the rational design and
optimization of drug candidates, as researchers adjust chemical moieties to enhance binding affinity,
selectivity, and functional efficacy while mitigating potential off-target effects. The eventual goal is
to develop therapeutic agents that are not only potent inhibitors of amyloid aggregation but are also
bioavailable, safe, and effective in clinical settings. Moreover, the application of biotinylated
peptides in patient-derived cellular models of Alzheimer's enables a closer evaluation of drug efficacy
in a more physiologically relevant context. The ability to evaluate drug effects on amyloid pathology
within living systems ensures a wider pharmacodynamic and pharmacokinetic understanding necessary for
eventual clinical application. Thus, Biotinyl-Amyloid β-Protein (1-40) serves as an essential component
in the drug discovery process for neurodegenerative diseases, supporting the development of novel
therapeutic interventions targeting amyloid pathologies.
What role does Biotinyl-Amyloid
β-Protein (1-40) play in diagnostics, and how might it be integrated into clinical practice?
Biotinyl-Amyloid β-Protein (1-40) holds potential beyond basic research, extending into the
diagnostic realm, particularly concerning Alzheimer's disease. The pathological aggregation of amyloid
beta peptides is a hallmark of Alzheimer's, and early detection of these peptides in patients can
facilitate timely diagnosis and intervention, potentially slowing disease progression. Biotinylated
peptides serve as crucial tools in developing diagnostic assays designed to detect low concentrations of
amyloid beta in biological samples such as cerebrospinal fluid (CSF) or blood serum. The robust
interaction between biotin and avidin or streptavidin makes biotinylated peptides ideal candidates for
highly sensitive detection systems capable of picking up minor changes in amyloid beta levels that might
indicate disease onset or progression. The peptides can be immobilized on assay platforms to capture
amyloid-specific antibodies from patient samples, thus establishing a basis for immunoassay techniques
like ELISA or lateral flow assays used in diagnostic testing. These immunoassays are integral to
identifying the presence and concentration of amyloid beta, facilitating early diagnosis of Alzheimer's
when therapeutic interventions are more likely to be effective. Integrating Biotinyl-Amyloid β-Protein
(1-40) into clinical practice presents an opportunity for developing robust diagnostic platforms that
can be employed routinely. Such platforms can be tailored for use in hospital laboratories or
potentially adapted for point-of-care testing, a critical aspect for widespread and accessible
Alzheimer's disease screening. Additionally, the sensitivity and specificity offered by biotinylated
peptides make them suitable for use in conjunction with imaging biomarkers. Techniques like positron
emission tomography (PET) utilize radiolabeled ligands that bind amyloid plaques; biotinylated peptides
can aid in the pre-screening or correlation with image-based findings, providing a more comprehensive
view of amyloid pathology within patients. Moreover, biotinylated peptides are amenable to multiplexed
diagnostics, where the simultaneous detection of multiple biomarkers is advantageous for comprehensive
disease profiling. This multiplexing capability expands the potential diagnostic applications beyond
Alzheimer's alone, possibly serving as a model for other amyloidogenic diseases where peptide
aggregation plays a critical role. Overall, while the transition of Biotinyl-Amyloid β-Protein (1-40)
from lab-based research to clinical diagnostics holds promise, it requires rigorous validation and
standardization processes, including understanding any potential variances introduced by biotinylation
that may affect biomarker fidelity. Collaborative efforts across research institutions, clinical labs,
and industry stakeholders are essential to ensure that technologies developed harnessing
Biotinyl-Amyloid β-Protein (1-40) can meet clinical diagnostic standards and be effectively translated
into routine patient care. The integration of such diagnostic strategies into clinical practice
signifies a move towards precision medicine, where early intervention strategies could profoundly impact
patient outcomes, reduce the societal burden of Alzheimer's disease, and improve the quality of life for
individuals at risk or suffering from neurodegenerative conditions.