DPE1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to DPEP1 and Its Antibodies

DPEP1 (dipeptidase 1) is a membrane-bound enzyme that hydrolyzes dipeptides, including leukotriene D4, and is implicated in kidney function and cancer biology . Antibodies against DPEP1 are used to study its expression, localization, and role in diseases. Two well-characterized DPEP1 antibodies include Proteintech 68081-1-Ig (mouse monoclonal) and Abcam ab121308 (rabbit polyclonal).

Proteintech 68081-1-Ig (Mouse Monoclonal)

ParameterDetails
Host Species/IsotypeMouse IgG2a
ReactivityHuman, Mouse, Rat, Pig, Rabbit
ApplicationsWB (1:5,000–1:50,000), IHC (1:500–1:2,000), IF-P (1:200–1:800), ELISA
ImmunogenDPEP1 fusion protein (Ag14591)
Molecular Weight46 kDa (observed and calculated)
Storage-20°C in PBS with 0.02% sodium azide and 50% glycerol
UniProt IDP16444
Key ValidationDetected in human testis, rabbit kidney, and rat pancreas tissues .

Abcam ab121308 (Rabbit Polyclonal)

ParameterDetails
Host Species/IsotypeRabbit IgG
ReactivityHuman
ApplicationsWB, IHC-P
ImmunogenRecombinant fragment (Human DPEP1 aa 250–400)
Validation DataWestern blot confirmed in human samples .

Western Blot (WB)

  • 68081-1-Ig: Detects DPEP1 in human testis, HepG2 cells, and rodent tissues at dilutions up to 1:50,000 .

  • ab121308: Validated in human samples with a predicted band at ~46 kDa .

Immunohistochemistry (IHC)

  • 68081-1-Ig:

    • Detects DPEP1 in human kidney and colon cancer tissues.

    • Optimal antigen retrieval: TE buffer (pH 9.0) or citrate buffer (pH 6.0) .

  • ab121308: Suitable for IHC-P but requires further validation for non-human samples .

Immunofluorescence (IF)

  • 68081-1-Ig: Validated in mouse kidney sections with a recommended dilution of 1:200–1:800 .

Comparative Analysis

FeatureProteintech 68081-1-IgAbcam ab121308
Host SpeciesMouseRabbit
ReactivityBroad (5 species)Human-specific
ApplicationsWB, IHC, IF-P, ELISAWB, IHC-P
Key StrengthCross-species validationSpecificity for human tissues
LimitationRequires titration for optimal performanceLimited validation in non-human models

Research Implications

DPEP1 antibodies enable studies on:

  • Renal Pathophysiology: DPEP1’s role in hydrolyzing nephrotoxic substrates like cisplatin .

  • Cancer Biomarkers: Overexpression in colorectal and pancreatic cancers .

  • Inflammatory Diseases: Association with leukotriene metabolism in asthma and allergies .

Validation and Quality Control

  • Proteintech 68081-1-Ig: Includes size-exclusion chromatography data confirming specificity for the 46 kDa band .

  • Abcam ab121308: Validated using recombinant protein and human tissue lysates .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
DPE1 antibody; At5g64860 antibody; MXK3.94-alpha-glucanotransferase DPE1 antibody; chloroplastic/amyloplastic antibody; EC 2.4.1.25 antibody; Amylomaltase antibody; Disproportionating enzyme antibody; D-enzyme antibody; Protein DISPROPORTIONATING ENZYME 1 antibody
Target Names
DPE1
Uniprot No.

Target Background

Function

DPE1 is a chloroplastic alpha-glucanotransferase involved in maltotriose metabolism. It likely utilizes maltotriose as a substrate to transfer a maltosyl unit from one molecule to another, resulting in the production of glucose and maltopentaose. The maltopentaose can then be further metabolized into maltose and maltotriose by beta-amylase. DPE1 plays a crucial role in the normal starch degradation process within leaves.

Gene References Into Functions
  1. AtDPE1 forms a homodimer, with each subunit containing a deep, long, and open-ended active site canyon. PMID: 26504082
Database Links

KEGG: ath:AT5G64860

STRING: 3702.AT5G64860.1

UniGene: At.7845

Protein Families
Disproportionating enzyme family
Subcellular Location
Plastid, chloroplast. Plastid, amyloplast.

Q&A

How can I verify the specificity of my antibody for the target antigen?

Antibody specificity verification requires a multi-method approach to ensure reliable experimental outcomes. Begin with western blotting to confirm the antibody recognizes a protein of the expected molecular weight. This should be complemented with immunoprecipitation followed by mass spectrometry to identify all proteins captured by the antibody. For definitive validation, perform parallel experiments with knockout/knockdown models lacking your target protein to confirm signal absence. Additionally, competitive binding assays with recombinant antigen can demonstrate specificity through signal reduction when the antibody binding site is blocked .

Cross-reactivity testing should include related proteins with similar sequences to your target antigen. In epitope mapping studies, techniques like phage display or peptide arrays can identify the specific sequence recognized by your antibody, providing further validation of specificity . When available, testing the antibody across multiple species can also demonstrate conservation of binding, which often correlates with functional relevance of the epitope.

What are the optimal storage conditions to maintain antibody functionality long-term?

Maintaining antibody functionality requires careful attention to storage conditions that prevent degradation and preserve binding capacity. Store purified antibodies at concentrations between 1-5 mg/mL in PBS buffer with stabilizing proteins (0.1-1% BSA) to prevent adsorption to container surfaces. For long-term storage, aliquot antibodies into small volumes (50-100 μL) and store at -80°C to minimize freeze-thaw cycles, as repeated freezing and thawing can cause antibody degradation and loss of binding capacity .

When working with research-grade antibodies, many researchers maintain a working stock at 4°C (with 0.02% sodium azide as preservative) for up to 1 month, while keeping the main stock frozen. Monitor antibody performance regularly through functional assays relevant to your experimental applications. Some antibody formats show better stability than others; for example, IgG antibodies generally display better stability compared to IgM antibodies due to their structural properties. Documentation of lot numbers, receipt dates, and performance in standardized assays is essential for tracking potential degradation over time .

How do epitope characteristics affect antibody binding and experimental applications?

The nature of the epitope recognized by an antibody significantly impacts its experimental utility and performance. Linear epitopes (consisting of sequential amino acids) versus conformational epitopes (formed by protein folding) determine which applications an antibody is suitable for. Antibodies recognizing linear epitopes typically perform well in western blotting where proteins are denatured, while conformational epitope-binding antibodies excel in applications using native protein structures such as immunoprecipitation or flow cytometry .

The location of the epitope on the antigen is equally critical. Epitopes in accessible regions of the native protein structure facilitate applications like immunohistochemistry and cell-surface staining. Conversely, antibodies targeting epitopes that become exposed only after protein denaturation may be limited to western blotting applications. Research has demonstrated that epitope characteristics can influence cross-reactivity patterns, with some conserved epitopes enabling cross-species reactivity while unique epitope regions provide species specificity . The development of competitive binding assays has enabled researchers to map epitopes and determine if multiple antibodies recognize the same or different regions of an antigen, which is particularly valuable for developing antibody panels for complex antigens .

What controls should be included when using antibodies for immunodetection methods?

Rigorous control implementation is essential for validating antibody-based experimental results. Primary controls must include a negative control omitting the primary antibody to assess non-specific binding of the secondary antibody or detection system. Isotype controls (non-specific antibodies of the same isotype, species, and concentration) help distinguish specific signal from binding through Fc receptors or other non-specific interactions .

For cellular or tissue-based assays, include positive control samples known to express your target protein and negative control samples that don't express it. Ideally, these would include genetic knockout/knockdown samples or cells/tissues naturally lacking your protein of interest. Competitive inhibition controls, where excess purified antigen is pre-incubated with the antibody before adding to the sample, provide evidence of binding specificity by showing reduced signal when the antibody is blocked by its target . When examining phosphorylation or other post-translational modifications, include treatment controls that modulate the modification status (e.g., phosphatase treatment to remove phosphorylation) .

How can I optimize antibody dilutions for different experimental techniques?

Systematic titration is the most reliable approach for determining optimal antibody concentrations across different applications. Begin with the manufacturer's recommended range, then perform a dilution series spanning at least one order of magnitude above and below this range. For each application, the optimal concentration balances maximum specific signal with minimal background .

For western blotting, start with dilutions between 1:500-1:5000 for primary antibodies and 1:1000-1:10000 for secondaries. In immunohistochemistry, typical starting dilutions range from 1:50-1:500 for primaries and 1:200-1:1000 for secondaries. Flow cytometry often requires higher concentrations (1:20-1:200) due to shorter incubation times. When optimizing, consider that different lots of the same antibody may require readjustment of dilutions, and sample preparation methods (fixation protocols, antigen retrieval methods) can significantly impact optimal concentrations . Document optimization parameters systematically to ensure reproducibility across experiments and to facilitate troubleshooting when new lots are obtained .

What factors influence antibody binding kinetics and how can they be optimized?

Antibody binding kinetics are governed by complex factors that can be manipulated to enhance experimental outcomes. Temperature significantly impacts binding: higher temperatures increase reaction rates but can reduce binding stability, while lower temperatures may increase specificity but require longer incubation times. Most protocols balance these considerations with room temperature or 4°C incubations .

Buffer composition plays a crucial role in optimizing antibody-antigen interactions. Ionic strength affects electrostatic interactions—high salt concentrations can reduce non-specific binding but may also weaken specific interactions. pH influences the charge state of both antibody and antigen, with most antibodies functioning optimally near physiological pH (7.2-7.4). Detergent inclusion (typically 0.05-0.1% Tween-20) reduces hydrophobic non-specific interactions, though excessive detergent can disrupt antibody binding .

Incubation time represents another critical variable. Binding follows saturation kinetics, where 80-90% of maximum binding typically occurs within the first 1-2 hours, with diminishing returns for longer incubations. For applications requiring maximum sensitivity, overnight incubations at 4°C may be beneficial. The antibody's intrinsic affinity (determined by its KD value) establishes the theoretical binding potential—higher affinity antibodies (lower KD) reach binding equilibrium faster and maintain stable binding during washing steps .

How do different antibody formats affect experimental applications and outcomes?

The structural and functional diversity of antibody formats creates distinct advantages and limitations across experimental contexts. Full-length IgG antibodies (150 kDa) provide excellent specificity and sensitivity in most applications but may have limited tissue penetration due to their size. In contrast, antibody fragments such as Fab (50 kDa) and scFv (25 kDa) offer improved tissue penetration and reduced non-specific binding through Fc receptors, though often with decreased avidity and stability .

Monoclonal antibodies provide consistent specificity to a single epitope, making them ideal for applications requiring precise targeting, such as therapeutic development or when background interference is problematic. Polyclonal antibodies recognize multiple epitopes on the target antigen, providing signal amplification that enhances detection sensitivity—particularly valuable in immunohistochemistry or when protein expression is low . Recent studies have demonstrated that recombinant antibodies offer superior reproducibility compared to hybridoma-derived monoclonals or serum-derived polyclonals, as their protein sequence remains consistent across production batches .

The choice of conjugated labels also significantly impacts experimental outcomes. Directly labeled primary antibodies simplify protocols and reduce non-specific binding but may sacrifice signal amplification. In contrast, detection systems using biotinylated antibodies with streptavidin-conjugated reporters or secondary antibody amplification can enhance sensitivity up to 10-fold, though with increased risk of background signal .

What strategies can resolve cross-reactivity issues in antibody-based experiments?

Cross-reactivity problems require systematic troubleshooting approaches to maintain experimental validity. Begin by increasing washing stringency through additional wash steps, higher detergent concentrations (0.1-0.5% Tween-20 or 0.1% Triton X-100), or higher salt concentrations (150-500 mM NaCl) in wash buffers. These modifications disrupt low-affinity non-specific interactions while preserving high-affinity specific binding .

Pre-absorption techniques can significantly reduce cross-reactivity. Incubate your antibody with tissues or cell lysates lacking your target protein but containing potential cross-reactive proteins before applying to your experimental sample. This pre-absorption depletes antibodies binding to non-target proteins. For immunohistochemistry applications, blocking with 5-10% serum from the same species as your secondary antibody prevents non-specific binding through endogenous immunoglobulins .

Epitope competition assays provide another powerful approach. By pre-incubating the antibody with excess purified antigen or synthetic peptides corresponding to the epitope region, specific binding is blocked while cross-reactive binding persists, allowing identification of non-specific signals. When persistent cross-reactivity occurs despite these measures, consider alternative antibody clones targeting different epitopes on your protein of interest or employ genetic approaches like CRISPR knockouts to definitively validate antibody specificity .

How can epitope mapping improve antibody characterization and experimental design?

Epitope mapping provides crucial insights that enhance experimental design and interpretation. Phage display techniques allow precise identification of linear epitopes by expressing peptide libraries on bacteriophage surfaces and selecting those that bind to the antibody of interest. For conformational epitopes, hydrogen-deuterium exchange mass spectrometry (HDX-MS) can identify regions protected from exchange by antibody binding. These approaches identify which regions of the antigen are recognized by the antibody, informing predictions about cross-reactivity, functionality, and application suitability .

Knowledge of the specific epitope enables rational experimental design. For instance, if an antibody targets an epitope that includes a phosphorylation site, it may only recognize the protein in specific activation states. Similarly, epitopes near protein-protein interaction domains suggest the antibody might interfere with these interactions, potentially altering cellular functions in live-cell experiments. Epitope information also facilitates the development of blocking peptides that can be used as controls to confirm signal specificity .

For therapeutic antibody development, epitope characterization is essential for predicting efficacy and specificity. Comprehensive epitope mapping through techniques like biolayer interferometry or surface plasmon resonance combined with structural analysis enables precise determination of binding kinetics and stoichiometry. This information helps predict antibody behavior in complex biological environments and guides the development of improved antibodies with enhanced specificity or affinity .

How can antibody engineering improve specificity and reduce background in challenging applications?

Advances in antibody engineering have created powerful tools for enhancing specificity in complex experimental systems. Site-directed mutagenesis of complementarity-determining regions (CDRs) can fine-tune antibody specificity by modifying amino acids critical for antigen recognition. This approach has been particularly valuable for distinguishing between highly similar proteins or protein isoforms that conventional antibodies cannot differentiate .

Framework modifications through humanization or germlining can reduce non-specific background by minimizing interactions with endogenous Fc receptors or complement proteins. These modifications involve replacing non-essential framework regions with human or germline sequences while preserving the antigen-binding regions, resulting in reduced immunogenicity and background binding .

The development of bispecific antibodies has enabled dual-epitope recognition, dramatically enhancing specificity through coincidence detection. These engineered formats require simultaneous binding to two different epitopes to generate a signal, effectively eliminating background from single-epitope cross-reactivity. Similarly, proximity-based detection systems like proximity ligation assays utilize antibody pairs that must bind near each other to generate signal, providing spatial resolution to antibody-based detection methods and further reducing background concerns .

What considerations are important when developing quantitative assays using antibodies?

Developing quantitative antibody-based assays requires careful attention to multiple factors affecting measurement accuracy. Standard curve design is fundamental—use purified recombinant proteins that closely match your experimental samples in terms of post-translational modifications and conformational state. The dynamic range should encompass expected sample concentrations, typically spanning 2-3 orders of magnitude, with standard points distributed to capture both high and low concentration regions accurately .

Antibody saturation kinetics significantly impact quantification reliability. Operate within the linear range of antibody binding where signal intensity directly correlates with antigen concentration. Excessive antigen can cause a prozone or hook effect where signal decreases despite increasing concentration. Conversely, at very low concentrations, non-specific binding may dominate signal generation. Validate that your assay maintains linearity across the concentration range of interest using dilution series of representative samples .

Matrix effects from sample composition can interfere with quantification accuracy. Components in biological samples (proteins, lipids, salts) may alter antibody binding kinetics or cause interference with detection systems. Address these issues by preparing standards in a matrix matching your samples or by sufficient sample dilution to minimize matrix effects. When developing multiplexed assays, additional validation is required to ensure antibodies do not cross-react or interfere with each other's binding or detection .

How can antibody-antigen binding kinetics be characterized and optimized for different research applications?

Understanding and optimizing antibody-antigen binding kinetics enables tailoring antibodies for specific research applications. Surface plasmon resonance (SPR) and bio-layer interferometry (BLI) provide real-time, label-free measurements of association (kon) and dissociation (koff) rates, allowing calculation of the equilibrium dissociation constant (KD). These parameters predict antibody performance in different applications—high kon values are critical for applications with short incubation times, while low koff values are essential when extensive washing is required .

Temperature dependency of binding kinetics reveals thermodynamic properties of the antibody-antigen interaction. Measuring binding constants at multiple temperatures allows calculation of enthalpy (ΔH) and entropy (ΔS) contributions to binding energy. Enthalpy-driven binding (large negative ΔH) generally indicates numerous hydrogen bonds and electrostatic interactions, creating highly specific binding that may be sensitive to buffer conditions. Entropy-driven binding often involves hydrophobic interactions that may be less specific but more tolerant of varying conditions .

Advanced kinetic characterization has revealed that many antibodies exhibit complex binding behaviors beyond simple 1:1 interactions. These include conformational changes upon binding, heterogeneous binding sites with different affinities, or avidity effects from bivalent binding. Mathematical modeling of these complex behaviors provides insights for optimizing experimental conditions—for example, selecting incubation times that favor high-affinity interactions while minimizing low-affinity cross-reactivity .

What approaches enable successful use of antibodies in multiplex detection systems?

Multiplex antibody applications require specialized strategies to maintain specificity while detecting multiple targets simultaneously. Species diversification represents a fundamental approach—selecting primary antibodies from different host species (mouse, rabbit, goat, etc.) allows discrimination through species-specific secondary antibodies. This approach typically limits multiplexing to 3-4 targets before cross-reactivity becomes problematic .

Isotype diversification offers another dimension for multiplexing within the same host species. Using primary antibodies of different isotypes (IgG1, IgG2a, IgG2b, etc.) with isotype-specific secondary antibodies enables detection of additional targets. Combining species and isotype diversification can extend multiplexing capability to 6-8 targets in carefully optimized systems .

Advanced multiplexing approaches incorporate spectral unmixing algorithms to distinguish overlapping fluorophore emissions, expanding the number of detectable targets by resolving signals that traditional bandpass filters cannot separate. This technology has enabled simultaneous detection of 30+ targets in specialized flow cytometry and imaging systems. For mass cytometry (CyTOF) applications, antibodies conjugated to isotopically pure metals can distinguish 40+ targets without signal overlap, revolutionizing high-dimensional cellular analysis .

How can computational methods enhance antibody validation and application development?

Computational approaches have transformed antibody research through enhanced prediction and validation capabilities. Sequence-based epitope prediction algorithms analyze antigen sequences to identify regions likely to be immunogenic and surface-exposed. These predictions guide epitope selection for antibody development, focusing efforts on regions most likely to yield functional antibodies. Machine learning algorithms trained on antibody-antigen crystal structures can predict binding affinities and cross-reactivity potential from sequence data alone .

Database integration has become essential for comprehensive antibody validation. Cross-referencing experimental results with proteomics databases validates detection of proteins at expected molecular weights and expression patterns. Specialized antibody validation repositories containing validated application data and known cross-reactivity profiles help researchers select appropriate antibodies and predict potential pitfalls .

Network analysis approaches model antibody binding in the context of the entire proteome, predicting off-target binding based on sequence or structural similarity to the intended target. These analyses identify potential cross-reactive proteins that should be included in validation experiments. The integration of these computational approaches with experimental validation creates a more robust framework for ensuring antibody specificity and functionality across diverse applications .

What factors should be considered when using antibodies for in vivo applications?

In vivo antibody applications present unique challenges requiring specialized considerations beyond in vitro experiments. Pharmacokinetic properties fundamentally determine antibody distribution and persistence in living systems. The antibody's molecular weight, charge, glycosylation pattern, and binding to FcRn receptors collectively influence circulation half-life, which typically ranges from hours for antibody fragments to weeks for full-length IgG antibodies. These properties must be optimized for the specific application—imaging may require rapid clearance to reduce background, while therapeutic applications typically benefit from extended circulation .

Tissue penetration barriers significantly impact antibody distribution in solid tissues. The 150 kDa size of intact IgG molecules limits extravasation and diffusion through tissues. Various strategies address this limitation, including using smaller antibody fragments (Fab, scFv), enhancing vascular permeability with adjuvants, or employing site-specific delivery methods. Recent studies have demonstrated that engineered antibodies with intermediate sizes (~80 kDa) often achieve optimal balance between tissue penetration and retention .

Immunogenicity represents a critical concern for repeated antibody administration in vivo. Anti-drug antibody (ADA) responses can neutralize the administered antibody and potentially cause adverse reactions. Strategies to mitigate immunogenicity include humanization of non-human antibodies, removing T-cell epitopes through sequence engineering, and PEGylation to mask immunogenic regions. For animal model experiments, species matching (using mouse antibodies in mice, etc.) reduces but doesn't eliminate immunogenicity concerns. Thorough monitoring for ADAs through serial sampling is essential for interpreting in vivo antibody studies, particularly in longitudinal experiments .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.