ALPHA-ADR Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Specificity

α-AR antibodies specifically bind to one or more subtypes of α-adrenergic receptors:

  • α1A-AR: Predominantly expressed in the heart, brain, and vascular smooth muscle .

  • α1B-AR: Found in liver, spleen, and kidneys .

  • α1D-AR: Localized to vascular smooth muscle and involved in blood pressure regulation .

Structurally, these antibodies are immunoglobulins composed of two heavy (α, γ, δ, ε, μ) and two light (κ, λ) chains, forming antigen-binding sites at their N-termini .

Key Validation Criteria

ParameterMethodologiesExample Data
SpecificitysiRNA knockdown, blocking peptides, overexpression models62–97% signal reduction in α1A-AR siRNA-treated cells vs. controls .
AffinitySurface plasmon resonance (SPR)Kd ∼50 nM for α1A-AR autoantibodies binding to extracellular loop peptides .
Functional ActivityCardiomyocyte chronotropy assays, vasoconstriction studiesDose-dependent ↑ in cardiomyocyte beating rate (1:200 dilution) .

Pathological Roles of α-AR Autoantibodies

  • Hypertension: Agonistic autoantibodies against α1-AR subtypes (e.g., α1A, α1D) are detected in 44% of primary hypertension cases, inducing Ca²⁺-dependent vasoconstriction and protein kinase C activation .

  • Cardiac Remodeling: α1A-AR antibodies activate ERK1/2 and upregulate PLA2-IIA and Cacna1c genes, promoting cardiomyocyte hypertrophy .

Subtype-Specific Effects

SubtypeKnockout (KO) PhenotypeOverexpression Phenotype
α1A-ARImpaired cardiac stress adaptation Adaptive cardiac hypertrophy, ischemic protection
α1B-ARReduced baroreflex response, insulin resistance Maladaptive hypertrophy, fibrosis
α1D-ARHypotension, reduced locomotion Sustained blood pressure elevation

Therapeutic Applications

  • Vaccine Development: The ADRQβ-004 vaccine targeting α1D-AR reduced blood pressure in hypertensive models for >10 weeks without immune injury .

  • Immunoadsorption Therapy: Removal of α1-AR autoantibodies via immunoadsorption lowered blood pressure in refractory hypertension .

Challenges and Limitations

  • Cross-Reactivity: Weak binding to non-target peptides (e.g., α1A-AR antibodies vs. α1D-AR epitopes) .

  • Validation Gaps: Only 70% of commercial antibodies meet stringent specificity benchmarks .

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
ALPHA-ADR antibody; At5g22770 antibody; MDJ22.19AP-2 complex subunit alpha-1 antibody; Adaptor protein complex AP-2 subunit alpha-1 antibody; Adaptor-related protein complex 2 subunit alpha-1 antibody; Alpha-adaptin 1 antibody; Clathrin assembly protein complex 2 alpha large chain 1 antibody; At-a-Ad antibody; At-alpha-Ad antibody
Target Names
ALPHA-ADR
Uniprot No.

Target Background

Function
ALPHA-ADR Antibody targets a subunit of the adaptor protein complex 2 (AP-2). Adaptor protein complexes play a crucial role in protein transport through transport vesicles, facilitating various membrane traffic pathways. These complexes are essential components of vesicle coats, actively participating in cargo selection and vesicle formation. AP-2 is specifically involved in clathrin-dependent endocytosis, a process where cargo proteins are incorporated into vesicles enveloped by clathrin (clathrin-coated vesicles, CCVs). These CCVs are subsequently destined for fusion with the early endosome. Notably, the complex exhibits binding affinity towards polyphosphoinositides.
Database Links

KEGG: ath:AT5G22770

STRING: 3702.AT5G22770.1

UniGene: At.46641

Protein Families
Adaptor complexes large subunit family
Subcellular Location
Membrane, coated pit; Peripheral membrane protein; Cytoplasmic side.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : Western Blot

Sample type: Cells

Review: The cells were incubated with mithramycin A (25 nM) for 30 min prior to high glucose treatment (25 mM) for 24 h. PICALM, AP2A1, CHC, and β-actin were subjected to western blot. n = 5 from independent experiments. *p < 0.05 vs. control, #p < 0.05 vs. high glucose.

Q&A

What are alpha-adrenergic receptor (ALPHA-ADR) antibodies and what receptor subtypes do they target?

Alpha-adrenergic receptor antibodies are immunological tools designed to detect and quantify alpha-adrenergic receptors in biological samples. These receptors are divided into two main families: alpha-1 and alpha-2 adrenergic receptors, each with multiple subtypes. Alpha-1 receptors include alpha-1A, alpha-1B, and alpha-1D subtypes, while alpha-2 receptors include alpha-2A, alpha-2B, and alpha-2C subtypes. Each receptor subtype has distinct physiological functions and tissue distribution patterns. Alpha-1D adrenergic receptors mediate their effects through the influx of extracellular calcium . In contrast, alpha-2 adrenergic receptors mediate catecholamine-induced inhibition of adenylate cyclase through G protein signaling . When selecting antibodies for research, it's crucial to consider which specific receptor subtype you need to target.

How do I validate the specificity of commercial ALPHA-ADR antibodies?

Validating antibody specificity is critical given documented issues with commercial ALPHA-ADR antibodies. The gold standard approach involves using knockout (KO) animal models where the gene for the specific receptor subtype has been deleted. A specific antibody should show immunoreactivity in wild-type samples but not in the corresponding KO samples. Research published in 2008 evaluated ten commercial antibodies for alpha-1 adrenergic receptor subtypes using western blot with tissues from wild-type mice and mice with knockout of one or all three alpha-1 subtypes. None of the tested antibodies demonstrated specificity for the targeted alpha-1 receptor subtypes .

Alternative validation approaches include:

  • Competitive binding assays with the immunizing peptide

  • Sibling selectivity testing (comparing reactivity across closely related receptor subtypes)

  • Transfection studies with receptor-expressing and non-expressing cell lines

  • Correlation with mRNA expression data

  • Multiple antibody approach (using different antibodies targeting different epitopes)

Remember that blocking peptide controls only confirm specificity for the immunizing peptide, not necessarily for the target receptor protein itself .

What techniques are commonly used with ALPHA-ADR antibodies in research?

ALPHA-ADR antibodies can be employed in various experimental techniques:

  • Immunohistochemistry/Immunofluorescence (IHC/IF): For localizing receptors in tissue sections. Alpha-1D adrenergic receptor antibodies have been validated for IHC-P (paraffin-embedded sections), as demonstrated in human prostate tissue .

  • Western blotting (WB): For detecting receptor proteins in tissue/cell lysates. Alpha-2A adrenergic receptor antibodies have been validated for western blotting of human pancreas samples .

  • Immunoprecipitation (IP): For isolating receptor proteins from complex mixtures.

  • Flow cytometry: For quantifying receptor expression on cell surfaces.

  • ELISA: For quantitative detection of solubilized receptors.

For each technique, optimization is essential. For western blotting, consider variables such as protein loading amount, primary and secondary antibody concentrations, incubation duration and temperature, and denaturing versus non-denaturing conditions .

Why might I observe discrepancies between ALPHA-ADR antibody results and functional studies?

Discrepancies between antibody-based detection and functional assays are common and may arise from several factors:

  • Antibody specificity issues: As documented in literature, many commercial antibodies for alpha-1 adrenergic receptors lack specificity, potentially detecting unrelated proteins . This fundamental issue undermines the correlation between antibody labeling and true receptor expression.

  • Receptor conformational states: Antibodies may preferentially recognize specific receptor conformations (active, inactive, or intermediate states) that don't correlate with functional measurements.

  • Receptor internalization and trafficking: Surface expression detected by antibodies may not reflect the functional receptor pool.

  • Post-translational modifications: Modifications affecting receptor function may alter antibody epitope accessibility.

  • Receptor-protein interactions: Interacting proteins may mask antibody epitopes while influencing receptor function.

When confronted with discrepancies, consider implementing complementary approaches:

  • Radioligand binding assays (currently the most reliable approach for quantifying alpha-1 adrenergic receptor subtype proteins)

  • RT-PCR for mRNA expression analysis

  • Pharmacological profiling with subtype-selective drugs

  • CRISPR-mediated receptor knockout controls

How do I address cross-reactivity issues with ALPHA-ADR antibodies?

Cross-reactivity remains a significant challenge with alpha adrenergic receptor antibodies. To address this issue:

  • Perform comprehensive validation: Test antibodies against tissues from receptor knockout models whenever possible . This approach provides the most definitive evidence of specificity.

  • Include appropriate negative controls: Use tissues or cells known not to express the target receptor or samples where the receptor has been knocked down.

  • Employ multiple antibody approach: Use antibodies targeting different epitopes on the same receptor. Consistent results across different antibodies provide stronger evidence of specificity.

  • Correlation testing: Compare antibody-based detection with mRNA expression patterns across tissues.

  • Competitive binding: Pre-incubation with the immunizing peptide should eliminate specific signals but leave non-specific signals intact.

  • Careful blot interpretation: For western blotting, verify that observed bands match the predicted molecular weight of the target receptor. Alpha-2A adrenergic receptors, for example, have a predicted band size of 48 kDa .

Research has shown that none of ten commonly cited commercial antibodies for alpha-1 adrenergic receptor subtypes demonstrated specific reactivity to their intended targets, highlighting the critical need for validation .

What strategies can improve detection sensitivity when working with low-abundance ALPHA-ADR expression?

Alpha adrenergic receptors are often expressed at relatively low levels, presenting detection challenges. Methodological approaches to enhance sensitivity include:

  • Sample enrichment techniques:

    • Membrane fraction isolation

    • Immunoprecipitation prior to western blotting

    • Signal amplification methods (tyramide signal amplification for IHC)

  • Optimized fixation and antigen retrieval for IHC:

    • Test multiple fixatives (formalin, Bouin's, acetone)

    • Compare different antigen retrieval methods (heat-induced, enzymatic)

  • Enhanced detection systems:

    • High-sensitivity ECL substrates for western blotting

    • Polymer detection systems for IHC

    • Fluorescent secondary antibodies with signal amplification

  • Technical optimization:

    • Increase primary antibody concentration and incubation time

    • Optimize blocking conditions to reduce background while preserving specific signal

    • Use highly sensitive imaging systems

  • Alternative approaches when antibody detection fails:

    • Radioligand binding assays remain the gold standard for receptor quantification

    • Consider epitope tagging approaches in experimental systems

For heart tissue, where alpha-1 adrenergic receptors are present at approximately 15 fmol/mg protein (with 30% alpha-1A and 70% alpha-1B), maximizing sensitivity is particularly important. Brain tissue contains higher levels (approximately 140 fmol/mg protein) with a different distribution pattern (55% alpha-1A, 35% alpha-1B, and 10% alpha-1D) .

How can I design studies to investigate ALPHA-ADR antibody-mediated signaling pathway activation?

Investigating antibody-mediated signaling pathway activation requires careful experimental design:

  • Pathway-specific readouts:

    • For alpha-1 receptors: Monitor calcium flux using fluorescent indicators (Fura-2, Fluo-4)

    • For alpha-2 receptors: Measure cAMP levels using ELISA or reporter systems

  • Comparative analysis with known agonists:

    • For alpha-2A receptors, compare antibody effects with the known agonist potency order: oxymetazoline > clonidine > epinephrine > norepinephrine > phenylephrine > dopamine > p-synephrine > p-tyramine > serotonin = p-octopamine

  • Antagonist blockade experiments:

    • For alpha-2A receptors, use antagonists following their rank order of potency: yohimbine > phentolamine = mianserine > chlorpromazine = spiperone = prazosin > propranolol > alprenolol = pindolol

  • Time-course analysis:

    • Track signaling events at multiple timepoints to distinguish between direct receptor activation and secondary effects

  • Dose-response relationships:

    • Establish the relationship between antibody concentration and signaling response to assess specificity

  • Knockout/knockdown controls:

    • Use cells with receptor depletion to confirm signaling specificity

  • Downstream effector analysis:

    • Monitor multiple pathway components (e.g., PKC activation, ERK phosphorylation) to characterize the full signaling cascade

Remember that some commercial antibodies may have partial agonist or antagonist properties themselves, potentially complicating the interpretation of results.

What are the considerations for designing antibodies with reduced anti-drug antibody (ADA) reactivity?

Designing antibodies with reduced anti-drug antibody reactivity is critical for therapeutic applications. Key considerations include:

  • Identification of problematic regions:

    • When reformatting antibodies to single-chain variable fragment format, the former variable/constant domain interface of the heavy chain becomes accessible for preexisting anti-drug antibody binding

    • This exposed region contains a previously hidden hydrophobic patch that can trigger immune responses

  • Strategic mutation approaches:

    • Introduce mutations in the exposed hydrophobic regions

    • Focus on threonine residues Thr101 and Thr146 in the variable heavy domain, which are critical for eliminating PE ADA reactivity

    • Aim to reduce hydrophobicity while maintaining structural integrity

  • Computational screening methods:

    • Use in silico approaches to pinpoint key residues for mutation

    • Evaluate designed molecules computationally before experimental testing

    • This approach can significantly reduce the number of molecules that need experimental characterization

  • Comprehensive biophysical characterization:

    • Ensure mutations don't compromise binding affinity, stability, or pharmacodynamic properties

    • Apply multiple biophysical methods to verify structural integrity

  • Balance between reducing immunogenicity and maintaining function:

    • Monitor how mutations affect binding to the target antigen

    • Assess potential impacts on manufacturability and stability

This approach has important implications for optimizing early drug development for antibody fragment-based therapeutics, potentially reducing immunogenicity risks while preserving therapeutic efficacy .

How do I develop and validate assays for detecting anti-drug antibodies against ALPHA-ADR antibodies?

Developing valid assays for anti-drug antibody (ADA) detection against alpha-adrenergic receptor antibodies requires a systematic approach:

  • Risk-based assessment:

    • Evaluate the potential consequences of immune responses to the therapeutic protein

    • Design immunogenicity tests to detect antibodies that could mediate unwanted biological or physiological consequences

  • Tiered testing strategy:

    • Screening assays (binding antibody assays): Detect antibodies that bind to the therapeutic protein

    • Confirmatory assays: Establish specificity through competition with the therapeutic protein

    • Titration assays: Characterize the magnitude of the ADA response, which may correlate better with clinical impact than mere incidence

    • Neutralization assays: Assess ADA for ability to interfere with target interactions

  • Critical assay parameters to optimize:

    • Sensitivity: Ability to detect low levels of ADA

    • Specificity: Ability to distinguish true ADA from non-specific binding

    • Drug tolerance: Ability to detect ADA in the presence of circulating drug

    • Reproducibility: Consistency across multiple runs and operators

  • Controls and reference materials:

    • Positive control antibodies with known characteristics

    • Negative control samples from drug-naïve individuals

    • System suitability controls to ensure assay performance

  • Validation considerations:

    • Cut-point determination through statistical analysis of drug-naïve samples

    • Precision assessment through intra-assay and inter-assay variability testing

    • Selectivity evaluation across different matrices and potential interfering substances

This comprehensive approach ensures that immunogenicity assessments provide meaningful data to evaluate the potential impact of ADA on pharmacokinetics, pharmacodynamics, safety, and efficacy of alpha-adrenergic receptor antibody therapeutics .

How do I address inconsistent results when using different batches of ALPHA-ADR antibodies?

Batch-to-batch variability is a significant challenge when working with alpha-adrenergic receptor antibodies. When facing inconsistent results:

  • Implement lot-specific validation:

    • Validate each new lot against your established positive controls

    • Document lot numbers in all experimental records

    • Consider testing multiple lots in parallel on the same samples

  • Maintain reference standards:

    • Store aliquots of well-characterized positive samples

    • Use these standards when testing new antibody lots

    • Create internal reference controls for quantitative comparisons

  • Normalize signal intensity:

    • Use loading controls (for western blots) or housekeeping proteins

    • Consider normalizing to receptor standard curves when possible

    • Implement tissue/sample normalization protocols

  • Antibody storage and handling:

    • Follow manufacturer's recommendations precisely

    • Aliquot antibodies upon receipt to minimize freeze-thaw cycles

    • Monitor storage conditions (temperature logs for freezers)

  • Standardize experimental protocols:

    • Use detailed standard operating procedures

    • Control for variables like incubation times and temperatures

    • Standardize sample preparation methods

Research has documented issues with multiple lot numbers of alpha-1 adrenergic receptor antibodies (sc-1475, sc-1476, sc-1477, and ab15851), indicating this is a widespread issue . When critical experiments are planned, consider reserving sufficient antibody from a single, validated lot.

What controls should I include when designing experiments with ALPHA-ADR antibodies?

Robust control systems are essential for reliable alpha-adrenergic receptor antibody research:

  • Biological controls:

    • Positive control tissues/cells known to express the target receptor

    • Negative control tissues/cells lacking the target receptor

    • Ideally, tissues from knockout animals for the specific receptor subtype

    • Samples with pharmacologically up/downregulated receptor expression

  • Technical controls:

    • Primary antibody omission controls

    • Isotype controls (antibodies of the same isotype but irrelevant specificity)

    • Blocking peptide controls (though these only confirm peptide binding, not receptor specificity)

    • Secondary antibody-only controls

  • Specificity controls:

    • Competition with purified receptor protein or immunizing peptide

    • Heterologous expression systems (cells transfected with the receptor versus non-transfected)

    • siRNA knockdown of the target receptor

  • Processing controls:

    • Standardized positive control samples processed in parallel with each experiment

    • Inter-assay calibrators for quantitative studies

    • Internal reference standards

  • Signal validation:

    • Multiple antibodies targeting different epitopes on the same receptor

    • Correlation with functional assays or mRNA expression data

    • For critical findings, validation with non-antibody-based methods like radioligand binding

The documented lack of specificity in many commercial alpha-1 adrenergic receptor antibodies emphasizes the importance of comprehensive controls for meaningful interpretation of results .

How can I optimize ALPHA-ADR antibody protocols for different tissue types?

Different tissues present unique challenges for alpha-adrenergic receptor antibody applications. Tissue-specific optimization strategies include:

  • Sample preparation adaptations:

    • For high-fat tissues: Modify extraction buffers with increased detergent concentrations

    • For fibrous tissues: Adjust homogenization methods and mechanical disruption

    • For highly vascular tissues: Include perfusion steps to remove blood proteins

  • Fixation and processing optimization:

    • For IHC, compare multiple fixatives (10% neutral buffered formalin, Bouin's, etc.)

    • Optimize fixation duration based on tissue density

    • Adjust antigen retrieval methods (heat-induced versus enzymatic)

  • Background reduction:

    • For tissues with high endogenous peroxidase (e.g., liver): Enhance blocking steps

    • For tissues with high biotin content: Use biotin-free detection systems

    • For tissues with high autofluorescence: Consider spectral unmixing or quenching agents

  • Signal enhancement strategies:

    • For tissues with low receptor expression: Employ signal amplification systems

    • Optimize antibody concentration specifically for each tissue type

    • Adjust incubation conditions (time, temperature) based on tissue penetration requirements

  • Validation in relevant contexts:

    • Heart tissue has approximately 15 fmol/mg protein total α1-ARs with 30% α1A, 70% α1B, and no detectable α1D

    • Brain tissue has approximately 140 fmol/mg protein total α1-ARs with 55% α1A, 35% α1B, and 10% α1D

When comparing alpha-adrenergic receptor expression across different tissues, it's essential to develop tissue-specific protocols while maintaining comparable sensitivity and specificity.

What are the alternatives to antibody-based detection for studying ALPHA-ADR receptors?

Given the documented limitations of commercial antibodies for alpha-adrenergic receptors, researchers should consider these alternative approaches:

  • Radioligand binding assays:

    • Currently the most reliable approach for quantifying alpha-1-AR subtype proteins

    • Can determine receptor density and pharmacological profiles

    • Allows for competition studies with subtype-selective ligands

    • Limitations include inability to provide cellular localization information

  • Fluorescent ligand-based approaches:

    • Fluorescently labeled receptor ligands for imaging studies

    • FRET/BRET-based interaction studies

    • Flow cytometry applications for cellular expression

  • Genetic approaches:

    • Receptor-reporter fusion proteins (GFP-tagged receptors)

    • CRISPR/Cas9 endogenous receptor tagging

    • Knock-in mouse models with tagged receptors

    • mRNA quantification via RT-PCR or RNA-Seq

  • Functional readouts:

    • Calcium mobilization assays for alpha-1 receptors

    • cAMP inhibition assays for alpha-2 receptors

    • Receptor-mediated signaling pathway activation

    • Pharmacological profiling with selective agonists/antagonists

  • Mass spectrometry-based proteomics:

    • Targeted proteomics for receptor quantification

    • Post-translational modification analysis

    • Receptor interactome characterization

Each approach has strengths and limitations, and combining multiple methods provides the most comprehensive understanding of alpha-adrenergic receptor biology.

How are emerging genetic technologies enhancing ALPHA-ADR antibody research?

Advanced genetic technologies are transforming alpha-adrenergic receptor research:

  • CRISPR/Cas9 applications:

    • Generation of receptor-specific knockout cell lines for antibody validation

    • Endogenous tagging of receptors with epitope tags or fluorescent proteins

    • Introduction of specific mutations to study structure-function relationships

    • Creation of reporter systems linked to receptor activation

  • Single-cell analysis technologies:

    • Single-cell RNA-Seq to correlate receptor mRNA expression with other cellular markers

    • Spatial transcriptomics to map receptor expression in tissue contexts

    • Examination of receptor expression heterogeneity within seemingly homogeneous populations

  • Advanced animal models:

    • Conditional and inducible receptor knockout models

    • Tissue-specific receptor expression systems

    • Humanized receptor mouse models for translational studies

  • Receptor variant analysis:

    • Identification of natural receptor variants and polymorphisms

    • Characterization of variant-specific antibodies

    • Study of variant-dependent signaling differences

  • Receptor trafficking and dynamics:

    • Live-cell imaging of receptors using genetic tags

    • Study of receptor internalization and recycling kinetics

    • Analysis of receptor-protein interactions in living cells

These technologies provide powerful tools to complement antibody-based approaches and overcome some of their limitations, enabling more comprehensive studies of alpha-adrenergic receptor biology and pharmacology.

What are the current challenges in developing therapeutic antibodies targeting ALPHA-ADR receptors?

Development of therapeutic antibodies targeting alpha-adrenergic receptors faces several significant challenges:

  • Antibody specificity limitations:

    • Documented lack of specificity in commercial antibodies raises concerns for therapeutic applications

    • Cross-reactivity between receptor subtypes could lead to off-target effects

    • Need for rigorous validation using knockout models and multiple approaches

  • Accessibility of target epitopes:

    • G-protein coupled receptors like alpha-adrenergic receptors have limited extracellular domains

    • Many functionally important regions are located in transmembrane or intracellular domains

    • Conformational states of the receptor may affect epitope accessibility

  • Functional modulation challenges:

    • Developing antibodies with specific agonist or antagonist properties

    • Addressing receptor subtype selectivity for targeted therapeutic effects

    • Controlling partial agonism versus full agonism/antagonism

  • Immunogenicity concerns:

    • Risk of anti-drug antibody (ADA) responses to therapeutic antibodies

    • Need for strategies to reduce ADA reactivity through:

      • Mutation of key residues (e.g., Thr101 and Thr146 in the variable heavy domain)

      • Reduction of exposed hydrophobic patches

      • Computational screening to identify optimal mutations

  • Tissue penetration and biodistribution:

    • Limited penetration of full-size antibodies into tissues like the brain

    • Need for engineered antibody fragments with improved tissue access

    • Challenges in maintaining half-life while reducing size

Addressing these challenges requires integrated approaches combining structural biology, computational design, advanced antibody engineering, and comprehensive validation methodologies to develop effective and safe therapeutic antibodies targeting alpha-adrenergic receptors.

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.