At1g59800 Antibody

Shipped with Ice Packs
In Stock

Description

Biological Context of At1g59800

The At1g59800 gene in Arabidopsis thaliana encodes a protein of unknown function. While direct studies on this gene are absent from the provided literature, antibodies against similar Arabidopsis proteins (e.g., AtCUL1, CLPP3, CKX6) are frequently used to investigate:

  • Protein localization (nucleus, cytoplasm, or organelles)

  • Gene expression patterns during development

  • Stress response mechanisms

Antibodies like At1g59800 are critical for Western blotting, immunolocalization, and protein-protein interaction studies in plant molecular biology .

Functional Genomics

Antibodies enable researchers to:

  • Track protein expression under varying conditions (e.g., light/dark cycles, pathogen exposure)

  • Validate CRISPR/Cas9 knockout lines by confirming protein absence

Comparative Analysis

Facilitates cross-species studies by comparing homologous proteins in other plants .

Research Challenges and Gaps

  • Functional annotation: The At1g59800 gene lacks experimental characterization, limiting antibody utility to exploratory studies .

  • Epitope specificity: Commercial listings provide minimal details about antigen design, raising questions about cross-reactivity .

  • Validation data: Absence of published validation experiments (e.g., knockout controls) in peer-reviewed literature .

Future Directions

Priority research areas include:

  1. Functional characterization: Linking At1g59800 to metabolic or regulatory pathways.

  2. Antibody validation: Publishing knockout validation data to confirm specificity .

  3. Structural modeling: Using antibody binding data to predict protein domains .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
At1g59800 antibody; F23H11.12Cullin-like protein 3 antibody; Cullin-5 antibody; AtCUL5 antibody
Target Names
At1g59800
Uniprot No.

Q&A

What is AT1R and why are antibodies against it significant in research?

AT1R (angiotensin receptor type 1) is a G-protein-coupled receptor that plays a crucial role in the renin-angiotensin system. Antibodies directed against AT1R are significant in research because they have been implicated in the pathogenesis of several diseases, including systemic sclerosis (SSc) . These autoantibodies can bind to the receptor and trigger various cellular responses, potentially contributing to inflammation, fibrosis, and vascular damage. Research on AT1R antibodies has expanded our understanding of autoimmune mechanisms and provided potential biomarkers for disease activity and progression. The significance of these antibodies extends to multiple organ systems, making them an important focus in both basic and translational research.

What methods are available for detecting AT1R antibodies in biological samples?

Several methods can be employed to detect AT1R antibodies in biological samples:

  • ELISA (Enzyme-Linked Immunosorbent Assay): This is the most commonly used method for quantifying AT1R antibody levels in serum or plasma samples . Commercial ELISA kits are available, typically using recombinant AT1R protein as the capture antigen.

  • Immunofluorescence: This technique can be used to visualize AT1R antibody binding to cells or tissues expressing AT1R . It is particularly useful for determining the tissue distribution of antibody binding.

  • Cell-based assays: Functional assays using cells expressing AT1R can measure the biological activity of the antibodies, such as their ability to induce signaling pathways or alter cellular responses .

  • Dynamic mass redistribution (DMR) technology: This label-free optical whole-cell biosensing assay can detect morphological changes in living cells as a consequence of AT1R antibody interaction with the receptor .

Each method has specific advantages and limitations regarding sensitivity, specificity, and the type of information provided, so the choice should be guided by the specific research question.

How can researchers differentiate between AT1R autoantibodies and other autoantibodies in experimental settings?

Differentiating AT1R autoantibodies from other autoantibodies requires several strategic approaches:

  • Receptor specificity testing: Using AT1R antagonists (such as losartan or candesartan) to block antibody binding. If the binding or functional effect is diminished in the presence of these antagonists, it suggests specificity for AT1R .

  • Cross-reactivity assessment: Testing the antibodies against related receptors (like AT2R) or unrelated G-protein-coupled receptors to confirm specificity.

  • Absorption studies: Pre-absorbing serum samples with recombinant AT1R or AT1R-expressing cells to deplete specific antibodies before testing.

  • Immunoprecipitation followed by mass spectrometry: This can identify the exact antigen being recognized.

  • Functional assays: Measuring specific cellular responses known to be mediated by AT1R activation, such as calcium flux, ERK phosphorylation, or NF-κB activation .

In research settings, it's also important to test for other relevant autoantibodies simultaneously, such as anti-endothelin receptor A (ETAR) antibodies, which often co-occur with AT1R antibodies .

What are the key considerations when designing an animal model to study AT1R antibody-mediated pathology?

Designing an effective animal model for studying AT1R antibody-mediated pathology requires careful consideration of multiple factors:

  • Species selection: C57BL/6J mice have been successfully used for AT1R immunization studies . Consider the homology between human and animal AT1R sequences when selecting a species.

  • Immunization approach: Several options exist:

    • Active immunization with membrane-embedded human AT1R

    • Immunization with specific AT1R peptides, such as the peptide 149-172 which has been shown to provoke lung inflammation

    • Passive transfer of purified AT1R antibodies or monoclonal AT1R antibodies

  • Control groups: Include appropriate controls such as immunization with empty membrane or irrelevant peptides .

  • Genetic modifications: Consider using AT1Ra/b knockout mice to validate the specificity of antibody effects .

  • Assessment timeline: Plan for both acute and chronic effects, as some manifestations (like fibrosis) may develop gradually.

  • Comprehensive phenotyping: Include histology, immunohistochemistry, immunofluorescence, apoptosis assays, and molecular analyses of relevant signaling pathways (e.g., Smad2/3 for fibrosis) .

  • Functional assessments: Include physiological measurements relevant to the pathology being studied (e.g., pulmonary function tests for lung involvement).

  • Cell-specific effects: Analyze effects on multiple cell types, as AT1R antibodies can activate different cells including cardiomyocytes, monocytes, and fibroblasts .

The model should be designed to allow investigation of both the immunological mechanisms of antibody generation and the pathological effects of the antibodies once present.

How can researchers distinguish between the pathological effects of AT1R antibodies and the effects of angiotensin II itself?

Distinguishing between effects mediated by AT1R antibodies and those mediated by angiotensin II (Ang II) requires sophisticated experimental approaches:

  • Receptor binding studies: AT1R antibodies can bind to epitopes distinct from the Ang II binding site. DMR technology can be used to characterize the binding characteristics and potential allosteric effects .

  • Dual blockade experiments: Compare the effects of:

    • AT1R antagonists alone (blocks Ang II but may not block all antibody effects)

    • Antibody-depleting interventions alone (such as immunoadsorption or B-cell depletion)

    • Combination of both approaches

  • Signal transduction analysis: AT1R antibodies may preferentially activate certain downstream pathways compared to Ang II. Detailed analysis of various signaling cascades (e.g., G-protein dependent vs. β-arrestin pathways) can help differentiate the effects .

  • Temporal characteristics: AT1R antibodies may have more prolonged effects compared to Ang II due to differences in receptor desensitization and internalization.

  • AT1R mutants: Using cells expressing AT1R with mutations in specific domains can help determine if antibodies and Ang II utilize different receptor regions for their effects.

  • Knockout models: Studies in AT1Ra/b knockout mice can definitively demonstrate AT1R-dependent effects .

Research has shown that AT1R antibodies can enhance Ang II-mediated AT1R activation, suggesting a synergistic relationship rather than merely parallel effects .

What methodological approaches can identify the key epitopes recognized by pathogenic AT1R antibodies?

Identifying key epitopes recognized by pathogenic AT1R antibodies requires a multi-faceted approach:

  • Peptide array analysis: Synthesizing overlapping peptides spanning the entire AT1R sequence to identify regions that bind to purified antibodies or patient serum.

  • Alanine scanning mutagenesis: Systematically replacing individual amino acids with alanine to identify critical residues for antibody binding.

  • Chimeric receptor constructs: Creating chimeric proteins between AT1R and related receptors to narrow down antibody-binding regions.

  • Competition assays: Using defined peptides to compete with the full receptor for antibody binding.

  • X-ray crystallography or cryo-EM: Structural analysis of antibody-receptor complexes for precise epitope mapping.

  • Phage display libraries: Screening random peptide libraries for sequences that bind to AT1R antibodies.

  • In vivo validation: Testing identified peptides in animal models, such as the AT1R peptide 149-172 which has been shown to provoke lung inflammation when used for immunization .

  • Computational modeling: Predicting potential epitopes based on receptor structure and accessibility, followed by experimental validation.

Understanding the specific epitopes recognized by pathogenic antibodies is critical for developing targeted therapeutic approaches and for gaining insight into the mechanisms of antibody-mediated pathology.

What is the evidence linking AT1R antibodies to COVID-19 severity and outcomes?

The relationship between AT1R antibodies and COVID-19 has yielded interesting but sometimes contradictory findings:

  • Increased prevalence in COVID-19: Studies have found significantly increased AT1R antibody titers in COVID-19 patients compared to controls . This suggests a potential role in the disease process.

  • Relationship with disease severity: The evidence is mixed:

    • Some research suggests AT1R antibodies might have a protective role in COVID-19 .

    • Other studies found no significant difference in AT1R antibody titers between patients with favorable versus unfavorable disease courses .

  • Comparison with other respiratory conditions: Intubated COVID-19 patients had significantly higher AT1R antibody titers compared to patients with acute respiratory distress syndrome (ARDS) due to other causes . This suggests a COVID-19-specific phenomenon rather than a general response to respiratory illness.

  • Correlation with markers and outcomes:

    • No correlation was found between AT1R antibody titers and baseline inflammatory markers during hospital admission .

    • Similarly, no correlation was observed with diffusion capacity, cognitive impairment, or fatigue measured at 3-month follow-up .

  • Co-occurrence with other autoantibodies: AT1R antibodies are often found alongside endothelin-1 type A receptor (ETAR) antibodies in COVID-19 patients , suggesting a broader autoimmune response.

These findings highlight the complex role of AT1R antibodies in COVID-19 pathogenesis. Further research is needed to clarify whether these antibodies contribute to disease mechanisms or represent an epiphenomenon.

How do AT1R antibodies contribute to the pathogenesis of systemic sclerosis and other autoimmune conditions?

AT1R antibodies contribute to the pathogenesis of systemic sclerosis (SSc) and other autoimmune conditions through multiple mechanisms:

  • Vascular effects:

    • Activation of endothelial cells, leading to increased expression of adhesion molecules and chemokines

    • Promotion of endothelial cell apoptosis, contributing to vascular damage

    • Perivascular inflammation, as observed in animal models

  • Inflammatory responses:

    • Stimulation of IL-8 release from leukocytes

    • Induction of CCL18 production, which can attract inflammatory cells

    • Promotion of T and B cell infiltration in affected tissues

  • Fibrotic processes:

    • Induction of TGFβ expression, a key profibrotic mediator

    • Direct stimulation of collagen type I production in fibroblasts

    • Activation of Smad2/3 signaling, central to fibrotic pathways

  • Immune system activation:

    • Involvement of CD4+ T cells and B cells in antibody generation, as evidenced by reduced pathology in mice deficient for these cell types

    • IgG deposition in affected tissues, indicating immune complex formation

  • Synergy with other pathways:

    • Enhancement of angiotensin II-mediated AT1R activation, amplifying natural receptor signaling

    • Potential interaction with other autoantibodies, such as anti-ETAR antibodies, which often co-occur

The pathogenicity of AT1R antibodies has been validated through both in vitro studies with patient-derived IgG and in vivo models where immunization with AT1R induces SSc-like manifestations including interstitial lung disease and skin fibrosis .

What are the primary challenges in developing highly specific monoclonal antibodies against AT1R, and how can they be addressed?

Developing highly specific monoclonal antibodies against AT1R presents several key challenges and potential solutions:

  • Receptor conformation and accessibility:

    • Challenge: AT1R is a seven-transmembrane G-protein-coupled receptor with limited exposed extracellular domains.

    • Solution: Use membrane-embedded full-length AT1R for immunization to maintain native conformation , or employ synthetic peptides corresponding to extracellular loops.

  • Species conservation and cross-reactivity:

    • Challenge: High conservation of AT1R sequence across species can limit immunogenicity.

    • Solution: Strategic immunization protocols in AT1R knockout animals or selection of less conserved epitopes for targeted approaches.

  • Functional validation:

    • Challenge: Ensuring antibodies have the desired functional properties (agonistic, antagonistic, or neutral).

    • Solution: Employ comprehensive functional screening assays such as DMR technology to characterize antibody effects on receptor activation . Test antibodies for their ability to modulate angiotensin II responses.

  • Hybridoma technique limitations:

    • Challenge: Traditional hybridoma techniques may yield limited diversity.

    • Solution: Utilize advanced antibody discovery platforms such as phage display or the Antibody-GAN approach, which employs generative adversarial networks to design diverse humanoid antibodies .

  • Epitope specificity:

    • Challenge: Developing antibodies against specific epitopes associated with pathogenic effects.

    • Solution: Employ epitope mapping techniques and rational design approaches to target specific receptor domains.

  • Stability and developability:

    • Challenge: Ensuring antibodies have favorable biophysical properties for research applications.

    • Solution: Through transfer learning approaches, bias antibody generation towards improved stability and developability characteristics .

The hybridoma technique has been successfully used to generate monoclonal AT1R antibodies for research purposes , but newer computational and synthetic biology approaches offer opportunities to overcome traditional limitations .

How can researchers address the issue of resistance development when using therapeutic antibodies targeting AT1R-related pathways?

Addressing resistance development in therapeutic antibodies targeting AT1R-related pathways requires strategic approaches based on lessons from other therapeutic antibody fields:

  • Antibody combinations:

    • Using non-competing antibody combinations targeting different epitopes of AT1R or related molecules can significantly reduce the risk of resistance development.

    • Evidence from other therapeutic antibody research shows that combination therapies can fully protect against resistance development, whereas monotherapies frequently lead to resistance .

    • In one study, resistance variants emerged in almost half (18/40) of animals treated with monotherapy antibodies versus none (0/20) of the animals treated with an antibody combination .

  • Epitope selection:

    • Targeting conserved, functionally critical regions of the receptor that cannot tolerate mutations without loss of function.

    • Designing antibodies that target multiple regions simultaneously.

  • Monitoring for resistance:

    • Regular sequencing to detect emerging variants in the AT1R gene (AGTR1) during treatment.

    • Establishing sensitive assays to detect reduced antibody binding or efficacy early.

  • Rational antibody design:

    • Utilizing computational approaches like the Antibody-GAN to design antibodies with improved properties .

    • Developing antibodies with higher binding affinities that remain effective even with partial epitope mutations.

  • Alternative mechanism targeting:

    • Simultaneously targeting different aspects of the pathway (e.g., combining receptor blockers with downstream signaling inhibitors).

    • Developing bispecific antibodies that can engage multiple targets.

  • Dynamic treatment protocols:

    • Implementing treatment rotation strategies to reduce selective pressure.

    • Dose optimization to achieve sufficient target coverage while minimizing resistance development.

The experience with SARS-CoV-2 therapeutic antibodies provides valuable lessons, demonstrating that while monotherapy commonly leads to selection of escape variants, properly designed antibody combinations can prevent resistance development .

How should researchers interpret contradictory findings regarding AT1R antibodies in different disease contexts?

Interpreting contradictory findings regarding AT1R antibodies across different disease contexts requires a systematic approach:

  • Methodological differences assessment:

    • Evaluate differences in antibody detection methods (ELISA, functional assays, etc.).

    • Compare cut-off values for positivity versus assessment of average values .

    • Consider sensitivity and specificity of different assays used.

  • Population heterogeneity analysis:

    • Stratify patient populations by disease subtype, duration, severity, and treatment history.

    • Consider genetic background differences that might influence antibody effects.

    • Evaluate age, gender, and comorbidity distributions across studies.

  • Contextual biology consideration:

    • AT1R antibodies may have different effects depending on the local tissue environment.

    • The same antibody might be protective in one context but pathogenic in another .

    • Consider interaction with other autoantibodies or mediators present in specific diseases.

  • Temporal dynamics evaluation:

    • Assess when samples were collected in the disease course.

    • Consider potential fluctuations in antibody levels over time.

  • Antibody characteristics comparison:

    • Different studies may be detecting antibodies with different epitope specificities.

    • Characterize antibody isotypes (IgG1, IgG2, etc.) as they may have different functional effects .

  • Statistical approach review:

    • Evaluate sample sizes and statistical power.

    • Consider correction for multiple testing and potential confounding variables.

  • Replication and validation:

    • Design studies specifically to resolve contradictions.

    • Use multiple methodologies within the same study to cross-validate findings.

For example, contradictory findings regarding AT1R antibodies in COVID-19 might be explained by differences in methodology—one study noted their findings were not in line with recent literature but emphasized they evaluated AT1R antibody positivity based on an actual positivity cut-off rather than average values evaluation .

What experimental design elements are crucial for accurately evaluating the functional effects of AT1R antibodies in in vitro systems?

Designing robust experiments to evaluate AT1R antibody functional effects in vitro requires attention to several critical elements:

  • Cell type selection:

    • Use multiple relevant cell types that naturally express AT1R (e.g., vascular smooth muscle cells, endothelial cells, cardiomyocytes, monocytes) .

    • Include AT1R-transfected cell lines with controlled receptor expression levels.

    • Consider species compatibility (human AT1R may respond differently than rodent AT1R).

  • Antibody preparation:

    • Purify IgG fractions from serum to remove potentially interfering factors.

    • Include both monoclonal antibodies and polyclonal preparations for comprehensive assessment .

    • Characterize antibody concentrations, subclasses, and binding properties.

  • Appropriate controls:

    • Include isotype-matched control antibodies.

    • Use antibodies depleted of AT1R reactivity (through absorption).

    • Include angiotensin II as a positive control for receptor activation.

  • Specificity validation:

    • Block effects with AT1R antagonists to confirm specificity .

    • Use AT1R knockout or silenced cells as negative controls.

    • Test effects on cells lacking AT1R expression.

  • Comprehensive readouts:

    • Assess multiple downstream signaling pathways (G-protein dependent, β-arrestin, etc.).

    • Measure both immediate (calcium flux, phosphorylation) and delayed responses (gene expression, protein synthesis).

    • Employ unbiased approaches like DMR technology to capture global cellular responses .

  • Physiological relevance:

    • Conduct experiments under conditions that mimic physiological or pathophysiological states.

    • Consider the influence of other factors present in disease states (cytokines, growth factors).

    • Evaluate concentration-dependent effects across a range relevant to in vivo conditions.

  • Time course analyses:

    • Assess both acute and chronic effects of antibody exposure.

    • Evaluate potential receptor desensitization or internalization over time.

  • Co-culture systems:

    • Develop co-culture models to assess intercellular communication effects.

    • For example, evaluate how AT1R antibody-activated monocytes influence fibroblast behavior .

These design elements will help ensure robust, reproducible, and physiologically relevant results when studying AT1R antibody functional effects in vitro.

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.