AICDA Antibody

Activation-induced cytidine deaminase, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Introduction to AICDA Antibody

The AICDA (Activation-Induced Cytidine Deaminase) antibody is a specialized immunological reagent designed to detect the AICDA protein, a key enzyme in antibody diversification processes. AICDA mediates somatic hypermutation (SHM), class-switch recombination (CSR), and gene conversion in B lymphocytes, enabling the immune system to generate high-affinity antibodies and diverse immune repertoires . The antibody serves as a critical tool for studying AICDA’s localization, expression levels, and functional roles in immune responses and diseases.

Applications in Immunological Research

AICDA antibodies are employed in diverse experimental contexts:

Immunohistochemistry (IHC)

  • Ramos Cell Line Staining: AICDA antibodies detect cytoplasmic and surface expression in Ramos human Burkitt’s lymphoma cells, as shown in fluorescence IHC .

  • Tissue Analysis: Used to study AICDA localization in lymphoid organs, such as germinal centers or bone marrow niches .

Western Blotting

  • Protein Validation: Confirms AICDA expression in lysates from B cell populations, including germinal center B cells or plasma cells .

Flow Cytometry

  • Cell-Specific Detection: Identifies AICDA+ B cells or T cells, enabling studies on AICDA’s role in tolerance or autoimmunity .

Case Study: AICDA in Central B-Cell Tolerance

AICDA antibodies have been used to investigate AICDA’s role in purging autoreactive B cells:

  • Murine Models: Aicda−/− mice exhibit impaired central tolerance, with elevated autoantibodies and reduced apoptosis of self-reactive B cells .

  • Human Studies: Patients with common variable immunodeficiency (CVID) and autoimmunity often carry AICDA mutations, highlighting its role in peripheral tolerance .

AICDA in Atherosclerosis

  • B Cell-Specific Knockout Models: AICDA antibodies help track B cell maturation markers (e.g., BAFFR) in atherosclerosis studies, linking class-switched antibodies to plaque formation .

Considerations for Use

  • Specificity: Ensure antibodies are pre-adsorbed against host serum proteins to minimize non-specific binding.

  • Optimal Dilution: Validate titers for each application (e.g., 10 µg/mL for IHC in Ramos cells) .

  • Fixation Protocols: Formalin-fixed, paraffin-embedded (FFPE) tissues require antigen retrieval for IHC .

Future Directions

Emerging research focuses on:

  • Epigenetic Regulation: AICDA’s role in DNA demethylation and its potential as a therapeutic target .

  • Cancer Immunology: Monitoring AICDA expression in B-cell lymphomas to guide treatment strategies .

Product Specs

Introduction
AICDA, a member of the cytidine deaminase family, functions as an RNA-editing deaminase. It plays a crucial role in immunoglobulin gene diversification processes, including somatic hypermutation, gene conversion, and class-switch recombination. Mutations in the AICDA gene can lead to autosomal recessive hyper-IgM immunodeficiency syndrome type 2 (HIGM2).
Formulation
The antibody is supplied at a concentration of 1 mg/ml in a solution of PBS at pH 7.4 with 0.1% sodium azide.
Storage Procedures
For short-term storage (up to 1 month), keep at 4°C. For long-term storage, store at -20°C. Avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The antibody is stable for 12 months when stored at -20°C and for 1 month at 4°C.
Applications
This AICDA antibody has undergone rigorous testing using ELISA and Western blot analysis to ensure its specificity and reactivity. However, optimal working dilutions should be determined empirically for each specific application. For Western blot analysis, a dilution range of 1:500 to 1:1000 is recommended, with a starting dilution of 1:500.
Synonyms
Activation-induced cytidine deaminase, Cytidine aminohydrolase, AICDA, AID, ARP2, CDA2, HIGM2.
Purification Method
AICDA antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
P2D3AT.
Immunogen
Anti-human AICDA mAb, is derived from hybridization of mouse SP2/0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human AICDA amino acids 1-54 purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and κ light chain.

Q&A

What is AICDA and why is it significant in immunological research?

AICDA (also known as AID) is an RNA-editing deaminase belonging to the cytidine deaminase family that plays a crucial role in three key processes of antibody diversification: somatic hypermutation (SHM), class switch recombination (CSR), and gene conversion. AICDA functions by deaminating deoxycytidine to deoxyuracil in single-stranded DNA, triggering error-prone repair processes that generate genetic diversity in immunoglobulin genes . This enzyme is fundamentally important for generating high-affinity antibodies during immune responses. Research interest in AICDA stems from its dual nature - while essential for adaptive immunity, its misregulation can contribute to genomic instability and cancer development . AICDA antibodies allow researchers to detect, localize, and study this protein's expression and function in various experimental systems.

How does AICDA expression vary across B cell developmental stages?

Contrary to earlier assumptions that AICDA expression was restricted to activated mature B cells in germinal centers, research has revealed that AICDA is also expressed at lower levels in bone marrow immature and transitional B cells . This expression pattern suggests a role in central tolerance, as Aicda^(-/-) mice show increased serum autoantibody levels and reduced capacity to eliminate self-reactive B cells during development . In the classic pathway, AICDA expression is highly upregulated in activated germinal center B cells, where it drives somatic hypermutation and class switching. For accurate experimental design, researchers should consider these developmental stage-specific expression patterns when selecting appropriate cellular models and interpreting antibody detection results.

What are the experimental advantages of monoclonal versus polyclonal anti-AICDA antibodies?

Monoclonal anti-AICDA antibodies, such as the P2D3AT clone, offer superior specificity and reproducibility for applications requiring precise epitope targeting . These antibodies are particularly valuable for comparing AICDA expression levels across different experimental conditions or for detecting specific structural variants. For example, monoclonal antibodies derived from hybridization of mouse SP2/0 myeloma cells with spleen cells from mice immunized with recombinant human AICDA amino acids 1-54 provide highly consistent results in Western blotting and immunofluorescence applications .

Polyclonal antibodies against AICDA, while potentially less specific, may offer advantages for detecting native protein conformations or when signal amplification is needed. The choice between monoclonal and polyclonal antibodies should be guided by the specific research question, with monoclonals preferred for quantitative comparisons and polyclonals sometimes favored for applications like immunoprecipitation where recognition of multiple epitopes may be beneficial.

How can AICDA antibodies be optimized for visualization of subcellular localization?

For optimal subcellular localization studies of AICDA, researchers should implement a strategic immunostaining protocol. Fluorescent immunocytochemistry using anti-AICDA antibodies at a concentration of 10 μg/mL with a 3-hour incubation at room temperature has proven effective for visualizing both cytoplasmic and surface localization in B cell lines such as Ramos (human Burkitt's lymphoma) . For dual-staining experiments, such as those examining the co-localization of AICDA with UBN1 or other interacting partners, double immunofluorescence assays followed by proximity ligation assays (PLA) provide definitive evidence of protein-protein interactions at subcellular resolution .

When preparing samples, both paraformaldehyde fixation followed by detergent permeabilization and methanol fixation protocols have been successful, though researchers should validate the optimal conditions for their specific cell types. For challenging samples, tyramide signal amplification may enhance detection of low-abundance AICDA, particularly in primary B cells or early developmental stages where expression levels are modest compared to germinal center B cells.

What approaches can be used to study AICDA-mediated DNA deamination activity in various experimental systems?

Studying AICDA-mediated DNA deamination requires specialized assays that go beyond simple protein detection. Researchers can employ several complementary approaches:

  • In vitro deamination assays: Using purified recombinant AICDA protein with single-stranded DNA substrates containing cytosine in various sequence contexts, followed by biochemical detection of deoxyuracil formation.

  • Cell-based mutation reporters: Transfection of target cells with specially designed plasmids containing reporter genes that can detect AICDA-induced mutations through changes in fluorescence or antibiotic resistance.

  • Endogenous mutation analysis: Deep sequencing of immunoglobulin loci or known off-target loci after AICDA induction, with or without AICDA antibody-mediated depletion experiments to confirm specificity.

For validating AICDA activity in experimental systems, researchers should combine antibody-based detection of the protein with functional readouts of deamination activity. When interpreting results, consider that AICDA activity is highly dependent on transcriptional status and chromatin accessibility of target loci, factors that should be controlled for in experimental designs .

How can chromatin immunoprecipitation (ChIP) with AICDA antibodies be optimized to identify genomic targets?

Optimizing ChIP protocols for AICDA requires addressing several technical challenges related to this protein's biology. AICDA-DNA interactions are often transient, occurring primarily during transcription of target genes. An effective ChIP protocol should include:

  • Crosslinking optimization: Standard formaldehyde crosslinking (1%, 10 minutes) should be supplemented with protein-protein crosslinkers like DSG (disuccinimidyl glutarate) to capture transient interactions.

  • Sonication parameters: Chromatin should be sheared to 200-300bp fragments, with sonication conditions empirically determined for each cell type.

  • Antibody selection: Use monoclonal antibodies with validated ChIP performance, typically requiring 5-10μg per immunoprecipitation reaction.

  • Controls: Include both input chromatin and immunoprecipitation with isotype-matched control antibodies as essential negative controls.

  • Sequential ChIP: For studying AICDA interaction with specific chromatin-associated factors (like the HIRA chaperon complex components), sequential ChIP (ChIP-reChIP) approaches can provide evidence for co-occupancy .

The highly methylated state of Region 4 in the Aicda gene in naïve B cells contrasts with its demethylated state in activated B cells, suggesting that ChIP experiments should be coupled with bisulfite sequencing or other epigenetic analyses to fully understand the regulatory context of AICDA targeting .

What are common causes of false negative results in AICDA immunodetection and how can they be addressed?

False negative results in AICDA detection can arise from multiple technical and biological factors:

  • Low endogenous expression levels: AICDA is expressed at very low levels outside of germinal center B cells. In bone marrow immature and transitional B cells, expression can be 10-100 fold lower than in activated germinal center B cells . Solution: Implement signal amplification techniques or use more sensitive detection methods such as RNAscope for mRNA detection followed by protein validation.

  • Epitope masking due to protein-protein interactions: AICDA interacts with numerous factors including UBN1 and other components of the HIRA chaperon complex, potentially obscuring antibody binding sites . Solution: Test multiple antibodies targeting different epitopes or use antigen retrieval methods optimized for nuclear proteins.

  • Nuclear-cytoplasmic shuttling: AICDA undergoes nucleocytoplasmic shuttling, which can affect detection depending on fixation and extraction methods. Solution: Use protocols that preserve both nuclear and cytoplasmic fractions, and consider dual immunostaining with markers of subcellular compartments.

  • Technical issues in immunoblotting: The relatively small size of AICDA (24 kDa) requires optimization of gel resolution and transfer conditions. Solution: Use gradient gels (4-20%) and optimize transfer conditions for small proteins (higher methanol concentration, lower voltage, longer transfer times).

A systematic approach to troubleshooting should include positive controls (such as Ramos cells or other AICDA-expressing B cell lines) and validation using multiple detection methods.

How can researchers distinguish between specific and non-specific signals when using AICDA antibodies in primary tissue samples?

Distinguishing specific from non-specific AICDA antibody signals in primary tissues requires rigorous experimental controls and validation steps:

  • Genetic controls: When possible, include tissues from Aicda^(-/-) mice as definitive negative controls that account for potential antibody cross-reactivity .

  • Blocking peptide controls: Pre-incubate the antibody with excess immunizing peptide before immunostaining to demonstrate signal specificity.

  • Multiple antibody validation: Use at least two antibodies raised against different AICDA epitopes to confirm staining patterns.

  • Correlation with known expression patterns: AICDA is predominantly expressed in germinal center B cells in secondary lymphoid tissues, with lower expression in specific developmental B cell stages. Staining patterns inconsistent with these known distributions warrant additional validation.

  • Complementary RNA detection: Correlate protein detection with AICDA mRNA detection using in situ hybridization or RT-PCR from isolated cell populations.

For immunohistochemistry applications, a dilution range of 1:500-1:1000 for Western blot analysis is recommended as a starting point, with each new antibody lot requiring titration to determine optimal conditions .

What strategies can address cross-reactivity issues between AICDA antibodies and other cytidine deaminase family members?

AICDA belongs to the cytidine deaminase family, which includes other members with structural similarity that may lead to cross-reactivity issues. To address these challenges:

  • Epitope selection: Choose antibodies raised against regions of AICDA with minimal sequence homology to other family members, particularly targeting the N-terminal region (amino acids 1-54) which contains unique structural elements .

  • Absorption controls: Pre-absorb antibodies with recombinant proteins of related family members to reduce cross-reactivity.

  • Expression pattern verification: Verify that the detected signal coincides with known AICDA expression patterns (e.g., germinal center B cells) rather than tissues known to express other family members.

  • Knockout/knockdown validation: Validate specificity using AICDA-deficient cells or siRNA knockdown experiments to confirm signal reduction.

  • Mass spectrometry validation: For critical experiments, confirm antibody specificity by immunoprecipitation followed by mass spectrometry identification of the captured proteins.

When studying tissues with potential expression of multiple cytidine deaminase family members, researchers should employ a combination of these approaches to ensure signal specificity.

How should researchers interpret AICDA localization data in the context of its nucleocytoplasmic shuttling properties?

AICDA undergoes complex nucleocytoplasmic shuttling, which significantly impacts the interpretation of localization data. Research has demonstrated that AICDA can be detected in both cytoplasmic and nuclear compartments, with the distribution varying based on cell type, activation state, and experimental conditions . When interpreting localization data:

  • Consider shuttling dynamics: AICDA is synthesized in the cytoplasm but functions in the nucleus. Its nuclear import is regulated while its export is constitutive, resulting in predominantly cytoplasmic steady-state localization despite its nuclear function.

  • Evaluate fixation artifacts: Different fixation methods can alter the apparent distribution of AICDA. Paraformaldehyde fixation may better preserve cytoplasmic pools, while methanol fixation might enhance nuclear detection.

  • Assess activation state: B cell activation status dramatically affects AICDA localization. In activated germinal center B cells, a greater proportion of AICDA may be nuclear compared to resting B cells.

  • Co-localization context: Interpret AICDA localization in relation to interaction partners. For example, co-localization with UBN1 and other HIRA complex components provides context for potential functional interactions .

  • Temporal considerations: AICDA localization is dynamic and changes during the cell cycle and in response to DNA damage, factors that should be controlled for in experimental designs.

Advanced imaging approaches such as live-cell tracking of fluorescently tagged AICDA provide more accurate insights into its shuttling dynamics than fixed-cell approaches alone.

What are the implications of AICDA's role in central tolerance for experimental design and interpretation?

The discovery that AICDA plays a role in central tolerance adds complexity to experimental design and data interpretation. Aicda^(-/-) mice exhibit increased serum autoantibody levels and reduced capacity to eliminate self-reactive B cells during development . When designing experiments:

  • Consider developmental timing: Experiments investigating B cell development must account for AICDA's expression and function in immature and transitional B cells, not just in mature activated B cells.

  • Recognize dual roles: AICDA functions differently in central tolerance versus peripheral antibody diversification, potentially through different targeting mechanisms or activity levels.

  • Account for autoimmunity confounders: When using Aicda^(-/-) models, increased autoimmunity may confound interpretation of other immunological phenotypes.

  • Design apoptosis assays: Since AICDA deficiency renders immature/transitional B cells more resistant to anti-IgM-induced apoptosis, experiments studying B cell selection should include appropriate apoptosis readouts .

  • Integrate epigenetic analyses: The regulation of AICDA through DNA methylation (particularly Region 4 of the Aicda gene) suggests that epigenetic analyses should complement protein expression studies .

This dual role highlights the importance of using developmental stage-specific markers when interpreting AICDA antibody staining in bone marrow and peripheral lymphoid tissues.

How can researchers leverage AICDA antibodies to study its role in pathological conditions like cancer?

AICDA antibodies provide valuable tools for investigating this enzyme's role in various pathological conditions, particularly in cancer development:

  • Oncogenic potential assessment: AICDA can contribute to genomic instability by introducing mutations in proto-oncogenes. Immunohistochemistry using AICDA antibodies (dilution 1:500) has been applied to examine expression in bladder urothelial cell carcinoma, revealing connections between AICDA expression and DNA demethylation in carcinogenesis networks .

  • Lymphoma studies: Given AICDA's high expression in germinal center B cells, antibody-based detection is critical for studying its role in B-cell lymphomagenesis. AICDA antibodies can help distinguish lymphoma subtypes and potentially correlate expression levels with genomic instability metrics.

  • Autoimmune disease research: The role of AICDA in central tolerance suggests applications in studying autoimmune pathologies. Researchers can assess whether aberrant AICDA expression or localization correlates with autoantibody production in diseases like lupus or rheumatoid arthritis.

  • Immunodeficiency diagnostics: AICDA antibodies can be used in diagnostic workflows for hyper-IgM syndrome type 2 (HIGM2), caused by AICDA deficiency . Immunoblotting or flow cytometry analysis of patient B cells can help identify protein expression defects.

  • Therapeutic target validation: As potential therapeutic approaches targeting AICDA emerge, antibodies will be essential for validating target engagement and monitoring treatment effects in preclinical models.

When studying pathological samples, researchers should implement appropriate controls and consider combining AICDA detection with markers of DNA damage, mutation signatures, or epigenetic alterations to establish mechanistic connections.

How are AICDA antibodies being utilized to investigate epigenetic mechanisms in B cell biology?

AICDA antibodies are increasingly employed to explore the intersection of epigenetic regulation and B cell function:

  • DNA methylation studies: Research has revealed that the Aicda gene itself is regulated by DNA methylation, with Region 4 being highly methylated in naïve B cells but demethylated in activated B cells . Combining chromatin immunoprecipitation using AICDA antibodies with bisulfite sequencing allows researchers to correlate AICDA binding with local epigenetic changes.

  • Histone modification interplay: The interaction between AICDA and the HIRA chaperone complex (which deposits H3.3 histone variants) suggests an important connection with chromatin architecture . Dual immunofluorescence and proximity ligation assays using antibodies against AICDA and various histone modifications help elucidate these relationships.

  • Epigenetic targeting mechanisms: AICDA's targeting to specific genomic regions likely involves recognition of particular chromatin states. ChIP-seq approaches using AICDA antibodies coupled with histone modification mapping can identify chromatin signatures associated with AICDA recruitment.

  • Off-target epigenetic effects: Beyond its role in DNA deamination, evidence suggests AICDA may influence DNA demethylation processes. Immunoprecipitation of AICDA followed by analysis of associated factors has revealed connections to DNA demethylation pathways in certain contexts, including cancer .

Researchers should design experiments that integrate AICDA protein detection with comprehensive epigenomic profiling to fully understand its multifaceted roles in chromatin regulation.

What methodological approaches can combine AICDA antibodies with cutting-edge genomic technologies?

Advanced methodological approaches integrating AICDA antibodies with genomic technologies offer new insights into this enzyme's function:

  • CUT&RUN or CUT&Tag approaches: These techniques provide higher resolution alternatives to traditional ChIP-seq for mapping AICDA genomic binding sites. By using antibody-directed nucleases rather than sonication, these methods can achieve better signal-to-noise ratios for factors with transient DNA interactions like AICDA.

  • Single-cell approaches: Combining AICDA immunostaining with single-cell RNA-seq or ATAC-seq enables correlation of AICDA protein levels with gene expression or chromatin accessibility profiles at the individual cell level, revealing heterogeneity in B cell populations.

  • CRISPR screening integration: AICDA antibodies can validate the effects of CRISPR perturbations of potential AICDA regulators or interactors, helping to establish genetic networks controlling its expression and function.

  • Spatial transcriptomics correlation: Combining AICDA immunohistochemistry with spatial transcriptomics in lymphoid tissues can reveal microenvironmental factors influencing its expression and activity in specific niches like germinal centers.

  • Mutation signature analysis: Correlating AICDA protein levels (detected via immunofluorescence) with whole-genome sequencing of single B cells can help establish direct relationships between AICDA expression and specific mutation signatures.

These integrated approaches require careful optimization of antibody performance characteristics for each specific technological application, with particular attention to specificity validation.

Product Science Overview

Introduction

Activation-induced cytidine deaminase (AID) is a crucial enzyme in the adaptive immune system, primarily involved in the diversification of antibodies. It is a member of the apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like (APOBEC) family of RNA/DNA editing enzymes . AID is encoded by the AICDA gene in humans and plays a central role in somatic hypermutation (SHM) and class switch recombination (CSR) of immunoglobulin genes .

Function and Mechanism

AID’s primary function is to convert cytidine to uracil in DNA, which is a key step in SHM and CSR . This process is essential for generating high-affinity antibodies and enhancing the specificity of the immune response . AID’s activity is tightly regulated to prevent genomic instability, as its dysregulation can lead to increased mutation load, translocations, and lymphomagenesis .

Role in B Cell Development

AID is predominantly expressed in B lymphocytes, where it plays a critical role in the diversification of antibodies . After naïve B cells encounter antigens in secondary lymphoid tissues, AID initiates SHM and CSR, leading to the production of antibodies with higher affinity and different isotypes . This process is vital for the adaptive immune system to effectively combat infections .

Implications in Disease

While AID is essential for immune function, its misregulation can have severe consequences. Overexpression or deregulation of AID has been implicated in various cancers, particularly B-cell lymphomas . Chronic inflammation can lead to AID overexpression, resulting in mutations and epigenetic changes that drive carcinogenesis . Additionally, AID has been implicated in non-lymphoid cancers and autoimmune diseases .

Mouse Anti Human AID Antibodies

Mouse anti-human AID antibodies are used in research to study the expression and function of AID in human cells. These antibodies are generated by immunizing mice with human AID protein, leading to the production of antibodies that specifically recognize and bind to human AID . These antibodies are valuable tools for investigating the role of AID in various biological processes and diseases .

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 2024 Thebiotek. All Rights Reserved.