ATO Antibody

Shipped with Ice Packs
In Stock

Description

ATO Overview

ATO (arsenic trioxide) is a chemotherapeutic agent primarily used in acute promyelocytic leukemia (APL) treatment. While no known "ATO antibody" exists in clinical or research contexts, ATO interacts with immune pathways and antibody-mediated processes in cancer therapy .

ATO’s Role in Immune Modulation

ATO enhances anticancer immune responses by promoting immunogenic cell death (ICD), which includes:

  • Calreticulin exposure: Surface exposure of calreticulin (CALR) on cancer cells, facilitating phagocytosis by dendritic cells .

  • ATP and HMGB1 release: Extracellular ATP acts as a chemoattractant for immune cells, while HMGB1 promotes antigen presentation .

  • STING pathway activation: ATO triggers cGAMP production and Type-1 interferon signaling, enhancing antitumor T-cell responses .

Table 1: ATO’s Immunomodulatory Effects

MechanismImpact on Immune SystemKey Sources
CALR exposureEnhances phagocytosis of cancer cells
ATP/HMGB1 releaseRecruits dendritic/T cells
STING pathway activationBoosts interferon-β1 production

ATO and Antibody-Related Therapeutics

ATO synergizes with antibody-mediated therapies:

  • PD-L1 upregulation: ATO induces PD-L1 expression in esophageal squamous cell carcinoma (ESCC), suggesting potential for combination with anti-PD-1/PD-L1 checkpoint inhibitors .

  • Cyclin D1 degradation: ATO reduces Cyclin D1 levels, suppressing CDK4/6 signaling and sensitizing tumors to immune checkpoint blockade .

  • Antioxidant pathway inhibition: ATO blocks Nrf2-mediated antioxidant responses, increasing leukemia cell susceptibility to retinoic acid (RA)-driven differentiation .

Table 2: ATO’s Synergistic Effects with Antibody Therapies

Combination PartnerMechanismOutcomeSources
Anti-PD-1/PD-L1ATO-induced PD-L1 upregulationEnhanced checkpoint inhibition
All-trans retinoic acidNrf2 inhibition and MAPK activationImproved leukemia differentiation

ATO in Autoimmune Thyroid Disease (AITD)

While not directly an antibody, ATO’s immunomodulatory effects intersect with thyroid autoantibodies:

  • Anti-TPO antibodies: Elevated in Hashimoto’s thyroiditis (HT) and Graves’ disease (GD), these antibodies correlate with ATO’s oxidative stress induction .

  • Anti-Tg antibodies: Linked to differentiated thyroid cancer (DTC) prognosis; declining TgAb levels post-ATO/RA therapy indicate tumor-free survival .

Table 3: Thyroid Antibody Dynamics in ATO-Exposed Contexts

Antibody TypeClinical RelevanceSource
Anti-TPO (TPOAbs)Higher prevalence in AITD vs. controls
Anti-Tg (TgAbs)Prognostic marker in DTC

Research Gaps and Future Directions

  • No studies explicitly identify an "ATO antibody."

  • ATO’s interplay with antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) remains unexplored.

  • Clinical trials combining ATO with monoclonal antibodies (e.g., rituximab) are warranted .

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
ATO antibody; At5g06160 antibody; MBL20.3Splicing factor SF3a60 homolog antibody; Protein ATROPOS antibody; Splicing factor ATO antibody
Target Names
ATO
Uniprot No.

Target Background

Function
ATO Antibody recognizes the Arabidopsis homologue of SF3a60, a splicing factor implicated in pre-spliceosome formation. This antibody is crucial for determining gametic cell fate.
Gene References Into Functions
  1. ATO encodes the Arabidopsis homologue of SF3a60, a protein that has been implicated in pre-spliceosome formation. PMID: 18702672
Database Links

KEGG: ath:AT5G06160

STRING: 3702.AT5G06160.1

UniGene: At.8012

Protein Families
SF3A3 family
Subcellular Location
Nucleus.
Tissue Specificity
Expressed at moderate levels in all sporophytic tissues with strongest expression in gametophytes.

Q&A

What is Arsenic Trioxide (ATO) and what are its primary applications in biomedical research?

Arsenic trioxide is an inorganic compound with the chemical formula As₂O₃ that has emerged as an important therapeutic agent in oncology research. It was initially approved for treating acute promyelocytic leukemia (APL), transforming this once fatal condition into a largely curable disease . Beyond APL, ATO has demonstrated clinical efficacy against hepatocellular carcinoma, with the State Food and Drug Administration of China approving its use for advanced hepatocellular carcinoma in 2004 . The compound's mechanism involves multiple cellular pathways, affecting both cancer cells directly and modulating immune responses. Researchers investigating ATO typically work with purified pharmaceutical-grade preparations in controlled laboratory environments to ensure safety and reproducibility of results.

How does ATO affect immune cell populations and antibody responses?

ATO demonstrates complex effects on various immune cell populations that directly influence antibody-mediated responses. Research has shown that ATO enhances natural killer (NK) cell cytotoxicity against acute promyelocytic leukemia cells, representing one of its key immunomodulatory effects . Studies have documented that ATO treatment leads to:

  • Decreased CD4+ T lymphocyte and regulatory T cell (Tregs) populations

  • Increased CD8+ T lymphocyte populations

  • Enhanced cytotoxic T lymphocyte activity

These shifts in lymphocyte populations can significantly impact antibody production, as T-helper cells play crucial roles in B-cell activation and subsequent antibody secretion. When designing experiments to investigate these effects, researchers should consider time-dependent sampling to capture the dynamic nature of immune responses following ATO administration.

What experimental approaches are most effective for studying ATO's effects on antibody production?

When investigating ATO's impact on antibody production, researchers should implement a multi-faceted experimental approach:

  • In vitro systems: Co-culture systems with B cells, T cells, and antigen-presenting cells exposed to varying concentrations of ATO can help determine direct effects on antibody-producing cells.

  • Flow cytometry: Essential for quantifying changes in immune cell populations (B cells, plasma cells, T helper cells) and activation markers following ATO exposure.

  • ELISA/multiplex assays: For measuring antibody titers and isotype distribution in response to ATO treatment.

  • Animal models: Mouse models treated with ATO followed by antigen challenge to assess in vivo antibody responses.

  • Human samples: Analysis of antibody profiles in patient samples before and after ATO therapy, with appropriate institutional approvals.

For optimal results, researchers should incorporate appropriate controls, including untreated groups and groups treated with other established immunomodulatory agents for comparison.

What are the molecular mechanisms behind ATO's immunomodulatory effects?

ATO's immunomodulatory effects occur through multiple molecular pathways that researchers continue to elucidate. At the cellular level, ATO induces increased reactive oxygen species (ROS) production and DNA damage in cancer cells . This oxidative stress triggers complex signaling cascades that influence immune function.

Key molecular mechanisms include:

  • CDK4/6 pathway modulation: ATO induces degradation of Cyclin D1, which suppresses the CDK4/6 pathway in esophageal squamous cell carcinoma (ESCC) cells .

  • PD-L1 regulation: ATO treatment causes upregulation of PD-L1 in cancer cells, with inverse correlation between Cyclin D1 and PD-L1 expression levels observed in human ESCC tissues .

  • Sumoylation and ubiquitination: ATO induces transient upregulation and nuclear translocation of Cyclin D1 through sumoylation, followed by increased ubiquitination and degradation via T286 phosphorylation .

  • STAT1 signaling: Cyclin D1 degradation is partly mediated by Stat1 Y701 phosphorylation, linking ATO to inflammatory signaling pathways .

When investigating these mechanisms, researchers should employ time-course experiments to capture the sequential nature of these molecular events, as some effects (like Cyclin D1 upregulation) are transient before subsequent degradation occurs.

How can researchers effectively study the synergy between ATO and immune checkpoint inhibitors?

Investigating the synergy between ATO and immune checkpoint inhibitors requires careful experimental design. Studies have demonstrated that ATO treatment significantly increases the efficacy of checkpoint inhibitors in mouse models of oral and esophageal squamous cell carcinoma . To effectively explore this synergy, researchers should consider:

  • Sequential vs. concurrent administration protocols: Testing different timing strategies to determine optimal therapeutic scheduling.

  • Multiparameter immune monitoring: Comprehensive immune profiling before, during, and after combination therapy to identify mechanistic biomarkers.

  • Tumor microenvironment analysis: Single-cell sequencing and spatial transcriptomics to characterize changes in the tumor immune landscape.

  • Pharmacokinetic/pharmacodynamic modeling: To understand drug-drug interactions and optimal dosing strategies.

  • Preclinical models that recapitulate human disease: Humanized mouse models or patient-derived xenografts may provide more translatable results.

A systematic approach using factorial experimental designs will help identify synergistic versus additive effects and determine the biological mechanisms underlying observed combinatorial benefits.

What methodologies should be used to investigate ATO-induced changes in antibody specificity and function?

Researching ATO's effects on antibody specificity and function requires sophisticated methodological approaches:

  • Repertoire sequencing: Next-generation sequencing of B-cell receptors before and after ATO treatment to assess changes in antibody diversity and clonal selection.

  • Affinity measurements: Surface plasmon resonance (SPR) or bio-layer interferometry (BLI) to quantify binding kinetics of antibodies from ATO-treated versus control samples.

  • Glycosylation analysis: Mass spectrometry to characterize changes in antibody glycosylation patterns, which can significantly impact antibody function.

  • Functional assays:

    • Complement-dependent cytotoxicity (CDC)

    • Antibody-dependent cellular cytotoxicity (ADCC)

    • Neutralization assays for relevant antigens

  • Structural biology approaches: Crystallography or cryo-EM to determine if ATO treatment alters the structural characteristics of produced antibodies.

These methodologies should be applied in integrated workflows to comprehensively characterize how ATO treatment modifies not just antibody quantities but their qualitative functional properties.

How should researchers address the contradictory data regarding ATO's effects on different immune cell populations?

Contradictory findings regarding ATO's effects on immune cells may arise from differences in experimental conditions, cell types, and concentrations used. To address these contradictions, researchers should:

  • Standardize experimental conditions: Use consistent ATO concentrations, exposure times, and cell culture conditions across experiments.

  • Perform dose-response and time-course studies: ATO's effects may be biphasic or concentration-dependent, necessitating comprehensive dosing studies.

  • Consider cellular context: Different immune cell subsets and activation states may respond differently to ATO. Flow cytometric analysis should include detailed phenotyping.

  • Account for microenvironmental factors: The tumor or tissue microenvironment may significantly alter ATO's effects on immune cells.

  • Use multiple complementary assays: Combine functional assays with molecular analyses to correlate phenotypic changes with underlying mechanisms.

  • Replicate key findings in different model systems: Validate observations across multiple in vitro systems and in vivo models.

A systematic meta-analysis approach to experimental design, where variables are systematically altered and documented, will help identify the specific conditions under which particular immune effects occur.

What are the critical considerations for designing ATO antibody studies in animal models?

When designing animal studies to investigate ATO's effects on antibody responses, researchers should address these critical considerations:

  • Species selection: Different species may metabolize ATO differently. Mouse models are common, but larger animals may better recapitulate human pharmacokinetics.

  • Dosing regimen: ATO dosage should be carefully calculated based on body surface area rather than simple weight conversion from human doses.

  • Route of administration: Intravenous, intraperitoneal, and oral routes may yield different pharmacokinetics and immune effects.

  • Timing of immunological assessments: Include both early and late timepoints to capture immediate effects and potential compensatory responses.

  • Antigen selection for antibody studies: Use both T-dependent and T-independent antigens to comprehensively assess B cell functionality.

  • Adjuvant effects: Consider whether ATO itself might function as an adjuvant, potentially confounding interpretation of antibody responses.

  • Genetic background: Different mouse strains may exhibit variable baseline immune responses and reactivity to ATO.

  • Ethical considerations: Implement the 3Rs (replacement, reduction, refinement) and ensure appropriate institutional approval.

A well-designed longitudinal study with multiple assessment points will provide the most comprehensive understanding of ATO's effects on antibody responses in vivo.

How can researchers differentiate between direct effects of ATO on antibody-producing cells versus indirect effects through other immune pathways?

Distinguishing direct from indirect effects requires systematic experimental approaches:

ApproachMethodologyKey Considerations
Direct B cell culturesIsolate purified B cells and expose to ATORequires assessment of B cell viability as ATO may be cytotoxic
Conditioned media experimentsTransfer supernatants from ATO-treated immune cells to B cellsControls needed to account for residual ATO in media
Transwell co-culture systemsPhysically separate B cells from other immune cellsAllows for study of soluble factor-mediated effects
Cell-specific genetic modelsUse conditional knockout models (e.g., B cell-specific vs. T cell-specific)Helps pinpoint the cellular origin of observed effects
In vivo depletion studiesSelectively deplete specific immune cell populations before ATO treatmentMust confirm depletion efficiency before interpretation
Gene expression profilingCompare transcriptional changes in different immune cell populations after ATO exposureRequires bioinformatic expertise to identify pathway signatures

Integration of these approaches with appropriate controls can help researchers delineate the complex network of direct and indirect effects of ATO on antibody production.

What techniques are most appropriate for analyzing ATO-induced post-translational modifications of antibodies?

Post-translational modifications (PTMs) of antibodies can significantly impact their function and are potentially altered by ATO exposure. The following analytical techniques are recommended:

  • Mass spectrometry-based approaches:

    • Liquid chromatography-mass spectrometry (LC-MS/MS) for identification and quantification of PTMs

    • Native mass spectrometry to preserve non-covalent interactions

    • Glycoproteomics to characterize complex glycan structures

  • Site-specific analysis:

    • Enzymatic digestion followed by peptide mapping

    • Electron transfer dissociation (ETD) for preserving labile modifications

    • Selected reaction monitoring (SRM) for targeted quantification of specific modifications

  • Functional correlation studies:

    • Surface plasmon resonance to correlate modifications with binding kinetics

    • Bioassays to link PTMs with biological activity

    • Thermal stability assessments to evaluate structural impacts of modifications

  • Visualization techniques:

    • Specific antibodies against PTMs (e.g., phosphorylation, glycosylation)

    • Lectin binding assays for carbohydrate modifications

    • Fluorescent labeling of modified residues

The combination of these techniques provides a comprehensive picture of how ATO exposure might alter the post-translational landscape of antibodies, potentially affecting their functionality and immunological properties.

What emerging technologies show promise for advancing our understanding of ATO-antibody interactions?

Several cutting-edge technologies are poised to revolutionize our understanding of how ATO influences antibody production and function:

  • Single-cell multi-omics: Integrating single-cell transcriptomics, proteomics, and epigenomics to comprehensively characterize individual cell responses to ATO.

  • CRISPR-based functional genomics: High-throughput CRISPR screens to identify genes essential for ATO's effects on antibody-producing cells.

  • Advanced biophysical techniques:

    • High-resolution cryo-electron microscopy to visualize structural changes in antibodies

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to probe conformational dynamics

  • Microfluidic systems: Organ-on-a-chip platforms that recapitulate complex tissue microenvironments for studying ATO effects under physiologically relevant conditions.

  • Computational approaches:

    • Molecular dynamics simulations to predict ATO binding to immune receptors

    • Machine learning algorithms to identify patterns in complex immunological datasets

    • Systems biology modeling to integrate multiple layers of biological information

  • In situ imaging technologies:

    • Multiplex ion beam imaging (MIBI)

    • Imaging mass cytometry

    • Spatial transcriptomics

These technologies will enable researchers to move beyond correlative observations toward mechanistic understanding of how ATO influences antibody responses at molecular and cellular levels.

How might understanding ATO-antibody interactions inform the development of novel immunotherapeutic approaches?

Insights into ATO-antibody interactions have several promising applications for next-generation immunotherapies:

  • Combination therapy optimization: Understanding the mechanisms behind ATO's synergy with checkpoint inhibitors could inform rational design of combination protocols that maximize efficacy while minimizing toxicity.

  • Novel adjuvant development: If ATO modulates antibody quality or specificity in beneficial ways, this knowledge could guide development of safer arsenical derivatives as vaccine adjuvants.

  • Targeted immunomodulation: Elucidating the specific signaling pathways through which ATO affects antibody-producing cells could identify novel druggable targets for more selective immunomodulation.

  • Overcoming treatment resistance: Knowledge of how ATO affects antibody-mediated immune responses could help address resistance mechanisms to existing immunotherapies.

  • Biomarker development: Characterizing antibody profiles associated with ATO response could yield predictive biomarkers for patient selection and monitoring.

  • Engineering enhanced antibody therapeutics: Understanding how ATO modifies antibody structure and function could inspire new approaches to antibody engineering.

The integration of multiple research approaches—from basic mechanistic studies to translational applications—will be essential for leveraging ATO-antibody interaction knowledge into clinical advances.

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.