ANO2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to ANO2 Antibody

ANO2 (anoctamin 2) antibodies are immunological tools targeting the ANO2 protein, a calcium-activated chloride channel involved in neuronal signaling and cellular ion transport. These antibodies are primarily used in research to study ANO2's role in physiological processes and diseases, particularly autoimmune conditions like multiple sclerosis (MS). ANO2 antibodies are critical for detecting protein expression, mapping epitopes, and investigating autoimmune reactivity in clinical and experimental settings .

Autoimmune Relevance in Multiple Sclerosis

ANO2 antibodies have identified ANO2 as an autoimmune target in MS. Key findings include:

  • Elevated Autoantibodies: MS patients show significantly higher ANO2 autoantibody reactivity compared to controls (p < 0.001) .

  • Epitope Specificity: Antibodies target cytoplasmic regions of ANO2, particularly residues 79–167 and 1–365 .

  • EBV Cross-Reactivity: Elevated ANO2 antibody titers in MS correlate with Epstein-Barr virus (EBV) exposure, suggesting molecular mimicry mechanisms .

Immunohistochemical Localization

  • Cellular Distribution: ANO2 antibodies localize to neurons and glial cells in human brain tissue, with heightened expression in MS lesion-associated macrophages/microglia .

  • Experimental Validation: Staining patterns in retina (rat/mouse) confirm antibody specificity .

Clinical and Experimental Implications

  • Diagnostic Potential: ANO2 autoantibodies may define an MS sub-phenotype, aiding patient stratification .

  • Therapeutic Targets: ANO2’s role in chloride flux highlights its relevance in neurodegenerative and inflammatory pathways .

  • Limitations: Current antibodies are research-grade only; clinical diagnostic use requires further validation .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery time may vary depending on your location and the method of purchase. Please consult your local distributor for specific delivery times.
Synonyms
Ano2 antibody; ANO2_HUMAN antibody; Anoctamin 2 antibody; Anoctamin-2 antibody; C12orf3 antibody; Chromosome 12 open reading frame 3 antibody; DKFZp434P102 antibody; Transmembrane protein 16B (eight membrane-spanning domains) antibody; Transmembrane protein 16B antibody
Target Names
ANO2
Uniprot No.

Target Background

Function
Ano2, also known as Calcium-activated chloride channel (CaCC), is a protein that plays a crucial role in olfactory signal transduction. Odorant molecules bind to odor-sensing receptors (OSRs), triggering an influx of calcium ions. This calcium increase activates Ano2, amplifying the depolarization of OSR cells. Ano2 is believed to be the underlying chloride channel involved in this process. Additionally, Ano2 may contribute to light perception amplification in the retina.
Gene References Into Functions
  • Residues facing the putative channel pore are responsible for both controlling the ion selectivity and gating of the channel. This provides an initial understanding of the molecular mechanism of ion permeation in TMEM16B. PMID: 28046119
  • Research has revealed a significant increase in autoantibody reactivity against the chloride-channel protein Ano2 (TMEM16B) in multiple sclerosis cases compared to controls. PMID: 26862169
  • Olfactory function impairment has been observed in Italian patients with type 3 von Willebrand disease who have a partial deletion of TMEM16B. PMID: 25635880
  • Ano2 is found in human myometrium. PMID: 24928056
  • The third intracellular loop of TMEM16B is responsible for calcium ion sensitivity, while the C-terminal part affects the rate of transition between the open and closed states of the channel. PMID: 23570556
  • Deletions in VWF and TMEM16B may play a role in severe von Willebrand disease type 3. PMID: 17371490
  • A study suggested that TMEM16B is a strong candidate for the long-sought Ca(2+)-dependent chloride channel in the photoreceptor synapse. PMID: 19474308
  • Ano2 likely amplifies the odor-induced generator potential in olfactory sensory neuron cilia by sensing elevated calcium levels, allowing the outward flow of chloride ions from the cell. PMID: 19561302
  • C12orf3, FLJ10261 (ORAOV2), C11orf25, and FLJ34272 constitute a family of eight-transmembrane proteins with N- and C-terminal tails facing the cytoplasm. PMID: 12739008
Database Links

HGNC: 1183

OMIM: 610109

KEGG: hsa:57101

STRING: 9606.ENSP00000314048

UniGene: Hs.148970

Protein Families
Anoctamin family
Subcellular Location
Cell membrane; Multi-pass membrane protein.
Tissue Specificity
Retina, especially in the photoreceptor synaptic terminals.

Q&A

What is ANO2 and why is it important in scientific research?

ANO2 (Anoctamin-2), also known as TMEM16B or C12orf3, belongs to the anoctamin family of calcium-activated chloride channels (CaCCs). It plays crucial roles in several physiological processes, particularly in sensory transduction and neuronal excitation .

ANO2 is predominantly expressed in:

  • Photoreceptor synaptic terminals in the retina

  • Olfactory sensory neurons

  • Various neuronal and glial cells in the central nervous system

The protein is particularly important in research because:

  • It mediates olfactory amplification in olfactory sensory neurons

  • It contributes to light perception amplification in the retina

  • It has been identified as a potential autoimmune target in multiple sclerosis

  • It demonstrates molecular mimicry with Epstein-Barr virus nuclear antigen 1, suggesting a potential link between viral infection and autoimmunity

What are the structural characteristics of ANO2 protein that influence antibody design?

ANO2 is a multi-pass membrane protein with several key structural features that influence antibody design and selection:

  • Protein size: Calculated molecular weight of 114 kDa

  • Structure: Contains eight transmembrane domains with both N and C termini located in the cytoplasm

  • Topology: Features intracellular domains that are more accessible for antibody targeting

  • Immunogenic regions: The N-terminal region (particularly amino acids 79-167) has been identified as highly immunogenic and is a common target for antibodies

When developing or selecting ANO2 antibodies, researchers should consider:

  • The specific epitope location (extracellular loops versus intracellular domains)

  • The accessibility of the target epitope in native conformations

  • The conservation of target sequences across species if cross-reactivity is desired

  • The specific fragment for immunization (e.g., some commercial antibodies target a 19-amino acid synthetic peptide from the amino terminus)

What species reactivity is typically available for ANO2 antibodies?

Based on the search results, commercially available ANO2 antibodies demonstrate reactivity with several species:

Antibody TypeCatalog ReferenceHostSpecies ReactivityApplications
PolyclonalA04801RabbitHuman, Mouse, RatELISA, WB, IHC-P, IF
Monoclonal67638-1-IgMouse IgG2aHuman, MouseWB, IHC, IF-P, ELISA
Polyclonal20647-1-APRabbit IgGHuman, Mouse, RatWB, IHC, IF, IP, ELISA
PolyclonalNBP2-81703Rabbit IgGHuman, Mouse, RatELISA, ICC/IF, IHC, IHC-P, WB
Monoclonal (D-2)sc-390956Mouse IgG2a κHumanWB, IP, IF, ELISA

When selecting an antibody for multi-species studies, researchers should verify cross-reactivity experimentally, as sequence homology varies across different regions of the ANO2 protein .

What are the optimal applications and dilutions for ANO2 antibody detection?

The optimal applications and working dilutions for ANO2 antibody depend on the specific antibody and application. Based on the search results, here are recommended dilutions and applications:

ApplicationRecommended Dilution RangeNotes
Western Blot (WB)1:500-1:6000Lower dilutions (1:500-1:1000) for polyclonal antibodies ; higher dilutions (1:1000-1:6000) for monoclonal antibodies
Immunohistochemistry (IHC)1:50-1:500Typically requires antigen retrieval with TE buffer pH 9.0 or citrate buffer pH 6.0
Immunofluorescence (IF)1:200-1:800Higher concentrations (around 20 μg/ml) may be needed for some antibodies
Immunoprecipitation (IP)0.5-4.0 μg per 1-3 mg proteinReported for specific polyclonal antibodies
ELISAVariableTypically follows manufacturer's specific protocols

For optimal results:

  • Titrate antibodies in each experimental system

  • Include positive and negative controls

  • Validate antibody specificity using ANO2-knockout tissues or cells when available

  • Follow manufacturer's recommended storage conditions (typically -20°C with 50% glycerol)

How can researchers validate the specificity of ANO2 antibodies?

Validating ANO2 antibody specificity is crucial for ensuring reliable experimental results. Based on the search results, recommended validation approaches include:

  • Multiple detection methods verification:

    • Compare results from Western blot, IHC, and IF to confirm consistent detection patterns

    • Observe expected molecular weight (approximately 114 kDa) in Western blot applications

  • Positive control samples:

    • Use tissues with known ANO2 expression, such as:

      • Mouse or rat brain tissue

      • Human brain tissue

      • Retinal tissue

      • Y79 cells, NCCIT cells, or U2OS cells

  • Advanced validation techniques:

    • Knockout/knockdown controls: Compare wild-type with ANO2-knockout or knockdown samples

    • Competing peptide blocking: Pre-incubate antibody with the immunizing peptide

    • Orthogonal validation: Correlate protein detection with mRNA expression data

    • Independent antibody validation: Use multiple antibodies targeting different epitopes of ANO2

  • Cross-reactivity assessment:

    • Test against closely related proteins (e.g., other anoctamin family members)

    • Especially important when studying ANO2 in tissues expressing multiple anoctamin proteins

Proper validation not only ensures specificity but also helps in identifying the optimal conditions for each application .

What methodological considerations are important in detecting ANO2 in brain and retinal tissues?

Detection of ANO2 in brain and retinal tissues requires special methodological considerations due to the complex nature of these tissues:

  • Tissue preparation:

    • For fixed tissues: Use appropriate fixation methods (typically 4% paraformaldehyde)

    • For frozen sections: Optimal cutting temperature (OCT) embedding is recommended

    • Section thickness: 5-10 μm sections are typically used for immunohistochemistry

  • Antigen retrieval:

    • Heat-induced epitope retrieval with TE buffer (pH 9.0) is often recommended

    • Alternative: Citrate buffer (pH 6.0) can also be effective

    • Duration and temperature are critical (typically 95-100°C for 15-20 minutes)

  • Background reduction:

    • Block endogenous peroxidase activity for IHC applications

    • Use appropriate blocking solutions (typically 5-10% normal serum)

    • Include detergents (0.1-0.3% Triton X-100) to improve antibody penetration for intracellular epitopes

  • Signal amplification considerations:

    • For weak signals: Consider tyramide signal amplification

    • For colocalization studies: Use fluorescent secondary antibodies with minimal spectral overlap

    • For quantitative analysis: Maintain consistent exposure settings across samples

  • Controls specific to neural tissue:

    • Include regions known to express ANO2 (e.g., olfactory epithelium) as positive controls

    • Use ANO2-negative regions as internal negative controls

    • For retina: Be aware of autofluorescence from lipofuscin granules

The antibody against ANO2 has been demonstrated to stain neuronal and glial cells (some GFAP+) from normal hippocampal and cortical regions, showing increased staining intensity near and inside MS lesions .

What is the evidence for ANO2 as an autoimmune target in multiple sclerosis?

Multiple studies have identified ANO2 as a significant autoimmune target in multiple sclerosis (MS), with several lines of evidence supporting this association:

  • Autoantibody prevalence in MS patients:

    • Large-scale screening of over 2,000 plasma samples revealed significantly increased autoantibody reactivity against ANO2 in MS patients compared to controls

    • The N-terminal region of ANO2 (residues 79-167) showed 5.3-fold higher median fluorescent intensity in MS cases versus controls (p = 1.5 × 10⁻¹⁶)

    • Positive reactivity percentages: 15.5% in all MS cases vs. 3.2% in controls (p = 4.3 × 10⁻²²)

    • This finding was replicated in independent validation cohorts (p = 2.3 × 10⁻²²)

  • Epitope mapping:

    • The minimal epitope was identified as a 12-amino acid sequence "HAGGPGDIELGP" (residues 136-147)

    • Fine-tuned epitope mapping confirmed this region using overlapping peptides

  • ANO2 expression in MS lesions:

    • Immunohistochemistry showed increased ANO2 staining intensity near and inside MS lesions

    • The protein forms cellular aggregates in these regions

    • Both neuronal and glial cells (some GFAP+) showed ANO2 expression

  • Genetic associations:

    • Strong interaction between ANO2 autoantibodies and the HLA complex MS-associated DRB1*15 allele

    • This genetic interaction reinforces the potential role of ANO2 autoreactivity in MS etiopathogenesis

These findings suggest that an ANO2 autoimmune subphenotype may exist in MS, potentially contributing to disease pathogenesis and providing a basis for subtyping MS patients .

How does molecular mimicry between ANO2 and Epstein-Barr virus contribute to autoimmunity?

The molecular mimicry between ANO2 and Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1) represents a significant mechanism potentially linking viral infection to autoimmunity in multiple sclerosis:

  • Evidence of molecular mimicry:

    • Antibodies against ANO2 recognize a fragment of EBV nuclear antigen 1 (EBNA1)

    • This cross-reactivity suggests that immune responses initially directed against EBV may target structurally similar epitopes in ANO2

  • Correlation with EBV serology:

    • Anti-EBNA1 antibody levels strongly influence anti-ANO2 antibody levels (p = 7.1 × 10⁻⁹⁵)

    • In multivariate analysis, anti-EBNA1 antibody levels had the strongest influence on anti-ANO2 antibody levels, followed by study type, MS status, and age

    • High correlation between anti-ANO2 and anti-EBNA1 reactivity in cerebrospinal fluid (CSF)

  • Proposed mechanism:

    • Initial immune response against EBV (particularly EBNA1)

    • Cross-reactive antibodies recognize structurally similar epitopes in ANO2

    • This cross-reactivity may promote central nervous system inflammation

    • T cells reactive with the same protein may be involved in the pathogenic process

  • Risk association:

    • The presence of ANO2 reactivity is associated with high MS risk, particularly in combination with:

      • HLA risk variants

      • High EBNA1 antibody titers

This molecular mimicry mechanism provides a potential explanation for how EBV infection, a known risk factor for MS, may lead to autoimmunity targeting CNS proteins like ANO2, creating a bridge between environmental triggers and autoimmune pathology .

What are the methodological approaches for detecting anti-ANO2 autoantibodies in patient samples?

Detection of anti-ANO2 autoantibodies in patient samples requires specialized methodologies optimized for autoantibody screening. Based on the search results, several approaches have been successfully employed:

  • Bead-based protein arrays:

    • Multiplex detection using protein fragments coupled to magnetic beads

    • Initial screening with 11,520 antigens, followed by focused analysis of 384 selected proteins

    • Measurement of median fluorescent intensity (MFI) values

    • Threshold setting: Median plus 3 × SD of control sample MFI values

  • Planar protein microarrays:

    • Arrays containing 21,120 protein fragments representing 12,412 unique human proteins

    • Used for confirmation of reactivity patterns identified in bead-based arrays

    • Provides independent validation on a different platform

  • Epitope mapping with peptide arrays:

    • Design of overlapping peptides (15 or 20 amino acids long with 12 or 10 amino acid overlaps)

    • N-terminally biotinylated peptides coupled to NeutrAvidin-coated magnetic beads

    • Preadsorption against NeutrAvidin-specific plasma antibodies

    • Multiplex mapping of IgG reactivity against specific ANO2 epitopes

  • Expression systems for antigen production:

    • Human Protein Atlas expression system

    • Alternative expression systems for independent validation

    • Production of both short fragments (e.g., ANO2 region 79-167) and longer constructs (e.g., region 1-365)

  • Cerebrospinal fluid (CSF) analysis:

    • Detection of anti-ANO2 antibodies in CSF samples

    • Correlation analysis between plasma and CSF reactivity

    • Parallel analysis of anti-EBNA1 antibodies in CSF

Statistical approaches include nonparametric Wilcoxon rank-sum tests for group comparisons, Fisher's exact test for evaluating differences in positive reactivity fractions, and multivariate analysis to assess the influence of various factors (e.g., anti-EBNA1 antibody levels, age, gender) .

How can active learning approaches improve ANO2 antibody-antigen binding prediction models?

Active learning (AL) represents an advanced approach to optimize experimental resources in antibody research, including studies of ANO2 antibodies. Based on search result , these methodologies can significantly enhance prediction models:

  • Active learning strategies for antibody-antigen binding prediction:

    • Model-based approaches:

      • Query-by-Committee (QBC): Trains multiple models and selects data points with greatest disagreement

      • Gradient-Based Uncertainty: Leverages model gradient as indicator of uncertainty

    • Diversity-based approaches:

      • Focus on selecting diverse sequence samples without relying on trained models

      • Ensures broad coverage of the sequence space

  • Implementation for ANO2 antibody research:

    • Training convolutional neural networks on available ANO2 binding data

    • Iteratively selecting the most informative new experiments to perform

    • Prioritizing uncertain ANO2 variants for additional experimental measurements

    • Comparing performance against random selection baselines

  • Performance evaluation metrics:

    • Receiver operating characteristic area under the curve (ROC AUC)

    • Active learning curve (ALC)

    • Area under ALC as final performance metric

  • Practical benefits for ANO2 antibody research:

    • Reducing the number of experiments needed to accurately predict ANO2 antibody binding

    • Optimizing selection of ANO2 variants for epitope mapping

    • Accelerating therapeutic ANO2 antibody development

    • Enabling more efficient characterization of autoantibody binding in MS patients

This approach is particularly valuable when researching ANO2 antibodies for diagnostic or therapeutic applications, as it can significantly reduce the experimental burden while maximizing information gain .

What are the key considerations when developing ANO2 antibodies for therapeutic or diagnostic applications?

Developing ANO2 antibodies for therapeutic or diagnostic applications, particularly in the context of multiple sclerosis, requires careful consideration of several critical factors:

  • Epitope selection and specificity:

    • Target the most immunogenic and disease-relevant epitopes

    • The N-terminal region (particularly residues 79-167) has been identified as highly relevant in MS

    • Consider the minimal epitope "HAGGPGDIELGP" (residues 136-147) for focused targeting

    • Ensure specificity against other anoctamin family members to prevent cross-reactivity

  • Accessibility considerations:

    • ANO2 is a multi-pass membrane protein with both intracellular and membrane domains

    • For therapeutic applications: Focus on accessible epitopes in disease contexts

    • For diagnostic applications: Both accessible and inaccessible epitopes may be relevant, depending on sample preparation methods

  • Cross-reactivity with viral proteins:

    • Consider the molecular mimicry between ANO2 and EBNA1

    • Assess whether therapeutic antibodies might cross-react with viral proteins

    • For diagnostics, this cross-reactivity could be leveraged to differentiate virus-induced versus primary autoimmunity

  • Subtype-specific considerations:

    • Different MS subtypes (relapsing-remitting vs. progressive) show varying rates of ANO2 autoantibody positivity

    • Relapsing MS: 15.4% positivity

    • Progressive MS: 16.4% positivity

    • Diagnostic applications should account for these subtype differences

  • Validation in relevant models:

    • Test candidate antibodies in appropriate MS models

    • Evaluate blood-brain barrier penetration for therapeutic applications

    • Assess specificity in tissues showing increased ANO2 expression in MS lesions

  • Technical considerations for diagnostics:

    • Define precise threshold values for positivity (e.g., median plus 3 × SD of control values)

    • Account for gender and age factors, though these do not appear to significantly affect ANO2 reactivity

    • Consider genetic backgrounds, particularly HLA-DRB1*15 status, which interacts with ANO2 autoantibody presence

These considerations can guide researchers in developing ANO2 antibodies that are optimized for their specific therapeutic or diagnostic applications in MS and other relevant conditions.

How can researchers reconcile contradictory findings when working with different ANO2 antibodies?

Researchers often encounter contradictory results when using different antibodies targeting the same protein. For ANO2 research, several approaches can help reconcile such discrepancies:

  • Epitope mapping and comparison:

    • Determine the exact epitopes recognized by each antibody

    • ANO2 has distinct domains (N-terminal region 79-167 vs. C-terminal region 932-1003)

    • Reactivity differences may be explained by epitope location and accessibility

    • Different epitopes may be exposed under different experimental conditions

  • Isoform-specific detection:

    • ANO2 has multiple isoforms resulting from alternative splicing

    • Verify which isoforms each antibody can detect

    • Some antibodies may only recognize specific isoforms (e.g., "At least two isoforms of EPAC2 are known to exist; this antibody will detect only the larger isoform")

  • Methodological standardization:

    • Compare experimental protocols in detail:

      • Fixation methods and duration

      • Antigen retrieval conditions

      • Blocking reagents and duration

      • Antibody concentrations and incubation times

      • Detection systems and signal amplification methods

    • Standardize protocols when comparing multiple antibodies

  • Cross-validation strategies:

    • Orthogonal techniques: Compare protein detection with mRNA expression

    • Multiple antibody validation: Use several antibodies targeting different epitopes

    • Knockout controls: Test antibodies in ANO2-knockout tissues/cells

    • Mass spectrometry validation: Confirm protein identity through proteomics

  • Statistical approaches to resolve contradictions:

    • Meta-analysis of multiple studies

    • Analysis of antibody performance across different tissues and cell types

    • Consideration of staining patterns and intensities rather than binary positive/negative results

  • Documentation and reporting:

    • Thoroughly document all experimental conditions

    • Report catalog numbers, lot numbers, and specific protocols

    • Include all controls used for validation

    • Consider publishing independent validation results to benefit the research community

By systematically addressing these factors, researchers can better understand and reconcile contradictory findings when working with different ANO2 antibodies, ultimately leading to more reliable and reproducible research outcomes.

What are the emerging applications of ANO2 antibodies in neurodegenerative disease research?

While the search results primarily focus on multiple sclerosis, they suggest several emerging applications for ANO2 antibodies in broader neurodegenerative disease research:

  • Biomarker development:

    • Anti-ANO2 autoantibodies as potential biomarkers for MS subtypes

    • Correlation with disease progression and treatment response

    • Development of diagnostic tests to identify patients with ANO2 autoimmune subphenotypes

    • Potential application in other neurodegenerative conditions with autoimmune components

  • Mechanistic studies of neurodegeneration:

    • Investigation of calcium-activated chloride channel dysfunction in neurodegeneration

    • Role of ANO2 in neuronal excitability and potential excitotoxicity

    • Contribution to sensory deficits in neurodegenerative conditions

    • Analysis of ANO2 expression and localization changes in disease states

  • Visualization of disease-associated protein aggregates:

    • ANO2 antibodies have revealed cellular aggregates near and inside MS lesions

    • Similar approaches could investigate protein aggregation in other neurodegenerative conditions

    • Colocalization studies with established markers of neurodegeneration

  • Therapeutic targeting:

    • Development of antibodies that modulate ANO2 function

    • Potential neutralization of pathogenic autoantibodies

    • Targeted immunotherapies for ANO2-reactive B cells

    • Neuroprotective strategies based on ANO2 function preservation

  • Investigation of viral-autoimmune connections:

    • Building on the EBV-ANO2 molecular mimicry model

    • Exploration of similar mechanisms in other neurodegenerative diseases

    • Development of antibodies that can differentiate between viral and autoimmune epitopes

  • Advanced imaging approaches:

    • Super-resolution microscopy of ANO2 distribution in neuronal subcellular compartments

    • In vivo imaging using labeled ANO2 antibodies

    • Correlation of ANO2 distribution with functional neuronal changes

These emerging applications demonstrate the potential breadth of ANO2 antibody utilization beyond current research focuses, particularly as our understanding of the role of ion channels and autoimmunity in neurodegenerative diseases continues to evolve.

What are common technical issues with ANO2 antibody staining and how can they be resolved?

Researchers working with ANO2 antibodies may encounter several technical challenges. Based on the search results and general immunostaining principles, here are common issues and their solutions:

  • High background staining:

    • Cause: Insufficient blocking, high antibody concentration, or non-specific binding

    • Solutions:

      • Increase blocking time (use 5-10% normal serum from the species of secondary antibody)

      • Optimize primary antibody dilution (start with manufacturer recommendations, e.g., 1:50-1:500 for IHC)

      • Add 0.1-0.3% Triton X-100 to improve antibody penetration for intracellular epitopes

      • Include additional blocking steps with bovine serum albumin (BSA)

  • Weak or absent signal:

    • Cause: Insufficient antigen retrieval, low antibody concentration, or masked epitopes

    • Solutions:

      • Optimize antigen retrieval: Use TE buffer pH 9.0 (recommended) or citrate buffer pH 6.0

      • Increase antibody concentration or incubation time

      • Try different detection systems (e.g., tyramide signal amplification)

      • Ensure proper storage of antibody (typically -20°C with 50% glycerol)

  • Inconsistent staining across samples:

    • Cause: Variability in fixation, processing, or antibody application

    • Solutions:

      • Standardize fixation protocols (type, duration, temperature)

      • Process all samples simultaneously when possible

      • Use automated staining platforms for consistency

      • Include positive controls in each experiment (e.g., brain or retinal tissue known to express ANO2)

  • Unexpected molecular weight in Western blots:

    • Cause: Post-translational modifications, alternative splicing, or degradation

    • Solutions:

      • Verify expected molecular weight (approximately 114 kDa for ANO2)

      • Use fresh samples and protease inhibitors during preparation

      • Test different sample preparation methods (e.g., different detergents for membrane proteins)

      • Consider native versus denaturing conditions

  • Cross-reactivity concerns:

    • Cause: Antibody recognizing related proteins (e.g., other anoctamin family members)

    • Solutions:

      • Use knockout/knockdown controls when available

      • Perform peptide competition assays

      • Compare staining patterns with known expression profiles

      • Consider using multiple antibodies targeting different ANO2 epitopes

These troubleshooting strategies can help researchers optimize ANO2 antibody applications and generate more reliable and reproducible results across different experimental systems.

How should researchers approach experimental design when studying ANO2 expression in complex tissues?

Studying ANO2 expression in complex tissues like brain and retina requires careful experimental design. Based on the search results, here is a comprehensive approach:

  • Tissue sampling and processing:

    • Fresh tissue collection:

      • Minimize post-mortem interval for human samples

      • Perform rapid fixation to preserve protein epitopes

      • Consider region-specific sampling based on known ANO2 expression patterns

    • Fixation optimization:

      • 4% paraformaldehyde is commonly used for ANO2 detection

      • Adjust fixation time based on tissue thickness (typically 24-48 hours for whole brain; shorter for smaller specimens)

      • Consider perfusion fixation for animal studies to improve tissue preservation

  • Comprehensive experimental controls:

    • Positive controls:

      • Include tissues with known ANO2 expression (e.g., olfactory epithelium, retina)

      • Use validated cell lines expressing ANO2 (e.g., Y79 cells)

    • Negative controls:

      • Primary antibody omission

      • Isotype controls

      • Tissues known to lack ANO2 expression

      • When available, ANO2 knockout tissues

  • Multi-method validation approach:

    • Protein detection methods:

      • Immunohistochemistry for localization

      • Western blot for size verification

      • Immunoprecipitation for interaction studies

    • Transcriptional validation:

      • In situ hybridization to detect ANO2 mRNA

      • qPCR for quantitative expression analysis

      • RNA-seq for comprehensive transcriptional profiling

  • Colocalization studies:

    • Cell-type markers:

      • Neuronal markers (e.g., NeuN, MAP2)

      • Glial markers (e.g., GFAP for astrocytes, as ANO2 has been detected in some GFAP+ cells)

      • Photoreceptor markers for retinal studies

    • Subcellular localization:

      • Membrane markers to confirm surface expression

      • Organelle markers to identify intracellular pools

      • Synaptic markers for neuronal studies

  • Quantitative analysis approaches:

    • Image analysis:

      • Use standardized acquisition parameters

      • Apply automated or semi-automated quantification

      • Measure both intensity and distribution patterns

    • Expression level quantification:

      • Western blot densitometry

      • Flow cytometry for cellular studies

      • Mass spectrometry for absolute quantification

This comprehensive approach enables robust characterization of ANO2 expression in complex tissues, reducing experimental variability and increasing confidence in the results.

How can researchers optimize ANO2 antibody-based detection in multiple sclerosis tissue samples?

Detecting ANO2 in multiple sclerosis tissue samples presents unique challenges due to disease-related tissue changes. Based on the search results, here are specialized recommendations:

  • MS lesion classification and sampling:

    • Lesion staging:

      • Sample both active and chronic lesions

      • Include normal-appearing white matter (NAWM)

      • Sample perilesional areas where ANO2 expression changes have been observed

    • Comprehensive tissue mapping:

      • Use serial sections to correlate ANO2 expression with demyelination markers

      • Map ANO2 expression relative to lesion borders

      • Consider 3D reconstruction for spatial understanding of ANO2 aggregates

  • Modified immunohistochemistry protocols:

    • Antigen retrieval optimization:

      • TE buffer pH 9.0 is recommended for ANO2 detection

      • Extended retrieval times may be necessary for heavily fixed MS tissue

      • Consider multiple retrieval methods if initial attempts yield poor results

    • Signal amplification:

      • Tyramide signal amplification for detecting low-level expression

      • Consider polymer-based detection systems

      • Optimize counterstaining to visualize tissue architecture around ANO2 aggregates

  • Double/triple immunolabeling strategies:

    • Inflammatory markers:

      • Combine ANO2 detection with CD68 (macrophages/microglia)

      • Include lymphocyte markers (CD4, CD8, CD20) to assess relationship with inflammation

      • Test for colocalization with complement components

    • Structural markers:

      • Myelin proteins (MBP, PLP) to define demyelinated areas

      • Axonal markers (neurofilament, SMI-31) to assess relationship with axonal damage

      • GFAP for astrocytes (ANO2 has been detected in some GFAP+ cells)

  • Controls specific to MS tissue:

    • Disease controls:

      • Include other neurological diseases as controls

      • Compare with non-inflammatory neurodegeneration

      • Age-matched normal controls

    • Technical controls:

      • Absorption controls with ANO2 peptides (particularly the immunogenic region 136-147)

      • Serial dilution of primary antibody to establish specificity

      • Comparison of multiple ANO2 antibodies targeting different epitopes

  • Quantitative analysis of ANO2 in MS context:

    • Lesion-specific quantification:

      • Measure ANO2+ cell density in different lesion types

      • Quantify ANO2 aggregate size and distribution

      • Analyze distance-dependent changes from lesion center to periphery

    • Cell-specific analysis:

      • Quantify proportion of different cell types expressing ANO2

      • Measure ANO2 expression intensity in different cellular populations

      • Correlate with markers of cellular stress or damage

These specialized approaches can help researchers optimize detection of ANO2 in MS tissue samples, enabling more detailed characterization of its role in MS pathology .

What are promising future directions for ANO2 antibody development and application?

Based on the search results and current trends in antibody research, several promising future directions emerge for ANO2 antibody development and application:

  • Advanced therapeutic antibody development:

    • Humanized ANO2 antibodies for therapeutic applications

    • Bispecific antibodies targeting ANO2 and inflammatory mediators

    • Antibody-drug conjugates for targeted delivery to ANO2-expressing cells

    • Antibodies that can modulate ANO2 channel function for neurological disorders

  • Diagnostic applications:

    • Point-of-care tests for ANO2 autoantibodies in MS

    • Multi-marker panels combining ANO2 with other MS-associated autoantibodies

    • Cerebrospinal fluid diagnostics to detect intrathecal anti-ANO2 antibody production

    • Imaging agents based on ANO2 antibodies for visualization of lesions

  • Technical innovations:

    • Single-domain antibodies (nanobodies) against ANO2 for improved tissue penetration

    • Recombinant antibody engineering for enhanced specificity and reduced background

    • AI-guided epitope selection and antibody design using active learning approaches

    • CRISPR-engineered cell lines for improved antibody validation

  • Research applications:

    • Tools for studying ANO2 trafficking and dynamics in living cells

    • Antibodies for super-resolution microscopy of ANO2 distribution

    • Conformation-specific antibodies to distinguish active vs. inactive channel states

    • Antibodies for studying ANO2 in human induced pluripotent stem cell-derived neurons

  • Understanding disease mechanisms:

    • Characterization of ANO2 autoimmune subphenotypes in MS

    • Investigation of ANO2's role in other neurodegenerative disorders

    • Further exploration of viral-autoimmune connections through molecular mimicry

    • Studies of ANO2's role in neuronal excitability and potential excitotoxicity

These future directions highlight the expanding potential of ANO2 antibodies beyond current applications, particularly in therapeutic development, diagnostics, and understanding disease mechanisms.

How might advances in computational methods enhance ANO2 antibody research?

Computational approaches are increasingly important in antibody research and offer significant potential for advancing ANO2 antibody development and applications:

  • Epitope prediction and antibody design:

    • Structure-based epitope prediction:

      • In silico modeling of ANO2's three-dimensional structure

      • Identification of surface-exposed, immunogenic regions

      • Design of antibodies targeting specific functional domains

    • Antibody-antigen docking simulations:

      • Predict binding modes between antibodies and ANO2

      • Optimize antibody complementarity determining regions (CDRs)

      • Evaluate binding affinity through molecular dynamics simulations

  • Machine learning approaches for binding prediction:

    • Active learning strategies:

      • Query-by-Committee (QBC) to select the most informative experiments

      • Gradient-Based Uncertainty measures to prioritize uncertain variants

      • Iterative improvement of predictive models with minimal experimental data

    • Deep learning for antibody-antigen interactions:

      • Convolutional neural networks for sequence-based binding prediction

      • Representation learning to capture complex binding patterns

      • Transfer learning from related proteins to improve ANO2-specific models

  • Bioinformatic analysis of sequence variation:

    • Cross-species conservation analysis:

      • Identify conserved regions that may be functionally important

      • Guide antibody development for cross-species reactivity

      • Understand evolutionary constraints on ANO2 structure

    • Variant effect prediction:

      • Assess the impact of genetic variants on antibody binding

      • Identify patient-specific variations that might affect diagnostic accuracy

      • Develop antibodies robust to common polymorphisms

  • Integration of multi-omics data:

    • Network analysis:

      • Map ANO2 interactions with other proteins

      • Identify potential co-targets for multiplexed antibody development

      • Understand ANO2's role in broader signaling networks

    • Expression correlation analysis:

      • Correlate ANO2 expression with disease biomarkers

      • Identify tissue-specific expression patterns to guide antibody application

      • Discover cell types with high ANO2 expression as targets for antibody therapy

These computational approaches can dramatically accelerate ANO2 antibody research by reducing experimental burden, optimizing experimental design, and providing mechanistic insights into antibody-antigen interactions .

What interdisciplinary approaches might advance our understanding of ANO2 autoimmunity in neurological diseases?

Advancing our understanding of ANO2 autoimmunity in neurological diseases will require interdisciplinary approaches that integrate multiple scientific disciplines:

  • Integrated immunology and neuroscience:

    • Neuroimmune interactions:

      • Study how ANO2 autoantibodies affect neuronal function and signaling

      • Investigate blood-brain barrier permeability to ANO2 antibodies

      • Examine inflammatory responses in ANO2-expressing neural tissues

    • Single-cell analysis:

      • Characterize ANO2-reactive B and T cell populations at single-cell resolution

      • Map ANO2 expression in different neuronal and glial subtypes

      • Correlate with functional outcomes and disease progression

  • Virology and autoimmunity connections:

    • Molecular mimicry mechanisms:

      • Further characterize the structural similarities between ANO2 and EBNA1

      • Identify other viral proteins with potential cross-reactivity to ANO2

      • Develop interventions targeting this molecular mimicry

    • Longitudinal studies:

      • Track EBV infection, anti-EBNA1 antibodies, and anti-ANO2 antibodies over time

      • Determine temporal relationships between viral infection and autoimmunity

      • Identify early biomarkers for developing ANO2 autoimmunity

  • Genetics and personalized medicine:

    • HLA and immune response genetics:

      • Expand on the known interaction between ANO2 autoantibodies and HLA-DRB1*15

      • Identify additional genetic factors influencing ANO2 autoimmunity

      • Develop genetic risk profiles for ANO2 autoimmune subphenotypes

    • Pharmacogenomics:

      • Predict response to therapies based on genetic profiles

      • Personalize treatment approaches for patients with ANO2 autoimmunity

      • Develop targeted therapies for specific genetic backgrounds

  • Advanced imaging and structural biology:

    • In vivo molecular imaging:

      • Develop tracers based on ANO2 antibodies for PET or SPECT imaging

      • Visualize ANO2 expression changes in living subjects

      • Monitor response to therapies targeting ANO2 autoimmunity

    • Structural characterization:

      • Determine crystal structures of ANO2-antibody complexes

      • Map conformational epitopes using hydrogen-deuterium exchange mass spectrometry

      • Understand structural basis of cross-reactivity with viral proteins

  • Systems biology and computational modeling:

    • Disease network modeling:

      • Map interactions between environmental factors, genetic predisposition, and ANO2 autoimmunity

      • Simulate disease progression under different intervention scenarios

      • Identify key nodes for therapeutic targeting

    • Multi-scale modeling:

      • Link molecular interactions to cellular, tissue, and organism-level outcomes

      • Predict long-term consequences of ANO2 autoimmunity

      • Model therapeutic interventions targeting different aspects of ANO2 autoimmunity

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.