CHL Antibody

Shipped with Ice Packs
In Stock

Description

Key CHL-1 Antibodies and Applications

Antibody NameHost SpeciesTargetApplicationsCross-Reactivity
Goat Anti-Mouse AF2147GoatMouse CHL-1WB, IHC, IP40% with human CHL-1
Rat Anti-Human MAB2126RatHuman CHL-1IHC, ICC, WBNo cross-reactivity

WB: Western Blot; IHC: Immunohistochemistry; IP: Immunoprecipitation; ICC: Immunocytochemistry .

Neural Development

  • CHL-1 suppresses neural progenitor cell proliferation via ERK1/2 inhibition, impacting cortical development .

  • Astrocytic CHL-1 expression is upregulated by PI3K/PKCδ signaling, suggesting roles in glial reactivity .

Cancer Biology

  • Elevated serum CHL-1 levels in GIST patients correlate with poor prognosis (HR = 2.1, p < 0.01) .

  • Nuclear CHL-1 localization in melanoma tissues implicates unconventional roles in tumorigenesis .

Therapeutic Potential

While CHL-1 antibodies are primarily research tools, their diagnostic utility is demonstrated in:

  • Stratifying GIST patients by risk profile .

  • Tracking CHL-1 dynamics in Alzheimer’s disease models .

Technical Considerations for CHL Antibodies

  • Species specificity: Anti-mouse CHL-1 (AF2147) shows partial human cross-reactivity, whereas anti-human MAB2126 is species-specific .

  • Detection sensitivity: Optimal results require 0.2 mg/mL lysate concentrations in Western blots .

  • Tissue localization: Heat-induced epitope retrieval enhances CHL-1 detection in formalin-fixed paraffin sections .

Emerging Directions

Recent advancements in antibody discovery, such as microfluidics-enabled single-cell screening, could accelerate therapeutic CHL-1 antibody development . Current challenges include elucidating CHL-1’s nuclear functions and validating its utility as a pan-cancer biomarker.

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
CHL antibody; At3g47860 antibody; T23J7.190Chloroplastic lipocalin antibody; AtCHL antibody
Target Names
CHL
Uniprot No.

Target Background

Function
CHL is a lipocalin protein that plays a crucial role in protecting thylakoidal membrane lipids from peroxidation and conferring resistance to oxidative stress. This protection is particularly important in response to high light conditions and other stressors, such as heat shocks, as it safeguards against damage caused by singlet oxygen. CHL is essential for seed longevity, ensuring the integrity of polyunsaturated lipids during seed development and storage.
Gene References Into Functions
  1. AtTIL and AtCHL exhibit overlapping functions in lipid protection, which are essential for stress resistance and survival. PMID: 23837879
  2. AtCHL is localized within the thylakoid lumen, where it mitigates membrane lipid peroxidation and enhances tolerance to oxidative stress induced by paraquat or drought treatment. PMID: 19674405
Database Links

KEGG: ath:AT3G47860

STRING: 3702.AT3G47860.1

UniGene: At.3254

Protein Families
Calycin superfamily, Lipocalin family
Subcellular Location
Plastid, chloroplast thylakoid lumen.
Tissue Specificity
Expressed in leaves at low levels (at protein levels). Present in seeds.

Q&A

What is CHL and what biological functions does it serve?

CHL (Chordin-Like) is a synonym for the CHRDL1 gene product, which encodes chordin-like 1 protein. This secreted protein plays crucial roles in cell differentiation and eye development pathways. The human version of CHL has a canonical amino acid length of 456 residues and a protein mass of approximately 52 kilodaltons, with four different isoforms identified to date. As a secreted protein, it functions in the extracellular environment, modulating various developmental processes. When designing experiments targeting CHL, researchers should account for its secretory nature, which affects sample preparation protocols and detection methodologies .

What are the primary applications for CHL antibodies in research?

CHL antibodies are routinely employed in several key laboratory techniques:

  • Western blotting for protein detection and semi-quantification

  • Enzyme-Linked Immunosorbent Assay (ELISA) for quantitative analysis

  • Immunocytochemistry for cellular localization studies

  • Immunohistochemistry for tissue distribution analysis

Each application requires specific optimization protocols and validation strategies to ensure specificity and sensitivity. For instance, Western blot applications typically require optimization of antibody dilution, blocking conditions, and detection systems specific to the cellular context being investigated .

How do I select the appropriate CHL antibody for my specific research needs?

Selection of an appropriate CHL antibody should be guided by several critical factors:

  • Experimental application (Western blot, ELISA, immunostaining)

  • Species reactivity requirements (human, mouse, etc.)

  • Antibody format (monoclonal vs. polyclonal)

  • Conjugate requirements (unconjugated or tagged with fluorophores, enzymes)

  • Evidence of validation in similar applications

Most importantly, researchers should verify that the antibody has been properly characterized using methods such as knockout validation, orthogonal approaches, or mass spectrometry confirmation. This is essential given that approximately 50% of commercial antibodies fail to meet basic standards for characterization, resulting in potentially misleading experimental results .

How can I optimize CHL antibody performance for immunohistochemistry in different tissue types?

Optimizing CHL antibody performance for immunohistochemistry requires systematic protocol adjustments based on tissue specificity:

  • Fixation optimization: Different fixation protocols significantly impact epitope availability. For CHL detection, compare paraformaldehyde (4%) versus alternative fixatives.

  • Antigen retrieval methods: Test both heat-induced (citrate buffer pH 6.0, EDTA pH 9.0) and enzymatic retrieval methods.

  • Blocking optimization: Use tissue-specific blocking protocols; for neural tissues, increase blocking duration to minimize background.

  • Antibody titration: Perform sequential dilutions (1:100, 1:500, 1:1000, 1:5000) to determine optimal signal-to-noise ratio.

  • Detection system selection: Compare amplification systems (TSA, polymer-based) for low-abundance targets.

Most importantly, always include appropriate controls, especially knockout or knockdown tissues, to verify specificity in the particular tissue context. The NeuroMab approach illustrates the importance of testing antibodies against actual tissue samples rather than relying solely on ELISA results with recombinant proteins .

What strategies can be employed for multiplex detection of CHL alongside other proteins?

For multiplex detection protocols involving CHL antibodies:

  • Antibody compatibility assessment:

    • Select primary antibodies from different host species

    • Verify sequential staining compatibility through pilot experiments

    • Test for cross-reactivity between detection systems

  • Multiplexing methodologies:

    • Fluorescent multiplexing: Use spectrally distinct fluorophores with minimal overlap

    • Chromogenic multiplexing: Employ enzyme systems with different substrates

    • Sequential multiplexing: Apply stripping/reprobing protocols with validation

  • Signal separation techniques:

    • Employ multispectral imaging for fluorescent overlap compensation

    • Utilize computational unmixing algorithms for complex signals

    • Implement sequential scanning strategies for confocal microscopy

Each multiplex combination requires dedicated validation to ensure signals represent true protein expression patterns rather than technical artifacts .

How should I design experiments to study CHL protein interactions using co-immunoprecipitation with CHL antibodies?

When designing co-immunoprecipitation experiments for CHL protein interactions:

  • Lysis buffer optimization:

    • Test multiple buffer compositions (RIPA, NP-40, digitonin-based)

    • Adjust salt concentration (150-500 mM) to balance specificity with interaction preservation

    • Evaluate detergent effects on preserving CHL protein complexes

  • Antibody coupling strategies:

    • Compare direct coupling to beads versus protein A/G approaches

    • Optimize antibody-to-bead ratios (typically 5-10 μg antibody per reaction)

    • Consider crosslinking antibodies to beads to prevent heavy chain interference

  • Controls implementation:

    • Include isotype control antibodies processed identically

    • Perform reverse co-immunoprecipitation when possible

    • Use knockout/knockdown samples as negative controls

  • Elution and detection optimization:

    • Compare harsh (SDS, low pH) versus mild (competitive peptide) elution conditions

    • Optimize Western blot protocols specifically for immunoprecipitated samples

    • Consider mass spectrometry validation of interacting partners

This methodological approach aligns with the "multiple pillars" concept of antibody validation, employing multiple independent methods to confirm interactions .

What are the recommended validation methods for ensuring CHL antibody specificity?

Following the "five pillars" of antibody validation framework, CHL antibodies should be validated through:

  • Genetic strategy validation:

    • Use CRISPR knockout cell lines expressing no CHL protein

    • Compare with siRNA/shRNA knockdown samples showing reduced expression

    • Test antibody in genetic models with known CHL mutations

  • Orthogonal strategy validation:

    • Compare antibody-based detection with mass spectrometry quantification

    • Correlate results with mRNA expression data

    • Verify findings with alternative protein detection methods

  • Independent antibody validation:

    • Test multiple antibodies targeting different CHL epitopes

    • Compare monoclonal and polyclonal antibody performance

    • Evaluate agreement between different antibody clones

  • Recombinant expression validation:

    • Test detection in systems with controlled CHL overexpression

    • Perform epitope mapping to confirm binding specificity

    • Assess cross-reactivity with related protein family members

  • Immunocapture mass spectrometry:

    • Analyze proteins captured by the CHL antibody

    • Confirm presence of target and identify potential cross-reactivities

    • Quantify specificity through enrichment factors

This comprehensive validation approach significantly increases confidence in antibody specificity and experimental reproducibility .

How do monoclonal and polyclonal CHL antibodies compare in research applications?

Comparative analysis of monoclonal versus polyclonal CHL antibodies shows distinct advantages for each type:

CharacteristicMonoclonal CHL AntibodiesPolyclonal CHL Antibodies
SpecificityHigher epitope specificityVariable batch-to-batch
SensitivityLower for single epitopesHigher for multiple epitopes
ReproducibilityHigher between batchesLower between preparations
ApplicationsSuperior for quantitative assaysBetter for detection in tissues
ProductionHybridoma or recombinantAnimal immunization
RenewalRenewable resourceNon-renewable resource
CostHigher production costGenerally lower cost

Polyclonal antibodies provide the advantage of recognizing multiple epitopes, potentially increasing sensitivity, but their non-renewable nature introduces significant batch variability that can compromise experimental reproducibility. Monoclonal antibodies offer consistency but may have reduced sensitivity for certain applications. Recent trends favor recombinant monoclonal antibodies which combine specificity with reproducibility advantages .

How should researchers address batch-to-batch variability in CHL antibody performance?

To address batch variability issues, researchers should implement:

  • Standardized validation protocols:

    • Perform side-by-side comparisons between batches

    • Document antibody performance metrics for each batch

    • Establish acceptance criteria before deploying new batches

  • Reference standard creation:

    • Create internal reference standards for key applications

    • Maintain positive control samples from validated experiments

    • Archive validation data for longitudinal comparison

  • Critical reagent management:

    • Purchase sufficient quantities of validated batches when possible

    • Aliquot and preserve antibody stocks to prevent freeze-thaw cycles

    • Implement antibody tracking systems with validation documentation

  • Alternative technology consideration:

    • Consider transitioning to recombinant antibodies for critical assays

    • Evaluate renewable alternatives to polyclonal antibodies

    • Implement orthogonal detection methods as controls

This approach is particularly important for polyclonal CHL antibodies, which demonstrate significant variability between production lots due to their biological origin and complexity of the antibody mixture present in serum .

What are common causes of false positive or negative results when using CHL antibodies, and how can they be addressed?

Common causes of false results with CHL antibodies include:

  • False positives:

    • Cross-reactivity with structurally similar proteins

    • Non-specific binding to highly abundant proteins

    • Secondary antibody cross-reactivity

    • Sample overloading in Western blots

  • False negatives:

    • Epitope masking by protein modifications

    • Insufficient antigen retrieval in fixed samples

    • Improper sample preparation destroying epitopes

    • Antibody degradation or denaturation

Mitigation strategies:

  • Implement multiple controls (positive, negative, isotype)

  • Validate results with orthogonal methods

  • Optimize protocols for each specific tissue/cell type

  • Include knockout/knockdown controls whenever possible

  • Verify antibody performance in your specific experimental system

The systematic approach to troubleshooting should include careful documentation of all experimental conditions and sequential modification of single variables to identify sources of variability .

How can researchers properly design experiments to study post-translational modifications of CHL protein?

When studying post-translational modifications (PTMs) of CHL protein:

  • Modification-specific antibody selection:

    • Verify antibody specificity for the particular PTM

    • Test against both modified and unmodified peptides

    • Evaluate cross-reactivity with similar PTM motifs

  • Control implementation:

    • Include samples with enzymatically removed modifications

    • Use treatments that induce or inhibit the specific modification

    • Generate site-directed mutants at modification sites

  • Enrichment strategies:

    • Implement PTM-specific enrichment protocols before detection

    • Use sequential immunoprecipitation approaches

    • Apply orthogonal enrichment methods for verification

  • Detection optimization:

    • Adjust sample preparation to preserve labile modifications

    • Optimize buffer conditions to maintain modification integrity

    • Consider specialized detection methods for quantification

  • Mass spectrometry validation:

    • Confirm antibody-detected modifications with MS/MS

    • Quantify modification stoichiometry

    • Map modification sites precisely

This systematic approach combines antibody-based detection with orthogonal validation methods to ensure reliable PTM identification and quantification .

What considerations are important when adapting CHL antibody protocols across different model organisms?

When adapting CHL antibody protocols across species:

  • Sequence homology assessment:

    • Perform sequence alignment analysis between species

    • Identify conserved epitope regions versus divergent domains

    • Predict potential cross-reactivity based on homology

  • Cross-reactivity testing:

    • Validate antibody performance in each species

    • Test knockout/knockdown controls from each organism

    • Perform peptide competition assays with species-specific peptides

  • Protocol adaptation:

    • Adjust fixation protocols based on tissue characteristics

    • Modify extraction methods for species-specific matrix effects

    • Optimize antibody concentration for each species

  • Species-specific controls:

    • Include appropriate positive controls from each species

    • Develop negative controls specific to each organism

    • Validate using orthogonal methods in each species

These considerations are critical since even small sequence variations between species can significantly impact antibody binding and specificity. Thorough validation in each model organism prevents misinterpretation of results .

How can CHL antibodies be effectively utilized in single-cell protein analysis techniques?

For single-cell protein analysis with CHL antibodies:

  • Flow cytometry applications:

    • Optimize fixation/permeabilization protocols for intracellular detection

    • Validate antibody performance in flow cytometry specifically

    • Develop compensation controls for multiplex applications

    • Establish quantification standards for expression level assessment

  • Mass cytometry (CyTOF) implementation:

    • Validate metal-conjugated antibodies separately from fluorescent versions

    • Optimize staining concentrations for single-cell resolution

    • Develop analysis pipelines for high-dimensional data interpretation

  • Imaging cytometry approaches:

    • Adapt immunofluorescence protocols for suspension cells

    • Optimize signal-to-noise ratios for quantitative imaging

    • Implement machine learning algorithms for automated analysis

  • Single-cell Western blot considerations:

    • Modify standard protocols for microfluidic platforms

    • Adjust antibody concentrations for microscale applications

    • Validate detection limits for low-abundance samples

These techniques enable researchers to examine CHL expression heterogeneity within populations and correlate expression with cellular phenotypes at unprecedented resolution .

What strategies should be employed when developing quantitative assays for CHL protein levels?

For developing quantitative CHL protein assays:

  • Calibration standard development:

    • Generate recombinant CHL protein standards

    • Verify standard purity and concentration

    • Create calibration curves spanning physiological ranges

  • Assay design considerations:

    • Select antibody pairs recognizing non-overlapping epitopes for sandwich assays

    • Optimize capture and detection antibody concentrations

    • Evaluate matrix effects on quantification accuracy

  • Validation parameters:

    • Determine assay sensitivity (limit of detection)

    • Assess linearity across the measurement range

    • Test precision (intra-assay and inter-assay variability)

    • Verify accuracy using spike-recovery experiments

    • Evaluate specificity against related proteins

  • Sample preparation standardization:

    • Develop consistent extraction protocols

    • Implement quality control measures for sample integrity

    • Standardize handling procedures to minimize variability

This methodical approach ensures that quantitative measurements of CHL protein are reliable, reproducible, and accurately reflect biological conditions .

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.