CALU Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Key Features of CALU Monoclonal Antibodies

CALU mAbs are developed using recombinant fragments or peptides of human calumenin. Two prominent clones are highlighted:

Clone 5D7-H21-C23 (Creative Diagnostics)

  • Host Species: Mouse (IgG1 isotype)

  • Immunogen: Recombinant C-terminal fragment of human calumenin (NP_001210.1)

  • Applications: Western blot (WB)

  • Reactivities: Human

Clone 67585-1-Ig (Proteintech)

  • Host Species: Mouse (IgG1 isotype)

  • Immunogen: Full-length calumenin fusion protein

  • Applications: WB, immunohistochemistry (IHC), immunofluorescence/immunocytochemistry (IF/ICC), ELISA

  • Reactivities: Human, mouse, rat

Table 1: Application-Specific Performance

ApplicationClone 5D7-H21-C23 Clone 67585-1-Ig
Western BlotConfirmed (HeLa)Confirmed (HeLa, NIH/3T3, HepG2, Jurkat, RAW264.7, HEK-293)
IHCNot testedValidated (human colon/kidney/cervical cancer, mouse placenta)
IF/ICCNot testedValidated (HeLa cells)
ELISANot testedCompatible

Table 2: Recommended Dilutions

ApplicationClone 67585-1-Ig
WB1:1000–1:4000
IHC1:250–1:1000
IF/ICC1:50–1:500

Research Implications

  • Disease Associations: CALU regulates vitamin K-dependent carboxylation and may influence thrombosis and cardiovascular diseases .

  • Therapeutic Potential: While no direct therapies currently target CALU, its interaction with clotting factors (e.g., Factor IX) positions it as a candidate for anticoagulant research .

Product Specs

Form
Purified mouse monoclonal antibody in a buffer containing 0.1M Tris-Glycine (pH 7.4), 150 mM NaCl, 0.2% sodium azide, and 51% glycerol.
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. For specific delivery estimates, please consult your local distributor.

Q&A

What is CALU and what are the basic characteristics of CALU monoclonal antibodies?

CALU (Calumenin) is a calcium-binding protein with Swiss-Prot accession number P10515 and Gene ID 1737. It belongs to the CREC family of Ca²⁺-binding proteins and plays roles in protein folding and sorting within the secretory pathway . CALU monoclonal antibodies are specifically engineered to recognize and bind to distinct epitopes of the CALU protein.

Currently available CALU monoclonal antibodies include clone 6C8, which targets full-length recombinant CALU, and clone 4C6-G10-B12, which specifically targets the C-terminus of CALU protein. These antibodies are typically mouse-derived IgG1 isotype and show reactivity toward human CALU, with some cross-reactivity to mouse and rat CALU proteins .

What experimental applications are CALU monoclonal antibodies validated for?

CALU monoclonal antibodies have been validated for several research applications:

  • Western Blotting: The recommended working dilution is typically 1-5 μg/ml, allowing for specific detection of CALU protein in tissue and cell lysates .

  • ELISA (Enzyme-Linked Immunosorbent Assay): CALU antibodies can be used for quantitative measurement of CALU in various sample types .

  • Immunohistochemistry: Although not explicitly specified in the provided information, monoclonal antibodies are generally applicable for tissue section analysis.

When designing experiments, researchers should consider that different antibody clones may have distinct performance characteristics across these applications, necessitating validation in each specific experimental system.

How should researchers optimize sample preparation for CALU detection?

For optimal CALU detection, sample preparation should account for the protein's characteristics. Since CALU is a calcium-binding protein, researchers should:

  • Consider calcium concentration in buffers: Ensure consistent calcium levels across samples, as this may affect protein conformation and antibody binding.

  • Protein extraction protocols: Use buffers containing protease inhibitors to prevent degradation of the target protein. CALU is primarily localized in the endoplasmic reticulum and secretory pathway, so extraction protocols should effectively solubilize these cellular compartments.

  • Sample denaturation conditions: For Western blot applications, standard reducing conditions (using β-mercaptoethanol or DTT) are generally suitable, though non-reducing conditions might preserve certain conformational epitopes if needed for specific antibody clones .

  • Control samples: Always include positive controls (samples known to express CALU) and negative controls (samples lacking CALU expression) to validate antibody specificity.

What are the recommended storage and handling procedures for CALU monoclonal antibodies?

To maintain antibody functionality and extend shelf-life:

  • Storage temperature: Store at -20°C for long-term storage. CALU antibodies are typically supplied in a stabilizing buffer containing glycerol (e.g., 51%) which prevents freezing at this temperature .

  • Working aliquots: Prepare small working aliquots to avoid multiple freeze-thaw cycles, which can degrade antibody performance.

  • Buffer composition: CALU antibodies are typically formulated in buffer containing 0.1M Tris-Glycine (pH 7.4), 150 mM NaCl, with 0.2% sodium azide as a preservative and glycerol for stabilization .

  • Handling precautions: Work with antibodies using clean pipettes and tubes to prevent contamination. Avoid vortexing antibody solutions, as this can cause protein denaturation.

  • Expiration considerations: While monoclonal antibodies generally have good stability, their performance should be validated periodically, especially for critical experiments.

How can researchers validate the specificity of CALU monoclonal antibodies in their experimental systems?

Rigorous validation of antibody specificity is crucial for generating reliable research data. For CALU antibodies, consider implementing these validation strategies:

  • Knockout/knockdown controls: Generate CALU knockout or knockdown samples to confirm antibody specificity. The absence of signal in these samples strongly supports antibody specificity.

  • Overexpression studies: Overexpress tagged CALU in cell systems with low endogenous expression, then confirm co-localization of antibody signal with the tag.

  • Peptide competition assays: Pre-incubate the antibody with purified recombinant CALU protein (such as the immunogen used to generate the antibody) before application to samples. Signal reduction indicates specific binding.

  • Mass spectrometry validation: Use immunoprecipitation with the CALU antibody followed by mass spectrometry to confirm pulled-down proteins match CALU.

  • Multiple antibody approach: Compare results using different CALU antibody clones (e.g., clone 6C8 versus 4C6-G10-B12) that recognize different epitopes .

What strategies can address cross-reactivity issues when using CALU monoclonal antibodies?

Cross-reactivity occurs when antibodies bind to proteins other than their intended target. For CALU antibodies, researchers can:

  • Epitope mapping: Determine the exact epitope recognized by your antibody. The 4C6-G10-B12 clone targets the C-terminus of CALU, which may have specific advantages for certain applications .

  • Species-specific considerations: Verify species cross-reactivity experimentally. While manufacturers indicate human, mouse, and rat reactivity, actual performance can vary across species due to sequence differences .

  • Pre-absorption protocols: For tissues with potential cross-reactivity, pre-absorb antibodies with recombinant proteins from the relevant family to reduce non-specific binding.

  • Dilution optimization: Titrate antibody concentrations to find the optimal balance between signal strength and background reduction.

  • Alternative detection methods: Complement antibody-based detection with non-antibody methods like mass spectrometry or PCR to confirm findings.

How do different immunization strategies affect the performance of CALU monoclonal antibodies?

The immunogen used to generate an antibody significantly impacts its characteristics. For CALU antibodies:

  • Full-length versus fragment immunization: Clone 6C8 was raised against full-length recombinant CALU, potentially recognizing multiple epitopes, while clone 4C6-G10-B12 was generated against C-terminal fragments, offering more targeted epitope recognition .

  • Recombinant protein quality: CALU antibodies are typically generated using E. coli-expressed recombinant proteins, which may lack post-translational modifications present in mammalian cells .

  • Implications for experimental design:

    • C-terminus-specific antibodies like 4C6-G10-B12 may be advantageous for detecting specific CALU isoforms

    • Full-length antibodies might offer broader detection capabilities across protein variants

    • Neither approach may effectively detect heavily modified forms of the protein

  • Choosing appropriate antibodies: Select antibodies based on the specific research question, considering whether detection of all CALU forms or specific variants is required.

How can researchers optimize CALU detection in multiplex immunoassay systems?

Multiplex detection systems allow simultaneous measurement of multiple proteins. For incorporating CALU antibodies:

  • Antibody conjugation strategies:

    • Direct labeling with fluorophores, ensuring the conjugation process doesn't affect the antigen-binding site

    • Biotinylation for streptavidin-based detection systems

    • Selection of compatible secondary antibodies based on host species and isotype (mouse IgG1 for both 6C8 and 4C6-G10-B12 clones)

  • Cross-reactivity assessment: Test for potential cross-reactivity with other antibodies in the multiplex panel by comparing signals from single-plex versus multiplex formats.

  • Standard curve optimization: Develop reliable standard curves using recombinant CALU protein across appropriate concentration ranges for quantitative applications.

  • Signal amplification techniques: For low-abundance detection, consider incorporating tyramide signal amplification or similar technologies, validating that these don't introduce artifacts.

  • Data normalization approaches: Implement appropriate controls and normalization methods to account for technical variability across samples.

What are the methodological considerations when developing a sandwich ELISA for CALU detection?

Sandwich ELISA development for CALU requires careful antibody pair selection:

  • Antibody pair selection: Identify compatible capture and detection antibody pairs that:

    • Recognize distinct, non-overlapping epitopes

    • Do not interfere with each other's binding

    • Function effectively in the ELISA format

  • Optimization steps:

    • Determine optimal coating concentration for capture antibody (typically 1-10 μg/ml)

    • Establish appropriate blocking conditions to minimize background

    • Titrate detection antibody concentration for maximum signal-to-noise ratio

    • Validate with recombinant CALU protein standards

  • Assay validation:

    • Determine limit of detection and quantification

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

    • Evaluate specificity against related calcium-binding proteins

    • Confirm linearity across the desired concentration range

  • Sample preparation considerations:

    • Optimize sample dilution to ensure measurements fall within the linear range

    • Evaluate matrix effects from different sample types (serum, tissue lysates, etc.)

    • Consider adding calcium chelators or stabilizers as appropriate

This methodological approach is similar to that used for developing other protein-specific sandwich ELISAs, such as those described for viral antigen detection .

What are common troubleshooting approaches for weak or absent CALU signal in Western blots?

When encountering weak or absent CALU signals in Western blotting, systematically address these potential issues:

  • Protein extraction efficiency:

    • Verify extraction protocol effectiveness for membrane/secretory pathway proteins

    • Consider alternative lysis buffers containing different detergents (RIPA, NP-40, Triton X-100)

    • Check total protein concentration using Bradford or BCA assays

  • Sample processing:

    • Evaluate heat denaturation conditions (temperature, duration)

    • Assess reducing agent concentration and effectiveness

    • Consider native versus denaturing conditions based on epitope characteristics

  • Antibody-specific factors:

    • Titrate antibody concentration beyond recommended range (1-5 μg/ml)

    • Extend primary antibody incubation time (overnight at 4°C)

    • Test alternative CALU antibody clones targeting different epitopes

  • Transfer and detection optimization:

    • Verify transfer efficiency using reversible protein stains

    • Increase exposure time during imaging

    • Consider more sensitive detection systems (enhanced chemiluminescence)

    • Optimize membrane blocking conditions to reduce background without impeding antibody binding

  • Positive controls:

    • Include recombinant CALU protein as a reference standard

    • Use cell lines known to express high levels of CALU

How can researchers address non-specific binding when using CALU monoclonal antibodies?

Non-specific binding can complicate data interpretation. To improve specificity:

  • Blocking optimization:

    • Test different blocking agents (BSA, milk, commercial blockers)

    • Extend blocking time or increase blocker concentration

    • Add 0.1-0.3% Tween-20 to wash buffers and antibody diluents

  • Antibody dilution:

    • Further dilute primary antibody beyond recommended range

    • Shorten incubation time if extended incubations increase background

  • Cross-adsorption techniques:

    • Pre-adsorb antibodies with tissues or lysates from species with expected cross-reactivity

    • Use highly purified secondary antibodies that have been cross-adsorbed against multiple species

  • Buffer modifications:

    • Add 0.1-0.5% non-ionic detergents to reduce hydrophobic interactions

    • Adjust salt concentration to optimize ionic strength

    • Include carrier proteins like BSA (0.1-1%) in antibody diluent

  • Alternative detection methods:

    • Switch between colorimetric, chemiluminescent, or fluorescent detection systems

    • Consider more specific detection technologies like proximity ligation assay

What methodological approaches can resolve contradictory results between different CALU antibody clones?

When different CALU antibody clones produce conflicting results:

  • Epitope mapping comparison:

    • Determine the precise epitopes recognized by each antibody clone

    • Assess whether epitopes might be differentially accessible in various experimental conditions

    • Consider whether post-translational modifications might affect epitope availability

  • Isoform-specific detection:

    • Evaluate whether antibodies detect different CALU isoforms or splice variants

    • Use RT-PCR to confirm which CALU transcript variants are expressed in your experimental system

  • Validation with orthogonal methods:

    • Confirm results with non-antibody-based techniques like mass spectrometry

    • Use genetic approaches (siRNA, CRISPR) to manipulate CALU expression and verify antibody specificity

    • Implement RNA-seq to correlate transcript levels with protein detection

  • Technical validation:

    • Have multiple researchers independently perform experiments

    • Blind sample identity during analysis to eliminate bias

    • Use statistical approaches to quantify and compare signals objectively

This approach mirrors validation strategies used for other monoclonal antibodies where multiple clones are available, as seen in research with viral antigen detection .

How can microfluidic-based approaches enhance the development and characterization of next-generation CALU monoclonal antibodies?

Emerging microfluidic technologies offer powerful approaches for antibody development:

  • Single B-cell isolation and screening:

    • Microfluidic platforms enable isolation of individual B cells from immunized animals

    • Each cell can be screened for antibody production and antigen specificity

    • This approach dramatically increases throughput compared to traditional hybridoma methods

    • Potential to identify rare high-affinity CALU-specific antibody-producing B cells

  • Antibody affinity measurement:

    • Microfluidic chambers can be used to measure antibody-antigen binding kinetics

    • Real-time monitoring of association/dissociation rates helps select antibodies with desired properties

    • Multiple antibody candidates can be tested simultaneously against CALU protein

  • Epitope mapping acceleration:

    • Microfluidics enables rapid testing of antibody binding to peptide arrays

    • Precise epitope identification helps understand antibody function and cross-reactivity

    • This information facilitates the rational selection of antibody pairs for assay development

  • Production optimization:

    • Microfluidic cell culture systems allow rapid optimization of expression conditions

    • Parameters affecting antibody yield and quality can be systematically evaluated

    • This approach accelerates development of production protocols

Implementing these technologies could significantly reduce development time from weeks to days while improving antibody quality, similar to advances seen in antibody development for viral targets .

What are the considerations for developing CALU monoclonal antibodies capable of distinguishing between post-translationally modified variants?

Post-translational modifications (PTMs) can dramatically affect protein function. For CALU-specific antibodies:

  • Modification-specific immunization strategies:

    • Generate antibodies against synthetic peptides containing specific PTMs

    • Use recombinant expression systems that reproduce relevant modifications

    • Create immunogens with defined modification states

  • Validation approaches for PTM-specific antibodies:

    • Use mass spectrometry to confirm modification status in samples

    • Compare reactivity against modified and unmodified recombinant proteins

    • Employ enzymatic treatments to remove specific modifications and confirm antibody specificity

  • Applications of PTM-specific CALU antibodies:

    • Study how calcium binding affects CALU conformation and function

    • Investigate glycosylation patterns of CALU in different tissues

    • Examine phosphorylation-dependent interactions with binding partners

  • Technical challenges:

    • PTMs may be substoichiometric, requiring sensitive detection methods

    • Some modifications may be labile and lost during sample processing

    • Multiple modification sites may create complex epitope patterns

This specialized approach requires rigorous validation but enables much more detailed functional studies of CALU biology.

How can CALU monoclonal antibodies be integrated into multiplexed diagnostic platforms?

Incorporating CALU antibodies into multiplex diagnostic systems requires:

  • Platform selection considerations:

    • Bead-based systems (similar to cytokine arrays) allow simultaneous detection of multiple proteins

    • Microarray formats enable spatial separation of capture antibodies

    • Microfluidic devices can integrate sample processing and multiplexed detection

  • Antibody compatibility assessment:

    • Evaluate cross-reactivity with other detection reagents in the multiplex panel

    • Optimize antibody concentrations to achieve balanced signal across all analytes

    • Validate performance in the multiplex format compared to single-analyte detection

  • Signal normalization methods:

    • Incorporate internal controls for system performance monitoring

    • Develop appropriate calibration standards for quantitative applications

    • Implement data normalization algorithms to account for technical variation

  • Clinical validation requirements:

    • Establish reference ranges in relevant populations

    • Determine clinical sensitivity and specificity for intended applications

    • Assess robustness across different sample types and clinical scenarios

These approaches mirror successful multiplex systems developed for other biomarkers, including viral antigen detection and autoantibody screening .

What are the methodological considerations for using CALU monoclonal antibodies in live-cell imaging applications?

Adapting CALU antibodies for live-cell imaging presents unique challenges:

  • Antibody format modification:

    • Convert to Fab fragments to improve tissue penetration and reduce Fc-mediated effects

    • Fluorophore conjugation strategies that maintain binding affinity

    • Consider single-chain variable fragments (scFvs) for reduced size

  • Cell delivery methods:

    • Protein transfection reagents for cytoplasmic delivery

    • Microinjection for precise delivery with minimal cellular disruption

    • Cell-penetrating peptide conjugation for enhanced membrane permeability

  • Imaging optimization:

    • Selection of appropriate fluorophores based on cellular autofluorescence profile

    • Photobleaching minimization strategies for extended imaging

    • Confocal or super-resolution techniques for improved spatial resolution

  • Controls and validation:

    • Parallel experiments with fixed cells using standard immunofluorescence

    • Correlation with CALU-GFP fusion protein localization

    • Verification that antibody binding doesn't alter normal CALU dynamics

  • Functional considerations:

    • Ensure antibody binding doesn't interfere with CALU's calcium-binding function

    • Verify that cellular processes dependent on CALU remain intact

    • Monitor potential antibody-induced aggregation or mislocalization

These methodological approaches require careful optimization but can provide valuable insights into dynamic CALU behavior in living systems.

What emerging technologies might enhance future CALU monoclonal antibody development and applications?

Several cutting-edge technologies hold promise for advancing CALU antibody research:

  • AI-driven antibody design:

    • Computational prediction of optimal epitopes for antibody generation

    • Machine learning algorithms to optimize antibody structure for improved affinity and specificity

    • In silico screening to predict cross-reactivity before experimental validation

  • Single-cell antibody discovery platforms:

    • Next-generation microfluidic systems for high-throughput screening of B cells

    • Integrated systems combining cell isolation, antibody sequencing, and functional testing

    • These approaches can dramatically accelerate development timelines compared to traditional methods

  • Advanced protein engineering:

    • Bispecific antibodies targeting CALU and interacting proteins

    • pH-sensitive antibodies for compartment-specific detection

    • Conformation-specific antibodies that distinguish calcium-bound from calcium-free CALU

  • Novel imaging applications:

    • Expansion microscopy compatible antibodies for super-resolution imaging

    • Antibody-based proximity labeling for interactome studies

    • Intrabodies optimized for specific cellular compartments

These emerging approaches will likely transform how researchers develop and utilize CALU monoclonal antibodies, enabling more sophisticated studies of this important calcium-binding protein.

What standardization efforts could improve reproducibility in CALU antibody-based research?

Reproducibility challenges in antibody-based research could be addressed through:

  • Comprehensive antibody validation standards:

    • Adoption of standardized validation protocols across the research community

    • Independent validation by reference laboratories

    • Public database of validation results for commercial CALU antibodies

  • Reference materials development:

    • Purified recombinant CALU protein standards with defined modifications

    • Cell and tissue reference samples with characterized CALU expression

    • Synthetic peptide arrays covering the complete CALU sequence

  • Reporting standards enhancement:

    • Detailed methods sections in publications, including clone identifiers, catalog numbers, and validation protocols

    • Antibody validation data repositories linked to publications

    • Sharing of raw imaging and blot data to enable independent analysis

  • Collaborative quality assessment:

    • Multi-laboratory studies comparing antibody performance across sites

    • Round-robin testing of antibody batches

    • Development of consensus protocols for specific applications

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.