COG0212 Antibody

Shipped with Ice Packs
In Stock

Description

COG2 Antibody Overview

COG2 (Conserved Oligomeric Golgi Complex Subunit 2) is a critical protein involved in Golgi apparatus structure and function. Antibodies targeting COG2 are essential tools for studying:

  • Golgi ribbon formation

  • Vesicular transport mechanisms

  • Glycosylation disorders

  • Genetic conditions linked to COG complex dysfunction (e.g., congenital disorders of glycosylation) .

Golgi Function and Disease Associations

  • COG2 antibodies detect aberrations in Golgi structure linked to impaired protein trafficking and secretion .

  • Mutations in COG2 correlate with congenital disorders of glycosylation (CDG), impacting enzyme trafficking and glycan synthesis .

  • Knockdown studies using COG2 antibodies reveal:

    • Disrupted Golgi ribbon formation

    • Accumulation of vesicles in the cytoplasm

    • Cell viability reduction due to defective glycosylation .

Western Blot Performance

Sample TypeCAB6251 PA5-30710
Human cell linesSW620, HepG2, MCF7Jurkat, Raji
Mouse tissuesKidneyNot tested
Observed bands88 kDa83 kDa

Immunohistochemistry

  • PA5-67442 localizes COG2 to the Golgi membrane in formalin-fixed paraffin-embedded (FFPE) human liver sections .

Technical Considerations

  • Storage: All COG2 antibodies require storage at -20°C in glycerol-containing buffers to prevent freeze-thaw damage .

  • Controls: Recommended positive controls include SW620 (human), Mouse kidney, and Rat testis .

  • Limitations: None show reactivity in ELISA or flow cytometry applications without optimization .

Related Research Context

While COG2 antibodies are unrelated to COVID-19 therapeutics, the search results highlight parallels in antibody development:

  • Broadly neutralizing antibodies (e.g., S309, BD-604) against SARS-CoV-2 employ similar epitope-mapping strategies .

  • Hybridoma technologies used for COG2 antibody production mirror methods for antiviral mAbs like LY-CoV555 and REGN-COV2 .

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
COG0212 antibody; At1g76730 antibody; F28O16.105-formyltetrahydrofolate cyclo-ligase-like protein COG0212 antibody; Protein CLUSTERS OF ORTHOLOGOUS GROUP 212 antibody
Target Names
COG0212
Uniprot No.

Target Background

Function
This antibody targets a protein potentially involved in thiamine metabolism. In vitro studies have demonstrated that it lacks 5-formyltetrahydrofolate cyclo-ligase activity.
Gene References Into Functions
  1. In vitro analysis of the 5-formyltetrahydrofolate cyclo-ligase protein from Arabidopsis thaliana revealed an absence of 5-formyltetrahydrofolate cyclo-ligase activity. PMID: 21538139
Database Links

KEGG: ath:AT1G76730

STRING: 3702.AT1G76730.1

UniGene: At.34623

Protein Families
5-formyltetrahydrofolate cyclo-ligase family
Subcellular Location
Plastid, chloroplast.

Q&A

What is the recommended validation protocol for confirming COG0212 antibody specificity?

A robust validation protocol should include multiple complementary techniques rather than relying on a single method. Begin with Western blot analysis using positive control samples known to express the target protein and negative controls where expression is absent. Critical control experiments should include:

  • Pre-adsorption tests with immunizing peptide to demonstrate binding specificity

  • Knockout/knockdown validation comparing staining in wild-type versus gene-depleted samples

  • Dual staining with independently raised antibodies targeting different epitopes

For polyclonal antibodies, validation across multiple detection methods is particularly important as different epitopes may be accessible depending on protein conformation and sample preparation techniques. When analyzing dual-fluorescence experiments, be aware that the statistical estimation of polyclonality requires adjustment using a k-parameter that is typically lower than the naïve 50% assumption to ensure accurate assessment of monoclonality .

How do monoclonal and polyclonal COG0212 antibodies differ in research applications?

The choice between monoclonal and polyclonal antibodies significantly impacts experimental outcomes and should be guided by specific research objectives:

AttributeMonoclonal AntibodiesPolyclonal AntibodiesApplication Consideration
Epitope RecognitionSingle epitopeMultiple epitopesPolyclonals provide higher detection sensitivity
Batch-to-batch VariabilityMinimalSignificantMonoclonals offer better reproducibility
Sensitivity to Target ModificationsHighly affectedLess affectedPolyclonals more robust for detecting modified proteins
Production ComplexityCell line immortalizationLess complexMonoclonals require more rigorous screening
Phenotypic Drift ImpactCritical concernLess criticalMonoclonals require stability monitoring

Polyclonal antibodies, derived from multiple B-cell lineages, recognize various epitopes on the target antigen. While this provides higher detection sensitivity and robustness against sample preparation variations, it increases the risk of cross-reactivity with structurally similar proteins . For COG0212 research requiring detection of low-abundance proteins or diverse isoforms, polyclonals might offer advantages despite their greater batch-to-batch variability.

What are the optimal storage conditions for preserving COG0212 antibody activity?

Maintaining antibody functionality requires careful attention to storage conditions. For COG0212 antibodies, like other research antibodies, implement these evidence-based practices:

  • Store stock solutions at -20°C to -80°C in small single-use aliquots to minimize freeze-thaw cycles, which can cause aggregation and denaturation

  • Include cryoprotectants such as glycerol (typically 30-50%) to prevent ice crystal formation

  • Maintain working dilutions at 4°C with preservatives (e.g., 0.02% sodium azide) for short-term storage only (1-2 weeks)

  • Avoid repeated freeze-thaw cycles; each cycle typically reduces activity by 5-10%

  • Document stability data through periodic validation assays comparing fresh and stored antibody performance

Monitoring antibody performance through regular validation assays is essential to establish lot-specific stability profiles, as storage stability may vary between different antibody preparations even when targeting the same protein. Similar antibodies like COG2 are typically stored at concentrations around 0.2 mg/ml, which provides a reference point for COG0212 storage concentration .

What experimental controls are essential when using COG0212 antibody in immunoassays?

Rigorous experimental design requires comprehensive controls to distinguish specific signal from technical artifacts:

  • Positive Controls: Include samples with confirmed target expression; for COG2-related proteins, human cell lines with known expression patterns serve as appropriate positive controls

  • Negative Controls:

    • Isotype controls using non-specific antibodies of the same isotype and concentration

    • Secondary antibody-only controls to assess background from secondary detection

    • Knockout/knockdown samples when available, which provide the most stringent specificity control

  • Absorption Controls: Pre-incubate antibody with immunizing antigen before staining to confirm signal specificity

  • Cross-reactivity Controls: Test antibody against closely related proteins or tissues known not to express the target

  • Technical Replicates: Perform at least three technical replicates to assess method reproducibility

Implementing these controls allows proper interpretation of results and enables troubleshooting if unexpected patterns emerge. For instance, when evaluating binding profiles of COG0212 antibodies to their target epitopes, include samples with known alterations in target protein structure to assess how conformational changes might affect antibody recognition .

How can machine learning approaches enhance the development of high-affinity COG0212 antibody variants?

Machine learning (ML) methodologies offer significant advantages for antibody optimization beyond traditional directed evolution techniques. A systematic ML approach to COG0212 antibody development would involve:

  • Training Data Collection: Generate sequence-function datasets from existing antibody libraries, capturing relationships between sequence variations and binding affinities

  • Model Development: Implement protein language models that can predict the functional impact of mutations by learning from evolutionary patterns across the antibody repertoire

  • In Silico Evolution: Rather than random mutation screening, use ML models to predict high-likelihood beneficial mutations, substantially reducing experimental burden

  • Experimental Validation: Test a small, manageable set (~10) of high-probability variants predicted by the model, focusing experimental resources on promising candidates

  • Iterative Refinement: Feed new experimental data back into the model to improve prediction accuracy over successive generations

This approach allows exploration of mutational space orders of magnitude larger than possible with traditional in vivo evolutionary trajectories, while computational prediction takes seconds compared to weeks required for traditional cell culture and sorting methods .

The Stanford technology for in silico antibody evolution exemplifies this approach, demonstrating that directed evolution guided by ML can identify improved antibody variants with higher binding affinity and thermostability . For COG0212 antibody optimization, integrating sequence data with structural modeling could enable even more precise epitope targeting and reduced cross-reactivity.

What methodologies best capture the conformational epitope binding properties of COG0212 antibodies?

Conformational epitope characterization requires sophisticated approaches that preserve native protein structure:

  • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS): This technique measures the rate of hydrogen-deuterium exchange in different protein regions when bound to the antibody, identifying protected regions that correspond to binding interfaces

  • X-ray Crystallography and Cryo-EM: These structural determination methods provide atomic-level resolution of antibody-antigen complexes, though they require significant sample preparation and optimization

  • Surface Plasmon Resonance with Epitope Mapping: Combining SPR kinetic analysis with systematic mutagenesis of target protein allows identification of critical binding residues

  • Circular Dichroism Spectroscopy: Measures changes in protein secondary structure upon antibody binding, useful for determining if binding induces conformational changes

  • Phage Display with Conformational Constraints: Using constrained peptide libraries that maintain secondary structure elements can identify conformational epitopes

For COG0212 antibodies, understanding conformational epitope binding is particularly important if the target protein undergoes structural rearrangements in different cellular contexts, similar to how proteins in the conserved oligomeric Golgi complex change conformation during trafficking . When designing such studies, researchers should implement controls that account for potential conformational heterogeneity in the target protein population.

How does phenotypic drift affect long-term stability in COG0212 antibody production cell lines?

Phenotypic drift presents a significant challenge for maintaining consistent antibody production over time and must be systematically monitored and mitigated:

  • Mechanisms of Drift: In monoclonal antibody production, even starting from a single progenitor cell, genomic mutations accumulate over cell generations, leading to heterogeneity in antibody expression, glycosylation patterns, and binding properties

  • Monitoring Strategies:

    • Regular sequence verification of the antibody genes in production cells

    • Periodic assessment of critical quality attributes (CQAs) such as binding affinity, specificity, and post-translational modifications

    • Flow cytometric analysis to detect emerging subpopulations with altered expression profiles

    • Functional binding assays to track changes in target recognition

  • Mitigation Approaches:

    • Implementation of good cell banking practices with master and working cell banks

    • Limited passage use for production cultures

    • Selection pressure maintenance to preserve antibody expression

    • Environmental parameter control to minimize stress-induced mutations

  • Statistical Modeling: Establish drift prediction models based on historical data to anticipate when cell line replacement might be necessary

The impact of phenotypic drift is particularly pronounced in polyclonal cell populations, where the divergence between different progenitor cells amplifies over time, causing the "fanning out" of product variability illustrated in monoclonal antibody production literature . For COG0212 antibody production, implementing robust cell line stability studies during development can help establish the maximum culture duration before critical changes in antibody properties occur.

What strategies can resolve cross-reactivity issues with COG0212 antibodies in multi-protein complexes?

Cross-reactivity challenges are particularly relevant for antibodies targeting components of multi-protein complexes like the conserved oligomeric Golgi complex, requiring systematic troubleshooting:

  • Epitope Refinement: Develop antibodies against unique, solvent-exposed regions of the target protein that show minimal sequence homology with related proteins

  • Absorption Protocols: Pre-absorb antibody preparations with recombinant versions of potential cross-reactive proteins to deplete non-specific binding populations

  • Competitive Binding Assays: Use unlabeled competing antigens at increasing concentrations to determine binding specificity profiles

  • Orthogonal Validation:

    • Combine antibody detection with mass spectrometry to confirm the identity of immunoprecipitated proteins

    • Use CRISPR-edited cell lines with epitope tags on the endogenous protein to benchmark antibody performance

  • Context-dependent Optimization: Adjust antibody concentration, incubation conditions, and washing stringency based on the specific experimental system

For protein complexes similar to the COG complex, where COG2 is known to interact with multiple other proteins in maintaining Golgi structure and function , cross-reactivity assessment should include evaluation against all complex components. When designing experiments involving COG0212 antibody application to complex biological samples, sequential immunodepletion strategies can help isolate specific signals from closely related proteins.

What protocol modifications are necessary when using COG0212 antibodies across different immunoassay platforms?

Transferring antibody applications between platforms requires systematic optimization to maintain specificity and sensitivity:

  • Western Blot to Immunohistochemistry (IHC) Transition:

    • Increase antibody concentration typically 2-5 fold due to lower antigen accessibility in fixed tissues

    • Optimize antigen retrieval methods (heat-induced vs. enzymatic) based on epitope characteristics

    • Incorporate longer incubation times (overnight at 4°C) to improve tissue penetration

  • IHC to Flow Cytometry Adaptation:

    • Reduce antibody concentration to minimize background in solution-phase binding

    • Optimize fixation/permeabilization protocols if targeting intracellular antigens

    • Include viability dyes to exclude non-specific binding to dead cells

  • ELISA to Multiplex Assay Conversion:

    • Validate for cross-platform interference using spike-recovery experiments

    • Adjust detection antibody concentration to account for different surface chemistries

    • Implement blocking optimizations to maintain signal-to-noise ratios

  • Cross-species Applications:

    • Perform epitope conservation analysis prior to application in non-validated species

    • Start with 2-3 fold higher concentration when testing in new species

    • Validate with appropriate positive and negative controls specific to the target species

For antibodies targeting proteins in the conserved oligomeric Golgi complex, like COG2, adjustments based on subcellular compartment accessibility are particularly important . When optimizing protocols for COG0212 antibodies, systematic titration experiments across each platform provide essential baseline data for establishing platform-specific working parameters.

How should researchers approach dual labeling experiments to verify COG0212 antibody specificity?

Dual labeling provides powerful validation of antibody specificity when properly designed and controlled:

  • Strategic Co-labeling Pairs:

    • Use antibodies targeting different epitopes on the same protein (confirming identity)

    • Select known interaction partners with established co-localization patterns

    • Include markers of the subcellular compartment where the target should localize

  • Technical Considerations:

    • Control for fluorophore spectral overlap with proper compensation controls

    • Order antibody application based on host species to prevent cross-reactivity

    • Optimize individual antibody concentrations before combining to maximize signal-to-noise ratios

  • Quantitative Colocalization Analysis:

    • Calculate Pearson's correlation coefficient between channels

    • Implement Manders' overlap coefficient for partial colocalization assessment

    • Use intensity correlation analysis to distinguish random overlap from true colocalization

  • Controls for Dual Fluorescence Experiments:

    • Single-labeled controls to establish bleed-through parameters

    • Competition controls where unlabeled antibody competes with labeled version

    • Cells expressing fluorescent protein-tagged versions of the target to benchmark antibody performance

The dual fluorescence approach can be particularly effective for detecting polyclonality in cell populations, where cells engineered to fluoresce either red or blue allow identification of wells containing multiple progenitor cells . For COG0212 antibody validation, comparing staining patterns with established markers of the target's known subcellular localization provides critical context for interpreting specificity.

What are the recommended controls for validating COG0212 antibody specificity in Western blot applications?

Robust Western blot validation requires a comprehensive control strategy:

  • Lysate Controls:

    • Positive control: Cell lines/tissues with confirmed high target expression

    • Negative control: Cell lines/tissues with confirmed absence of target

    • Gradient controls: Serial dilutions of positive control to assess detection linearity

  • Antibody Controls:

    • Pre-immune serum control (for polyclonal antibodies)

    • Isotype control at equivalent concentration (for monoclonals)

    • Peptide competition assay with immunizing antigen

  • Technical Controls:

    • Loading control with antibody against housekeeping protein

    • Molecular weight marker to confirm target band corresponds to predicted size

    • Secondary antibody-only control to assess non-specific binding

  • Genetic Validation:

    • Overexpression lysates to confirm band intensity increases

    • Knockdown/knockout lysates to confirm band disappears

    • Tagged-protein expression to provide band shift confirmation

For Western blot applications targeting proteins in the conserved oligomeric Golgi complex, like COG2, additional controls should account for potential protein-protein interactions that might affect migration patterns . When validating COG0212 antibodies specifically, researchers should analyze samples from multiple cell types to ensure detection across varied expression levels and post-translational modification states.

How can directed evolution techniques be applied to optimize COG0212 antibody binding affinity?

Directed evolution offers powerful approaches for antibody optimization that can be implemented through systematic workflows:

  • Library Generation Strategies:

    • Error-prone PCR to introduce random mutations in CDR regions

    • CDR shuffling to combine beneficial mutations from different clones

    • Targeted mutagenesis of specific residues identified through structural analysis

  • Selection Methodologies:

    • Phage display with increasingly stringent washing steps to select high-affinity variants

    • Yeast display combined with fluorescence-activated cell sorting for quantitative affinity screening

    • Ribosome display for larger library diversity when working with complex targets

  • High-throughput Screening Integration:

    • Microfluidic platforms for single-cell antibody secretion analysis

    • Next-generation sequencing to track enrichment of specific mutations across selection rounds

    • Machine learning predictions to focus on high-probability beneficial mutations

  • Computational Enhancements:

    • Protein language models trained on antibody sequences to predict improved variants

    • In silico evolution to explore a mutational space orders of magnitude larger than possible with traditional methods

    • Structure-based simulations to predict binding energy changes from mutations

The Stanford technology for in silico antibody evolution demonstrates how machine learning can dramatically accelerate this process by predicting a small, manageable set of high-likelihood protein variants using the predictive capabilities of protein language models, reducing the experimental burden of testing large libraries . For COG0212 antibody optimization, combining computational prediction with focused experimental validation enables efficient evolution of improved binding properties while maintaining specificity.

How should researchers interpret inconsistent staining patterns when using COG0212 antibodies in immunohistochemistry?

Inconsistent immunohistochemistry staining requires systematic investigation to distinguish technical artifacts from biological variability:

  • Sample Preparation Variables:

    • Evaluate fixation time effects: Overfixation can mask epitopes while underfixation preserves antigenicity but compromises morphology

    • Compare different antigen retrieval methods systematically (heat-induced vs. enzymatic)

    • Assess section thickness influence on antibody penetration and signal intensity

  • Antibody Validation Approach:

    • Test multiple antibody concentrations in a standardized dilution series

    • Compare different antibody clones targeting different epitopes of the same protein

    • Implement peptide competition controls to confirm specific binding

  • Biological Interpretation Considerations:

    • Evaluate if heterogeneous staining correlates with known biological variables (cell type, differentiation state)

    • Compare with mRNA expression data from parallel samples when available

    • Consider post-translational modifications that might affect epitope availability

  • Quantification Strategies:

    • Implement digital image analysis with consistent thresholding parameters

    • Use H-score or Allred scoring systems for standardized intensity reporting

    • Generate tissue microarrays for high-throughput comparative analysis

For proteins involved in dynamic cellular processes, like those in the Golgi apparatus, staining patterns may legitimately vary depending on the cell's physiological state . When troubleshooting COG0212 antibody staining, correlation with functional assays can help determine whether pattern variability reflects technical issues or biologically meaningful differences in protein localization or expression.

What statistical approaches are most appropriate for analyzing variability in COG0212 antibody binding assays?

Robust statistical analysis of antibody binding data requires methods that account for the unique characteristics of binding assays:

  • Assay Validation Statistics:

    • Calculate Z-factor to assess assay quality and separation between positive and negative controls

    • Determine minimum detectable concentration (MDC) through precision profile analysis

    • Compute coefficients of variation (%CV) for intra- and inter-assay variability

  • Dose-Response Analysis:

    • Fit binding data to four-parameter logistic regression models rather than linear models

    • Use AIC/BIC criteria to select appropriate binding models when comparing different antibody clones

    • Implement bootstrapping to generate confidence intervals for EC50 values

  • Comparative Analysis Approaches:

    • ANOVA with post-hoc tests for multi-group comparisons of binding parameters

    • Mixed-effects models when analyzing repeated measures or hierarchical data structures

    • Equivalence testing rather than difference testing when comparing antibody performance to reference standards

  • Advanced Statistical Considerations:

    • Account for the k-parameter in dual fluorescence experiments to correctly estimate polyclonality rates

    • Implement Bayesian approaches for small sample sizes with informative priors based on previous experiments

    • Use power analysis to determine appropriate sample sizes for detecting meaningful differences in binding parameters

When analyzing binding data from COG0212 antibodies across multiple experimental conditions, visualizing data using techniques like principal component analysis can help identify patterns in variability that might be missed by univariate statistical approaches. For quantitative comparisons between different antibody preparations, standardization to a reference standard is essential for meaningful statistical comparisons.

How can researchers distinguish between true COG0212 signal loss and technical artifacts in degraded tissue samples?

Differentiating biological signal loss from technical artifacts requires implementing multiple complementary approaches:

For proteins involved in complex cellular structures like the Golgi apparatus, comparison with other structural components can provide context for distinguishing between specific protein loss and general structural degradation . When evaluating COG0212 signal in potentially degraded samples, gradual decay patterns affecting multiple proteins similarly suggest technical artifacts, while selective loss of specific signals may indicate true biological differences.

What strategies can resolve cross-reactivity issues with COG0212 antibodies in multi-protein complexes?

Addressing cross-reactivity in complex protein environments requires methodical investigation and optimization:

  • Epitope Mapping and Refinement:

    • Perform detailed epitope mapping to identify unique regions with minimal homology to related proteins

    • Design blocking peptides specifically for regions suspected of causing cross-reactivity

    • Consider developing antibodies against post-translationally modified epitopes unique to the target

  • Immunoaffinity Purification:

    • Use stringent washing conditions in immunoprecipitation to disrupt weak cross-reactive binding

    • Implement sequential immunodepletion to remove specific cross-reactive proteins

    • Optimize detergent composition to maintain target protein conformation while minimizing non-specific interactions

  • Validation in Engineered Systems:

    • Test antibody specificity in cell lines overexpressing potential cross-reactive proteins

    • Generate epitope-tagged versions of suspected cross-reactive proteins to track specific interactions

    • Create domain deletion/mutation constructs to map precise regions of cross-reactivity

  • Analytical Separation Strategies:

    • Employ two-dimensional electrophoresis to resolve proteins of similar molecular weight

    • Use native gel electrophoresis to separate intact protein complexes before antibody probing

    • Implement chromatographic fractionation prior to immunodetection to simplify the protein mixture

For proteins functioning in multi-component complexes like the conserved oligomeric Golgi complex, where proteins share structural and functional similarities, cross-reactivity assessment should include evaluation against all complex components . When optimizing detection of COG0212 in such environments, competitive binding experiments with recombinant proteins can help quantify the relative affinities for target versus potentially cross-reactive proteins.

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.