BGAL12 Antibody

Shipped with Ice Packs
In Stock

Description

Role in Cellular Senescence

β-Galactosidase antibodies are pivotal in identifying senescent cells via SA-β-gal staining. Studies demonstrate that SA-β-gal activity, detected at pH 6.0, correlates with lysosomal β-gal accumulation rather than a unique isoform . This has been leveraged in cancer and aging research to track cellular senescence in vitro and in vivo.

Antibody Engineering and Production

Recent advances in recombinant antibody production have enhanced consistency and reduced batch variability. For example, the β-Galactosidase (E2U2I) Rabbit mAb (#27198) employs a recombinant format for superior lot-to-lot reproducibility . Glycosylation engineering in CHO cells has further optimized antibody efficacy, with modifications like bisecting GlcNAc increasing antibody-dependent cellular cytotoxicity (ADCC) by 3-fold .

Table 2: Engineering Strategies for β-Galactosidase Antibodies

StrategyOutcomeSource
Recombinant ProductionImproved consistency and animal-free manufacturing
Glycosylation ModulationEnhanced ADCC via reduced core fucosylation
Bispecific FormatsIncreased target specificity and lysosomal trafficking efficiency

Antibody-Drug Conjugates (ADCs)

β-Galactosidase antibodies have been explored in bispecific ADCs (bsADCs) to improve tumor targeting. For instance, co-targeting HER2 and APLP2 receptors using bsADCs enhanced cytotoxicity in cancer models by 1.36-fold compared to monospecific ADCs .

Autoimmune and Fibrotic Diseases

Galectin-3 (Gal-3), a β-galactoside-binding lectin, is neutralized using monoclonal antibodies (e.g., D11 and E07) to treat systemic sclerosis (SSc). These antibodies reduced skin thickening and collagen deposition in murine models, highlighting their therapeutic potential .

Challenges and Future Directions

While β-galactosidase antibodies are well-established, challenges persist in specificity for plant-specific isoforms like BGAL12. Future research should address cross-reactivity and develop isoform-selective tools. Advances in single-cell sequencing and in silico epitope mapping could refine antibody design for precision applications .

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
BGAL12 antibody; At4g26140 antibody; F20B18.250Beta-galactosidase 12 antibody; Lactase 12 antibody; EC 3.2.1.23 antibody
Target Names
BGAL12
Uniprot No.

Target Background

Database Links

KEGG: ath:AT4G26140

STRING: 3702.AT4G26140.1

UniGene: At.2962

Protein Families
Glycosyl hydrolase 35 family
Subcellular Location
Secreted, extracellular space, apoplast.
Tissue Specificity
Ubiquitous, with higher expression levels in roots and siliques.

Q&A

What is BGAL12 antibody and what forms is it available in?

BGAL12 is a polyclonal antibody raised in rabbits against recombinant purified E. coli Beta-galactosidase (~16 kDa). It is available in multiple forms to accommodate different experimental applications: unpurified antiserum (BGAL12-S), purified IgG (BGAL12-A), and conjugated versions including HRP-conjugated (BGAL12-HRP), biotin-conjugated (BGAL12-BTN), and FITC-conjugated (BGAL12-FITC). Each format allows researchers to select the appropriate reagent based on their detection method requirements, whether for Western blotting, ELISA, immunoprecipitation, or immunofluorescence applications .

How does BGAL12 antibody specifically recognize beta-galactosidase?

BGAL12 antibody recognizes epitopes on the E. coli beta-galactosidase protein through antibody-antigen interactions. The antibody was generated by immunizing rabbits with purified recombinant E. coli beta-galactosidase, resulting in the production of polyclonal antibodies that bind to multiple epitopes on the protein. This polyclonal nature enables robust detection across various experimental platforms. The antibody binding involves specific molecular interactions between complementarity-determining regions (CDRs) of the antibody and structural epitopes on the beta-galactosidase protein, similar to the mechanisms observed in other antibody-antigen interactions where germline-encoded residues play crucial roles in recognition .

What are the primary research applications for BGAL12 antibody?

BGAL12 antibody serves multiple critical research functions: (1) Detection of beta-galactosidase fusion proteins in expression systems, enabling verification of successful cloning and expression; (2) Immunoaffinity chromatography purification of beta-galactosidase fusion proteins directly from crude bacterial lysates; (3) Immunoprecipitation of beta-galactosidase and its fusion partners; (4) Immunocytochemical detection of beta-galactosidase in cells and tissues expressing the transfected bacterial lacZ gene, particularly valuable in reporter gene assays and lineage tracing experiments; and (5) Various immunoassays to identify the expression of beta-galactosidase fusion proteins in experimental systems .

What dilution ranges are recommended for different experimental applications?

For optimal results with BGAL12 antibody, application-specific dilution ranges should be employed. For Western blot analyses, the recommended dilution range is 1:1,000 to 1:5,000 of the antibody, which provides sufficient sensitivity while minimizing background. For ELISA applications, researchers should use dilutions between 1:1,000 and 1:10,000, with the exact dilution requiring optimization based on antigen concentration and detection system. For immunocytochemistry and immunohistochemistry, starting dilutions of 1:500 to 1:2,000 are recommended, followed by optimization based on signal-to-noise ratio. When using the conjugated versions (HRP, biotin, or FITC), slightly higher concentrations may be required due to potential reduction in antibody activity following conjugation .

How should I design controls for experiments using BGAL12 antibody?

Robust experimental design with BGAL12 antibody requires appropriate controls. Positive controls should include known beta-galactosidase-expressing samples (e.g., E. coli extracts expressing the lacZ gene or purified beta-galactosidase). Negative controls should include samples lacking beta-galactosidase expression, such as untransfected cells or tissues from non-transgenic animals. For specificity verification, include pre-adsorption controls where the antibody is pre-incubated with purified beta-galactosidase before applying to samples, which should significantly reduce or eliminate specific staining. When detecting fusion proteins, include controls expressing beta-galactosidase alone to differentiate between beta-galactosidase epitopes and epitopes on the fusion partner. For immunohistochemistry applications, include secondary antibody-only controls to assess non-specific binding .

What sample preparation techniques maximize BGAL12 antibody detection sensitivity?

To optimize detection sensitivity with BGAL12 antibody, sample preparation techniques should be tailored to the experimental context. For protein extraction, use fresh samples and maintain cold conditions (4°C) throughout extraction to minimize protein degradation. A standard lysis buffer (50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% NP-40 or Triton X-100, 0.5% sodium deoxycholate) supplemented with protease inhibitors effectively extracts beta-galactosidase while preserving its structure. For fixed tissue sections or cells, optimize fixation conditions (typically 4% paraformaldehyde for 10-20 minutes) to maintain antigen accessibility while preserving cellular morphology. When working with fusion proteins, avoid harsh denaturing conditions that might disrupt the conformational epitopes recognized by the polyclonal antibody. For Western blotting, mild denaturation conditions (heating at 70°C for 10 minutes in standard Laemmli buffer) often provide better results than boiling samples .

How can BGAL12 antibody be used for tracking lacZ-transfected cells in vivo?

Tracking lacZ-transfected cells in vivo using BGAL12 antibody involves a sophisticated multi-step approach. First, cells should be stably transfected with the lacZ gene under an appropriate promoter to ensure consistent expression. Prior to in vivo implementation, validate beta-galactosidase expression in vitro using both enzymatic activity assays and immunostaining with BGAL12 antibody. For in vivo tracking, harvest tissues at appropriate timepoints and prepare cryosections (typically 10-20 μm). Fix sections with 4% paraformaldehyde for 10-15 minutes at room temperature, followed by careful permeabilization with 0.2% Triton X-100. Block with 5-10% normal serum from the species of the secondary antibody in PBS containing 1% BSA for 1 hour. Incubate with BGAL12 antibody at 1:1,000 dilution overnight at 4°C. For visualization, use fluorescently labeled secondary antibodies or HRP-conjugated secondaries with appropriate substrates. Counterstain with DAPI to visualize nuclei. To distinguish transfected cells from potential background, perform parallel X-gal staining on adjacent sections to confirm the presence of enzymatically active beta-galactosidase .

How can I optimize purification of beta-galactosidase fusion proteins using BGAL12 antibody?

Optimizing immunoaffinity purification of beta-galactosidase fusion proteins requires a methodical approach. Begin by immobilizing purified BGAL12-A (IgG fraction) to an appropriate matrix such as CNBr-activated Sepharose or commercial protein A/G columns with crosslinking. Use approximately 5-10 mg of antibody per mL of resin. After immobilization, block remaining active sites with ethanolamine or Tris buffer. For bacterial expression systems, lyse cells under non-denaturing conditions using buffer containing 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1 mM EDTA, and 0.5-1% Triton X-100 with protease inhibitors. Clarify lysate by centrifugation at 15,000 × g for 30 minutes at 4°C. Pre-clear the lysate with unmodified Sepharose to reduce non-specific binding. Apply the pre-cleared lysate to the antibody column and incubate with gentle rotation for 2-4 hours at 4°C. Wash extensively with PBS containing 0.1% Triton X-100, followed by PBS alone. Elute bound proteins using a pH gradient (pH 3.0-2.5 glycine buffer) or competitive elution with excess beta-galactosidase peptide. Immediately neutralize acidic fractions with 1M Tris-HCl pH 9.0. Analyze fractions by SDS-PAGE and Western blotting to identify fusion protein-containing fractions. For higher purity, consider a second purification step such as size exclusion chromatography .

What strategies can optimize co-immunoprecipitation of beta-galactosidase fusion proteins and their interaction partners?

For effective co-immunoprecipitation (co-IP) of beta-galactosidase fusion proteins and their interaction partners, several optimization strategies should be considered. First, cell lysis conditions must preserve protein-protein interactions; use non-denaturing buffers (typically 25-50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1 mM EDTA, 0.5-1% NP-40 or Triton X-100) supplemented with protease and phosphatase inhibitors. Cross-linking with formaldehyde (0.1-1%) prior to lysis can stabilize transient interactions. When using BGAL12 antibody for immunoprecipitation, couple it to Protein A/G beads using standard protocols or commercially available conjugation kits to minimize antibody contamination in the final eluate. Pre-clear lysates with naked beads to reduce non-specific binding. Optimize antibody concentration (typically 2-5 μg per mg of total protein) and incubation conditions (4°C for 2-16 hours with gentle rotation). For washing, use increasingly stringent buffers while carefully monitoring to avoid disrupting specific interactions. Elute complexes using low pH buffer (100 mM glycine pH 2.5), competitive elution with excess antigen, or direct boiling in SDS sample buffer. Analyze precipitated complexes using Western blotting, mass spectrometry, or other appropriate techniques to identify interaction partners .

What are common causes of high background when using BGAL12 antibody in immunohistochemistry?

High background in immunohistochemistry with BGAL12 antibody can stem from multiple sources. Insufficient blocking is a primary cause; increase blocking duration (2 hours at room temperature or overnight at 4°C) and concentration (5-10% normal serum with 1-2% BSA) to reduce non-specific binding. Excessive antibody concentration can increase background; perform titration experiments (1:500 to 1:5,000 dilutions) to determine optimal concentration. Inadequate washing between steps allows residual unbound antibody to generate background; implement at least 3-5 washes of 5-10 minutes each with gentle agitation. Endogenous peroxidase activity can cause false-positive signals with HRP detection systems; pre-treat samples with 0.3-3% hydrogen peroxide in methanol for 10-30 minutes. Tissue autofluorescence can interfere with fluorescent detection; pre-treat sections with 0.1-1% sodium borohydride or commercial autofluorescence quenching solutions. Cross-reactivity with endogenous proteins can be assessed using pre-adsorption controls. Overfixation may cause non-specific antibody trapping; optimize fixation time and consider antigen retrieval methods if necessary. Finally, using detergent-based antibody diluents (PBS with 0.05-0.1% Tween-20 or Triton X-100) can reduce non-specific hydrophobic interactions .

How can I troubleshoot weak or absent signals in Western blots using BGAL12 antibody?

Weak or absent signals in Western blots using BGAL12 antibody can be addressed through systematic troubleshooting. First, verify protein transfer efficiency using reversible total protein stains (Ponceau S or SYPRO Ruby). Check antibody activity with a positive control (purified beta-galactosidase). Increase protein loading (25-50 μg total protein) to enhance detection of low-abundance fusion proteins. Optimize antibody concentration by testing a range of dilutions (1:500 to 1:5,000). Extend primary antibody incubation time (overnight at 4°C) and secondary antibody incubation (2 hours at room temperature). For enhanced sensitivity, switch to more sensitive detection systems such as enhanced chemiluminescence (ECL) plus reagents or fluorescent secondary antibodies with digital imaging. Consider mild denaturation conditions (70°C for 10 minutes) rather than boiling samples to preserve conformational epitopes. For membrane proteins or difficult-to-extract proteins, adjust lysis conditions or consider membrane-specific extraction buffers. If working with fixed tissues, optimize antigen retrieval methods such as heat-induced epitope retrieval in citrate buffer (pH 6.0) or Tris-EDTA buffer (pH 9.0). Finally, fresh antibody aliquots may be necessary if repeated freeze-thaw cycles have compromised antibody activity .

What are optimal storage conditions for maintaining BGAL12 antibody activity long-term?

To maintain BGAL12 antibody activity long-term, implement proper storage practices. Store the antibody at -20°C for extended periods or at 4°C with 0.05% sodium azide for up to one month. Aliquot the antibody into single-use volumes (20-50 μL) upon receipt to minimize freeze-thaw cycles; each freeze-thaw cycle can reduce activity by 10-20%. Use sterile techniques when handling the antibody to prevent microbial contamination. For longer-term storage (>1 year), consider storing aliquots at -80°C. When diluting for use, use high-quality, pure water and sterile buffers. PBS with 0.05-0.1% sodium azide and 1-5% carrier protein (BSA or normal serum) can help stabilize diluted antibody. Avoid repeatedly freezing and thawing diluted working solutions; instead, prepare fresh dilutions for each experiment. If antibody precipitation occurs, centrifuge at 10,000 × g for 5 minutes before use. Do not add glycerol or other cryoprotectants unless specifically recommended by the manufacturer, as these may interfere with binding efficiency. Regularly validate antibody activity using positive controls at 6-month intervals during long-term storage .

How does BGAL12 antibody performance compare with other beta-galactosidase detection methods?

A methodological comparison of BGAL12 antibody with other beta-galactosidase detection approaches reveals distinct advantages and limitations for different experimental contexts. The table below summarizes key performance characteristics:

Detection MethodSensitivitySpecificityTime RequiredCell/Tissue ViabilityQuantification PotentialKey Limitations
BGAL12 Antibody (IHC/ICC)High (can detect 0.1-1 ng)Very High4-24 hoursRequires fixationGood with image analysisCannot detect enzymatic activity
BGAL12 Antibody (Western)High (1-5 ng)Very High4-8 hoursN/AGood with densitometryDenatures protein
X-gal StainingModerateHigh0.5-16 hoursCompatible with fixed or living tissuesPoor (qualitative)Diffusion artifacts
ONPG/CPRG AssaysHighHigh0.5-2 hoursRequires cell lysisExcellent (quantitative)No spatial information
FDG (Fluorescein di-β-D-galactopyranoside)Very HighHigh0.5-1 hourCompatible with living cellsGood with flow cytometrySubstrate loading variability

How can I analyze conflicting results between BGAL12 antibody detection and enzymatic activity assays?

When faced with discrepancies between BGAL12 antibody detection and enzymatic activity assays, a systematic analytical approach is required. First, determine if the fusion protein structure might be impacting enzymatic activity while preserving antibody epitopes. Beta-galactosidase fusion at the N-terminus often preserves activity better than C-terminal fusions. Next, assess whether post-translational modifications or proteolytic processing in your experimental system might affect either enzymatic activity or antibody recognition. Verify antibody specificity using Western blots alongside enzyme activity assays from the same sample to determine if the antibody recognizes enzymatically inactive forms of the protein. Consider performing time-course experiments, as protein stability may differ between detection methods - enzymatic activity may decrease more rapidly than antibody epitope availability. Evaluate assay pH conditions, as beta-galactosidase activity is optimal at pH 7.2-7.5, while some fixation protocols for antibody detection may alter protein conformation. If working with fixed tissues, test different fixation protocols, as some fixatives may preserve antigenicity while inactivating the enzyme. When interpreting results, remember that antibody detection measures protein presence (both active and inactive forms), while enzyme assays detect only catalytically active protein. In cases where antibody detection is positive but enzyme activity is negative, consider protein misfolding or denaturation. Conversely, if enzyme activity is detected without antibody signal, epitope masking or antibody sensitivity limitations may be responsible .

What advanced data analysis approaches can enhance interpretation of BGAL12 antibody results in co-localization studies?

Advanced data analysis of BGAL12 antibody-based co-localization studies requires sophisticated quantitative approaches beyond visual assessment. Begin with proper image acquisition using confocal microscopy with appropriate filter sets to minimize spectral overlap between fluorophores. Acquire z-stack images at Nyquist sampling rates to enable three-dimensional co-localization analysis. For quantification, implement pixel intensity correlation analyses using Pearson's correlation coefficient (PCC), which ranges from -1 (perfect negative correlation) to +1 (perfect positive correlation), with values above 0.5 generally indicating significant co-localization. Manders' overlap coefficient provides complementary information by calculating the fraction of each protein that co-localizes with the other. Object-based approaches identify discrete structures labeled by each antibody and determine spatial relationships between them, particularly valuable for punctate staining patterns. Implement randomization controls by analyzing artificially shifted or rotated images to establish thresholds for significant co-localization. For dynamic studies, consider fluorescence resonance energy transfer (FRET) analyses to determine molecular proximity beyond the diffraction limit of light microscopy. When working with tissue sections, employ tissue-specific autofluorescence subtraction algorithms to enhance signal-to-noise ratios. For multiple sample comparisons, utilize standardized intensity thresholding approaches across all images. Finally, present co-localization data with both representative images and quantitative metrics with appropriate statistical analyses to demonstrate reproducibility and significance of findings .

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.