GLB1L3 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Our standard lead time for dispatching GLB1L3 Antibody is 1-3 business days following receipt of your order. The specific delivery timeframe may vary depending on the chosen purchasing method and destination. For detailed delivery information, please contact your local distributor.
Synonyms
GLB1L3 antibody; Beta-galactosidase-1-like protein 3 antibody; EC 3.2.1.- antibody
Target Names
GLB1L3
Uniprot No.

Q&A

What is GLB1L3 and what cellular functions has it been implicated in?

GLB1L3 (beta-galactosidase-1-like protein 3) is a 653 amino acid protein belonging to the glycosyl hydrolase 35 family. The protein displays hydrolase activity that catalyzes the cleavage of lactose, as well as galactosyl residues from gangliosides, glycoproteins, and glycosaminoglycans . GLB1L3 exists in three isoforms produced by alternative splicing (75kDa, 36kDa, and 35kDa) . Recent studies implicate GLB1L3 in various cellular processes including lysosomal function and potential roles in neurodegeneration . Structurally, the protein shares highest homology with GLB1L2 (56% identity) and approximately 38% identity with both GLB1 and GLB1L proteins .

How does GLB1L3 expression vary across different tissue types and developmental stages?

Analysis of GLB1L3 expression patterns shows significant tissue specificity. In ocular tissues, GLB1L3 mRNA is predominantly expressed in retinal layers and the RPE/choroid, while other GLB-related genes exhibit ubiquitous expression patterns across different ocular tissues, including the cornea and lens . Developmental studies demonstrate that GLB1L3 expression is strongly induced during postnatal retinal development, with age-related increased expression persisting throughout adulthood and aging . This distinct expression pattern suggests a specialized role for GLB1L3 in retinal tissues compared to its related family members.

What evolutionary conservation exists for GLB1L3 across species?

Phylogenetic analysis reveals significant evolutionary conservation of GLB1L3 among mammals. Following pairwise alignment of mouse GLB1L3 against homologous sequences from other species, the highest degree of identity at the protein level was observed for the rat homolog (88%), followed by the human homolog (67%) . This high degree of conservation suggests critical functional roles for this protein that have been maintained throughout mammalian evolution. The close evolutionary relationship between GLB1L3 and GLB1L2 proteins further indicates possible functional redundancy or complementary roles between these family members.

Which detection methods are most appropriate for GLB1L3 localization in retinal tissues?

For GLB1L3 localization in retinal tissues, a combined approach using in situ hybridization and immunohistochemistry provides the most comprehensive results. Research demonstrates that GLB1L3 is expressed in the outer nuclear layer (ONL), inner nuclear layer (INL), and ganglion cell layer (GCL) of both healthy and diseased retinas . When conducting immunohistochemistry, optimal dilution ranges from 1:100 to 1:300 for most commercial antibodies . For in situ hybridization, antisense probes targeting the unique regions of GLB1L3 mRNA prove most effective in distinguishing GLB1L3 from other beta-galactosidase family members. This combined approach allows researchers to correlate mRNA expression with protein localization, providing more robust evidence of GLB1L3's spatial distribution within complex retinal architecture.

What are the optimal conditions for Western blot detection of GLB1L3 in different tissue samples?

For Western blot detection of GLB1L3, the following protocol optimizations are recommended based on experimental validations:

ParameterRecommended ConditionsNotes
Antibody Dilution1:500-1:2000Sample-dependent, requires titration
Expected Molecular Weight75 kDa (full length)Alternative isoforms at 36kDa and 35kDa
Observed Molecular Weight35 kDa (most common)Depends on tissue type and isoform
Positive Control TissuesMouse/rat testis tissueReliable positive expression
Protein Loading25-50 μg total proteinHigher loading may be needed for low expression tissues
Blocking Solution5% non-fat milk in TBSTBSA alternatives may reduce background

Tissue-specific considerations include higher GLB1L3 expression in retinal samples compared to other tissues, which may require adjustment of antibody concentration . Importantly, the observed molecular weight of 35 kDa in most applications represents one of the alternatively spliced isoforms rather than the full-length protein, necessitating careful interpretation of Western blot results .

How can researchers distinguish between different GLB1 family members in experimental applications?

Distinguishing between GLB1 family members (GLB1, GLB1L, GLB1L2, and GLB1L3) requires careful consideration of antibody specificity and experimental controls. The following approach is recommended:

  • Select antibodies targeting unique epitopes: Choose antibodies raised against regions with lowest sequence homology between family members. Most commercial GLB1L3 antibodies target the C-terminal region (residues 291-340), which shows minimal overlap with other family members .

  • Include parallel validation controls: Run side-by-side experiments with positive controls for each family member to identify potential cross-reactivity.

  • Employ RNA expression analysis: Complement protein studies with RT-PCR using primers designed to amplify unique regions of each family member. For GLB1L3, real-time PCR demonstrates that expression is predominantly restricted to retinal tissues, while other family members show more ubiquitous expression patterns .

  • Use knockout or knockdown models: When available, utilize genetic models with selective depletion of specific family members as definitive controls for antibody specificity.

The multiple alignment of GLB-related amino acid residues reveals that GLB1L3 protein has the highest degree of homology with GLB1L2 (56% identity) and only 38% with both GLB1 and GLB1L proteins, making these differences useful targets for selective detection .

How is GLB1L3 expression altered in the Rpe65(-/-) mouse model of Leber congenital amaurosis?

In the Rpe65(-/-) mouse model of Leber congenital amaurosis (LCA), GLB1L3 shows distinctive expression changes that differentiate it from other beta-galactosidase family members. Comprehensive studies using oligonucleotide microarray, real-time PCR, and RT-PCR analyses demonstrate that GLB1L3 mRNA expression is strongly downregulated at all ages in Rpe65(-/-) mice compared to wild-type controls (p<0.001) . This downregulation occurs before the onset and persists during progression of retinal degeneration.

In contrast, expression of other family members (GLB1, GLB1L) remains unchanged in the Rpe65(-/-) model, with only GLB1L2 showing some alterations . This selective dysregulation suggests a specific relationship between GLB1L3 and retinal pathophysiology in this disease model. In situ hybridization confirms reduced GLB1L3 mRNA levels across all retinal layers (ONL, INL, and GCL) in Rpe65(-/-) retinas, corroborating the quantitative PCR findings .

What evidence exists for GLB1L3's potential role in cancer and other pathological conditions?

Emerging research indicates potential associations between GLB1L3 and various pathological conditions, particularly in cancer. Studies suggest that GLB1L3 expression is altered in certain malignancies, potentially contributing to lysosomal dysfunction and altered glycosylation patterns . The protein's role in glycan metabolism makes it a candidate for involvement in metabolic adaptations observed in cancer cells.

GLB1L3's location on human chromosome 11q25 places it in a genomic region associated with various diseases . Chromosome 11 is considered gene and disease association dense, with approximately 135 million base pairs and 1,400 genes making up around 4% of human genomic DNA . While direct causal relationships between GLB1L3 and specific cancers remain under investigation, its functional classification within glycosyl hydrolases suggests potential involvement in altered cellular glycobiology, a hallmark of numerous malignancies and metabolic disorders.

What is the proposed mechanism linking GLB1L3 downregulation to retinal degeneration in the Rpe65(-/-) mouse model?

The mechanistic link between GLB1L3 downregulation and retinal degeneration in Rpe65(-/-) mice appears to involve chromophore deficiency. Research indicates that impaired GLB1L3 expression in this model is primarily due to the absence of the chromophore 11-cis retinal, suggesting that Rpe65 deficiency may have cascading metabolic consequences in the underlying neuroretina .

The temporal pattern of GLB1L3 downregulation—occurring before the onset of detectable retinal degeneration—positions it as a potential early biomarker or contributor to disease pathogenesis rather than merely a consequence of generalized cell death . The specificity of GLB1L3 downregulation compared to other family members further supports a selective role in retinal pathophysiology.

Current hypotheses suggest that as a glycosyl hydrolase, reduced GLB1L3 activity could lead to abnormal accumulation of specific galactosyl-containing substrates, potentially disrupting glycan metabolism in photoreceptors and other retinal neurons . This metabolic disruption may contribute to cellular stress and eventual degeneration, though definitive causative evidence requires further experimental validation.

How can researchers optimize immunoprecipitation protocols for GLB1L3 protein complex studies?

Optimizing immunoprecipitation (IP) for GLB1L3 protein complex studies requires several methodological considerations based on the protein's biochemical properties:

  • Lysis buffer selection: Due to GLB1L3's potential association with membrane fractions and lysosomal compartments, use a lysis buffer containing 1% NP-40 or 0.5% Triton X-100 supplemented with protease inhibitors to maintain protein-protein interactions while efficiently extracting the protein .

  • Antibody selection: Choose antibodies targeting the C-terminal region (residues 291-340) of GLB1L3 for IP applications, as this region shows greater accessibility and specificity .

  • Cross-linking considerations: For transient or weak interactions, consider using reversible cross-linking agents (DSP or formaldehyde at 0.1-1%) prior to cell lysis to stabilize protein complexes.

  • Validation controls: Include isotype-matched control antibodies and lysates from tissues known to have low GLB1L3 expression (e.g., cornea) as negative controls .

  • Elution conditions: For mass spectrometry applications, elute with acidic glycine buffer (pH 2.5) rather than SDS-containing buffers to maintain compatibility with downstream applications.

This optimized approach facilitates investigation of GLB1L3's potential interactions with other glycan-processing enzymes or trafficking machinery, providing insights into its functional network within cellular compartments.

What are the critical considerations when designing knockdown or knockout models for GLB1L3 functional studies?

When designing knockdown or knockout models for GLB1L3 functional studies, several critical factors must be considered:

  • Genetic compensation mechanisms: Due to 56% sequence homology with GLB1L2, consider potential functional redundancy that might mask phenotypes in single-gene knockout models . Design experiments to measure compensatory upregulation of other family members.

  • Tissue-specific versus global approach: Given GLB1L3's enrichment in retinal tissues, both tissue-specific (using retina-specific promoters) and global knockout approaches offer complementary insights . Retina-specific knockouts may better isolate direct effects from systemic compensatory mechanisms.

  • Temporal control considerations: Since GLB1L3 expression is developmentally regulated with sustained expression into adulthood, inducible knockout systems allow temporal dissection of acute versus developmental roles .

  • Validation strategy: Confirm knockdown/knockout efficiency at both mRNA (qPCR) and protein levels (Western blot), particularly distinguishing between different isoforms (75kDa, 36kDa, 35kDa) .

  • Phenotypic assessment: Include comprehensive retinal phenotyping including electroretinography, optical coherence tomography, and histological analysis to detect subtle functional or structural changes that might precede overt degeneration.

For siRNA/shRNA approaches, targeting the unique regions of GLB1L3 (regions with lowest homology to other family members) maximizes specificity and minimizes off-target effects on related family members.

What advanced techniques can be employed to assess GLB1L3 enzymatic activity in tissue samples?

Assessing GLB1L3 enzymatic activity requires specialized techniques that distinguish it from other beta-galactosidases. The following advanced methodological approaches are recommended:

  • Fluorogenic substrate assays: Utilize 4-methylumbelliferyl-β-D-galactopyranoside (4-MUG) as substrate with conditions optimized for GLB1L3 (pH 5.5-6.0, different from the pH 4.5 optimum for GLB1) . Measure fluorescence at excitation/emission wavelengths of 365/448 nm.

  • Isoform-specific immunocapture: Pre-clear lysates with antibodies against GLB1 and GLB1L2 before assessing residual beta-galactosidase activity attributable to GLB1L3 .

  • Mass spectrometry-based activity profiling: Employ activity-based protein profiling with galactose-configured cyclophellitol aziridine probes, followed by mass spectrometry to distinguish GLB1L3 activity from other family members.

  • In situ activity assessment: Combine X-gal histochemistry with immunofluorescence using GLB1L3-specific antibodies to correlate enzyme activity with protein localization in tissue sections.

  • Recombinant protein controls: Express and purify recombinant GLB1L3 isoforms (all three variants) as positive controls for activity assays and for generating activity standard curves.

These techniques provide complementary approaches to assess not only the presence but also the functional activity of GLB1L3 in complex biological samples, enabling correlation between expression levels and enzymatic function in normal and pathological states.

How can researchers address nonspecific binding when using GLB1L3 antibodies for immunohistochemistry?

Nonspecific binding is a common challenge when using GLB1L3 antibodies for immunohistochemistry. Based on experimental validations, the following troubleshooting approaches are recommended:

  • Epitope retrieval optimization: For formalin-fixed paraffin-embedded tissues, test both citrate buffer (pH 6.0) and EDTA buffer (pH 9.0) for antigen retrieval, as GLB1L3 epitopes show differential accessibility depending on fixation conditions .

  • Blocking protocol enhancement: Implement a sequential blocking strategy using 10% normal serum from the same species as the secondary antibody, followed by 1% BSA and 0.3% Triton X-100 in PBS for 2 hours at room temperature .

  • Antibody validation with appropriate controls:

    • Positive control: Include mouse/rat testis tissue, known to express GLB1L3

    • Negative control: Omit primary antibody while maintaining all other steps

    • Competitive inhibition: Pre-incubate antibody with immunizing peptide to confirm specificity

  • Dilution optimization: Titrate antibody concentrations from 1:50 to 1:300, as the optimal dilution varies significantly depending on tissue type and fixation method .

  • Secondary antibody selection: Use highly cross-adsorbed secondary antibodies to minimize cross-reactivity with endogenous immunoglobulins in the tissue sample.

These methodological refinements significantly improve signal-to-noise ratio when detecting GLB1L3 in complex tissues like retina, where endogenous peroxidase activity and high protein density can confound results.

What approaches can resolve discrepancies between observed and predicted molecular weights of GLB1L3 in Western blot applications?

Discrepancies between observed and predicted molecular weights of GLB1L3 in Western blot applications arise from several biological and technical factors. The following approaches help resolve these inconsistencies:

  • Isoform consideration: GLB1L3 exists in three alternatively spliced isoforms (75kDa, 36kDa, and 35kDa) . Different antibodies may preferentially detect specific isoforms depending on epitope location. The observed 35kDa band represents the most commonly detected isoform rather than the full-length protein .

  • Post-translational modification analysis: Treat samples with glycosidases (PNGase F) prior to SDS-PAGE to determine if glycosylation contributes to molecular weight shifts. GLB1L3, as a glycosyl hydrolase, may itself be glycosylated.

  • Sample preparation optimization:

    • For membrane-associated fractions: Include specialized extraction buffers containing 0.5% SDS or 0.5% sodium deoxycholate

    • For full denaturation: Heat samples at 95°C for 10 minutes in Laemmli buffer containing fresh reducing agents

  • Gradient gel utilization: Employ 4-15% gradient gels to better resolve the full range of potential GLB1L3 isoforms simultaneously.

  • Validation with recombinant standards: Run purified recombinant GLB1L3 isoforms alongside tissue samples to establish migration patterns of each isoform.

This comprehensive approach accounts for the complexity of GLB1L3 biology and provides more accurate interpretation of Western blot results, particularly when comparing expression across different tissue types or disease models.

How should researchers interpret contradictory findings regarding GLB1L3 tissue distribution across different studies?

Contradictory findings regarding GLB1L3 tissue distribution across studies may stem from methodological differences, species variations, or developmental stage differences. The following interpretative framework helps researchers navigate these discrepancies:

  • Methodological differences assessment: Compare detection methods (qPCR vs. immunodetection) across studies. RNA levels (measured via qPCR) may not directly correlate with protein levels due to post-transcriptional regulation. Studies using in situ hybridization demonstrate GLB1L3 mRNA expression in multiple retinal layers (ONL, INL, GCL) , which may not perfectly align with protein localization studies.

  • Antibody epitope considerations: Different antibodies targeting distinct epitopes may detect different GLB1L3 isoforms. Catalog the specific antibody clones used across studies and their target epitopes (N-terminal vs. C-terminal) .

  • Species-specific variation analysis: Consider species differences in expression patterns. Human GLB1L3 shows 67% identity with mouse GLB1L3 at the protein level , which may translate to different tissue distribution patterns between species.

  • Developmental timing examination: GLB1L3 expression increases during postnatal retinal development and continues rising with age . Studies conducted at different developmental timepoints may yield apparently contradictory results.

  • Resolution approach: When designing experiments to resolve contradictions, include:

    • Multiple detection methods (protein and mRNA)

    • Developmental time series

    • Standardized positive and negative control tissues

    • Cross-validation with at least two independent antibodies targeting different epitopes

This analytical framework helps researchers reconcile apparently contradictory findings and construct a more unified understanding of GLB1L3 biology across different experimental contexts.

What emerging technologies could advance our understanding of GLB1L3 function and interactions?

Several emerging technologies hold promise for deepening our understanding of GLB1L3 function and interactions:

  • CRISPR-based proximity labeling: Combining CRISPR-mediated tagging of endogenous GLB1L3 with proximity labeling technologies (BioID or APEX2) would enable identification of transient interaction partners in living cells, providing insights into GLB1L3's protein interaction network in relevant tissue contexts .

  • Single-cell multi-omics: Integration of single-cell transcriptomics with proteomics would reveal cell type-specific expression patterns of GLB1L3 within heterogeneous tissues like retina, potentially uncovering specialized functions in specific cell populations .

  • Cryo-electron microscopy: Structural characterization of GLB1L3 using cryo-EM would elucidate substrate binding sites and catalytic mechanisms, informing rational design of specific inhibitors or activity probes.

  • Glycan microarrays: High-throughput screening of GLB1L3 against diverse glycan substrates would define its substrate specificity compared to other beta-galactosidases, clarifying its functional niche in glycan metabolism.

  • Organoid models: Development of retinal organoids from iPSCs with modified GLB1L3 expression would provide physiologically relevant 3D models to study its function in human retinal development and disease.

These technological approaches would address current knowledge gaps regarding GLB1L3's precise substrate specificity, interaction partners, and cell type-specific functions, potentially revealing novel therapeutic targets for retinal degenerative diseases.

What experimental approaches could elucidate the potential therapeutic relevance of GLB1L3 in retinal degeneration disorders?

Elucidating the therapeutic relevance of GLB1L3 in retinal degeneration disorders requires multifaceted experimental approaches:

  • Therapeutic modulation studies: Test whether restoring GLB1L3 expression in Rpe65(-/-) mice via AAV-mediated gene delivery can attenuate retinal degeneration, providing proof-of-concept for therapeutic potential .

  • Patient-derived iPSC models: Generate retinal organoids from LCA patient-derived iPSCs to determine if GLB1L3 downregulation is conserved in human disease models and whether this represents a common pathway in multiple forms of retinal degeneration.

  • Metabolomic profiling: Identify accumulating metabolic substrates in GLB1L3-deficient retinas using targeted and untargeted metabolomics, revealing potential toxic intermediates that could be therapeutic targets.

  • Small molecule screening: Develop high-throughput screening assays for compounds that can either enhance residual GLB1L3 activity or bypass the metabolic block caused by GLB1L3 deficiency.

  • Conditional rescue experiments: Implement temporally controlled restoration of GLB1L3 expression at different disease stages to determine therapeutic windows for intervention.

These approaches would establish whether GLB1L3 represents a therapeutic target or biomarker in retinal degeneration and potentially other neurodegenerative conditions where glycan metabolism might be dysregulated.

How might novel antibody engineering approaches improve GLB1L3 detection and functional studies?

Novel antibody engineering approaches offer significant potential for advancing GLB1L3 research through improved detection specificity and expanded functional applications:

  • Single-domain antibodies (nanobodies): Development of camelid-derived single-domain antibodies against GLB1L3 would provide improved tissue penetration for in vivo imaging and potentially distinguish between closely related family members with greater specificity .

  • Intrabodies with organelle targeting: Engineer antibody fragments with subcellular localization signals to detect GLB1L3 in specific organelles (lysosomes, Golgi) in living cells, providing dynamic information about trafficking and processing.

  • Bispecific antibodies: Create bispecific antibodies targeting GLB1L3 alongside markers for specific retinal cell types to improve histological characterization of expression patterns in complex retinal architecture.

  • Antibody-enzyme fusions: Develop antibody-luciferase or antibody-HRP fusions specific to GLB1L3 to enable ultrasensitive detection in samples with low expression levels.

  • Recombinant antibody libraries: Implement Golden Gate-based dual-expression vector systems for rapid screening of recombinant monoclonal antibodies against different GLB1L3 epitopes, accelerating the development of research tools .

Implementation of these advanced antibody engineering approaches would overcome current limitations in GLB1L3 detection sensitivity and specificity, particularly in distinguishing between closely related beta-galactosidase family members in complex biological samples.

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.