POGLUT1 Antibody, Biotin conjugated

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
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the method of purchase and your location. Please contact your local distributor for specific delivery time information.
Synonyms
C3orf9 antibody; CAP10 like 46 kDa protein antibody; CAP10 like protein, 46 kDa antibody; CAP10-like 46 kDa protein antibody; CLP46 antibody; hCLP46 antibody; hRumi antibody; KDELC family like 1 antibody; KDELCL1 antibody; KTEL (Lys Tyr Glu Leu) containing 1 antibody; KTEL motif containing protein 1 antibody; KTEL motif-containing protein 1 antibody; KTEL1 antibody; KTELC1 antibody; MDS010 antibody; MDSRP antibody; Myelodysplastic syndromes relative protein antibody; O-glucosyltransferase rumi homolog antibody; PGLT1_HUMAN antibody; Poglut1 antibody; protein O glucosyltransferase 1 antibody; Protein O-glucosyltransferase 1 antibody; Protein O-xylosyltransferase antibody; Rumi antibody; x 010 protein antibody
Target Names
POGLUT1
Uniprot No.

Target Background

Function
POGLUT1, a dual specificity glycosyltransferase, catalyzes the transfer of glucose and xylose from UDP-glucose and UDP-xylose, respectively, to a serine residue within the consensus sequence C-X-S-X-P-C. Notably, POGLUT1 specifically targets extracellular EGF repeats of proteins such as CRB2, F7, F9, and NOTCH2. It functions as a positive regulator of Notch signaling by mediating O-glucosylation of Notch, thereby modulating muscle development. Importantly, Notch glucosylation does not interfere with Notch ligand binding. POGLUT1 is essential during early development to promote gastrulation, as it mediates O-glucosylation of CRB2, a process critical for CRB2 localization to the cell membrane.
Gene References Into Functions
  1. This study presents the generation and characterization of an iPSC line (CSCRMi001-A) derived from a LGMD-2Z patient harboring a missense mutation in POGLUT1. This iPSC line serves as a valuable tool for in vitro disease modeling. PMID: 29034878
  2. The data indicate that overexpression of hCLP46 (human CAP10-like protein 46 kDa) in colorectal cancer is associated with advanced tumor-node-metastasis stage, lymph node metastasis, and reduced survival time. PMID: 28481732
  3. These findings broaden the spectrum of mutations in POGLUT1 and solidify its status as a third candidate gene, alongside KRT5 and POFUT1, to consider in the diagnosis of GGD/DDD. PMID: 27479915
  4. These data suggest that a key pathogenic mechanism for this novel form of muscular dystrophy linked to POGLUT1 mutations is Notch-dependent loss of satellite cells. PMID: 27807076
  5. hCLP46 enhances Smad3 protein stability by inhibiting its ubiquitin-proteasomal degradation. PMID: 26058784
  6. miR-134 effectively inhibits proliferation and migration of human endometrial cancer stem cells by targeting protein O-glucosyltransferase 1 (POGLUT1) expression. PMID: 25528443
  7. Mutations within POFUT1, encoding protein O-fucosyltransferase 1, have been identified as the underlying cause of Dowling-Degos disease. PMID: 24387993
  8. Overexpression of hCLP46 effectively inhibits proliferation of 293TRexs cells, accompanied by an increase in cyclin-dependent kinase inhibitors p21 and p27, while reduced hCLP46 expression modestly enhances cell proliferation. PMID: 23692084
  9. Absence of hCLP46 leads to impaired ligand-induced Notch activation in mammalian cells. Moreover, hCLP46 regulates the proliferation of U937 cells through the CDKI-RB signaling pathway, potentially playing a role in the pathogenesis of leukemia. PMID: 21458412
  10. Elevated CLP46 expression is observed in AML, T-ALL, and leukemic cell lines. Considering CLP46's ability to modify the Notch pathway, this finding underscores the potential significance of Notch signaling in the pathogenesis of AML and T-ALL. PMID: 20143914
  11. Altered C3ORF9 expression in myelodysplastic syndrome could be attributed to distinct gene regulation in these patients and/or an increase in CD34+ cells. PMID: 19822096

Show More

Hide All

Database Links

HGNC: 22954

OMIM: 615618

KEGG: hsa:56983

STRING: 9606.ENSP00000295588

UniGene: Hs.231750

Involvement In Disease
Dowling-Degos disease 4 (DDD4); Limb-girdle muscular dystrophy 2Z (LGMD2Z)
Protein Families
Glycosyltransferase 90 family
Subcellular Location
Endoplasmic reticulum lumen.
Tissue Specificity
Expressed in most adult tissues at different intensities. Abundantly expressed in liver. Expressed also in brain, heart, skeletal muscle, spleen, kidney, placenta, lung and peripheral blood leukocyte. Not detectable in colon, thymus and small intestine. E

Q&A

What is POGLUT1 and why is it important for developmental and cellular processes?

POGLUT1 (Protein O-Glucosyltransferase 1) is an enzyme critical for adding O-glucose to EGF repeats in the extracellular domain of various proteins, most notably in the Notch signaling pathway . POGLUT1 contains a carboxy-terminal KTEL motif that functions as an endoplasmic reticulum retention signal . Its importance extends beyond Notch signaling, as demonstrated by mouse models where POGLUT1 deficiency causes more severe developmental defects than Notch pathway disruption alone .

The significance of POGLUT1 is evident in multiple biological processes. It plays a crucial role in embryonic development, particularly during gastrulation, by modifying the extracellular domain of CRUMBS2, an apical transmembrane protein essential for epithelial polarity . POGLUT1 also influences protein trafficking from the endoplasmic reticulum to the cell surface, as demonstrated in studies of DLK1 transport . Mutations in POGLUT1 have been linked to limb-girdle muscular dystrophy (LGMD R21), characterized by distinctive patterns of muscle degeneration .

What are the key applications for biotin-conjugated POGLUT1 antibodies in research?

Biotin-conjugated POGLUT1 antibodies offer several advantages for research applications due to their high sensitivity and versatility in detection systems. The primary applications include:

Western Blotting: Biotin-conjugated POGLUT1 antibodies are effective in Western blot analyses for detecting POGLUT1 protein expression in cell or tissue lysates. The biotin conjugation enables signal amplification through streptavidin-based detection systems, providing enhanced sensitivity for detecting low-abundance POGLUT1 variants .

ELISA Applications: These antibodies demonstrate particular utility in enzyme-linked immunosorbent assays, where the biotin-streptavidin interaction significantly increases detection sensitivity. This makes them valuable for quantifying POGLUT1 levels in complex biological samples .

Immunohistochemistry: For both frozen and paraffin-embedded tissue sections, biotin-conjugated POGLUT1 antibodies enable detailed localization studies of POGLUT1 expression patterns across different tissue types and developmental stages .

Multi-color Immunofluorescence Studies: The biotin conjugation allows researchers to design complex immunostaining protocols where multiple proteins can be visualized simultaneously, particularly useful when studying POGLUT1's interactions with substrate proteins like NOTCH1 or CRUMBS2 .

How should researchers properly store and handle biotin-conjugated POGLUT1 antibodies?

Proper storage and handling of biotin-conjugated POGLUT1 antibodies is essential for maintaining their functionality and extending their useful lifespan. The following methodological approaches are recommended:

Storage Temperature: Store biotin-conjugated POGLUT1 antibodies at -20°C for long-term storage. For antibodies targeting specific amino acid regions (e.g., AA 132-392 or AA 201-300), avoid repeated freeze-thaw cycles which can cause degradation of the antibody and loss of biotin conjugation integrity .

Aliquoting Protocol: Upon receipt, divide the antibody into small single-use aliquots before freezing. This minimizes freeze-thaw cycles and reduces contamination risk. Use sterile, low-protein binding microcentrifuge tubes for aliquoting.

Working Solution Preparation: When preparing working dilutions, use high-quality, sterile PBS supplemented with 0.1% BSA as a diluent. Working dilutions can be stored at 4°C for up to one week, but extended storage at this temperature may reduce antibody performance, particularly in sensitive applications like immunofluorescence.

Contamination Prevention: Avoid introducing microbial contamination by using sterile technique when handling the antibody. Consider adding sodium azide (0.02%) to working solutions intended for short-term storage, but note that azide can interfere with HRP-based detection systems.

Quality Control Monitoring: Periodically verify antibody performance using positive control samples with known POGLUT1 expression levels. This is particularly important after extended storage periods or when using the antibody in new experimental systems.

What controls should be included when using biotin-conjugated POGLUT1 antibodies?

Implementing appropriate controls is crucial for ensuring the validity and interpretability of results obtained with biotin-conjugated POGLUT1 antibodies. The following controls should be systematically included:

Positive Controls: Include samples with verified POGLUT1 expression. For human POGLUT1 studies, HEK293T cells transfected with POGLUT1 expression vectors provide reliable positive controls . For mouse studies, embryonic tissue samples from wild-type mice at appropriate developmental stages can serve as positive controls .

Negative Controls: Two types of negative controls are essential:

  • Primary Antibody Omission: Process samples without adding the biotin-conjugated POGLUT1 antibody to assess non-specific binding of detection reagents.

  • Biological Negative Controls: When available, include POGLUT1-knockout or knockdown samples to confirm antibody specificity .

Isotype Controls: Include control samples treated with biotin-conjugated non-specific antibodies of the same isotype (e.g., rabbit IgG for polyclonal POGLUT1 antibodies) to identify potential non-specific binding due to Fc receptor interactions or other non-target-specific mechanisms .

Peptide Competition: Pre-incubate the biotin-conjugated POGLUT1 antibody with excess purified POGLUT1 peptide (corresponding to the immunogen sequence) before application to samples. This verifies binding specificity by blocking specific antibody-antigen interactions .

Endogenous Biotin Blocking: For tissues with high endogenous biotin levels (e.g., liver, kidney), implement biotin blocking steps using avidin/biotin blocking kits prior to applying the biotin-conjugated POGLUT1 antibody, particularly for immunohistochemistry applications.

How can researchers optimize immunofluorescence protocols for biotin-conjugated POGLUT1 antibodies in co-localization studies?

Optimizing immunofluorescence protocols for co-localization studies involving biotin-conjugated POGLUT1 antibodies requires careful consideration of multiple experimental parameters. The following methodological approach has been validated in studies investigating POGLUT1's role in protein trafficking and subcellular localization:

Fixation Optimization: Comparative testing of fixation methods is essential, as POGLUT1's ER localization can be affected by fixation conditions. For co-localization with ER markers:

  • For cells: 4% paraformaldehyde for 10-15 minutes at room temperature preserves POGLUT1 epitopes while maintaining cellular architecture.

  • For tissue sections: 4% paraformaldehyde overnight at 4°C provides consistent results .

Antigen Retrieval Protocol: For paraffin-embedded tissues or challenging samples, implement heat-induced epitope retrieval using Vector unmasking solution at 98°C for 10 minutes, followed by acetone treatment at -20°C for 8 minutes to enhance antibody accessibility to POGLUT1 epitopes .

Sequential Detection Strategy: To avoid cross-reactivity in multiple labeling experiments, employ a sequential detection approach:

  • Apply primary antibodies individually with thorough washing between applications

  • Use streptavidin conjugated to spectrally distinct fluorophores for detecting biotin-conjugated POGLUT1 antibodies

  • Apply other detection reagents for co-localization partners

Blocking Protocol Refinement: Implement extended blocking (overnight at 4°C) with a high-concentration blocking buffer (10% goat serum, 5% BSA, 0.3% Triton-X100 in PBS) to minimize background, especially critical when studying POGLUT1 in relation to ER markers like BFP-KDEL or Golgi markers like GM130 .

Signal Amplification System: For detection of low-abundance POGLUT1 in certain cellular compartments, employ a tyramide signal amplification system compatible with biotin-streptavidin interactions, which can provide up to 100-fold signal enhancement without compromising resolution for co-localization analyses.

Imaging Parameters: Use point-scanning confocal microscopy with appropriate channel separation settings to minimize bleed-through. Z-stack acquisition with optimal step size (typically 0.3-0.5 μm) is essential for accurate three-dimensional co-localization analysis .

What are the key considerations when using biotin-conjugated POGLUT1 antibodies in studies of muscular dystrophy models?

When investigating muscular dystrophy models, particularly those related to LGMD R21 caused by POGLUT1 mutations, several critical methodological considerations must be addressed:

Muscle-Specific Sample Processing: Skeletal muscle tissue requires specialized handling for optimal results with biotin-conjugated POGLUT1 antibodies:

  • Flash-freeze biopsies in isopentane cooled in liquid nitrogen to preserve tissue architecture

  • Section at 8-10 μm thickness for immunohistochemistry

  • Implement extended antigen retrieval protocols to counteract the dense connective tissue matrix in dystrophic samples

Quantitative Analysis Parameters: For comparative studies between normal and dystrophic tissues:

  • Establish standardized imaging parameters across all samples

  • Implement automated quantification methods to assess POGLUT1 staining intensity and distribution patterns

  • Correlate POGLUT1 levels with clinical severity metrics and muscle imaging findings showing the distinctive "inside-to-outside" fatty degeneration pattern characteristic of POGLUT1-related muscular dystrophy

Downstream Signaling Assessment: Since POGLUT1 mutations affect Notch signaling, parallel analysis of Notch pathway components provides mechanistic insights:

  • Quantify NOTCH1 intracellular domain levels using specific antibodies against the cleaved active form (Val 1744)

  • Assess satellite cell populations, which are consistently decreased in POGLUT1-related muscular dystrophy

  • Evaluate α-dystroglycan glycosylation status, as hypoglycosylation is a hallmark of POGLUT1-related muscular dystrophy

Mutation-Specific Considerations: Different POGLUT1 mutations (R183W, Y57C, I129T, R98W, C102F, W308L) affect enzyme activity and protein stability to varying degrees. When studying specific mutations:

  • Implement Western blotting protocols optimized for detecting both wild-type and mutant POGLUT1 proteins

  • Compare subcellular localization patterns of mutant versus wild-type POGLUT1

  • Correlate biochemical findings with clinical phenotypes to establish genotype-phenotype correlations

How can researchers effectively use biotin-conjugated POGLUT1 antibodies in protein trafficking studies?

Biotin-conjugated POGLUT1 antibodies can be strategically employed in protein trafficking studies, particularly when investigating the role of POGLUT1 in facilitating protein movement through the secretory pathway. The following methodological approach enables robust trafficking analysis:

Pulse-Chase Experimental Design: To study dynamic trafficking processes:

  • Establish stably transfected cell lines expressing tagged reporter proteins (e.g., Reporter DLK1 del) in wild-type and POGLUT1-deficient backgrounds

  • Implement biotin labeling at specific time points using membrane-impermeable biotinylation reagents

  • Use biotin-conjugated POGLUT1 antibodies in combination with organelle-specific markers to track protein movement

Subcellular Fractionation Protocol: For biochemical assessment of protein distribution:

  • Perform differential centrifugation to isolate ER, Golgi, and plasma membrane fractions

  • Analyze POGLUT1 substrates across fractions using biotin-conjugated POGLUT1 antibodies

  • Quantify the relative distribution of target proteins in wild-type versus POGLUT1-deficient conditions

Live Cell Imaging Approach: For real-time trafficking analysis:

  • Express fluorescently tagged POGLUT1 substrates (e.g., CRUMBS2, DLK1) in appropriate cell models

  • Apply cell-permeable biotin conjugates for pulse-labeling newly synthesized proteins

  • Use streptavidin-based detection of biotin-conjugated POGLUT1 antibodies in fixed time-point samples to correlate with live imaging data

Co-localization Analysis Parameters:

  • Implement automated co-localization algorithms (e.g., Pearson's correlation coefficient, Manders' overlap coefficient)

  • Quantify percentage of POGLUT1 substrates co-localizing with ER markers (BFP-KDEL) versus Golgi markers (GM130) in wild-type and POGLUT1-deficient conditions

  • Track temporal changes in co-localization patterns following biotin pulse-labeling

Transport Rate Quantification: To measure the kinetics of protein trafficking:

  • Establish a standardized protocol using biotin supplementation to trigger protein transport

  • Collect samples at defined time points (e.g., 0, 15, 30, 50 minutes) post-stimulation

  • Quantify the progressive shift from ER to Golgi to plasma membrane localization

  • Compare transport rates between wild-type and POGLUT1-deficient cells

What methodological approaches can resolve contradictory findings when using biotin-conjugated POGLUT1 antibodies across different experimental systems?

Resolving contradictory findings when using biotin-conjugated POGLUT1 antibodies across experimental systems requires systematic troubleshooting and methodological refinement. The following approaches address common sources of discrepancy:

Epitope Accessibility Assessment: Different experimental systems may affect epitope accessibility:

  • Conduct comparative analysis of multiple fixation and permeabilization protocols

  • Test native versus denaturing conditions for Western blot applications

  • Implement parallel analysis using antibodies targeting different POGLUT1 epitopes (e.g., AA 132-392, AA 201-300, AA 353-382)

Species-Specific Optimization: When contradictions arise between human and mouse studies:

  • Verify sequence homology between human and mouse POGLUT1 in the antibody target region

  • Validate each biotin-conjugated POGLUT1 antibody in species-specific positive controls

  • Implement species-matched negative controls to confirm specificity

Post-Translational Modification Analysis: POGLUT1 itself may undergo modifications affecting antibody recognition:

  • Treat samples with appropriate deglycosylation enzymes prior to antibody application

  • Compare results between reducing and non-reducing conditions

  • Implement mass spectrometry analysis to characterize POGLUT1 modifications in each experimental system

Context-Dependent Expression Level Adjustment: Optimize antibody concentration based on expression levels:

  • Perform titration experiments to determine optimal antibody concentration for each experimental system

  • For systems with high endogenous POGLUT1 expression, dilute primary antibody to prevent signal saturation

  • For systems with low expression, implement signal amplification systems while maintaining controlled background levels

Validation Through Complementary Approaches: Confirm findings using orthogonal methods:

  • Complement antibody-based detection with reporter-based systems (e.g., POGLUT1-GFP fusion proteins)

  • Validate antibody specificity using CRISPR/Cas9-mediated POGLUT1 knockout models

  • Implement rescue experiments with wild-type POGLUT1 in deficient systems to confirm specificity of observed phenotypes

How should researchers design experiments to investigate POGLUT1's role in Notch signaling pathways?

Designing robust experiments to investigate POGLUT1's role in Notch signaling requires careful consideration of multiple experimental parameters. The following methodological framework provides a comprehensive approach:

Experimental Model Selection:

  • Cell-based systems: HEK293T cells provide a reliable system for biochemical assays of POGLUT1 activity, while mammalian cell lines with active Notch signaling (e.g., C2C12 myoblasts) offer insights into functional consequences .

  • Animal models: Mouse embryonic development studies using timed matings provide critical in vivo insights, particularly focusing on embryonic days E7.5-E9.5 when Notch signaling is crucial for somitogenesis .

  • Patient-derived materials: For translational relevance, primary cells from patients with POGLUT1 mutations enable direct assessment of pathophysiological mechanisms .

Genetic Manipulation Strategy:

  • Generate isogenic cell lines with POGLUT1 deletion using CRISPR/Cas9

  • Create rescue lines expressing either wild-type POGLUT1 or specific mutants (R183W, Y57C, I129T, R98W, C102F, W308L)

  • Implement inducible expression systems to study temporal aspects of POGLUT1 function

Notch Pathway Activity Assessment:

  • Protein-level analysis: Implement Western blotting with antibodies specifically detecting cleaved (active) NOTCH1 intracellular domain (Val 1744)

  • Transcriptional readouts: Quantify expression of canonical Notch target genes (e.g., Hes1, Hey1) using qRT-PCR

  • Reporter assays: Utilize Notch-responsive luciferase reporters to quantify pathway activity

  • Immunolocalization: Perform whole-mount immunostaining for active NOTCH1 in embryonic tissues using the antigen unmasking protocol (Vector unmasking solution at 98°C for 10 minutes followed by acetone treatment)

Functional Assay Design:

  • In vitro glycosylation assays: Assess POGLUT1 enzymatic activity using purified EGF repeat substrates

  • Notch ligand binding assays: Quantify the effect of POGLUT1 deficiency on Notch-ligand interactions

  • Developmental phenotyping: Analyze embryonic patterning defects, particularly in somitogenesis

  • Cell fate decisions: Monitor myogenic differentiation, which depends on precise Notch signaling

Controls and Validation:

  • Compare POGLUT1 deficiency phenotypes with established Notch pathway inhibition (e.g., γ-secretase inhibitors, dominant-negative RBPJ constructs)

  • Include parallel analysis of known POGLUT1 substrates beyond Notch (e.g., CRUMBS2) to distinguish Notch-specific effects

  • Implement rescue experiments with constitutively active NOTCH1 (NICD) to determine which phenotypes are Notch-dependent versus Notch-independent

What are the critical parameters for using biotin-conjugated POGLUT1 antibodies in developmental biology research?

Developmental biology research involving biotin-conjugated POGLUT1 antibodies requires careful consideration of stage-specific, tissue-specific, and technical parameters. The following methodological approach addresses these critical considerations:

Developmental Stage Selection:

  • For mouse embryonic studies, focus on E7.5-E9.5 stages where POGLUT1 function is critical for gastrulation and early organogenesis

  • Implement precise staging criteria beyond chronological age (e.g., somite number, morphological landmarks)

  • Design time-course experiments with narrow collection windows (4-hour intervals) to capture dynamic developmental processes

Tissue Fixation and Processing Protocol:

  • For whole-mount applications: Fix embryos overnight in 4% PFA/PBS at 4°C, followed by dehydration in methanol for storage at -20°C

  • For sectioning: Fix embryos for 1 hour at room temperature for immunostaining or overnight at 4°C for in-situ hybridization before OCT embedding

  • For antigen preservation: Implement antigen unmasking in Vector unmasking solution at 98°C for 10 minutes, followed by acetone treatment at -20°C for 8 minutes

Detection System Optimization:

  • For whole-mount applications: Use extended antibody incubation periods (2 days for primary antibody at 1:1000 dilution) in high-concentration blocking buffer (10% goat serum, 5% BSA, 0.3% Triton-X100 in PBS)

  • For sections: Apply primary antibodies diluted in blocking buffer overnight at 4°C, followed by secondary antibody incubation for 1 hour at room temperature

  • For multi-protein detection: Implement sequential staining protocols with complete antibody elution between rounds

Imaging Parameters:

  • Utilize point-scanning confocal microscopy (Leica-Inverted SP5 or Leica-Upright SP5) for high-resolution imaging

  • Implement standardized laser power, gain, and offset settings for comparative analyses

  • Collect z-stacks with appropriate step size (10-12 μm section thickness) for three-dimensional reconstruction

  • Analyze confocal datasets using appropriate software packages (e.g., Volocity) for quantitative assessment

Phenotypic Analysis Framework:

  • Implement parallel analysis of multiple POGLUT1 substrates (NOTCH1, CRUMBS2) in the same developmental stages

  • Correlate POGLUT1 expression patterns with tissue-specific developmental abnormalities

  • Compare phenotypes between POGLUT1-deficient models and substrate-specific mutants (e.g., Crumbs2 null) to establish mechanistic relationships

  • Assess both cell-autonomous and non-cell-autonomous effects through tissue-specific conditional knockout approaches

How can researchers use biotin-conjugated POGLUT1 antibodies to investigate post-translational modifications of substrate proteins?

Investigating post-translational modifications of POGLUT1 substrate proteins requires sophisticated biochemical approaches leveraging the specificity and sensitivity of biotin-conjugated POGLUT1 antibodies. The following methodological framework enables comprehensive analysis:

Substrate Protein Isolation Strategy:

  • Implement immunoprecipitation protocols using antibodies against known POGLUT1 substrates (NOTCH1, CRUMBS2, DLK1)

  • For challenging substrates, express epitope-tagged versions (V5, FLAG, MycHis6) to facilitate efficient pulldown

  • Design cell-based expression systems for wild-type and mutant substrate proteins with mutations in EGF repeats to identify specific modification sites

Glycan Detection Approaches:

  • Western blot mobility shift analysis: Compare electrophoretic mobility of substrates from wild-type versus POGLUT1-deficient samples

  • Enzymatic deglycosylation: Treat immunoprecipitated substrates with specific glycosidases to remove O-glucose modifications

  • Lectins with O-glucose specificity: Use in blot overlay assays to directly detect O-glucose modifications

  • Mass spectrometry analysis: Implement specialized glycoproteomics workflows to map specific O-glucose modification sites on EGF repeats

Functional Correlation Analysis:

  • Generate substrate constructs with mutations in specific EGF repeats to prevent O-glucosylation

  • Assess subcellular localization of wild-type versus O-glucosylation-deficient substrates

  • Quantify trafficking rates of modified versus unmodified substrates using pulse-chase approaches

  • Evaluate ligand-binding properties of glycosylated versus unglycosylated substrates using solid-phase binding assays

Visualization of Modified Substrates:

  • Implement dual-labeling strategies with biotin-conjugated POGLUT1 antibodies and substrate-specific antibodies

  • Use proximity ligation assays to detect close association between POGLUT1 and its substrates in situ

  • Apply super-resolution microscopy techniques to visualize subcellular compartments where modification occurs

  • Develop FRET-based reporters to monitor substrate modification in living cells

Comparative Cross-Species Analysis:

  • Assess conservation of O-glucosylation sites across species (human, mouse, Drosophila)

  • Compare POGLUT1 substrate specificity between vertebrate and invertebrate systems

  • Evaluate functional consequences of substrate modification in different model organisms

  • Implement cross-species rescue experiments to determine functional conservation of O-glucosylation

What technical challenges might researchers encounter when using biotin-conjugated POGLUT1 antibodies in mass spectrometry-based proteomics studies?

Mass spectrometry-based proteomics involving biotin-conjugated POGLUT1 antibodies presents several technical challenges that require specialized methodological approaches. The following strategies address these challenges:

Sample Preparation Considerations:

  • Antibody Interference Management: The biotin conjugation on POGLUT1 antibodies can generate confounding peptides during proteolytic digestion. Implement size-exclusion chromatography or immunodepletion steps to remove antibody fragments before analysis.

  • Cross-linking Optimization: When using cross-linking approaches to capture transient POGLUT1-substrate interactions, carefully titrate cross-linker concentration and reaction time to minimize non-specific aggregation while maximizing specific complex formation .

  • Enrichment Strategy Selection: For low-abundance POGLUT1 substrates, implement sequential enrichment protocols combining biotin-streptavidin capture with substrate-specific immunoprecipitation.

Mass Spectrometric Analysis Parameters:

  • Glycopeptide Fragmentation Optimization: O-glucose modifications on EGF repeats require specialized fragmentation approaches. Implement electron-transfer dissociation (ETD) or electron-capture dissociation (ECD) in addition to collision-induced dissociation (CID) to preserve the glycan-peptide linkage .

  • Data Acquisition Settings: Design targeted acquisition methods (parallel reaction monitoring or targeted MS/MS) focusing on predicted glycopeptides from EGF repeats of known POGLUT1 substrates.

  • Internal Standards Implementation: Develop synthetic glycopeptide standards incorporating stable isotope labels to enable absolute quantification of O-glucose occupancy at specific EGF repeats.

Data Analysis Challenges:

  • Modified Database Searches: Create customized search parameters that account for O-glucose modifications (+162.05 Da) and potential extended glycan structures.

  • False Discovery Rate Control: Implement stringent filters for glycopeptide identification, requiring detection of diagnostic glycan oxonium ions and retention time alignment with standards.

  • Site Localization Algorithms: Develop scoring algorithms that assess the confidence of O-glucose site assignment within EGF repeats containing multiple potential modification sites.

Quantification Approach Selection:

  • Stable Isotope Labeling: Implement SILAC or TMT labeling to compare glycopeptide abundance between wild-type and POGLUT1-deficient samples.

  • Label-free Quantification: Develop specialized extraction ion chromatogram (XIC) methods accounting for the altered chromatographic behavior of glycopeptides.

  • Glycoform Profile Analysis: Quantify the distribution of different glycoforms (O-glucose, O-glucose-xylose) at individual modification sites to assess glycosylation pathway dynamics.

Validation Requirements:

  • Orthogonal Glycopeptide Characterization: Confirm MS-based identifications using synthetic glycopeptide standards with identical retention time and fragmentation patterns.

  • Site-directed Mutagenesis: Generate substrate proteins with mutations at putative O-glucosylation sites to verify MS-based site assignments through comparative analysis.

  • Complementary Glycomics Approaches: Implement orthogonal glycan analysis after enzymatic release to confirm glycan compositions identified in glycoproteomic workflows .

What are common sources of false positives/negatives when using biotin-conjugated POGLUT1 antibodies, and how can they be mitigated?

Biotin-conjugated POGLUT1 antibodies can produce misleading results due to several technical factors. The following comprehensive approach addresses common sources of error and provides effective mitigation strategies:

Sources of False Positives and Mitigation Strategies:

  • Endogenous Biotin Interference:

    • Problem: Tissues rich in endogenous biotin (liver, kidney, brain) can produce false positive signals.

    • Solution: Implement avidin/biotin blocking steps before applying biotin-conjugated antibodies. Use commercially available endogenous biotin blocking kits with sequential avidin and biotin incubation steps .

  • Non-specific Binding of Detection Reagents:

    • Problem: Streptavidin conjugates can bind non-specifically to certain tissue components.

    • Solution: Include blocking steps with 0.1% BSA and 0.3% Triton X-100 in PBS. Extend blocking time to overnight at 4°C for challenging samples. Use highly purified streptavidin conjugates and titrate to optimal concentration .

  • Cross-reactivity with Related Proteins:

    • Problem: Antibodies may recognize proteins with similar epitopes to POGLUT1.

    • Solution: Validate with POGLUT1 knockout/knockdown samples. Perform peptide competition assays using the specific immunogen sequence (e.g., AA 160-280) to confirm binding specificity .

  • Biotin Conjugate Degradation:

    • Problem: Degraded biotin conjugates can produce spurious signals.

    • Solution: Store antibodies at -20°C in single-use aliquots. Periodically verify conjugate integrity using positive control samples with known POGLUT1 expression patterns .

Sources of False Negatives and Mitigation Strategies:

  • Epitope Masking Due to Fixation:

    • Problem: Certain fixation protocols can mask the POGLUT1 epitope.

    • Solution: Implement antigen retrieval using Vector unmasking solution at 98°C for 10 minutes, followed by acetone treatment at -20°C for 8 minutes. Compare multiple fixation protocols to identify optimal conditions .

  • Insufficient Permeabilization:

    • Problem: POGLUT1's ER localization requires effective membrane permeabilization.

    • Solution: Optimize detergent concentration and incubation time. For challenging samples, consider dual detergent approaches combining Triton X-100 (0.3%) with saponin (0.1%) .

  • Suboptimal Antibody Concentration:

    • Problem: Too low antibody concentration can yield false negatives.

    • Solution: Perform systematic titration experiments for each experimental system. For tissues with low POGLUT1 expression, extend primary antibody incubation to 48-72 hours at 4°C with gentle agitation .

  • Signal Quenching by Sample Autofluorescence:

    • Problem: Tissue autofluorescence can mask specific signals.

    • Solution: Implement autofluorescence reduction protocols (e.g., Sudan Black B treatment). Use fluorophores with emission spectra distinct from autofluorescence profiles. Consider spectral unmixing during image acquisition and analysis .

Quality Control Implementation:

  • Systematic Validation Protocol:

    • Include parallel staining with non-conjugated POGLUT1 antibodies detected with secondary antibodies

    • Compare staining patterns between different POGLUT1 antibodies targeting distinct epitopes

    • Implement dual detection using antibodies against known POGLUT1 interaction partners

  • Quantitative Assessment Methods:

    • Develop standardized signal-to-noise measurement protocols

    • Establish threshold criteria for positive versus negative staining

    • Implement automated image analysis algorithms for objective signal quantification

How can researchers validate the specificity of biotin-conjugated POGLUT1 antibodies when studying novel substrate proteins?

Validating the specificity of biotin-conjugated POGLUT1 antibodies for studies of novel substrate proteins requires a systematic, multi-pronged approach. The following methodological framework provides comprehensive validation:

Expression System Validation:

  • Recombinant Protein Expression Controls:

    • Generate expression constructs for both POGLUT1 and the putative substrate protein

    • Create epitope-tagged versions (e.g., MycHis6, FLAG) for parallel detection with tag-specific antibodies

    • Express in multiple cell types to identify optimal expression systems

  • Inducible Expression Systems:

    • Implement tetracycline-inducible or similar systems to precisely control expression levels

    • Create dose-response curves to correlate antibody signal with controlled protein expression

    • Compare wild-type POGLUT1 with catalytically inactive mutants (e.g., R183W, Y57C) to distinguish enzymatic versus structural interactions

Genetic Manipulation Approaches:

  • CRISPR/Cas9-Mediated Knockout Validation:

    • Generate POGLUT1 knockout cell lines as negative controls

    • Create substrate protein knockout lines to confirm signal specificity

    • Implement rescue experiments with wild-type and mutant constructs

  • siRNA/shRNA Knockdown Controls:

    • Utilize siRNA pools targeting different regions of POGLUT1 mRNA

    • Establish dose-dependent knockdown to correlate with signal reduction

    • Include non-targeting control siRNAs to assess non-specific effects

Biochemical Validation Strategies:

  • Immunoprecipitation-Western Blot Confirmation:

    • Perform immunoprecipitation with substrate-specific antibodies followed by Western blot with biotin-conjugated POGLUT1 antibodies

    • Conduct reciprocal immunoprecipitation with POGLUT1 antibodies followed by substrate protein detection

    • Include multiple control antibodies of the same isotype to assess non-specific binding

  • Mass Spectrometry Validation:

    • Isolate protein complexes using biotin-conjugated POGLUT1 antibodies

    • Analyze by LC-MS/MS to identify interacting proteins

    • Compare protein profiles from wild-type versus POGLUT1-deficient samples

    • Confirm the presence of the substrate protein and characterize modification sites

Functional Validation Approaches:

  • In Vitro Glycosylation Assays:

    • Express and purify recombinant EGF repeats from the putative substrate

    • Perform in vitro glycosylation assays with purified POGLUT1

    • Analyze glycosylation using mass spectrometry or radioactive UDP-glucose incorporation

  • Subcellular Co-localization Analysis:

    • Perform dual immunofluorescence with markers for subcellular compartments

    • Quantify co-localization using Pearson's correlation coefficient or Manders' overlap coefficient

    • Compare wild-type versus POGLUT1-deficient cells to assess substrate protein localization changes

  • Functional Rescue Experiments:

    • Express catalytically active POGLUT1 in deficient cells

    • Assess restoration of substrate protein function

    • Compare with expression of catalytically inactive POGLUT1 mutants as controls

What strategies can researchers employ to overcome batch-to-batch variability in biotin-conjugated POGLUT1 antibodies?

Batch-to-batch variability in biotin-conjugated POGLUT1 antibodies can significantly impact experimental reproducibility. The following methodological framework provides comprehensive strategies to mitigate this variability:

Standardized Antibody Qualification Protocol:

  • Reference Standard Establishment:

    • Create a master reference sample with verified POGLUT1 expression

    • Aliquot and store at -80°C for long-term stability

    • Use this reference standard to qualify each new antibody batch

  • Multi-parameter Performance Assessment:

    • Develop a standardized testing protocol evaluating:

      • Signal-to-noise ratio in Western blotting

      • Specific band pattern and molecular weight verification

      • Immunostaining pattern in control cell lines and tissues

      • Quantitative signal intensity compared to reference standards

  • Titration Curve Analysis:

    • Perform systematic dilution series (typically 1:100 to 1:10,000)

    • Identify optimal working concentration for each batch

    • Document batch-specific optimal concentrations for different applications

Internal Control Implementation:

  • Positive Control Panel Development:

    • Create a panel of cell lines or tissue samples with varying POGLUT1 expression levels

    • Include samples with:

      • Endogenous POGLUT1 expression

      • POGLUT1 overexpression (wild-type and tagged versions)

      • POGLUT1 knockout/knockdown models

  • Normalization Standards Integration:

    • Include invariant markers as internal loading controls

    • Implement housekeeping proteins for Western blot normalization

    • Use tissue-specific structural markers for immunohistochemistry normalization

  • Multiplex Detection Approaches:

    • Design experimental protocols with simultaneous detection of multiple targets

    • Include at least one well-characterized marker with stable detection properties

    • Normalize POGLUT1 signal to these internal reference markers

Biotin Conjugation Consistency:

  • In-house Conjugation Option:

    • Purchase consistent lots of unconjugated antibodies

    • Perform standardized in-house biotinylation using commercial kits

    • Verify biotinylation efficiency using avidin-binding assays

  • Biotin-to-Antibody Ratio Assessment:

    • Implement HABA assay or similar methods to quantify biotin incorporation

    • Target consistent biotin:antibody ratios across batches

    • Adjust working concentrations based on conjugation efficiency

  • Storage Stability Monitoring:

    • Establish a quality control testing schedule (e.g., every 3 months)

    • Monitor signal intensity and specificity over time

    • Document stability profiles to predict shelf-life for each batch

Cross-Validation Approaches:

  • Multi-antibody Consensus Strategy:

    • Use multiple antibodies targeting different POGLUT1 epitopes

    • Implement antibodies from different host species or different clones

    • Consider results valid only when consistently observed across antibodies

  • Orthogonal Detection Methods:

    • Complement antibody-based detection with:

      • mRNA expression analysis (qRT-PCR, RNA-seq)

      • Metabolic labeling of glycosylated proteins

      • Activity-based enzyme assays for POGLUT1 function

  • Bridging Study Design:

    • When transitioning to a new antibody batch, perform parallel experiments

    • Generate conversion factors to normalize historical data

    • Maintain a small reserve of previous batches for critical comparative studies

How should researchers interpret changes in POGLUT1 expression patterns in relation to substrate glycosylation status?

Interpreting changes in POGLUT1 expression patterns in relation to substrate glycosylation requires a sophisticated analytical framework that integrates multiple data types. The following methodological approach provides comprehensive guidance:

Correlation Analysis Framework:

  • Quantitative Expression Profiling:

    • Implement Western blot densitometry with standardized exposure and analysis settings

    • Normalize POGLUT1 expression to appropriate housekeeping proteins

    • Establish dose-response relationships between POGLUT1 levels and substrate glycosylation

  • Temporal Dynamics Assessment:

    • Design time-course experiments capturing POGLUT1 expression changes

    • Monitor corresponding changes in substrate glycosylation with appropriate lag times

    • Implement mathematical modeling to characterize the relationship between enzyme expression and substrate modification

  • Spatial Co-localization Quantification:

    • Perform dual immunofluorescence for POGLUT1 and substrate proteins

    • Quantify co-localization using appropriate algorithms (Pearson's coefficient, Manders' overlap)

    • Correlate spatial overlap with functional evidence of substrate modification

Substrate-Specific Analysis Approaches:

  • NOTCH1 Pathway Correlation:

    • Assess NOTCH1 cleavage (using Val 1744-specific antibodies) as a functional readout of proper glycosylation

    • Quantify downstream target gene expression (Hes1, Hey1) as indicators of functional Notch signaling

    • Correlate POGLUT1 levels with both NOTCH1 processing and signaling output

  • CRUMBS2 Localization Assessment:

    • Monitor apical membrane localization of CRUMBS2 as an indicator of proper glycosylation

    • Quantify the ratio of membrane to intracellular CRUMBS2 as a function of POGLUT1 expression

    • Assess epithelial polarity markers as functional readouts of CRUMBS2 activity

  • DLK1 Trafficking Analysis:

    • Measure the rate of DLK1 transport from ER to cell surface in relation to POGLUT1 levels

    • Quantify co-localization with compartment markers (BFP-KDEL for ER, GM130 for Golgi)

    • Establish threshold levels of POGLUT1 required for efficient trafficking

Glycan Structure Characterization:

  • Glycoform Profiling Approach:

    • Implement lectin blotting or mass spectrometry to characterize O-glucose structures

    • Assess changes in glycan complexity (e.g., addition of xylose to O-glucose) as a function of POGLUT1 expression

    • Correlate specific glycoforms with functional properties of the substrate proteins

  • Site Occupancy Analysis:

    • Determine the percentage of substrate molecules modified at specific EGF repeats

    • Assess whether POGLUT1 expression affects modification site preference

    • Correlate modification patterns with functional outcomes

Integrated Multi-parameter Analysis:

  • Multivariate Statistical Approaches:

    • Implement principal component analysis to identify patterns across multiple parameters

    • Develop regression models relating POGLUT1 expression to multiple substrate characteristics

    • Identify threshold effects and non-linear relationships in the data

  • Systems Biology Integration:

    • Incorporate POGLUT1 expression data into broader pathway models

    • Account for competing enzymatic activities and substrate availability

    • Develop predictive models relating enzyme expression to functional outcomes

  • Context-Dependent Interpretation Framework:

    • Consider cell type-specific factors that may influence the relationship between POGLUT1 expression and substrate glycosylation

    • Account for developmental stage-specific effects in embryonic studies

    • Integrate findings across experimental systems to identify conserved versus context-dependent relationships

What analytical approaches can distinguish between direct and indirect effects of POGLUT1 on cellular processes?

Distinguishing between direct and indirect effects of POGLUT1 on cellular processes requires sophisticated analytical approaches that isolate specific molecular interactions. The following methodological framework provides comprehensive strategies:

Temporal Sequence Analysis:

  • High-Resolution Time Course Studies:

    • Implement tightly spaced temporal sampling following POGLUT1 perturbation

    • Identify the chronological order of molecular and cellular changes

    • Primary (direct) effects should precede secondary (indirect) effects

    • Utilize mathematical modeling to establish causality relationships

  • Pulse-Chase Experimental Design:

    • Apply acute POGLUT1 inhibition or activation systems

    • Monitor immediate versus delayed responses

    • Direct targets should show rapid response kinetics

    • Implement metabolic labeling to track newly synthesized proteins versus existing pools

  • Reversibility Testing:

    • Utilize inducible expression systems to restore POGLUT1 function

    • Monitor the temporal sequence of recovery

    • Direct targets should show more immediate normalization

    • Compare recovery kinetics across multiple cellular processes

Substrate-Specific Manipulation Approaches:

  • Comparative Phenotypic Analysis:

    • Directly compare phenotypes between POGLUT1-deficient models and substrate-specific mutants

    • Processes affected similarly in both models likely represent direct POGLUT1 effects

    • Implement quantitative phenotypic scoring to identify subtle differences

  • Genetic Epistasis Testing:

    • Perform epistasis analysis with POGLUT1 and putative substrate mutations

    • Generate double mutants and compare to single mutants

    • Direct relationships typically show non-additive effects in double mutants

    • Extend analysis to downstream pathway components to map hierarchical relationships

  • Substrate-Specific Rescue Approaches:

    • Express modified substrate proteins that bypass the need for POGLUT1 modification

    • For NOTCH1, express the constitutively active intracellular domain

    • For CRUMBS2, implement membrane-targeting strategies that bypass ER quality control

    • Assess which POGLUT1-dependent phenotypes are rescued by each substrate

Biochemical Interaction Characterization:

  • In Vitro Enzymatic Assays:

    • Reconstitute POGLUT1 activity with purified components

    • Test candidate substrates directly in controlled reactions

    • Implement kinetic analysis to determine substrate preferences

    • Compare in vitro results with in vivo observations

  • Proximity Labeling Approaches:

    • Utilize BioID or APEX2 fusion proteins to identify proteins in close proximity to POGLUT1

    • Compare proximity proteomes across different cellular compartments

    • Direct substrates should show consistent proximity to POGLUT1

    • Validate candidate interactions using independent methods

  • Structure-Function Analysis:

    • Generate catalytically inactive POGLUT1 mutants that maintain protein interactions

    • Compare phenotypes between null mutants and catalytically inactive mutants

    • Processes requiring enzymatic activity represent direct effects

    • Processes maintained with inactive POGLUT1 suggest scaffold functions

Integrated Systems Approaches:

  • Multi-omics Data Integration:

    • Combine transcriptomics, proteomics, and glycoproteomics data

    • Apply network analysis to identify direct POGLUT1 interactors

    • Implement machine learning approaches to distinguish primary from secondary effects

    • Validate predictions using targeted experimental approaches

  • Pathway-Specific Reporter Systems:

    • Develop reporters for NOTCH, WNT, TGF-β and other key developmental pathways

    • Monitor pathway activity changes following POGLUT1 manipulation

    • Direct targets should show more immediate and consistent responses

    • Cross-validate findings using pharmacological pathway inhibitors

  • Cell Type-Specific Analysis:

    • Compare POGLUT1 effects across different cell types

    • Direct targets should show consistent responses across cellular contexts

    • Context-dependent effects likely represent indirect mechanisms

    • Implement single-cell analysis to resolve heterogeneous responses

How might biotin-conjugated POGLUT1 antibodies be used in emerging single-cell glycoproteomics approaches?

Single-cell glycoproteomics represents a frontier for understanding heterogeneity in protein glycosylation at unprecedented resolution. Biotin-conjugated POGLUT1 antibodies offer unique capabilities in this emerging field through the following methodological approaches:

Single-Cell Protein Glycosylation Profiling:

  • Mass Cytometry (CyTOF) Integration:

    • Develop metal-tagged streptavidin for detecting biotin-conjugated POGLUT1 antibodies

    • Combine with antibodies against known POGLUT1 substrates and their glycoforms

    • Implement computational clustering to identify cell subpopulations with distinct glycosylation profiles

    • Correlate POGLUT1 levels with substrate modification efficiency at single-cell resolution

  • Microfluidic-Based Single-Cell Western Blotting:

    • Adapt biotin-conjugated POGLUT1 antibodies for microfluidic platforms

    • Implement parallel detection of POGLUT1 and its substrates

    • Quantify cell-to-cell variability in enzyme-substrate relationships

    • Correlate protein expression heterogeneity with functional phenotypes

  • In Situ Glycan Imaging Technologies:

    • Combine biotin-conjugated POGLUT1 antibodies with glycan-specific probes

    • Implement multiplexed imaging approaches (CODEX, MIBI-TOF)

    • Achieve subcellular resolution of POGLUT1 activity zones

    • Map spatial relationships between enzyme localization and substrate modification

Single-Cell Glycoproteogenomics Integration:

  • Combined Protein and Transcript Analysis:

    • Pair antibody-based POGLUT1 protein detection with single-cell RNA-seq

    • Implement CITE-seq or similar technologies for simultaneous protein and RNA analysis

    • Correlate POGLUT1 protein levels with transcript abundance of glycosylation machinery components

    • Identify regulatory relationships governing glycosylation heterogeneity

  • Lineage Tracing with Glycosylation Profiling:

    • Monitor changes in POGLUT1 expression and substrate glycosylation during cellular differentiation

    • Implement genetic barcoding to track clonal relationships

    • Identify developmental branch points where glycosylation patterns diverge

    • Correlate glycosylation changes with cell fate decisions in developmental processes

  • Spatial Transcriptomics Integration:

    • Combine in situ analysis of POGLUT1 and substrate localization with spatial transcriptomics

    • Map microenvironmental influences on glycosylation patterns

    • Identify tissue niches with distinctive glycosylation signatures

    • Correlate spatial glycosylation patterns with local gene expression programs

Novel Technical Implementations:

  • Nanobody-Based Detection Systems:

    • Develop anti-biotin nanobodies for improved penetration in tissue sections

    • Create bispecific nanobodies targeting both biotin and glycan structures

    • Implement nanobody-based proximity labeling for glycoprotein interactome mapping

    • Enable super-resolution imaging of glycosylation machinery and substrates

  • Glyco-Focused Spatial Proteomics:

    • Apply multiplexed ion beam imaging with biotin-conjugated POGLUT1 antibodies

    • Achieve subcellular resolution of POGLUT1 and substrate co-localization

    • Map the spatial distribution of glycosylation machinery components

    • Correlate enzyme distribution with substrate modification patterns

  • Single-Cell Glycoproteome Analysis:

    • Develop microfluidic platforms for single-cell protein extraction

    • Implement specialized glycopeptide enrichment strategies compatible with limited material

    • Combine with ultrasensitive mass spectrometry approaches

    • Quantify cell-to-cell variability in site-specific glycosylation patterns

Data Analysis and Integration Frameworks:

  • Machine Learning Classification Approaches:

    • Develop algorithms to classify cells based on glycosylation signatures

    • Implement unsupervised clustering to identify novel cell states defined by glycosylation patterns

    • Create predictive models relating POGLUT1 expression to substrate modification profiles

    • Enable phenotypic prediction from glycosylation data

  • Network Analysis of Glycosylation Regulation:

    • Construct regulatory networks linking transcriptional control to glycosylation outcomes

    • Identify key nodes controlling glycosylation heterogeneity

    • Map feedback mechanisms between glycosylation status and cellular responses

    • Develop integrated models of glycosylation regulation at single-cell resolution

What are potential applications of biotin-conjugated POGLUT1 antibodies in investigating disease mechanisms beyond muscular dystrophy?

Biotin-conjugated POGLUT1 antibodies offer valuable tools for investigating disease mechanisms across multiple pathological contexts beyond muscular dystrophy. The following approaches highlight their potential applications:

Neurodevelopmental and Neurodegenerative Disorders:

  • Notch-Dependent Neurogenesis Dysregulation:

    • Investigate POGLUT1's role in cortical development through Notch signaling

    • Analyze neuronal progenitor maintenance and differentiation timing

    • Assess potential contributions to microcephaly and cortical malformation disorders

    • Implement lineage tracing studies to monitor neural stem cell fate decisions

  • Protein Misfolding Mechanisms:

    • Examine POGLUT1's role in ER quality control for EGF repeat-containing proteins

    • Investigate potential contributions to protein aggregation disorders

    • Assess ER stress responses in neurodegenerative disease models

    • Explore therapeutic approaches targeting POGLUT1-dependent glycosylation

  • Blood-Brain Barrier Development and Integrity:

    • Study POGLUT1-dependent glycosylation of Notch receptors in endothelial cells

    • Investigate barrier formation during development and maintenance in adulthood

    • Explore implications for cerebrovascular disorders and neurodegenerative conditions

    • Develop targeted delivery approaches for crossing the blood-brain barrier

Cancer Biology Applications:

  • Tumor Heterogeneity Analysis:

    • Characterize POGLUT1 expression across tumor subpopulations

    • Correlate expression patterns with cancer stem cell markers

    • Assess glycosylation heterogeneity and its relationship to therapy resistance

    • Implement single-cell approaches to resolve intratumoral variability

  • Metastasis Pathway Investigation:

    • Examine POGLUT1's role in epithelial-to-mesenchymal transition

    • Analyze basement membrane interactions of metastatic cells

    • Investigate migration and invasion capabilities in relation to substrate glycosylation

    • Develop therapeutic strategies targeting metastasis-specific glycosylation patterns

  • Therapy Response Prediction:

    • Develop glycosylation-based biomarkers for therapy selection

    • Assess whether POGLUT1-dependent glycosylation affects drug sensitivity

    • Monitor glycosylation changes during treatment as indicators of response

    • Identify combination approaches targeting both glycosylation and downstream pathways

Developmental and Congenital Disorders:

  • Laterality Defect Analysis:

    • Investigate POGLUT1's role in left-right patterning through Notch signaling

    • Assess ciliary functions in relation to POGLUT1-dependent glycosylation

    • Explore potential contributions to heterotaxy and related disorders

    • Develop diagnostic approaches based on glycosylation profiles

  • Congenital Heart Defect Mechanisms:

    • Analyze cardiac development with focus on valve formation and septation

    • Investigate cardiomyocyte differentiation and organization

    • Assess endocardial-myocardial signaling mediated by Notch

    • Explore therapeutic approaches for POGLUT1-related cardiac phenotypes

  • Craniofacial Development Disorders:

    • Examine neural crest specification and migration

    • Investigate patterning of pharyngeal arches and derivatives

    • Assess signaling networks coordinating craniofacial morphogenesis

    • Develop diagnostic markers for craniofacial disorders with altered glycosylation

Metabolic and Inflammatory Conditions:

  • Diabetes and Insulin Signaling:

    • Investigate POGLUT1's role in pancreatic islet development and maintenance

    • Assess β-cell regeneration and turnover in relation to Notch signaling

    • Explore potential contributions to insulin resistance mechanisms

    • Develop therapeutic approaches targeting pancreatic Notch signaling

  • Fibrotic Disease Mechanisms:

    • Analyze POGLUT1-dependent regulation of myofibroblast differentiation

    • Investigate extracellular matrix production and remodeling

    • Assess epithelial-mesenchymal communication in fibrotic disorders

    • Explore anti-fibrotic therapeutic approaches targeting glycosylation pathways

  • Inflammatory Signaling Regulation:

    • Examine POGLUT1's role in regulating immune cell differentiation

    • Investigate potential modifications of inflammatory signaling receptors

    • Assess macrophage polarization in relation to glycosylation patterns

    • Develop immunomodulatory approaches based on glycosylation targeting

How might researchers integrate biotin-conjugated POGLUT1 antibodies with emerging glycobiology technologies?

Integration of biotin-conjugated POGLUT1 antibodies with emerging glycobiology technologies offers unprecedented opportunities to advance our understanding of protein O-glucosylation. The following methodological framework outlines innovative integration approaches:

CRISPR/Cas9-Based Glycoengineering Platforms:

  • Precise Glycosylation Site Editing:

    • Engineer EGF repeats in substrate proteins to modify O-glucosylation sites

    • Generate isogenic cell lines with specific glycosylation patterns

    • Use biotin-conjugated POGLUT1 antibodies to verify modified glycosylation

    • Correlate site-specific glycosylation changes with functional outcomes

  • Glycosylation Machinery Screens:

    • Implement CRISPR/Cas9 screens targeting glycosylation pathway components

    • Use biotin-conjugated POGLUT1 antibodies as readouts for altered substrate modification

    • Identify novel regulators of O-glucosylation pathways

    • Develop pathway maps linking glycosylation regulators to functional outcomes

  • Endogenous Tagging of Glycoproteins:

    • Insert epitope tags into endogenous POGLUT1 substrates

    • Develop dual detection strategies combining biotin-conjugated POGLUT1 antibodies with tag-specific antibodies

    • Enable live-cell imaging of glycoprotein trafficking and localization

    • Correlate glycosylation status with real-time protein dynamics

Advanced Microscopy Applications:

  • Super-Resolution Glycoprotein Imaging:

    • Apply STORM, PALM, or STED microscopy with biotin-conjugated POGLUT1 antibodies

    • Achieve nanoscale resolution of glycosylation machinery organization

    • Visualize glycoprotein clustering and membrane microdomain association

    • Map spatial relationships between POGLUT1 and substrate proteins at molecular scale

  • Live-Cell Glycosylation Monitoring:

    • Develop split-fluorescent protein systems reporting POGLUT1-substrate interactions

    • Create FRET-based biosensors for detecting O-glucosylation events

    • Implement optogenetic control of POGLUT1 activity

    • Monitor real-time glycosylation dynamics during developmental processes

  • Volumetric Tissue Imaging:

    • Adapt biotin-conjugated POGLUT1 antibodies for cleared tissue imaging (CLARITY, CUBIC)

    • Implement light-sheet microscopy for high-speed 3D imaging

    • Develop computational approaches for analyzing glycosylation patterns across tissue volumes

    • Map organwide patterns of substrate glycosylation during development

Glycoproteomics Integration Strategies:

  • O-Glycopeptide-Focused Mass Spectrometry:

    • Develop enrichment strategies combining biotin-conjugated POGLUT1 antibodies with glycopeptide-specific capture

    • Implement electron-transfer dissociation for precise glycosite mapping

    • Quantify site occupancy at individual EGF repeats

    • Correlate site-specific glycosylation with protein structure and function

  • Intact Glycoprotein Analysis:

    • Apply native mass spectrometry to analyze intact POGLUT1 substrates

    • Quantify the distribution of glycoforms across molecular populations

    • Assess how POGLUT1 levels affect glycoform heterogeneity

    • Correlate glycoform patterns with functional properties

  • Temporal Glycoproteomics:

    • Implement pulse-chase approaches with metabolic labeling of glycans

    • Track glycosylation kinetics of POGLUT1 substrates

    • Assess competition between different substrates for POGLUT1 activity

    • Develop mathematical models of glycosylation dynamics

Synthetic Biology and Glycoengineering:

  • Engineered Glycosylation Systems:

    • Create simplified in vitro glycosylation platforms using purified components

    • Develop cell-free expression systems with controlled glycosylation

    • Use biotin-conjugated POGLUT1 antibodies to validate engineered glycoproteins

    • Apply to therapeutic glycoprotein production and optimization

  • Glycosylation Circuit Design:

    • Engineer feedback systems regulating POGLUT1 expression and activity

    • Create synthetic genetic circuits linking glycosylation to cellular responses

    • Develop reporter systems monitoring glycosylation efficiency

    • Apply to directed evolution of optimized glycosylation pathways

  • Orthogonal Glycosylation Machinery:

    • Develop modified POGLUT1 variants with altered substrate specificity

    • Create orthogonal sugar-nucleotide donors for selective labeling

    • Implement bioorthogonal chemistry for specific detection of modified glycans

    • Enable selective tracking of specific substrate populations

Translational Glycobiology Applications:

  • Glycosylation-Based Biomarker Development:

    • Identify disease-specific glycoform patterns of POGLUT1 substrates

    • Develop multiplexed detection systems using biotin-conjugated antibodies

    • Create diagnostic platforms for muscular dystrophy and related disorders

    • Implement machine learning for pattern recognition in complex glycoform data

  • Therapeutic Glycoengineering:

    • Design approaches to restore proper glycosylation in disease models

    • Develop small molecule modulators of POGLUT1 activity

    • Create gene therapy vectors for glycosylation correction

    • Assess therapeutic outcomes using biotin-conjugated antibodies as monitoring tools

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.