POGLUT1 Human

Protein O-Glucosyltransferase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

POGLUT1 Human Recombinant produced in in Sf9 Baculovirus cells is a single, non-glycosylated polypeptide chain containing 377 amino acids (24-392a.a) and having a molecular mass of 44.5kDa (Migrates at 40-57kDa on SDS-PAGE under reducing conditions). POGLUT1 is fused to a 8 amino acid His-tag at C-Terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
POGLUT1, a homologue of Rumi from Drosophila, is an endoplasmic reticulum (ER)-retaining glucosyltransferase. This enzyme catalyzes the transfer of glucose and xylose from UDP-glucose and UDP-xylose, respectively, to EGF repeats containing the consensus sequence C-X-S-X-P-C. POGLUT1 demonstrates a positive regulatory effect on Notch signaling pathways without impacting Notch ligand binding.
Description
Recombinant human POGLUT1, expressed in Sf9 Baculovirus cells, is a non-glycosylated polypeptide chain. This single chain protein consists of 377 amino acids (24-392a.a) and exhibits a molecular weight of 44.5kDa. Under reducing conditions on SDS-PAGE, it migrates between 40-57kDa. The protein is fused with an 8 amino acid His-tag at the C-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The POGLUT1 solution is provided at a concentration of 0.25mg/ml in Phosphate Buffered Saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
Purity is determined to be greater than 90% by SDS-PAGE analysis.
Synonyms
POGLUT1, C3orf9, CLP46, hCLP46, KDELCL1, KTELC1, Protein O-glucosyltransferase 1, CAP10-like 46 kDa protein, KTEL motif-containing protein 1, Myelodysplastic syndromes relative protein, O-glucosyltransferase Rumi homolog, hRumi, Protein O-xylosyltransferase, MDSRP.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
RQKESGSKWK VFIDQINRSL ENYEPCSSQN CSCYHGVIEE DLTPFRGGIS RKMMAEVVRR KLGTHYQITK NRLYRENDCM FPSRCSGVEH FILEVIGRLP DMEMVINVRD YPQVPKWMEP AIPVFSFSKT SEYHDIMYPA WTFWEGGPAV WPIYPTGLGR WDLFREDLVR SAAQWPWKKK NSTAYFRGSR TSPERDPLIL LSRKNPKLVD AEYTKNQAWK SMKDTLGKPA AKDVHLVDHC KYKYLFNFRG VAASFRFKHL FLCGSLVFHV GDEWLEFFYP QLKPWVHYIP VKTDLSNVQE LLQFVKANDD VAQEIAERGS QFIRNHLQMD DITCYWENLL SEYSKFLSYN VTRRKGYDQI IPKMLKTELL EHHHHHH.

Q&A

What is POGLUT1 and what is its primary function in human cells?

POGLUT1 (Protein O-glucosyltransferase 1) is an enzyme that catalyzes the addition of O-glucose to serine residues within specific EGF-like domains of target proteins. Its primary function involves post-translational modification of Notch receptors and other EGF repeat-containing proteins through O-glucosylation, which significantly influences protein functionality. POGLUT1 contains a signal peptide, a CAP10 domain (a fungal glycosyltransferase domain), and exhibits high specificity for properly folded EGF-like domains . In mammalian cells, POGLUT1 is essential for proper Notch signaling and has been shown to be critical for early embryonic development, muscle regeneration, and cellular differentiation processes .

To study POGLUT1's primary function, researchers typically employ enzymatic activity assays using purified wild-type and mutant POGLUT1 proteins with EGF repeat substrates. For instance, scientists have demonstrated POGLUT1's enzymatic activity by expressing wild-type and mutant forms in HEK293T cells, followed by purification from culture media and assessment of O-glucosyltransferase activity toward EGF repeats from human coagulation factor IX or mouse Notch1 . These biochemical approaches have revealed that POGLUT1 transfers glucose from UDP-glucose (donor substrate) to serine residues within the consensus sequence C₁XSXPC₂ in EGF repeats (acceptor substrate) .

What protein domains does POGLUT1 target and what is the consensus sequence it recognizes?

POGLUT1 specifically targets EGF-like domains in various proteins, with a strong preference for properly folded domains. The canonical consensus sequence recognized by POGLUT1 is C₁XSXPC₂, where C₁ and C₂ represent the first and second conserved cysteines of an EGF repeat, X can be any amino acid, S is the serine residue that gets modified, and P is proline . This specificity for the region between the first and second cysteines of EGF repeats distinguishes POGLUT1 from other recently identified O-glucosyltransferases (POGLUT2 and POGLUT3), which modify sites between different cysteine pairs .

Researchers investigating POGLUT1 target sites typically employ mass spectrometry-based approaches to identify glycosylated residues. For example, studies have used tryptic digestion of modified proteins followed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) to precisely locate O-glucose modifications on target proteins . Experimental evidence shows that POGLUT1 can modify multiple EGF repeats in Notch receptors, including EGF12 and EGF13 of NOTCH1, demonstrating its ability to recognize the consensus sequence across different EGF domains . Additionally, POGLUT1 has been shown to possess O-xylosyltransferase activity toward certain EGF repeats containing a diserine motif within the O-glucose consensus sequence (e.g., EGF16 of mouse Notch2) .

How does POGLUT1 contribute to Notch signaling pathways?

POGLUT1 plays a critical role in Notch signaling by modifying EGF repeats in the extracellular domain of Notch receptors with O-linked glucose. These glycosylation events are essential for proper Notch receptor folding, trafficking, and interaction with ligands. Research has demonstrated that POGLUT1-mediated O-glucosylation directly impacts Notch receptor cleavage and subsequent signal transduction . In mouse models, POGLUT1 deficiency leads to decreased Notch1 cleavage and reduced Notch signaling, confirming its importance in vivo .

To investigate POGLUT1's contribution to Notch signaling, researchers employ multiple complementary approaches:

  • Biochemical assays with purified components to assess O-glucosyltransferase activity on Notch EGF repeats

  • Cell-based Notch reporter assays to measure signaling output in response to POGLUT1 manipulation

  • In vivo models with tissue-specific POGLUT1 deletion or mutation to evaluate developmental and physiological consequences

Studies have shown that the D233E mutation in POGLUT1, associated with muscular dystrophy, significantly reduces O-glucosyltransferase activity toward Notch EGF repeats, resulting in decreased Notch signaling in patient muscles . Importantly, researchers have demonstrated that increasing Notch signaling can rescue myogenesis defects in cells with POGLUT1 mutations, establishing a direct mechanistic link between POGLUT1 activity, Notch signaling, and muscle development .

What are the known disease associations of POGLUT1 mutations in humans?

Mutations in POGLUT1 have been associated with two distinct human disorders: a novel form of muscular dystrophy and Dowling-Degos Disease type 4. The muscular dystrophy phenotype is linked to a homozygous D233E mutation that reduces POGLUT1's O-glucosyltransferase activity on Notch EGF repeats . This autosomal recessive condition is characterized by decreased Notch signaling, dramatic reduction in satellite cell pool, and muscle-specific α-dystroglycan hypoglycosylation . Patients present with slow myoblast proliferation, facilitated differentiation, and a decreased pool of quiescent PAX7+ cells, consistent with impaired muscle regeneration capacity .

Dowling-Degos Disease type 4, an autosomal dominant disorder characterized by hyperpigmentation, has also been linked to mutations in POGLUT1 . While initially attributed to altered Notch signaling, recent research suggests that effects on CRUMBS proteins may also contribute to the disease pathogenesis . This is supported by evidence that POGLUT1 modifies CRUMBS proteins, which are essential for epithelial cell polarity and tissue organization.

Researchers investigating these disease associations typically employ a combination of:

  • Genetic analyses to identify and characterize pathogenic mutations

  • Biochemical assays to assess mutant protein activity

  • Patient-derived cell models to study cellular phenotypes

  • Animal models to understand developmental and tissue-specific consequences

These approaches have revealed that the pathomechanisms differ between the two disorders, with muscular dystrophy primarily involving Notch-dependent loss of satellite cells, while Dowling-Degos Disease may involve more complex effects on multiple glycosylation targets .

How is POGLUT1 structurally organized and what are its key functional domains?

POGLUT1 is structurally organized with several distinct domains that contribute to its enzymatic function and cellular localization. The protein contains an N-terminal signal peptide for targeting to the secretory pathway, a CAP10 domain (a fungal glycosyltransferase domain) that constitutes the catalytic core, and a C-terminal KDEL-like ER retention signal that keeps the enzyme in the endoplasmic reticulum . This domain organization is conserved from Drosophila (where the homolog is called Rumi) to humans, highlighting its evolutionary importance .

Structural studies of human POGLUT1 have revealed mechanistic insights into its substrate specificity and catalytic mechanism. POGLUT1 shows specificity for folded EGF-like domains, which also leads to its serine specificity within the C₁XSXPC₂ motif . The enzyme possesses the unusual ability to transfer both glucose and xylose to target substrates, which appears to be mediated by two distinct local conformational states of the protein .

To investigate POGLUT1's structure-function relationships, researchers employ several complementary approaches:

  • X-ray crystallography to determine three-dimensional structures of POGLUT1 alone and in complex with substrates or products

  • Site-directed mutagenesis to assess the roles of specific amino acids in substrate binding and catalysis

  • Enzymatic assays with different substrate analogs to probe the determinants of specificity

  • Comparative analyses across species to identify conserved functional regions

These studies have shown that POGLUT1 has co-evolved with EGF-like domains of the type found in Notch proteins, suggesting a long-standing functional relationship between the enzyme and its substrates .

What experimental approaches are most effective for studying POGLUT1's dual substrate specificity for glucose and xylose transfer?

POGLUT1 exhibits the uncommon ability to utilize both UDP-glucose and UDP-xylose as donor substrates for glycosylation of EGF repeats. To effectively study this dual substrate specificity, researchers employ a multi-faceted experimental approach combining biochemical, structural, and cellular techniques.

The most informative experimental strategy includes:

  • In vitro enzymatic assays with purified components: This approach involves expressing and purifying recombinant wild-type and mutant POGLUT1 proteins, along with EGF repeat substrates. Reactions are performed with either UDP-glucose or UDP-xylose as donor substrates, and product formation is monitored by techniques such as HPLC, mass spectrometry, or radioactive assays. For example, researchers have demonstrated that POGLUT1 can transfer glucose to multiple EGF repeats from Notch1 and also possesses O-xylosyltransferase activity toward EGF repeats with a diserine motif within the O-glucose consensus sequence (e.g., EGF16 of mouse Notch2) .

  • Structural studies of enzyme-substrate complexes: X-ray crystallography of POGLUT1 in complex with donor substrates (UDP-glucose or UDP-xylose) and acceptor substrates (EGF repeats) provides atomic-level insights into the basis for dual specificity. These studies have revealed that POGLUT1 likely adopts two distinct local conformational states that accommodate the different donor substrates .

  • Kinetic analyses comparing glucose versus xylose transfer: Detailed enzyme kinetics comparing the efficiency of glucose versus xylose transfer to various EGF repeat substrates helps quantify substrate preferences. Parameters such as Km, Vmax, and kcat/Km provide insights into the relative efficiency of the two reactions.

  • Mass spectrometry to identify and quantify modification sites: High-resolution mass spectrometry techniques, including electron transfer dissociation (ETD) and higher-energy collisional dissociation (HCD), are essential for identifying and quantifying O-glucose and O-xylose modifications on specific serine residues within EGF repeats .

  • Cellular assays with metabolic labeling: Incorporating isotopically labeled glucose or xylose into cell culture media, followed by isolation of POGLUT1 target proteins and mass spectrometric analysis, allows for tracking of these modifications in a cellular context.

This comprehensive approach has revealed that POGLUT1 can xylosylate a much broader range of EGF-like domain substrates than was previously thought , highlighting the importance of employing multiple complementary techniques when investigating dual substrate specificity.

How do different POGLUT1 mutations affect its enzymatic activity and what cellular assays best quantify these effects?

Different POGLUT1 mutations can have distinct effects on enzymatic activity, ranging from complete loss of function to partial activity reduction or altered substrate specificity. The D233E mutation identified in muscular dystrophy patients serves as an excellent case study, as it retains residual enzymatic activity but shows significantly lower O-glucosyltransferase activity than wild-type POGLUT1 .

To systematically assess how POGLUT1 mutations affect enzymatic activity, researchers employ a multi-tiered approach:

Biochemical Assays:

  • Enzyme activity assays with purified proteins: Wild-type and mutant POGLUT1 proteins are expressed in systems like HEK293T cells, purified, and tested for O-glucosyltransferase activity using EGF repeat substrates and UDP-glucose. Activity is typically quantified by measuring the incorporation of glucose into the substrate using techniques like HPLC or mass spectrometry .

  • Substrate dependence studies: Enzymatic activity is measured across a range of substrate concentrations to determine kinetic parameters (Km, Vmax) for both donor (UDP-glucose) and acceptor (EGF repeat) substrates. The D233E mutant showed lower activity than wild-type toward five different single EGF repeats from mouse Notch1, indicating that the mutation affects O-glucosylation of all EGF repeats containing the O-glucose consensus sequence .

Cellular Assays:

  • Notch reporter assays: Since POGLUT1 directly affects Notch signaling, luciferase-based Notch reporter assays provide a functional readout of POGLUT1 activity in cells. Decreased reporter activity correlates with reduced POGLUT1 function.

  • Flow cytometry for cell surface Notch: The level of properly folded Notch receptors on the cell surface can be quantified by flow cytometry using conformation-specific antibodies, providing insights into POGLUT1's effect on Notch trafficking.

  • Immunoblotting for Notch processing: Western blot analysis of Notch cleavage products (particularly NICD, the Notch intracellular domain) can reveal defects in Notch activation caused by POGLUT1 mutations. Muscles from patients with the D233E mutation showed decreased Notch signaling and reduced Notch1 cleavage .

  • Myoblast proliferation and differentiation assays: For mutations associated with muscular dystrophy, assays measuring myoblast proliferation rates, differentiation capacity (via myotube formation), and maintenance of PAX7+ quiescent cells provide functional readouts. Primary myoblasts from patients with the D233E mutation showed slow proliferation, facilitated differentiation, and a decreased pool of quiescent PAX7+ cells .

  • Rescue experiments: Perhaps the most informative cellular assay involves attempting to rescue phenotypes by increasing Notch signaling through complementary approaches. A robust rescue of myogenesis defects in patient cells was demonstrated by increasing Notch signaling, confirming that impaired Notch signaling is a key consequence of the D233E mutation .

These combined approaches provide a comprehensive assessment of how different POGLUT1 mutations affect enzymatic activity and downstream cellular functions.

What are the current challenges in differentiating between the roles of POGLUT1, POGLUT2, and POGLUT3 in human tissues?

Recent research has identified KDELC1 and KDELC2 as novel protein O-glucosyltransferases, now renamed POGLUT2 and POGLUT3, respectively . These enzymes modify sites distinct from those targeted by POGLUT1, creating significant challenges in differentiating their specific roles in human tissues. Understanding these challenges and developing strategies to address them is crucial for advancing our knowledge of protein O-glucosylation.

Key Challenges:

  • Overlapping substrate specificities: While POGLUT1 specifically modifies serine residues within the C₁XSXPC₂ motif (between cysteines 1 and 2 of EGF repeats), POGLUT2 and POGLUT3 can modify serine residues between cysteines 3 and 4, such as S435 in NOTCH1 EGF11 . This overlap complicates the attribution of observed O-glucose modifications to specific enzymes in complex biological samples.

  • Limited availability of enzyme-specific inhibitors: Currently, there are few selective inhibitors that can specifically target one POGLUT family member without affecting others, making pharmacological approaches to differentiation challenging.

  • Compensatory mechanisms: Genetic knockout or knockdown of one POGLUT enzyme may trigger compensatory upregulation of other family members, potentially masking phenotypes and confounding interpretation.

  • Tissue-specific expression patterns: Each POGLUT enzyme may have distinct tissue expression patterns that are not fully characterized, requiring comprehensive expression profiling across diverse human tissues.

  • Differential substrate preferences in vivo: While in vitro studies have defined consensus sequences for each enzyme, the actual substrate preferences in complex cellular environments may be influenced by additional factors such as protein-protein interactions or subcellular localization.

Methodological Approaches to Address These Challenges:

  • CRISPR-Cas9 genome editing: Generate single, double, and triple knockout cell lines for POGLUT1, POGLUT2, and POGLUT3 to systematically assess their individual and combined roles in O-glucosylation of target proteins.

  • Mass spectrometry-based glycoproteomics: Develop targeted mass spectrometry approaches to distinguish O-glucose modifications at different positions within EGF repeats, allowing attribution to specific POGLUT enzymes. Analysis of the tryptic peptide 429-CINTLGSFECQCLQGYTGPR-448 from NOTCH1 EGF11 revealed that this region is efficiently modified with a single O-Glc by POGLUT2 or POGLUT3, but not by POGLUT1 .

  • Site-directed mutagenesis of consensus sequences: Systematically mutate different O-glucosylation sites in target proteins (e.g., Notch receptors) to assess the functional consequences of losing modifications at specific positions.

  • Development of enzyme-specific antibodies and activity assays: Generate tools that can specifically detect and measure the activity of each POGLUT family member in complex biological samples.

  • Conditional tissue-specific knockout mouse models: Create mouse models with tissue-specific deletion of individual POGLUT enzymes to assess their roles in development and homeostasis without confounding compensatory effects.

By combining these approaches, researchers can begin to disentangle the specific roles of POGLUT1, POGLUT2, and POGLUT3 in human tissues and better understand their contributions to normal physiology and disease states.

How does POGLUT1-mediated glycosylation specifically impact satellite cell maintenance in muscular dystrophy models?

POGLUT1-mediated glycosylation plays a critical role in satellite cell maintenance, with significant implications for muscular dystrophy pathogenesis. Patients with the D233E mutation in POGLUT1 exhibit a novel form of muscular dystrophy characterized by a dramatic reduction in the satellite cell pool, highlighting the importance of this enzyme in muscle stem cell biology .

Mechanistic Impact on Satellite Cells:

  • Notch Signaling Dependency: POGLUT1-mediated O-glucosylation is essential for proper Notch receptor function, which in turn is critical for satellite cell quiescence and self-renewal. Muscles from patients with the D233E POGLUT1 mutation showed decreased Notch signaling concurrent with dramatic reduction in the satellite cell pool . This establishes a direct mechanistic link between impaired POGLUT1 activity, reduced Notch signaling, and satellite cell depletion.

  • Cell Cycle Regulation: Primary myoblasts derived from patients with the D233E mutation exhibited slow proliferation rates, suggesting that POGLUT1-mediated glycosylation influences cell cycle progression in muscle progenitor cells . This effect is likely mediated through Notch signaling, which regulates cyclins and cyclin-dependent kinase inhibitors.

  • Premature Differentiation: Patient-derived myoblasts showed facilitated differentiation, indicating that POGLUT1 deficiency disrupts the normal balance between proliferation and differentiation . This premature differentiation exhausts the stem cell pool and compromises long-term muscle regenerative capacity.

  • Maintenance of Quiescent PAX7+ Cells: A defining feature of the POGLUT1-associated muscular dystrophy is a decreased pool of quiescent PAX7+ cells . PAX7 is a transcription factor critical for satellite cell identity and self-renewal, suggesting that POGLUT1-mediated glycosylation is required for maintaining the undifferentiated state of muscle stem cells.

Experimental Approaches to Study This Relationship:

  • Immunohistochemical Analysis of Muscle Biopsies: Quantification of PAX7+ satellite cells in muscle sections from patients versus controls provides direct evidence of satellite cell depletion. Complementary staining for activated Notch (NICD) can establish the correlation between reduced Notch signaling and satellite cell loss .

  • Primary Myoblast Cultures: Isolation and culture of primary myoblasts from patient biopsies allows for detailed analysis of proliferation kinetics, differentiation capacity, and maintenance of stemness markers. These cultures can also be used for rescue experiments to establish causality .

  • Notch Signaling Manipulation: The most definitive evidence for the Notch-dependent mechanism comes from rescue experiments. A robust rescue of myogenesis was demonstrated by increasing Notch signaling in patient-derived cells, confirming that impaired Notch signaling is the key pathomechanism linking POGLUT1 dysfunction to satellite cell depletion .

  • Single-Cell Transcriptomics: This approach can reveal the molecular signatures of satellite cells with impaired POGLUT1 function, identifying downstream effectors that mediate the observed phenotypes.

  • In Vivo Lineage Tracing: Using mouse models with conditional POGLUT1 deletion and satellite cell-specific reporters allows for tracking of satellite cell fate decisions in vivo during muscle regeneration.

These findings establish that a key pathomechanism for POGLUT1-associated muscular dystrophy is Notch-dependent loss of satellite cells , distinguishing it from other forms of muscular dystrophy and suggesting potential therapeutic strategies aimed at restoring Notch signaling or preserving the satellite cell pool.

What are the mechanistic differences between POGLUT1's role in Notch signaling versus its impact on CRUMBS proteins?

POGLUT1 modifies both Notch receptors and CRUMBS proteins through O-glucosylation of their EGF-like repeats, but research has revealed significant mechanistic differences in how this glycosylation affects the function of these distinct protein families . Understanding these differences is essential for interpreting tissue-specific phenotypes associated with POGLUT1 mutations.

Differential Roles in Notch versus CRUMBS:

  • Developmental Timing: While POGLUT1 is required for Notch signaling in mouse embryos after gastrulation (around E8.0), its modification of CRUMBS2 is essential for gastrulation itself (around E7.5) . This temporal difference suggests that POGLUT1's earliest developmental role is mediated through CRUMBS proteins rather than Notch receptors.

  • Mechanistic Impact on Protein Function:

    • Notch Receptors: POGLUT1-mediated O-glucosylation affects Notch receptor cleavage and subsequent signal transduction. The absence of POGLUT1 leads to decreased NOTCH1 cleavage and reduced Notch signaling .

    • CRUMBS Proteins: For CRUMBS2, POGLUT1-mediated glycosylation is essential for proper protein localization to the apical membrane. The unmodified CRUMBS2 protein fails to localize correctly, compromising epithelial polarity and gastrulation movements .

  • Species-Specific Requirements:

    • In Drosophila, modification of Crumbs by the POGLUT1 homolog Rumi is not required for Crumbs function .

    • In contrast, mouse POGLUT1 is absolutely required for CRUMBS2 activity, highlighting an evolutionary divergence in the functional importance of this modification .

  • Combined Glycosylation Effects: CRUMBS proteins contain both O-glucose and O-fucose modification sites. The stronger phenotype of mouse Poglut1 mutants compared to Pofut1 (protein O-fucosyltransferase 1) mutants suggests that the two types of O-glycosylation have distinct effects on CRUMBS proteins .

Experimental Approaches to Differentiate These Roles:

  • Comparative Phenotypic Analysis: The gastrulation defects of Poglut1 mutants are indistinguishable from those of Crumbs2 mutants, providing strong evidence that CRUMBS2 is the critical POGLUT1 target during early development .

  • Protein Localization Studies: Immunofluorescence analysis of CRUMBS2 localization in wild-type versus POGLUT1-deficient tissues reveals the requirement of O-glucosylation for proper apical membrane targeting .

  • Modification Site Mutagenesis: Systematic mutation of O-glucose sites in Notch versus CRUMBS proteins, followed by functional assays, can reveal which modifications are critical for each protein's function.

  • Rescue Experiments: Testing whether expressing constitutively active Notch or properly localized CRUMBS2 can rescue POGLUT1 deficiency phenotypes helps establish causality and distinguish the relative contributions of each pathway.

  • Tissue-Specific Conditional Knockout Models: Creating conditional POGLUT1 knockout models in tissues where either Notch or CRUMBS functions predominate can help isolate the specific effects on each pathway.

Understanding these mechanistic differences has important implications for interpreting human diseases associated with POGLUT1 mutations. For example, while the hyperpigmentation in Dowling-Degos Disease type 4 was previously attributed to altered Notch signaling, the findings regarding CRUMBS proteins suggest that this and other POGLUT1-associated phenotypes may involve more complex mechanisms affecting multiple glycosylation targets .

What techniques are most reliable for analyzing the structural basis of POGLUT1's specificity for folded EGF-like domains?

POGLUT1 exhibits a remarkable specificity for folded EGF-like domains, which is central to its biological function and distinguishes it from other glycosyltransferases. Understanding the structural basis of this specificity is crucial for elucidating POGLUT1's role in development and disease. Several complementary techniques have proven reliable for analyzing this structural specificity.

Key Experimental Approaches:

  • X-ray Crystallography of Enzyme-Substrate Complexes: The most definitive approach involves determining high-resolution crystal structures of POGLUT1 in complex with folded EGF-like domains and donor substrates. These structures have revealed how POGLUT1 recognizes the three-dimensional conformation of properly folded EGF repeats rather than simply recognizing a linear amino acid sequence . The availability of structures showing POGLUT1 bound to both substrates and products provides detailed insights into the catalytic mechanism and the basis for specificity.

  • Site-Directed Mutagenesis Coupled with Activity Assays: Systematic mutation of residues at the enzyme-substrate interface, followed by activity assays, helps identify critical contacts that mediate specificity. For POGLUT1, mutations that disrupt the folding of EGF repeats abolish glycosylation, confirming the requirement for properly folded domains .

  • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS): This technique probes the dynamics and solvent accessibility of proteins and can reveal conformational changes in both POGLUT1 and EGF repeats upon complex formation. HDX-MS is particularly valuable for mapping interface regions and identifying flexibility changes associated with substrate recognition.

  • Surface Plasmon Resonance (SPR) or Bio-Layer Interferometry (BLI): These techniques provide quantitative measurements of binding kinetics and affinities between POGLUT1 and various EGF repeat substrates. Comparing binding parameters for properly folded versus misfolded or linear peptide variants of EGF repeats can quantitatively demonstrate the specificity for folded domains.

  • Comparative Structural Analysis Across Species: POGLUT1 has co-evolved with EGF-like domains of the type found in Notch . Comparative structural analysis of POGLUT1 orthologs from different species alongside their cognate substrates can reveal conserved recognition elements and species-specific adaptations.

  • Molecular Dynamics Simulations: Computational approaches can model the dynamic interactions between POGLUT1 and EGF repeats, providing insights into transient contacts and conformational changes that may not be captured in static crystal structures.

  • Nuclear Magnetic Resonance (NMR) Spectroscopy: For smaller EGF repeats, NMR can provide detailed information about their three-dimensional structure in solution and how this structure changes upon interaction with POGLUT1. Chemical shift perturbation experiments can map the interaction interface at atomic resolution.

The combined application of these techniques has revealed that POGLUT1's requirement for folded EGF-like domains leads to its serine specificity within the C₁XSXPC₂ motif . The enzyme can only glycosylate serine residues that are presented in the correct three-dimensional context within a properly folded EGF repeat. This structural specificity ensures that POGLUT1 only modifies its targets after they have achieved the correct fold, potentially serving as a quality control mechanism in the secretory pathway.

How can researchers effectively distinguish between direct POGLUT1 effects and secondary consequences in cellular models?

Distinguishing between direct effects of POGLUT1 activity and secondary downstream consequences represents a significant challenge in cellular models. This distinction is crucial for accurately interpreting phenotypes associated with POGLUT1 mutations or deficiency and for identifying true mechanistic links. Several methodological approaches can help researchers effectively differentiate between these effects.

Methodological Approaches:

  • Acute versus Chronic Manipulation of POGLUT1:

    • Acute inhibition using inducible systems (e.g., tetracycline-regulated expression or degradation-tag approaches) can reveal immediate effects before secondary adaptations occur.

    • Comparison with chronic knockout models helps distinguish primary effects from compensatory responses.

  • Substrate-Specific Rescue Experiments:

    • One of the most powerful approaches involves selective rescue experiments targeting specific POGLUT1 substrates. For example, in muscular dystrophy models with the D233E POGLUT1 mutation, researchers demonstrated a robust rescue of myogenesis defects by specifically increasing Notch signaling .

    • This approach established that impaired Notch signaling (rather than other potential POGLUT1 substrates) is the primary mechanism underlying the observed phenotype.

  • Direct Glycosylation Site Mutagenesis:

    • Mutating specific O-glucosylation sites in individual POGLUT1 substrates (e.g., Notch receptors or CRUMBS proteins) and comparing the resulting phenotypes with POGLUT1 deficiency helps identify which substrates mediate specific aspects of the POGLUT1-dependent phenotype.

    • This approach revealed that the gastrulation defects of POGLUT1-deficient mouse embryos are due to CRUMBS2 dysfunction rather than impaired Notch signaling .

  • Temporal Analysis of Molecular and Cellular Events:

    • Establishing a detailed timeline of molecular and cellular changes following POGLUT1 inhibition helps distinguish early (likely direct) effects from later (potentially secondary) consequences.

    • Time-course analyses of Notch signaling components, CRUMBS localization, and cellular phenotypes can reveal the sequence of events following POGLUT1 disruption.

  • Comparative Analysis with Pathway-Specific Inhibitors:

    • Comparing POGLUT1 inhibition phenotypes with those resulting from specific inhibition of downstream pathways (e.g., Notch signaling inhibitors like γ-secretase inhibitors) helps identify which aspects of the phenotype are attributable to specific pathways.

  • Cell Type-Specific Analyses:

    • Different cell types may utilize POGLUT1 for distinct substrates. For example, muscles from POGLUT1 D233E patients showed α-dystroglycan hypoglycosylation, but this was not present in patients' fibroblasts .

    • This cell type-specific effect helps distinguish direct POGLUT1 targets from indirect consequences of pathway dysregulation.

  • Single-Cell Analysis Approaches:

    • Single-cell transcriptomics or proteomics can reveal cell-specific responses to POGLUT1 manipulation, helping to distinguish direct cellular targets from bystander effects.

  • In Vitro Reconstitution with Purified Components:

    • Reconstituting POGLUT1-dependent processes with purified components in vitro eliminates the complexity of cellular systems and allows for direct assessment of POGLUT1's activity on specific substrates.

By applying these complementary approaches, researchers can build a more accurate picture of POGLUT1's direct effects versus secondary consequences. This distinction is essential for developing targeted interventions for POGLUT1-associated diseases and for understanding the basic biology of O-glucosylation in development and homeostasis.

What are the current contradictions in the literature regarding POGLUT1's tissue-specific functions?

The literature on POGLUT1 contains several apparent contradictions regarding its tissue-specific functions and the relative importance of different substrates across developmental contexts and species. Understanding these contradictions is essential for developing a complete picture of POGLUT1 biology and for designing appropriate experimental approaches to resolve remaining questions.

Key Contradictions and Their Potential Resolutions:

  • Notch versus CRUMBS as Primary POGLUT1 Substrates:

    • Contradiction: While early studies emphasized POGLUT1's role in Notch signaling, subsequent research revealed that its earliest developmental function in mice involves CRUMBS2 rather than Notch receptors .

    • Resolution Approach: Temporal and tissue-specific analysis of POGLUT1 activity on different substrates can help establish context-dependent hierarchies of substrate importance. The finding that POGLUT1 is required for CRUMBS2 function during gastrulation (E7.5) and for Notch signaling slightly later (E8.0) suggests developmental stage-specific substrate prioritization .

  • Species-Specific Requirements for CRUMBS Glycosylation:

    • Contradiction: In Drosophila, modification of Crumbs by the POGLUT1 homolog Rumi is not required for Crumbs function, whereas mouse POGLUT1 is absolutely required for CRUMBS2 activity .

    • Resolution Approach: Comparative structural and functional studies of CRUMBS proteins across species can reveal how evolutionary divergence in protein structure may have altered dependence on glycosylation. Domain swapping experiments between Drosophila Crumbs and mouse CRUMBS2 could identify regions responsible for differential glycosylation dependence.

  • Cell Type-Specific α-Dystroglycan Hypoglycosylation:

    • Contradiction: Patients with the D233E POGLUT1 mutation exhibit muscle-specific α-dystroglycan hypoglycosylation that is not present in fibroblasts from the same patients .

    • Resolution Approach: Investigation of tissue-specific glycosylation machinery and compensatory mechanisms could explain why some tissues are more vulnerable to POGLUT1 dysfunction than others. Analysis of the expression levels of other glycosyltransferases across tissues might reveal compensatory mechanisms present in fibroblasts but absent in muscle.

  • Dual Substrate Specificity Mechanisms:

    • Contradiction: While POGLUT1 shows specificity for the C₁XSXPC₂ motif, it also exhibits O-xylosyltransferase activity toward certain EGF repeats with a diserine motif within the O-glucose consensus sequence .

    • Resolution Approach: Structural studies suggest that two distinct local conformational states are likely responsible for POGLUT1's ability to transfer both glucose and xylose . Further structural and biochemical analyses focusing on the transition between these states could clarify the mechanistic basis for dual specificity.

  • Relative Contributions to Disease Phenotypes:

    • Contradiction: While muscular dystrophy associated with the D233E mutation appears to be primarily mediated through impaired Notch signaling and satellite cell depletion , the pathogenesis of Dowling-Degos Disease type 4 is less clear and may involve multiple POGLUT1 substrates .

    • Resolution Approach: Comprehensive analysis of glycoproteomes in affected tissues from patients with different POGLUT1 mutations could reveal disease-specific alterations in substrate glycosylation patterns. Creating mutation-specific mouse models would allow for systematic assessment of tissue-specific phenotypes.

  • Relationship Between POGLUT Family Members:

    • Contradiction: The recent identification of POGLUT2 and POGLUT3 raises questions about potential functional redundancy or compensation between family members .

    • Resolution Approach: Generation of single, double, and triple knockout models for POGLUT family members, coupled with comprehensive glycoproteome analysis, could reveal the extent of overlap and compensation between these enzymes.

Resolving these contradictions requires integrated approaches combining structural biology, biochemistry, developmental biology, and disease modeling. Understanding the context-dependent functions of POGLUT1 will provide important insights into the role of protein glycosylation in development and disease.

Product Science Overview

Structure and Function

POGLUT1 is an endoplasmic reticulum (ER)-retaining glucosyltransferase. The human recombinant form of POGLUT1 is typically produced in a Chinese Hamster Ovary (CHO) cell line and is characterized by a molecular mass of approximately 44 kDa . The enzyme’s activity is measured by its ability to hydrolyze UDP-Glucose, with a specific activity of more than 2 pmol/min/μg .

Biological Significance

The addition of glucose to serine residues by POGLUT1 is a critical step in the Notch signaling pathway, which is involved in various cellular processes, including differentiation, proliferation, and apoptosis . Dysregulation of this pathway can lead to several diseases, including cancer and congenital disorders.

Applications in Research

Recombinant POGLUT1 is widely used in research to study its role in the Notch signaling pathway and its potential implications in various diseases. The recombinant protein is supplied as a 0.2 μm filtered solution in Tris and NaCl and is typically stored at -20 to -70 °C to maintain its stability .

Storage and Stability

The recombinant POGLUT1 protein is shipped with polar packs and should be stored immediately upon receipt at the recommended temperature. It is stable for six months from the date of receipt when stored at -20 to -70 °C and for three months under sterile conditions after opening .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.