Expression System: Sf9 insect cells infected with recombinant baculovirus .
Purification: Proprietary chromatographic techniques, yielding sterile, endotoxin-free protein (<1 EU/μg) .
Formulation: Lyophilized in phosphate-buffered saline (pH 7.4) with 10% glycerol .
Ligand Repertoire: Binds AGEs, HMGB1, S100 proteins, and amyloid-β .
Bioactivity:
Glycosylation Impact:
In Vivo Models:
Mechanistic Studies: Used to dissect RAGE signaling in diabetes, neurodegeneration, and vascular inflammation .
Decoy Receptor Therapy: Explored for blocking AGE-mediated pathologies, though limited by rapid clearance .
Structural Biology: Aids in crystallography and ligand-binding assays .
Advanced Glycosylation End-Product Specific Receptor, Advanced Glycosylation End Product-Specific Receptor, RAGE, Receptor for Advanced Glycation End-Products Variant 20, Receptor for Advanced Glycosylation End Products, Receptor for Advanced Glycation End-Products, RAGE Isoform NtRAGE-Delta, RAGE Isoform SRAGE-Delta, SCARJ1.
Sf9, Baculovirus cells.
ADLQNITARI GEPLVLKCKG APKKPPQRLE WKLNTGRTEA WKVLSPQGGG PWDSVARVLP NGSLFLPAVG IQDEGIFRCQ AMNRNGKETK SNYRVRVYQI PGKPEIVDSA SELTAGVPNK VGTCVSEGSY PAGTLSWHLD GKPLVPNEKG VSVKEQTRRH PETGLFTLQS ELMVTPARGG DPRPTFSCSF SPGLPRHRAL RTAPIQPRVW EPVPLEEVQL VVEPEGGAVA PGGTVTLTCE VPAQPSPQIH WMKDGVPLPL PPSPVLILPE IGPQDQGTYS CVATHSSHGP QESRAVSISI IEPGEEGPTA GSVGGSGLGT LALEPKSCDK THTCPPCPAP ELLGGPSVFL FPPKPKDTLM ISRTPEVTCV VVDVSHEDPE VKFNWYVDGV EVHNAKTKPR EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK VSNKALPAPI EKTISKAKGQ PREPQVYTLP PSRDELTKNQ VSLTCLVKGF YPSDIAVEWE SNGQPENNYK TTPPVLDSDG SFFLYSKLTV DKSRWQQGNV FSCSVMHEA HNHYTQKSLS LSPGKHHHHH H.
RAGE produced in mammalian cells (like CHO cells) and insect Sf9 cells differs primarily in their N-glycosylation patterns. While both cell types can glycosylate RAGE at the same N-glycosylation sites (Asn25 and Asn81 in human RAGE), the specific glycoforms attached are substantially different. Mammalian-expressed RAGE (sRAGE CHO) predominantly carries complex-type N-glycoforms with multivalent sialylations, with approximately 70% of its N-glycans being anionic species. In contrast, RAGE expressed in Sf9 cells (sRAGE Sf9) mainly carries neutral paucimannose glycans lacking sialic acid residues . This fundamental difference in post-translational modification significantly impacts the protein's bioactivity.
Soluble RAGE exists in several isoforms that are generated through different mechanisms:
Endogenous secretory RAGE (esRAGE/RAGEv1): Produced by alternative splicing of intron 9 and removal of exon 10, resulting in a truncated protein with a unique C-terminal sequence of 16 amino acids that lacks both transmembrane and cytoplasmic domains .
Cleaved RAGE (cRAGE): Generated through proteolytic shedding of the membrane-bound full-length RAGE (FL-RAGE) by metalloproteinases, which cleave the extracellular domain from the cell surface .
Both esRAGE and cRAGE contain the intact extracellular domain capable of binding RAGE ligands, thereby acting as decoy receptors that block RAGE signaling. Interestingly, the mechanisms regulating these isoforms differ between humans and mice, with shedding being the predominant mechanism in mice while humans produce significant amounts of both esRAGE and cRAGE .
N-glycosylation is a critical determinant of RAGE functionality. The two N-glycosylation sites on RAGE's V-domain (Asn25 and Asn81 in human RAGE) significantly impact the protein's ligand binding capacity and downstream signaling. Research has demonstrated that polymorphisms enhancing N-glycosylation of RAGE increase its signaling capacity . Furthermore, RAGE enriched with anti-carboxylated glycan antibodies exhibits higher binding capacity to RAGE ligands such as high mobility group box 1 (HMGB1) and S100A family proteins .
When N-glycosylation is removed through site-directed mutagenesis (as in sRAGE CHO(N25T/N81T)), the protein loses its ability to inhibit NF-κB activation and cellular migration, demonstrating that N-glycosylation is not merely a structural modification but is essential for RAGE's functional interactions with its ligands .
For comprehensive N-glycoform characterization of sRAGE preparations, researchers should employ multiple complementary analytical techniques:
High-Performance Anion-Exchange Chromatography (HPAEC): This technique effectively resolves PNGase F-released N-glycans from sRAGE preparations. When comparing chromatographic patterns of N-glycans from sRAGE CHO and sRAGE Sf9, HPAEC reveals distinct profiles - with sRAGE CHO predominantly showing anionic species (approximately 70%) while sRAGE Sf9 displays mainly neutral species .
SDS-PAGE with Glycosidase Treatments: Treatment of sRAGE preparations with neuraminidase (which removes sialic acids from N-glycans) followed by SDS-PAGE analysis can confirm the presence of sialic acid modifications. This technique shows migration differences for sRAGE CHO after neuraminidase treatment but not for sRAGE Sf9, confirming that only the mammalian-produced variant carries sialylated complex-type N-glycoforms .
Mass Spectrometry: Although not explicitly mentioned in the search results, mass spectrometry (particularly MALDI-TOF MS and LC-MS/MS) is crucial for detailed structural characterization of specific glycan compositions and sequences.
Lectin Binding Assays: Different lectins with specific glycan-binding properties can be used to qualitatively assess glycan composition differences between sRAGE preparations.
These combined approaches provide a comprehensive profile of the carbohydrate components that distinguish different sRAGE preparations and correlate with their biological activities.
When designing experiments to compare bioactivity of different sRAGE preparations, researchers should implement the following methodological approaches:
Cellular Assays:
NF-κB Activation Assays: Utilize NF-κB-driven luciferase reporter systems in cells expressing human RAGE (such as transfected HEK293 cells). Compare how different sRAGE preparations inhibit NF-κB activation induced by RAGE ligands (HMGB1, S100B) when added in equimolar ratios. This approach has demonstrated that sRAGE CHO blocks 58.6% of HMGB1-induced and 40.2% of S100B-induced NF-κB activity, while sRAGE Sf9 shows significantly lower inhibition (19.7% and 20.4%, respectively) .
Cell Migration Assays: Use vascular smooth muscle cell (VSMC) migration assays to assess how different sRAGE preparations inhibit cellular responses to RAGE ligands.
Dosage Standardization:
Implement molar equivalent comparisons rather than weight-based dosing to account for potential differences in molecular weight due to varying glycosylation patterns.
Include unglycosylated variants (e.g., sRAGE CHO(N25T/N81T)) as controls to isolate the effects of glycosylation.
In Vivo Models:
In rodent injury models (such as carotid artery balloon injury), compare different sRAGE preparations using escalating doses to establish dose-response relationships.
Measure multiple endpoints including neointimal hyperplasia, inflammatory marker expression, and recruitment of administered sRAGE to injury sites.
Use single-dose administration to determine immediate efficacy differences between preparations.
Control Experiments:
Ensure protein quality and integrity of all preparations using identical purification procedures.
Verify that observed differences are not due to preparation artifacts by testing binding capacities to known RAGE ligands in cell-free assays.
This multi-faceted approach allows for robust comparison of bioactivity differences attributable specifically to N-glycoform variations between sRAGE preparations .
When designing in vivo therapeutic studies with different sRAGE preparations, researchers must consider several critical factors:
Dosage Determination:
Previous studies with sRAGE Sf9 required high doses (5 μg/g body weight/day in mice, 1.25 μg/g body weight/day in rats) administered daily for 6-7 days to achieve therapeutic effects in vascular injury models .
Comparative studies should include a wide dose range, as sRAGE CHO has demonstrated efficacy at dramatically lower doses (3 ng/g in a single administration) compared to sRAGE Sf9 .
Include dose-response analyses to determine minimum effective doses for each preparation.
Administration Regimen:
Species-Specific Considerations:
Account for species differences in RAGE biology - humans and mice utilize different mechanisms to regulate and generate sRAGE isoforms .
Mice express a distinct transcript variant (mRAGEv4) not found in humans that is resistant to shedding .
These differences may significantly impact how RAGE signaling operates in rodent models versus human disease.
Potential Immunogenicity:
Insect cell-derived paucimannose glycoforms can be immunogenic in mammals, potentially confounding therapeutic outcomes in longer-term studies .
For chronic disease models, consider immunogenicity testing of different sRAGE preparations.
Monitor inflammatory markers that might indicate immune responses to the administered proteins.
Mechanistic Readouts:
Include analyses of tissue recruitment of administered sRAGE to determine if biodistribution differs between preparations.
Measure both disease outcomes and molecular signaling pathways (like NF-κB activation) in target tissues.
These considerations are essential for generating translatable research findings that account for the distinct biological properties of different sRAGE preparations .
The therapeutic potentials of sRAGE CHO and sRAGE Sf9 in vascular injury models differ dramatically:
These comparative results demonstrate that the N-glycoform of sRAGE is a key determinant of its therapeutic efficacy, with mammalian cell-produced sRAGE showing dramatically enhanced potency compared to insect cell-produced variants .
Developing human therapeutic applications of sRAGE through N-glycoform engineering requires attention to several key considerations:
These considerations highlight the importance of glycobiology in the development of sRAGE therapeutics and suggest that glycoengineering approaches will be central to maximizing clinical potential .
Addressing species-specific differences in RAGE biology presents significant challenges for translational research. Researchers should implement the following strategies:
Characterize Species-Specific RAGE Isoforms:
Humans and mice utilize different mechanisms to generate soluble RAGE isoforms .
Mice express the mRAGEv4 transcript variant that lacks exon 9 and is resistant to shedding, whereas this variant is not found in humans .
Comprehensive characterization of species-specific RAGE isoform expression patterns in targeted tissues is essential before extrapolating findings across species.
Develop Humanized Models:
Consider using humanized mouse models expressing human RAGE variants to better predict human responses.
Alternatively, employ ex vivo human tissue systems or organoids to validate findings from animal models in human tissues.
Comparative Signaling Analysis:
Investigate whether downstream signaling pathways activated by RAGE are conserved between species.
The RAGE intracellular domain (RAGE-ICD) plays a critical role in signal transduction, particularly in regulating cell migration . Researchers should determine if these mechanisms are conserved across species.
Adjustment of Therapeutic Dosing:
Due to species differences in RAGE biology, dosing regimens established in rodent models may not directly translate to humans.
Calculations should account for differences in:
a) RAGE isoform distribution
b) Constitutive shedding rates (higher in murine FL-RAGE than human FL-RAGE)
c) Tissue-specific expression patterns
Biomarker Development:
Develop species-appropriate biomarkers that reflect RAGE activity.
For human studies, consider the ratio of different sRAGE isoforms (esRAGE vs. cRAGE) as potential biomarkers, since these may not be relevant in mouse models.
Mechanistic Validation Across Species:
Validate key mechanistic findings in multiple species, including human tissues or cells whenever possible.
Focus on conserved aspects of RAGE biology while acknowledging species-specific differences.
By systematically addressing these species differences, researchers can develop more robust translational approaches that account for the unique features of human RAGE biology .
To identify the specific N-glycan structures in sRAGE that are most critical for enhanced bioactivity, researchers should employ a multi-faceted approach combining glycoanalytical techniques with functional assays:
Glycoform Fractionation and Analysis:
Use lectin affinity chromatography to separate sRAGE preparations into fractions with different glycan compositions.
Analyze each fraction using high-performance anion-exchange chromatography (HPAEC) and mass spectrometry to characterize specific glycan structures .
Test each glycoform-enriched fraction for bioactivity in standardized assays (NF-κB inhibition, VSMC migration) to correlate specific structures with function.
Site-Specific Glycan Analysis:
Perform site-specific glycoproteomic analysis using tandem mass spectrometry to determine which glycans are attached to each of the two N-glycosylation sites (Asn25 and Asn81).
Create single-site glycosylation mutants (N25 only or N81 only) to determine if glycosylation at specific sites has differential effects on bioactivity.
Glycoengineering Approaches:
Express sRAGE in CHO cell lines with modified glycosylation machinery (glycosylation-deficient CHO mutants or genetically engineered lines with human glycosyltransferases).
Generate sRAGE variants with defined glycan structures by using in vitro enzymatic remodeling of purified proteins with specific glycosidases and glycosyltransferases.
Test these defined glycoforms in functional assays to establish structure-activity relationships.
Molecular Dynamics Simulations:
Use computational approaches to model how different glycan structures might affect:
a) Protein conformation and stability
b) Ligand binding interface accessibility
c) Receptor-ligand interaction dynamics
Binding Studies with Defined Glycoforms:
Use surface plasmon resonance or bio-layer interferometry to measure binding kinetics of different glycoforms with RAGE ligands (HMGB1, S100 proteins).
Compare on/off rates and binding affinities to identify glycan features that enhance ligand interactions.
In Vivo Structure-Function Studies:
Test defined glycoforms in animal models at equivalent molar doses.
Monitor both efficacy (e.g., reduction in neointimal hyperplasia) and pharmacokinetic parameters.
Use imaging techniques with labeled sRAGE variants to track tissue distribution and retention.
Through this comprehensive approach, researchers can identify the specific structural features of complex-type N-glycans (branching patterns, terminal sialylation, core modifications) that confer enhanced bioactivity to sRAGE and use this information to develop optimized therapeutic candidates .
Emerging approaches for studying RAGE signaling regulation through different isoforms in human disease contexts encompass several innovative methodologies:
Isoform-Specific Detection Systems:
Develop antibodies or aptamers that specifically recognize different RAGE isoforms (FL-RAGE, esRAGE, cRAGE) to enable precise quantification in human samples.
Use digital PCR or targeted RNA-seq to accurately measure transcript variants encoding different RAGE isoforms in disease tissues.
These approaches can help establish isoform ratios as potential biomarkers for disease progression or treatment response.
CRISPR-Based Functional Genomics:
Employ CRISPR/Cas9 genome editing to introduce specific mutations that affect:
a) Alternative splicing sites that regulate esRAGE production
b) Proteolytic cleavage sites that affect cRAGE generation
c) N-glycosylation sites that influence ligand binding and signaling
Create isogenic cell lines differing only in RAGE isoform expression to isolate isoform-specific effects.
Single-Cell Multi-Omics:
Use single-cell RNA sequencing combined with protein analysis to understand cell-type-specific expression patterns of RAGE isoforms in diseased tissues.
This approach can reveal how cellular heterogeneity in RAGE isoform expression contributes to disease pathophysiology.
Domain-Specific Functional Analysis:
Advanced Imaging Techniques:
Use super-resolution microscopy or proximity ligation assays to visualize interactions between different RAGE isoforms and their ligands at the subcellular level.
Implement intravital imaging to monitor RAGE signaling dynamics in vivo in disease models.
Integration of Glycomics with Proteomics:
Develop integrated analytical platforms that simultaneously characterize protein expression, glycosylation patterns, and functional activities of RAGE isoforms.
Apply these platforms to analyze clinical samples from patients with RAGE-associated diseases to identify disease-specific patterns.
Humanized Disease Models:
Create models that recapitulate human-specific aspects of RAGE biology, addressing the limitations of traditional animal models that exhibit different RAGE isoform regulation mechanisms .
Patient-derived organoids or tissue constructs may offer more translatable systems for studying human RAGE signaling.
These emerging approaches offer promising avenues to dissect the complex regulation of RAGE signaling through its various isoforms in human diseases, potentially leading to more precise therapeutic targeting strategies .
The Advanced Glycosylation End Product-Specific Receptor (AGER), commonly known as the Receptor for Advanced Glycation End Products (RAGE), is a member of the immunoglobulin superfamily of cell surface receptors. This receptor plays a crucial role in various physiological and pathological processes, including inflammation, development, and homeostasis .
RAGE is a 35 kilodalton transmembrane receptor that was first characterized in 1992. It is known for its ability to bind advanced glycation end products (AGEs), which are nonenzymatically glycosylated proteins that accumulate in vascular tissues during aging and at an accelerated rate in diabetes . The receptor has a broad ligand repertoire, including AGEs, S100 proteins, high-mobility group box 1 protein (HMGB1), amyloid beta, nucleic acids, phospholipids, and glycosaminoglycans .
RAGE is implicated in the pathogenesis of various diseases, such as diabetes, Alzheimer’s disease, vascular complications, neurodegenerative disorders, and cancers. The interaction between RAGE and its ligands results in the activation of pro-inflammatory gene expression, contributing to the progression of these diseases .
The human recombinant form of RAGE is often expressed in Sf9 cells, which are derived from the fall armyworm Spodoptera frugiperda. Sf9 cells are commonly used in the baculovirus expression system, which is a powerful tool for producing recombinant proteins. This system allows for high-level expression and proper post-translational modifications, making it suitable for producing functional human proteins .