The glucagon-like peptide-1 receptor (GLP1R) is a class B G protein-coupled receptor (GPCR) encoded by the GLP1R gene located on chromosome 6p21.2 in humans . It plays a central role in glucose homeostasis, appetite regulation, and metabolic health by mediating the effects of the incretin hormone GLP-1 (glucagon-like peptide-1). GLP1R is expressed in pancreatic β-cells, neurons, adipose tissue, and cardiovascular systems, enabling diverse physiological functions .
GLP1R Human regulates:
Insulin Secretion:
Glucagon Suppression:
Appetite Regulation:
Cardiovascular Effects:
GLP1R agonists (GLP-1RAs) are first-line therapies for type 2 diabetes (T2D) and obesity:
Reduced MACE: GLP-1RAs lower myocardial infarction, stroke, and mortality in T2D patients .
Vascular effects: Coronary microvascular dilation independent of systemic blood pressure .
Structural Insights:
Genetic Studies:
Novel Tools:
The glucagon-like peptide-1 receptor (GLP1R) is a protein found on certain brain cells and pancreatic beta cells. When activated, it triggers a cascade that ultimately increases intracellular cAMP levels. In humans, the GLP1R gene, located on chromosome 6, is responsible for its production. GLP1R plays a crucial role in regulating blood sugar by stimulating insulin release. It belongs to the glucagon receptor family, a class of G protein-coupled receptors. Structurally, GLP1R consists of two domains: a transmembrane domain (TMD) that binds to a specific region of GLP-1, and an extracellular domain (ECD) that binds to another region of GLP-1. The TMD houses a specific arrangement of amino acids that influence the receptor's signaling, while the ECD and areas surrounding the TMD contribute to the activation of specific signaling pathways.
This product consists of the GLP1R protein from humans, produced in E. coli bacteria. It is a single chain of 132 amino acids, including a 10 amino acid tag, and has a molecular weight of 15.55 kDa. This protein is not glycosylated.
This product is supplied as a freeze-dried powder. The GLP1R protein was filtered through a 0.4 micrometer filter and freeze-dried from a solution containing 0.5 mg/ml GLP1R in a 50 mM acetate buffer at a pH of 4.0.
To prepare the product for use, it is recommended to first reconstitute the freeze-dried powder in 100 mM acetate buffer to a concentration of approximately 0.5 mg/ml. Any further dilutions should be done using a buffer solution that contains protein.
For long-term storage, the lyophilized protein should be kept at -20°C. After reconstituting the protein, it is recommended to divide it into smaller portions to avoid repeatedly freezing and thawing the product. Once reconstituted, the protein can be stored at 4°C for up to two weeks without degradation.
Purity is greater than 90% as determined by SDS-PAGE analysis.
GLP1, GLP2, GRPP
Escherichia Coli.
MKHHHHHHAS RPQGATVSLW ETVQKWREYR RQCQRSLTED PPPATDLFCN RTFDEYACWP DGEPGSFVNV SCPWYLPWAS SVPQGHVYRF CTAEGLWLQK DNSSLPWRDL SECEESKRGE RSSPEEQLLF LY
The glucagon-like peptide-1 receptor (GLP1R) is a class B1 G-protein coupled receptor (GPCR) expressed in various tissues throughout the human body. It is prominently expressed in the pancreas (particularly in β-cells), brain (especially in the basolateral amygdala and hypothalamic regions), and in multiple tissues outside the central nervous system . In the pancreatic islets, studies using validated GLP-1R antibodies have demonstrated that more than 90% of β-cells express GLP1R protein, though single-cell RNA sequencing data suggests heterogeneity in gene expression levels . GLP1R's endogenous ligand, glucagon-like peptide-1 (GLP-1), is fully conserved across mammals and plays crucial roles in energy homeostasis .
GLP1R signaling exhibits tissue-specific effects that likely reflect differences in downstream signaling pathways. In pancreatic β-cells, GLP1R activation primarily stimulates insulin secretion under hyperglycemic conditions through the "incretin effect" . In the central nervous system, particularly in the hypothalamus and amygdala, GLP1R signaling regulates satiety, food intake, and has neuroprotective functions . These diverse physiological responses may result from differences in the spatial concentration gradients of GLP-1 in target tissues and variations in signaling partners across different cell types . Interestingly, the apparent EC50 for GLP1R activation differs markedly between direct receptor conformational changes and downstream cAMP generation, suggesting complex signal transduction mechanisms across tissues .
Recent advances have yielded sophisticated tools for studying GLP1R dynamics. Most notably, GLPLight1 is a novel genetically encoded fluorescent sensor that provides real-time visualization of GLP1R conformational activation. This sensor was engineered by inserting a circularly permuted green fluorescent protein (cpGFP) into the third intracellular loop (ICL3) of the human GLP1R, between residues K336 and T343 . GLPLight1 offers several advantages:
High sensitivity with a maximum ΔF/F response of approximately 456% to GLP-1 application in primary neurons
Spectral properties similar to other GPCR-sensors with peak excitation around 500nm and emission at 512nm
Accurate reporting of receptor conformational changes in response to various pharmacological ligands
Ability to detect activation by clinical GLP1R agonists including liraglutide and semaglutide
This tool allows unprecedented spatiotemporal resolution for investigating GLP1R dynamics under various physiological and pharmacological conditions, significantly advancing our understanding of this receptor's activation mechanisms.
Creating humanized GLP1R models involves precise genetic engineering approaches. A successful strategy involves generating knock-in mice that express the human GLP1R from the murine Glp-1r locus . The key methodology includes:
Designing a targeting construct containing the human GLP1R sequence flanked by LoxP sites
Including a C-terminal FLAG epitope tag to facilitate biochemical studies
Using homologous recombination to integrate the construct into the murine Glp-1r locus
Confirming proper expression through physiological testing (glucose tolerance, insulin secretion)
This approach ensures the human receptor is expressed in physiologically relevant tissues and at native levels. The resulting model exhibits normal glucose metabolism, insulin secretion, and gastric transit responses similar to wild-type littermates, while enabling human-specific drug testing . Additionally, the FLAG epitope enables biochemical studies through immunoprecipitation and mass spectrometry to identify potential GLP1R interacting proteins, advancing our understanding of receptor regulation mechanisms.
Developing specific antibodies for human GLP1R has been historically challenging due to the complex structure of this class B GPCR and the high sequence conservation between species. Effective strategies include:
Using humanized GLP1R mouse models as validation tools for antibody specificity
Creating global Glp-1r−/− animals (by crossing hGLP-1R lines with Rosa26Cre mice) as negative controls
Validating antibody specificity through comparative immunohistochemistry across:
This comprehensive validation approach has successfully identified human GLP1R-specific antibodies that reliably detect the receptor in both human pancreas and humanized mouse models . When developing new antibodies, targeting unique extracellular epitopes of human GLP1R that differ from murine sequences can improve specificity, though extensive validation across multiple tissue types remains essential.
Detailed kinetic analysis of GLP1R activation reveals important mechanistic insights about this class B1 GPCR. The GLPLight1 sensor has demonstrated that GLP1R exhibits slower activation kinetics compared to class A GPCRs . Key methodological approaches include:
Real-time fluorescence measurements using GLPLight1 in various cell types
Comparison of GLP1R activation kinetics to those of class A GPCR-based sensors
Manipulation of activation conditions through photocaged ligands (e.g., photo-GLP1)
These studies reveal that GLP1R activation involves a distinct two-step binding mechanism:
Initial "capture" of the ligand by the receptor's extracellular domain (ECD)
Subsequent ligand insertion into the receptor binding pocket to initiate signaling
This mechanism explains the slower activation kinetics observed for GLP1R compared to class A GPCRs. Interestingly, pre-incubation with an inactive form of GLP-1 peptide (photo-GLP1) significantly influences receptor activation kinetics, likely because the inactive peptide occupies the receptor's ECD and facilitates the subsequent activation step . These findings provide critical insights for rational drug design targeting GLP1R.
The observed discordance between GLP1R gene expression (scRNASeq data) and protein detection (antibody-based methods) necessitates careful experimental approaches . To address these discrepancies, researchers should:
Implement complementary methods for GLP1R detection:
FACS coupled with quantitative RT-PCR
Validated GLP-1R antibodies
Flow cytometry
GLP1R reporter systems
Consider technical limitations of each approach:
scRNASeq may miss low-abundance transcripts
Antibody specificity must be rigorously validated
Reporter systems may not perfectly mirror endogenous expression
Examine both transcriptional and post-transcriptional regulation:
Assess mRNA stability
Evaluate protein translation efficiency
Measure receptor turnover rates
A comprehensive experimental design should incorporate multiple detection methods and include appropriate controls to avoid misinterpretation. When studying GLP1R in pancreatic β-cells, for instance, researchers should not rely solely on transcriptomic data, as this may underestimate the proportion of GLP1R-positive cells .
Developing photocaged GLP-1 derivatives (like photo-GLP1) for precise temporal control of GLP1R activation requires careful molecular design considerations:
Strategic placement of photolabile protecting groups:
Must block peptide activity until light exposure
Should not interfere with initial receptor binding
Must be efficiently removed upon illumination
Characterization of uncaging efficiency:
Determine optimal wavelength and intensity
Measure kinetics of peptide activation
Assess completeness of uncaging
Verification of biological activity:
Compare EC50 of uncaged peptide to native GLP-1
Confirm specificity for GLP1R over related receptors
Measure downstream signaling activation
Optimization for combined use with optical sensors:
When used in conjunction with the GLPLight1 sensor, photocaged GLP-1 can facilitate all-optical control and visualization of GLP1R activation, creating powerful experimental paradigms for studying receptor dynamics with unprecedented precision .
The GLPLight1 sensor provides significant advantages for pharmacological screening of GLP1R-targeting compounds. Implementation strategies include:
High-throughput fluorescence-based assays:
Real-time detection of GLP1R conformational changes
Direct optical readout with high sensitivity
Ability to measure both agonist and antagonist effects
Detailed pharmacological characterization:
Determination of ligand potency (EC50) values
Assessment of ligand specificity against related receptors
Measurement of activation and deactivation kinetics
Comparative analysis of different compound classes:
Peptide-based GLP1R agonists (GLP-1, exendin-4)
Clinical GLP1RAs (liraglutide, semaglutide)
Novel small molecule modulators
GLPLight1 has already demonstrated utility in characterizing the pharmacological profile of various GLP1R ligands, showing responses to clinical drugs like liraglutide and semaglutide comparable to native GLP-1 . The sensor also enables detection of antagonist activity, as demonstrated by its response to exendin-9, which partially reversed GLP-1 activation within minutes . This direct readout of receptor conformational changes provides higher temporal resolution than conventional downstream signaling assays, accelerating drug discovery efforts for GLP1R-targeted therapeutics.
Addressing species differences in GLP1R structure and function requires specialized experimental approaches:
Humanized mouse models expressing human GLP1R:
Comparative pharmacology studies:
Side-by-side testing of compounds on human versus rodent GLP1R
Assessment of species-specific differences in binding affinity
Evaluation of potential differences in signaling bias
Structure-guided drug design:
Targeting conserved binding pockets to minimize species differences
Focusing on human receptor conformations for rational drug design
Accounting for species-specific receptor-ligand interactions
Humanized GLP1R mice have demonstrated particular value for testing novel non-peptide, orally available molecules targeting GLP1R. Unlike peptide therapeutics that make numerous receptor contacts, small molecules make fewer contacts, thus increasing the impact of amino acid differences across species . These models provide more predictive preclinical data for human clinical outcomes while maintaining physiologically relevant expression patterns.
Identifying GLP1R interacting proteins in physiologically relevant contexts requires sophisticated biochemical approaches:
Epitope-tagged GLP1R mouse models:
Affinity purification strategies:
Anti-FLAG immunoprecipitation from specific tissues (islets, lung, stomach)
Gentle solubilization conditions to preserve protein-protein interactions
Stringent washing procedures to minimize false positives
Mass spectrometry-based identification:
Quantitative proteomics to identify specific interactors
Comparison between different activation states
Validation of hits through reciprocal co-immunoprecipitation
The FLAG-tagged humanized GLP1R mouse model provides a valuable tool for these approaches, enabling biochemical studies through immunoprecipitation and mass spectrometry analyses from physiologically relevant tissues . This strategy overcomes limitations of previous approaches that relied on overexpressed receptors in heterologous systems, allowing identification of authentic interacting partners that regulate GLP1R function in vivo.
Distinguishing between GLP1R conformational changes and downstream signaling requires complementary methodological approaches:
Direct conformational change detection:
Proximal signaling measurements:
MiniGs recruitment assays: Directly measure G protein coupling to the activated receptor
Arrestin recruitment assays: Detect receptor interaction with arrestin proteins
Downstream signaling quantification:
cAMP assays: Measure second messenger production following receptor activation
Calcium imaging: Detect changes in intracellular calcium levels
ERK phosphorylation: Assess activation of downstream kinase pathways
Comparative analysis reveals important insights. For example, GLPLight1 fluorescence response and miniGs recruitment to hmGLP1R show similar EC50 values for GLP-1, while the EC50 for cAMP response is approximately three orders of magnitude lower . This discrepancy highlights fundamental differences between direct receptor conformational changes and enzymatically amplified downstream signals, suggesting that GLP-1 may elicit different functional responses based on local concentration gradients in target tissues .
The distinctive two-domain structure of class B1 GPCRs like GLP1R necessitates specialized experimental approaches for studying ligand binding:
Domain-specific binding studies:
Isolated extracellular domain (ECD) binding assays
Transmembrane domain (TMD) interaction assessments
Full-length receptor studies for complete binding profiles
Two-step binding mechanism characterization:
Structure-guided mutation approaches:
Alanine scanning of key binding residues
Domain-specific mutations to isolate effects
Creation of chimeric receptors to map binding regions
Cutting-edge approaches for studying GLP1R dynamics in intact systems include:
In vivo application of optical sensors:
Tissue-specific manipulation of humanized GLP1R models:
Advanced imaging techniques for endogenous GLP-1 and GLP1R dynamics:
Multiplex imaging to simultaneously track ligand and receptor
High-resolution approaches to visualize subcellular receptor trafficking
Longitudinal imaging to monitor adaptive changes in receptor expression
While GLPLight1 has shown promise in cultured cells, its potential for in vivo application remains to be fully explored . The high sensitivity and lack of interference with intracellular signaling suggest it could be employed to investigate endogenous GLP-1 dynamics directly in living systems, though successful in vivo implementation will require careful optimization of expression systems and imaging approaches .
Single-cell approaches offer powerful insights into GLP1R heterogeneity within pancreatic islets:
Integrated multi-omics analysis:
Combined single-cell RNA sequencing and proteomics
Correlation of GLP1R transcript and protein levels in the same cells
Assessment of post-transcriptional regulatory mechanisms
Functional heterogeneity characterization:
Single-cell calcium imaging in response to GLP1R activation
Patch-clamp electrophysiology of identified GLP1R-positive and negative cells
Insulin secretion measurements from sorted cell populations
Spatial transcriptomics and proteomics:
Mapping GLP1R expression patterns within intact islets
Correlating with anatomical location and microenvironment
Identifying potential neighbor effects on receptor expression
This approach is particularly important given the discordance between GLP1R gene and protein expression observed in β-cells . While scRNASeq data suggests heterogeneous expression patterns, protein-level detection indicates more uniform expression . Integrated single-cell approaches could resolve this contradiction and provide insights into the functional significance of any true heterogeneity in GLP1R expression or activity among β-cells.
Approach | Advantages | Limitations | Key Applications |
---|---|---|---|
scRNASeq | Unbiased transcriptome-wide analysis, High throughput | May miss low-abundance transcripts, No protein information | Cell type identification, Transcriptional heterogeneity |
Flow cytometry with GLP1R antibodies | Direct protein detection, Quantitative, High throughput | Requires validated antibodies, Limited to surface proteins | Protein expression quantification, Cell sorting |
GLPLight1 sensor | Real-time activity detection, Functional readout, High sensitivity | Requires genetic modification, Expression variability | Functional heterogeneity, Drug response studies |
Spatial transcriptomics | Preserves tissue context, Maps expression patterns | Lower resolution than scRNASeq, Technical challenges | Anatomical distribution, Niche effects |
The separation of GLP1R conformational changes from downstream signaling cascades has significant implications for drug development:
Biased agonist development potential:
Targeting specific receptor conformations that activate particular signaling pathways
Designing drugs that favor therapeutic effects while minimizing side effects
Creating compounds with unique pharmacodynamic properties
Understanding of structure-activity relationships:
Correlation between receptor conformational changes and specific signaling outcomes
Identification of critical molecular determinants for pathway-specific activation
Rational design of compounds with precise signaling profiles
Novel therapeutic strategies:
Development of allosteric modulators that influence specific aspects of receptor function
Creation of partial agonists with defined efficacy profiles
Design of compounds with altered receptor desensitization or trafficking properties
The GLPLight1 sensor, which shows minimal coupling to endogenous signaling pathways even at high GLP-1 concentrations, provides a valuable tool for these approaches . The significant differences observed between the EC50 for receptor conformational changes and downstream cAMP responses (approximately three orders of magnitude) further highlight the potential for developing compounds that differentially affect these aspects of receptor function . This could lead to more precise and effective GLP1R-targeted therapeutics with improved specificity and reduced side effects.
GLP-1R is composed of two main domains: an extracellular domain (ECD) that binds the C-terminal helix of GLP-1, and a transmembrane domain (TMD) that binds the N-terminal region of GLP-1 . The receptor is primarily found on beta cells of the pancreas and neurons in the brain . It plays a significant role in enhancing insulin secretion in response to elevated blood glucose levels .
The receptor is involved in several biological processes, including: