The GLP1R recombinant monoclonal antibody was produced by CUSABIO using the following steps: First, B cells were separated from the spleen of the immunized animal. The immunization process involved using recombinant human GLP1R protein as the immunogen. Next, RNA was isolated from the B cells, followed by reverse transcription to synthesize cDNA. Using the cDNA as a template, the gene encoding the GLP1R antibody was extended using a degenerate primer. The GLP1R antibody was inserted into a vector. The recombinant vector was introduced into host cells through transfection, allowing the host cells to express the GLP1R recombinant monoclonal antibody. The GLP1R recombinant monoclonal antibodies were harvested from the cell culture supernatant and subsequently purified using affinity chromatography. Finally, these purified antibodies have been demonstrated to recognize the human GLP1R protein in ELISA.
CUSABIO developed the GLP1R recombinant monoclonal antibody through a rigorous process. Initially, B cells were isolated from the spleen of an immunized animal. The immunization process utilized recombinant human GLP1R protein as the immunogen. Subsequently, RNA was extracted from the B cells and reverse transcribed to synthesize cDNA. Using this cDNA as a template, the gene encoding the GLP1R antibody was amplified using a degenerate primer. The GLP1R antibody gene was then inserted into a suitable vector. The recombinant vector was introduced into host cells via transfection, enabling the expression of the GLP1R recombinant monoclonal antibody. These antibodies were harvested from the cell culture supernatant and purified using affinity chromatography. Finally, the purified GLP1R recombinant monoclonal antibodies were confirmed to recognize the human GLP1R protein through ELISA assays.
Applications : SPR
Sample type: Purified protein
Sample dilution: Immobilized 1nm
Review: CSB-RA009514MA1HU This Human GLP1R antibody runs a very stable profile in SPR affinity detection assays with high 1E-10 affinity, making it ideal for evaluating the activity of purchased proteins.
GLP1R (glucagon-like peptide-1 receptor) is a G protein-coupled receptor belonging to the secretin receptor super-family, also known as class B GPCRs . This receptor is widely distributed throughout the body, with significant expression in pancreatic islets, muscles, gastrointestinal tract, lung, liver, and pancreas . The importance of GLP1R stems from its critical role in glucose homeostasis, particularly in mediating glucose-dependent insulin secretion from pancreatic beta cells .
Beyond diabetes research, GLP1R has emerged as a significant target for studying obesity, cardiovascular diseases, non-alcoholic fatty liver disease (NAFLD), neurodegenerative conditions, musculoskeletal inflammation, and various forms of cancer . The receptor's broad tissue distribution and multifaceted physiological functions make it a crucial target for both basic research and therapeutic development.
Selection of a GLP1R antibody should be guided by your experimental requirements and the specific application. Consider these research-validated criteria:
Application | Recommended Antibody Characteristics | Dilution Range | Validation Requirements |
---|---|---|---|
Western Blot | Monoclonal with specified molecular weight detection (~53 kDa) | 1:500-1:2000 | Positive control tissue (pancreas), KO validation |
Immunohistochemistry | High specificity with low background | 1:50-1:500 | KO tissue validation, antigen retrieval optimization |
Immunofluorescence | High signal-to-noise ratio | Application-dependent | Co-localization with reporter systems |
Immunoprecipitation | High affinity for native protein | Application-dependent | Validation with mass spectrometry |
For antagonistic properties, Glp1R0017 has been extensively characterized in multiple species with demonstrated specificity . For general detection purposes, antibodies like 26196-1-AP have been validated in multiple applications and species (human, mouse, rat) . Before investing in an antibody, verify that it has been validated in your species of interest and that knockout/knockdown validation data exists to confirm specificity .
Rigorous validation is critical when working with GLP1R antibodies, as many commercially available options lack genuine specificity. A comprehensive validation strategy should include:
Genetic validation: The gold standard involves testing in GLP1R knockout tissues or cells. An authentic GLP1R antibody should show positive staining in wildtype tissue but complete absence of signal in knockout samples . This approach was successfully employed with Glp1R0017, showing specific staining in islets of Langerhans that was entirely absent in Glp1r knockout tissue .
Reporter system correlation: Validation through co-localization with GLP1R reporter systems (e.g., GLP1RCre;R26-tdRFP) can provide additional confirmation of specificity .
Cross-reactivity testing: Confirm that the antibody does not cross-react with similar receptors such as GIPR, glucagon-like peptide-2 receptor, or glucagon receptor .
Functional validation: For antagonistic antibodies, functional assays (cAMP production, insulin secretion) should demonstrate specific inhibition of GLP1R signaling .
Multi-application consistency: Consistent results across different techniques (WB, IHC, IF) with expected molecular weight and localization patterns further supports specificity .
Optimizing immunohistochemistry for GLP1R detection requires careful attention to several technical parameters:
Antigen retrieval: For optimal results with antibodies like 26196-1-AP, use TE buffer at pH 9.0 for antigen retrieval. Alternatively, citrate buffer at pH 6.0 may be employed, though comparative testing is recommended for your specific tissue .
Antibody dilution: Begin with a dilution range of 1:50-1:500 for IHC applications and optimize based on signal-to-noise ratio . The optimal dilution is typically tissue-dependent and may require titration experiments.
Positive controls: Always include pancreatic tissue as a positive control, as it contains high levels of GLP1R expression in beta cells .
Specificity controls: Include GLP1R knockout tissue as a negative control whenever possible to confirm signal specificity .
Signal amplification: For tissues with lower expression levels, consider using tyramide signal amplification or other sensitive detection methods.
Blocking optimization: Use comprehensive blocking solutions containing both serum (5-10%) and BSA (1-3%) to minimize background, particularly in tissues with high endogenous biotin or peroxidase activity.
Incubation conditions: For monoclonal antibodies like Glp1R0017, overnight incubation at 4°C often yields superior results compared to shorter incubations at room temperature.
When investigating GLP1R signaling with monoclonal antibodies, consider these critical experimental design elements:
Antibody selection based on experimental goals:
Pathway-specific readouts: Design experiments with appropriate downstream signaling markers:
Controls for signaling experiments:
Positive control: GLP-1 or GLP-1 mimetics (e.g., liraglutide) to confirm receptor activation
Negative control: Unrelated peptide hormones
Competitive control: Co-administration of GLP-1 with antagonistic antibody to demonstrate specific inhibition
Temporal considerations: GLP1R signaling has both acute and chronic components. Design experiments to capture both immediate signaling (minutes to hours) and long-term effects (hours to days) when relevant.
Cell-specific responses: Different cell types may exhibit distinct signaling outcomes downstream of GLP1R activation. Always validate findings across multiple relevant cell types when possible.
Quantification methods: Employ both qualitative (e.g., western blot) and quantitative (e.g., ELISA, reporter assays) methods to comprehensively characterize signaling responses.
The primary advantages of antagonistic monoclonal antibodies include their exceptional specificity and extended half-life. For example, Glp1R0017 has been specifically validated to antagonize GLP1R without affecting GIPR, glucagon like peptide-2 receptor, or glucagon receptor . This specificity is particularly valuable for investigations of extrapancreatic tissues where off-target effects could confound results.
Investigating GLP1R distribution in extrapancreatic tissues presents unique challenges due to generally lower expression levels compared to pancreatic beta cells. A comprehensive approach using GLP1R monoclonal antibodies should include:
Tissue preparation optimization: Different fixation protocols significantly impact GLP1R epitope preservation. For tissues with lower expression, perfusion fixation often yields superior results compared to immersion fixation.
Multiplex immunofluorescence strategies: Combine GLP1R antibody staining with cell type-specific markers to precisely identify which cell populations express the receptor. This approach is particularly valuable in heterogeneous tissues like brain, intestine, or lung.
Super-resolution microscopy: For subcellular localization studies, techniques like STORM or STED microscopy combined with high-specificity antibodies can reveal receptor distribution patterns not visible with conventional microscopy.
Parallel validation approaches: Complement antibody-based detection with alternative methods:
In situ hybridization for GLP1R mRNA
GLP1R reporter mouse models
Functional assays measuring cAMP responses to GLP-1 in isolated cells
Quantitative analysis: Implement digital image analysis workflows to quantify receptor density across different tissues and experimental conditions.
Species considerations: When investigating novel extrapancreatic GLP1R expression sites, confirm findings across multiple species using antibodies validated to work across species boundaries, such as Glp1R0017, which has been characterized in mouse, human, rat, cynomolgus monkey, and dog models .
Contradictory findings regarding GLP1R expression are common in the literature, often stemming from methodological differences. To resolve such discrepancies:
Antibody validation hierarchy: Establish a clear hierarchy of validation methods, with genetic validation (knockout controls) as the gold standard . Re-evaluate previous studies based on the rigor of their antibody validation.
Multi-method confirmation: Implement orthogonal detection methods:
Antibody-based detection (IHC/IF)
RNA-based detection (qPCR, RNA-seq, in situ hybridization)
Functional assays (cAMP response to GLP-1)
Binding studies with labeled GLP-1
Standardized reporting: Document all methodological details:
Antibody source, catalog number, and lot
Detailed staining protocol including antigen retrieval method
Image acquisition parameters
Quantification methodology
Cross-laboratory validation: When possible, exchange tissue samples or have multiple labs perform independent analyses using their established protocols.
Developmental and physiological context: GLP1R expression can vary with developmental stage, nutritional status, and disease state. Carefully control and document these variables.
Single-cell approaches: For heterogeneous tissues, single-cell RNA-seq combined with receptor localization techniques can resolve contradictions arising from bulk tissue analysis.
In vivo experiments with GLP1R antagonistic antibodies require careful design to maximize scientific insight:
Dose optimization: Establish dose-response relationships in pilot studies. For Glp1R0017, previous research demonstrated efficacy in reversing glucose-lowering effects of liraglutide during IPGTTs and reducing glucose tolerance by blocking endogenous GLP-1 in OGTTs .
Administration route: Consider the biodistribution profile based on the target tissue:
Systemic administration (IV/IP) for broad GLP1R blockade
Tissue-specific delivery methods for localized effects (e.g., intracerebroventricular for central effects)
Temporal considerations:
Experimental readouts:
Tissue-specific functional assays beyond glucose homeostasis
Molecular signaling analysis in target tissues (phosphoprotein analysis)
Physiological readouts relevant to the tissue of interest
Controls and comparisons:
IgG isotype control to account for non-specific antibody effects
Peptide antagonist (exendin 9-39) comparison group
GLP1R knockout mice as a reference for complete receptor absence
Combinatorial approaches: Consider combining GLP1R antagonistic antibodies with:
Tissue-specific knockout models for mechanistic dissection
Pharmacological modulators of downstream pathways
Environmental or dietary interventions
Technical validation: Confirm antibody engagement with the receptor in target tissues through ex vivo analysis after in vivo administration.
To ensure reliable results:
Always include positive control tissues with known GLP1R expression (e.g., pancreas) .
Whenever possible, include negative controls such as GLP1R knockout tissue .
Validate new lots of antibodies against previously established protocols.
For critical experiments, confirm findings using two different antibodies targeting distinct epitopes.
Quantitative assessment of GLP1R expression requires standardized approaches:
Immunoblotting quantification:
Immunohistochemistry/immunofluorescence quantification:
Establish standardized image acquisition parameters
Use automated cell counting and intensity measurement software
Include calibration standards in each experiment
Analyze multiple fields and tissue sections to account for heterogeneity
Flow cytometry:
Use fluorophore-conjugated GLP1R antibodies for single-cell quantification
Include calibration beads to convert fluorescence to absolute receptor numbers
Implement compensation controls for multicolor experiments
Gene expression analysis:
Complement protein detection with qPCR for GLP1R mRNA
Use absolute quantification with standard curves
Normalize to validated reference genes appropriate for the tissue/condition
Receptor binding assays:
Use radiolabeled or fluorescently labeled GLP-1 ligands
Perform competition binding with increasing concentrations of unlabeled ligand
Calculate receptor density from Bmax values
For comparative studies across tissues or conditions, it is essential to process all samples simultaneously using identical protocols to minimize technical variability.
Developing new GLP1R antibodies requires strategic planning:
Epitope selection strategies:
Target extracellular domains for applications requiring native protein recognition
For the antagonistic antibody Glp1R0017, naive phage display libraries were used in selections on biotinylated human GLP1R extracellular domains (ECDs)
Consider species conservation if cross-reactivity is desired
Avoid regions with high homology to related receptors (GIPR, GLP2R, GCGR)
Validation pipeline design:
Implement a sequential validation approach starting with binding assays
Progress to specificity testing against related receptors
For antagonistic antibodies, confirm functional effects in cAMP assays and insulin secretion studies
The single-chain variable fragment (ScFv) with highest affinity (Glp1R0017) can be converted to human IgG1 format for enhanced potency
Format optimization:
Consider developing multiple formats (full IgG, Fab, ScFv) for different applications
Engineer modifications for specific needs (fluorophore conjugation, enzyme linkage)
Cross-species reactivity testing:
Systematic testing across relevant species (mouse, rat, human, NHP)
For therapeutic development, confirm activity in both rodent models and higher species
Application-specific optimization:
For imaging: Optimize fluorophore:antibody ratio
For functional studies: Confirm consistent antagonistic/agonistic properties
For detection: Validate across multiple assay platforms (IHC, IF, WB, IP)
The development process should include rigorous benchmarking against established antibodies like Glp1R0017, which shows specific staining in the islets of Langerhans and is absent in Glp1r knockout tissue .
GLP1R antibodies offer unique tools for investigating complex receptor biology:
Conformation-specific antibodies: Developing antibodies that recognize distinct receptor conformational states could enable:
Direct visualization of active vs. inactive receptor populations
Tracking of receptor state changes in response to different ligands
Identification of tissue-specific receptor conformations
Biased signaling investigation: Antibodies that preferentially trigger or inhibit specific signaling pathways could help dissect:
G protein vs. β-arrestin pathway contributions to physiological outcomes
Tissue-specific signaling preferences
Pathway-specific therapeutic effects
Receptor trafficking studies: Antibodies targeting different receptor epitopes could illuminate:
Internalization pathways
Recycling vs. degradation sorting mechanisms
Ligand-dependent trafficking differences
Receptor complex formation: Proximity ligation assays using GLP1R antibodies could reveal:
Homodimerization patterns
Heterodimerization with other receptors (e.g., GIPR)
Interaction with signaling scaffold proteins
Single-molecule imaging approaches: Using highly specific antibodies conjugated to quantum dots or other bright fluorophores could enable:
Real-time tracking of individual receptors in living cells
Measurement of diffusion constants and confinement zones
Observation of individual signaling events
These approaches could significantly advance our understanding of how GLP1R signaling differs across tissues and potentially explain the diverse physiological effects of GLP-1-based therapies.
GLP1R antibodies have significant potential to advance therapeutic development:
Antibody-drug conjugates (ADCs):
GLP1R-targeted delivery of therapeutic payloads to specific cell populations
Beta cell-specific delivery of growth factors or protective agents
Targeted delivery to GLP1R-expressing tumors
Bispecific antibodies:
Combined targeting of GLP1R and GIPR for enhanced metabolic effects
GLP1R targeting combined with immune cell engagement for oncology applications
Dual targeting of GLP1R and inflammatory mediators for metabolic inflammation
Imaging and theranostic applications:
GLP1R antibodies conjugated to imaging agents for beta cell mass quantification
Combined diagnostic and therapeutic applications
Patient stratification based on receptor expression patterns
Tissue-selective GLP1R modulation:
Development of antibodies with tissue-biased biodistribution
Engineering antibodies that preferentially recognize tissue-specific receptor conformations
Creation of antibodies that trigger signaling pathways enriched in target tissues
Structure-guided antibody optimization:
Using structural biology insights to design antibodies with precisely tuned effects
Rational engineering of partial agonists/antagonists
Development of allosteric modulators that fine-tune receptor response to endogenous GLP-1
The extensive cross-species reactivity of antibodies like Glp1R0017 (effective in mouse, human, rat, cynomolgus monkey, and dog models) can facilitate translational research from preclinical models to clinical applications .