GLP1R monoclonal antibody

Shipped with Ice Packs
In Stock

Description

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.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

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.

Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days after receiving them. Delivery times may vary depending on the purchase method and destination. For specific delivery timeframes, please consult your local distributors.
Synonyms
GLP1R monoclonal antibody
Target Names
Uniprot No.

Target Background

Function
The Glucagon-like peptide 1 receptor (GLP-1R) is a G-protein coupled receptor responsible for recognizing glucagon-like peptide 1 (GLP-1). Upon ligand binding, the receptor initiates a signaling cascade that activates adenylyl cyclase, leading to an increase in intracellular cyclic adenosine monophosphate (cAMP) levels. This signaling pathway plays a crucial role in regulating insulin secretion in response to GLP-1.
Gene References Into Functions
  1. This research explores the role of GLP-1 and its receptor in the spinal cord, focusing on the expression of GLP-1R and the innervation of PPG neurons. It examines the potential therapeutic benefits of activating GLP-1R. PMID: 29329976
  2. This study provides evidence that zfGIP's insulinotropic action in mammalian systems involves activation of both the GLP-1 and the GIP receptors, but not the glucagon receptor. PMID: 29157578
  3. Clinical trials conducted on non-diabetic patients with neurodegenerative disorders demonstrate neuroprotective effects following administration of GLP-1 receptor agonists, highlighting that these effects are independent of blood glucose levels. PMID: 29412810
  4. While clinical trials involving GLP-1 receptor agonists (GLP-1RA) for type 2 diabetes mellitus (T2DM) may not have been specifically designed to investigate bone fracture, initial results suggest that these agonists may not exacerbate the abnormal bone quality often observed in T2DM. PMID: 29412811
  5. This research delves into recent findings concerning the signaling and trafficking of the GLP-1R in pancreatic beta cells. It explores how targeting specific signaling pathways mediated by the receptor, like cAMP generation versus beta-arrestin recruitment and ERK1/2 activation, could lead to the development of novel GLP-1R analogues with improved clinical efficacy. PMID: 29412835
  6. This study suggests a potential mechanism for GLP-1R agonist-induced cardioprotection in type 2 diabetes, as increases in fatty acid oxidation and decreases in glucose oxidation are frequently observed in the hearts of animals and humans with T2DM. PMID: 29412838
  7. In a Han Chinese population of type 2 diabetic patients, certain variations in the GLP-1R gene were associated with a reduced risk of developing coronary artery disease. PMID: 30271789
  8. Activation of endogenous GLP-1 has been linked to sepsis in patients with type 2 diabetes. PMID: 29334697
  9. Increased GLP-1R innervation in the intestines of individuals with inflammatory bowel disease (IBD) could potentially mediate enhanced visceral afferent signaling, presenting a peripheral target for therapeutic intervention. PMID: 29813107
  10. GLP1R mRNA transcripts, encompassing the entire open reading frame, were detected in all four cardiac chambers of 15 human hearts, at levels comparable to those observed in the human pancreas. PMID: 29444223
  11. Low active GLP-1 secretion has been associated with hypertriglyceridemia. PMID: 29135069
  12. This study investigated the role of glucagon and glucagon-like peptide-1 (GLP-1), hormones produced by alpha cells, in insulin secretion in INS-1 cells, a beta cell line. The study found that co-treatment with glucagon and exendin-4 (Ex-4), a GLP-1 receptor agonist, additively increased glucose-stimulated insulin secretion in INS-1 cells. PMID: 29725251
  13. Genetic association studies in a Korean population suggest that SNPs in PAX4 and GLP1R are associated with type 2 diabetes (T2D). The PAX4 Arg192His variant increased the risk of T2D, while the GLP1R Arg131Gln variant decreased the risk. (PAX4 = paired box 4 protein; GLP1R = glucagon-like peptide 1 receptor) PMID: 29941447
  14. LINC01121 acts as a tumor promoter by engaging in translational repression of GLP1R and inhibiting the cAMP/PKA signaling pathway. PMID: 29843149
  15. The cryo-EM structure of the human GLP-1 receptor in complex with the G protein-biased peptide exendin-P5 and a Galphas heterotrimer has been determined at a global resolution of 3.3 Å. PMID: 29466332
  16. This research demonstrates that exendin-4 induces a partial reduction in triglycerides in steatotic hepatocytes within 12 hours through GLP-1 receptor-mediated activation of protein kinase A. The reduction in hepatocyte triglyceride accumulation is likely driven primarily by downregulation of lipogenesis and upregulation of beta-oxidation of free fatty acids. PMID: 28707223
  17. Data, including findings from studies in knockout mice, suggest that MIR204 (highly enriched in beta-cells) directly targets the 3'-untranslated region of GLP1R, thereby down-regulating GLP1R expression in beta-cells. Further studies were conducted in primary human and mouse beta-cells and in a rat insulinoma cell line. PMID: 29101219
  18. This research suggests that GLP1R signaling in pancreatic beta-cells leading to insulin secretion involves interactions between GLP1R and HIP1, SNX1, and SNX27. HIP1 appears to regulate the coupling of cell surface GLP1R activation with endocytosis, while SNX1 and SNX27 seem to control the balance between GLP1R plasma membrane recycling and lysosomal degradation. PMID: 29284659
  19. The GLP-1R was found to be abundantly expressed in various brain regions, including the septal nucleus, hypothalamus, and brainstem. PMID: 29095968
  20. This study reports the novel finding that human epicardial adipose tissue expresses both GLP-1R and GLP-2R genes. PMID: 28514806
  21. Changes in GLP-1 levels were observed to be associated with weight loss in newly diagnosed Chinese diabetic patients receiving acarbose. PMID: 27717194
  22. This study demonstrates that GLP1R overexpression significantly reduces proliferation, migration, and cytokine release in airway smooth muscle cells from COPD patients. This reduction was accompanied by a significant increase in ABCA1 expression levels. The findings suggest that GLP1R may be a potential therapeutic target for COPD treatment. PMID: 28560433
  23. IL-33, GLP-1R, and CCL20 are deregulated in human inflammatory bowel disease. GLP-1 receptor agonists upregulate IL-33, mucin 5b, and CCL20 in murine Brunner's glands. These agonists impact gut homeostasis in both proximal and distal parts of the gastrointestinal tract. PMID: 27542128
  24. This research shows that exendin-4 (Ex-4) could attenuate breast cancer cell proliferation through activation of the glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) and subsequent inhibition of nuclear factor kappaB (NF-kappaB) activation. PMID: 29045658
  25. The crystal structure of the full-length GLP-1 receptor bound to a truncated peptide agonist has been determined. PMID: 28562585
  26. The crystal structures of the human GLP-1R transmembrane domain in complex with two different negative allosteric modulators, PF-06372222 and NNC0640, have been determined at resolutions of 2.7 Å and 3.0 Å, respectively. PMID: 28514449
  27. Data indicate that pancreatic GLP1R levels are highest in insulin-secreting cells. Immunostaining revealed the highest intensity of GLP1R in beta-cells in pancreatic tissues obtained from organ-donor cadavers with type 2 diabetes. PMID: 28094469
  28. This study reports the successful purification of glucagon-like peptide-1 (GLP-1) receptor (GLP1R) in nanodiscs, which demonstrated binding to GLP-1 and exendin-4, activating Gs protein. PMID: 28609478
  29. In real-world clinical settings, dapagliflozin, when added to patients with T2DM already treated with GLP1-R agonists, induced a further significant, albeit modest, improvement in A1C and additional weight loss. PMID: 28077257
  30. This research analyzed the biological binding site of the exendin-4 peptide within the N-terminal domain of the intact human glucagon-like peptide-1 receptor. PMID: 28283573
  31. GLP1-R may represent a novel therapeutic target for addressing bronchial hyperresponsiveness. PMID: 27447052
  32. The findings of this study indicate that GLP-1R is widely expressed throughout the human hypothalamus. Reduced GLP-1R expression in the paraventricular nucleus (PVN) and infundibular nucleus (IFN) of T2DM patients may be associated with dysregulation of feeding behavior and glucose homeostasis in type 2 diabetes mellitus. PMID: 26672638
  33. This study reveals that exenatide significantly improves coronary endothelial function in patients with newly diagnosed type 2 diabetes. The effect is likely mediated through activation of the AMPK/PI3K-Akt/eNOS pathway, involving a GLP-1R/cAMP-dependent mechanism. PMID: 27072494
  34. A higher probability of achieving A1c goal levels was observed when GLP-1R agonists were initiated. PMID: 28230449
  35. Immunohistochemistry of human ileum tissues revealed co-localization of TAS2R38 with glucagon-like peptide 1 (GLP-1) in enteroendocrine L-cells. PMID: 27208775
  36. This research suggests that three conserved positively charged residues located at the extracellular ends of transmembrane helices 3, 4, and 5 of GLP1R are crucial for high-affinity agonist binding and conformational transitions linked to pleiotropic effector coupling through stabilization of extracellular domains. PMID: 27569426
  37. The rate of homologous desensitization and internalization of the GLP-1R has been determined using a transgenic cell line system. PMID: 28035964
  38. In cells expressing the glucagon-like peptide receptor (GLP-1R), small molecule agonists induced cAMP production but did not cause intracellular Ca2+ accumulation, ERK phosphorylation, or hGLP-1R internalization. PMID: 27100083
  39. This study aimed to investigate whether genetic variations in the glucagon-like peptide receptor are associated with responses to dipeptidyl peptidase-4 inhibitors in patients with type 2 diabetes. Polymorphism in the GLP-1 receptor may influence the response to DPP-4 inhibitors. PMID: 27858848
  40. Results suggest that pancreatic ductal adenocarcinoma (PDAC) cells or their precursor lesions do not overexpress the glucagon-like peptide-1 receptor (GLP-1R) compared to non-neoplastic pancreatic cells. PMID: 26495786
  41. Molecular dynamics simulations of wild-type and mutant GLP-1R-ligand complexes provided molecular insights into GLP-1R-specific recognition mechanisms for the N terminus of GLP-1 by residues in the 7TM pocket. These simulations also explained how glucagon-mimicking GLP-1 mutants restored binding affinity for (glucagon receptor -mimicking) GLP-1R mutants. PMID: 27059958
  42. The NMR-determined structure of a high-potency cyclic conformationally-constrained 11-residue analogue of GLP-1 was also docked into the receptor-binding site. PMID: 26598711
  43. No association was found between the rs6923761 GLP-1 R polymorphism and weight loss. PMID: 26015316
  44. An association was observed between the rs6923761 GLP-1 receptor polymorphism and basal GLP-1 levels in diabetes mellitus type 2 patients. PMID: 25200998
  45. Although GLP-1R is not an independent prognostic factor in PDAC patients, it seems to have some implications for pancreatic ductal adenocarcinoma metastatic ability. PMID: 26238361
  46. Retinal GLP1R expression was found to be similar in patients with diabetes and healthy controls. PMID: 26384381
  47. A reduced level of GLP-1R was observed in the renal arteries of hypertensive patients. PMID: 25915883
  48. These findings demonstrate that the hGLP-1R has distinct regions within its C-terminal domain that are essential for cell surface expression, activity, and agonist-induced internalization. PMID: 26116235
  49. The GLP-1R rs10305420 polymorphism explained some of the inter-individual differences in response to liraglutide regarding weight loss in obese women with polycystic ovary syndrome (PCOS). PMID: 25991051
  50. The 168Ser (rs6923761) variant was nominally associated with alcohol use disorder. The 168 Ser/Ser genotype was associated with increased alcohol administration and a higher BOLD response in the right globus pallidus. PMID: 26080318

Show More

Hide All

Database Links

HGNC: 4324

OMIM: 138032

KEGG: hsa:2740

STRING: 9606.ENSP00000362353

UniGene: Hs.351883

Protein Families
G-protein coupled receptor 2 family
Subcellular Location
Cell membrane; Multi-pass membrane protein.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

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.

Q&A

What is GLP1R and why is it an important research target?

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.

How do I select the most appropriate GLP1R antibody for my specific research application?

Selection of a GLP1R antibody should be guided by your experimental requirements and the specific application. Consider these research-validated criteria:

ApplicationRecommended Antibody CharacteristicsDilution RangeValidation Requirements
Western BlotMonoclonal with specified molecular weight detection (~53 kDa)1:500-1:2000Positive control tissue (pancreas), KO validation
ImmunohistochemistryHigh specificity with low background1:50-1:500KO tissue validation, antigen retrieval optimization
ImmunofluorescenceHigh signal-to-noise ratioApplication-dependentCo-localization with reporter systems
ImmunoprecipitationHigh affinity for native proteinApplication-dependentValidation 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 .

What validation methods are essential to confirm antibody specificity for GLP1R?

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 .

How can I optimize immunohistochemistry protocols for detecting endogenous GLP1R in tissue samples?

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.

What are the key considerations when designing experiments to study GLP1R signaling pathways using monoclonal antibodies?

When investigating GLP1R signaling with monoclonal antibodies, consider these critical experimental design elements:

  • Antibody selection based on experimental goals:

    • For signaling blockade: Use antagonistic antibodies like Glp1R0017 that have been functionally validated to block GLP-1-induced cAMP production and insulin secretion .

    • For localization/expression studies: Use validated detection antibodies with knockout-confirmed specificity .

  • Pathway-specific readouts: Design experiments with appropriate downstream signaling markers:

    • cAMP production (primary GLP1R signaling pathway)

    • PKA activation and substrate phosphorylation

    • Insulin secretion in beta cells

    • Calcium mobilization

  • 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.

What are the advantages and limitations of using antagonistic monoclonal antibodies versus peptide antagonists like exendin 9-39 for GLP1R inhibition?

FeatureAntagonistic mAbs (e.g., Glp1R0017)Peptide Antagonists (e.g., exendin 9-39)
SpecificityHighly specific for GLP1R with minimal off-target effects Known to have off-target effects on other related receptors
Half-lifeExtended half-life (days) suitable for subchronic studies Short half-life (hours) requiring continuous administration for prolonged studies
AdministrationSingle injection sufficient for extended periods Requires continuous infusion or frequent dosing
Research applicationsIdeal for investigating extrapancreatic GLP1R signaling where specificity is crucial Useful for acute inhibition experiments
Species cross-reactivityWell-characterized antibodies like Glp1R0017 work across multiple species (mouse, human, rat, cynomolgus monkey, dog) Similar activity across species
CostHigher initial cost but potentially economical for extended studiesLower cost per dose but potentially higher for prolonged studies
StabilityHigh stability in vivoMore susceptible to proteolytic degradation

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.

How can GLP1R monoclonal antibodies be employed to investigate receptor distribution in extrapancreatic tissues?

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 .

What methodological approaches can resolve contradictory findings in GLP1R expression studies?

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.

How can I design in vivo experiments using GLP1R antagonistic antibodies to study extrapancreatic GLP1R functions?

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:

    • Acute studies: Take advantage of the immediate antagonistic effects

    • Chronic studies: Utilize the extended half-life of antibodies compared to peptide antagonists

  • 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.

What are common technical pitfalls when working with GLP1R antibodies and how can they be addressed?

Common IssuePossible CausesSolutions
False positive signalsNon-specific antibody bindingUse GLP1R knockout tissue as negative control ; Pre-adsorb antibody with immunizing peptide if available
Secondary antibody cross-reactivityInclude secondary-only controls; Use species-appropriate secondary antibodies
Inconsistent stainingVariable epitope accessibilityOptimize antigen retrieval (compare TE buffer pH 9.0 vs. citrate buffer pH 6.0)
Fixation-related epitope maskingTest multiple fixation protocols (PFA concentrations, fixation duration)
Weak or absent signalLow expression levelsImplement signal amplification techniques; Increase antibody concentration
Epitope degradationMinimize sample processing time; Maintain consistent protocols
High backgroundInsufficient blockingOptimize blocking with 5-10% serum and 1-3% BSA; Consider adding 0.1-0.3% Triton X-100
Antibody concentration too highTitrate antibody to optimal concentration; 1:50-1:500 for IHC
Discrepant molecular weight in WBPost-translational modificationsUse positive control tissue (pancreas) ; Consider deglycosylation treatment
Protein degradationAdd protease inhibitors; Maintain samples at 4°C during preparation

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.

How can I quantitatively assess GLP1R expression levels across different tissues or experimental conditions?

Quantitative assessment of GLP1R expression requires standardized approaches:

  • Immunoblotting quantification:

    • Use recombinant GLP1R protein standards for calibration

    • Normalize to housekeeping proteins and total protein loading

    • Implement digital image analysis with appropriate software

    • For WB applications, a dilution range of 1:500-1:2000 is typically effective

  • 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.

What are the technical considerations for developing new GLP1R antibodies for specialized research applications?

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 .

How might GLP1R antibodies enable new approaches to studying receptor biased signaling and conformational dynamics?

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.

How can GLP1R antibodies contribute to the development of next-generation targeted therapeutics?

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 .

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.