ANGPTL4 is a 45-kDa protein (calculated molecular weight: 45,214 Da ) with a fibrinogen-like C-terminal domain. It inhibits lipoprotein lipase (LPL), elevating circulating triglycerides (TG) . ANGPTL4 is hypoxia-inducible and plays roles in:
Monoclonal antibodies targeting ANGPTL4, such as MOR022770-LALA and 14D12, have been engineered for high specificity and affinity. Key features include:
High binding affinity: Equilibrium dissociation constant (K<sub>D</sub>) ≤100 pM .
Epitope specificity: Antibodies like 14D12 bind the N-terminal domain (amino acids Gln<sup>29</sup>–His<sup>53</sup>), critical for blocking ANGPTL4-LPL interactions .
Functional inhibition: These antibodies reduce plasma TG levels by >50% in preclinical models .
HRP-conjugated ANGPTL4 antibodies enable sensitive detection in assays. Examples include:
Specificity: Commercial antibodies show no cross-reactivity with other ANGPTL family proteins (e.g., ANGPTL3) .
Sensitivity: Effective in detecting ANGPTL4 at concentrations as low as 1–15 pg/mL in inflammatory models .
Functional assays: Antibodies like 14D12 inhibit >80% of ANGPTL4-LPL binding in vitro, reducing TG accumulation .
ANGPTL4-knockout mice exhibit severe systemic inflammation under high-fat diets, detectable via elevated serum amyloid A (SAA) and hepatic acute-phase proteins .
Anti-ANGPTL4 antibodies mitigate hypertriglyceridemia in murine models, mimicking genetic knockout phenotypes .
ANGPTL4 is upregulated in lung adenocarcinoma (H441, A549) and mammary carcinoma (MCF7) cells under PMA stimulation .
Antibodies detect ANGPTL4 in hypoxic tumor microenvironments, correlating with angiogenesis and metastasis .
Antigen retrieval: EDTA buffer (pH 8.0) optimizes detection in formalin-fixed tissues .
Blocking: 10% goat serum reduces nonspecific binding in IHC .
Controls: Recombinant human ANGPTL4 is used for validation .
ANGPTL4 antibodies are therapeutic candidates for:
ANGPTL4 (Angiopoietin-Like Protein 4) is a secreted glycoprotein that functions as a critical regulator of lipid metabolism. Its primary biological functions include:
Mediating inactivation of lipoprotein lipase (LPL), thus regulating triglyceride clearance from blood serum
Playing a significant role in lipid metabolism through its inhibitory effect on LPL activity
Potentially regulating glucose homeostasis and insulin sensitivity
Inhibiting proliferation, migration, and tubule formation of endothelial cells
Reducing vascular leakage and affecting endothelial cell adhesion to the extracellular matrix
Potentially modulating tumor-related angiogenesis in certain contexts
The protein has a molecular weight of approximately 45.2 kDa, though it is often detected at around 65 kDa in Western blot applications due to post-translational modifications .
ANGPTL4 is a two-domain protein with distinct structural and functional components:
N-terminal domain: A coiled-coil structure that mediates oligomerization of the protein and contains the LPL inhibitory activity. This domain is responsible for ANGPTL4's ability to inhibit LPL and regulate lipid metabolism .
C-terminal domain: A fibrinogen-like domain with distinct biological functions from the N-terminal region .
After secretion, these domains can be cleaved apart by pro-protein convertases. Importantly, this cleavage enhances the inhibitory effect of the N-terminal domain on LPL activity . When designing experiments with ANGPTL4 antibodies, researchers should consider whether their antibody targets the full-length protein, the N-terminal domain, or the C-terminal domain, as this will affect experimental outcomes and interpretation.
HRP-conjugated ANGPTL4 antibodies function through enzymatic amplification of detection signals in immunoassays. The process follows these methodological steps:
Primary binding: The ANGPTL4 antibody portion binds specifically to ANGPTL4 protein in the sample.
Signal generation: The conjugated HRP enzyme catalyzes the oxidation of substrate molecules (such as TMB or DAB) in the presence of hydrogen peroxide.
Visualization: This enzymatic reaction produces a colored, fluorescent, or chemiluminescent product that can be measured to quantify ANGPTL4 levels.
In sandwich ELISA configurations, the system typically employs:
A capture antibody pre-coated on a microplate surface
The sample containing ANGPTL4
A biotinylated detection antibody specific for ANGPTL4
The substrate solution is then added, and only wells containing the complete sandwich complex will develop color. The optical density measured at 450 nm is proportional to the ANGPTL4 concentration in the sample .
Optimizing Western blot protocols for ANGPTL4 detection requires careful attention to several parameters. Based on experimental validation data, the following methodology is recommended:
Sample Preparation:
Load 30 μg of protein lysate per lane under reducing conditions
ANGPTL4 can be detected in various human tissues including placenta and hepatocellular carcinoma
Electrophoresis Conditions:
Use a 5-20% gradient SDS-PAGE gel
Run at 70V for stacking gel and 90V for resolving gel
Continue electrophoresis for 2-3 hours for optimal separation
Transfer Protocol:
Blocking and Antibody Incubation:
Block membrane with 5% non-fat milk/TBS for 1.5 hours at room temperature
Incubate with primary ANGPTL4 antibody at 0.5 μg/mL overnight at 4°C
Wash with TBS-0.1% Tween three times, 5 minutes each
Incubate with HRP-conjugated secondary antibody at 1:5000 dilution for 1.5 hours at room temperature
Detection:
Develop using enhanced chemiluminescence detection system
Expected band size for ANGPTL4 is approximately 45 kDa, though it often appears at approximately 65 kDa due to post-translational modifications
These conditions have been experimentally validated to produce specific detection with minimal background signal.
Successful immunohistochemical detection of ANGPTL4 in tissue sections requires specific optimization steps:
Antigen Retrieval Protocol:
Heat-mediated antigen retrieval in EDTA buffer (pH 8.0) is recommended for optimal epitope exposure
Complete retrieval is critical for consistent and reproducible results
Blocking Procedure:
Block tissue sections with 10% goat serum to minimize nonspecific binding
Ensure complete coverage of the tissue section
Antibody Concentration and Incubation:
Use ANGPTL4 primary antibody at 2 μg/ml concentration
Incubate overnight at 4°C to ensure complete antigen binding
Secondary Detection System:
Apply peroxidase-conjugated secondary antibody specific to the primary antibody host species
Incubate for 30 minutes at 37°C
Visualization using DAB (3,3'-diaminobenzidine) as the chromogen
This methodology has been successfully employed in detecting ANGPTL4 in various human tissues including placenta and spleen, with high specificity and minimal background staining .
ANGPTL4 inhibits lipoprotein lipase (LPL) through a reversible, noncompetitive inhibition mechanism, rather than through the previously proposed "unfolding molecular chaperone" model. To study this interaction:
Experimental Approach to Study ANGPTL4-LPL Interaction:
Co-immunoprecipitation assays to demonstrate ANGPTL4-LPL complex formation
Activity recovery assays showing that LPL regains activity after dissociation from ANGPTL4
Kinetic studies to determine the noncompetitive nature of inhibition
Key Mechanistic Findings:
LPL inhibited by ANGPTL4 exists in a complex with ANGPTL4
Upon dissociation from ANGPTL4, LPL can regain its lipase activity
The inhibition is noncompetitive, not involving the catalytic conversion of LPL dimers to inactive monomers
Methodological Considerations:
Researchers can use divalent cation-dependent variants of ANGPTL4 to create regulatable systems for studying this interaction
Chelation treatment can reverse the inhibition in these systems, confirming the reversible nature of the inhibition
This mechanistic understanding is crucial for research aimed at developing therapeutic strategies targeting hypertriglyceridemia and related disorders.
Proper preparation of HRP conjugate working solutions is critical for sensitive and reproducible ANGPTL4 detection, particularly in ELISA applications. The following methodological approach ensures optimal reagent performance:
HRP Conjugate Preparation Protocol:
Calculate the required volume based on the experimental design (100 μL/well)
Always prepare slightly more than calculated to account for pipetting errors
Centrifuge the concentrated HRP conjugate at 800×g for 1 minute to collect all liquid
Dilute the 100× concentrated HRP conjugate to 1× working solution using appropriate diluent
Use precise dilution ratio of 1:99 (Concentrated HRP Conjugate:HRP Conjugate Diluent)
Prepare the working solution immediately before use to maintain optimal enzyme activity
Storage Considerations:
Store stock concentrated HRP conjugate according to manufacturer recommendations
Do not store diluted working solutions for extended periods
Avoid repeated freeze-thaw cycles of concentrated conjugate
Following these precise methodological steps ensures consistent enzymatic activity, minimizing inter-assay variability and maximizing detection sensitivity.
Ensuring high specificity in ANGPTL4 detection requires careful consideration of several technical factors:
Antibody Selection Criteria:
Validation status: Select antibodies with validated specificity against ANGPTL4 in relevant species (human, mouse, rat)
Domain specificity: Determine whether the antibody recognizes the N-terminal domain, C-terminal domain, or full-length protein based on experimental needs
Cross-reactivity profile: Verify absence of significant cross-reactivity with related proteins
Experimental Controls to Ensure Specificity:
Positive controls: Include known ANGPTL4-expressing tissues (placenta, hepatocellular carcinoma tissues)
Negative controls: Include samples where primary antibody is omitted
Competitive inhibition: Pre-incubation with recombinant ANGPTL4 should abolish signal
Sample Preparation Considerations:
Optimal buffer systems to maintain ANGPTL4 conformational epitopes
Appropriate blocking agents to minimize background
Implementing these methodological considerations significantly enhances the specificity and reliability of ANGPTL4 detection in research applications.
Generating a reliable standard curve is essential for accurate quantification of ANGPTL4 in biological samples. The following methodology ensures optimal standard curve preparation:
Standard Preparation Protocol:
Reconstitute lyophilized ANGPTL4 standard according to manufacturer instructions
Prepare a two-fold serial dilution series spanning 1.56-100 ng/mL concentration range
Standard Curve Performance Characteristics:
| ANGPTL4 Concentration (ng/mL) | Typical OD450 Value | %CV |
|---|---|---|
| 100 | 2.1-2.4 | <8% |
| 50 | 1.5-1.8 | <8% |
| 25 | 1.0-1.3 | <8% |
| 12.5 | 0.7-0.9 | <8% |
| 6.25 | 0.4-0.6 | <8% |
| 3.125 | 0.2-0.3 | <10% |
| 1.56 | 0.1-0.15 | <12% |
| 0 | <0.05 | N/A |
Analytical Considerations:
Working range: The assay provides reliable quantification between 1.56-100 ng/mL
Repeatability: Intra-assay coefficient of variation should be <10%
For accurate quantification, all standards and samples should be assayed in duplicate, and standard curves should be generated for each ELISA plate to account for plate-to-plate variations.
Interpreting Western blot data for ANGPTL4 requires careful consideration of post-translational modifications and proteolytic processing. Researchers commonly observe multiple bands on ANGPTL4 Western blots that require proper interpretation:
Common ANGPTL4 Western Blot Patterns:
Full-length ANGPTL4: Often detected at ~65 kDa, despite theoretical molecular weight of 45.2 kDa due to glycosylation and other post-translational modifications
N-terminal domain: ~26-28 kDa after proteolytic cleavage
C-terminal domain: ~17-20 kDa after proteolytic cleavage
Interpretation Guidelines:
Multiple bands may represent physiologically relevant processed forms rather than non-specific binding
Post-translational modifications (particularly glycosylation) can cause significant shifts in apparent molecular weight
Pro-protein convertase activity in the biological sample affects the ratio of full-length to cleaved forms
Sample preparation conditions (reducing vs. non-reducing) can affect observed banding patterns
Methodological Approaches to Confirm Band Identity:
Compare detection patterns using antibodies targeting different ANGPTL4 domains
Include recombinant ANGPTL4 (full-length and domains) as positive controls
Test lysates from tissues known to express different ANGPTL4 forms (placenta, liver, adipose tissue)
Understanding these considerations allows for accurate interpretation of ANGPTL4 Western blot data in experimental contexts.
Reconciling discrepancies between ELISA and Western blot quantification of ANGPTL4 requires understanding the fundamental differences between these techniques:
Common Sources of Discrepancies:
Epitope Accessibility Differences:
ELISA typically detects native conformations
Western blot detects denatured epitopes
Some antibodies may preferentially recognize certain conformational states
Domain-Specific Detection:
Sample Matrix Effects:
ELISA may be subject to matrix interference from complex biological samples
Western blot involves SDS-PAGE separation that can reduce matrix effects
Methodological Approach to Reconcile Discrepancies:
Determine domain specificity of both ELISA and Western blot antibodies
Consider using recombinant standards containing both full-length and cleaved forms
Validate results using orthogonal methods such as mass spectrometry
When possible, use antibody pairs targeting the same epitopes for both methods
When significant discrepancies occur, researchers should report results from both methods with appropriate caveats about the limitations of each approach.
Analysis of ANGPTL4-lipoprotein lipase (LPL) interactions in complex biological systems requires consideration of several critical factors that influence experimental outcomes:
Physiological Variables Affecting ANGPTL4-LPL Interactions:
Oligomerization State:
Proteolytic Processing:
Reversibility of Inhibition:
Methodological Approaches for Studying These Interactions:
In Vitro Systems:
Purified component assays to determine direct interactions
Kinetic studies to establish inhibition mechanisms
Structural analyses to identify interaction domains
Cellular Systems:
Cell lines expressing defined ANGPTL4 and LPL variants
Analysis of triglyceride processing in presence of ANGPTL4
Co-immunoprecipitation to verify complex formation
In Vivo Models:
Tissue-specific expression systems
Conditional knockout models
Physiological challenges (fasting, lipid loading) to assess dynamic regulation
Understanding these considerations enables researchers to design experiments that accurately reflect the complex regulatory mechanisms of lipid metabolism mediated by ANGPTL4-LPL interactions.
Advanced multi-parameter imaging approaches using HRP-conjugated ANGPTL4 antibodies enable simultaneous visualization of ANGPTL4 with other proteins or cellular structures. These methodologies provide valuable insights into protein co-localization and functional interactions.
Multiplexed Immunohistochemistry Protocols:
Sequential Multiplex IHC:
Apply ANGPTL4 primary antibody and HRP-conjugated secondary
Develop with spectrally distinct chromogens (DAB for brown, AEC for red)
Strip or quench the first round of antibodies
Apply subsequent antibodies for other targets with different detection systems
Tyramide Signal Amplification (TSA) Multiplexing:
Use HRP-conjugated antibodies with tyramide-fluorophore conjugates
HRP catalyzes covalent binding of fluorescent tyramides to nearby proteins
Heat-inactivate HRP after each round
Repeat with different antibodies and fluorophores
Enables detection of 5-7 proteins on a single tissue section
Methodological Considerations:
Optimize antigen retrieval for all target proteins simultaneously
Carefully validate antibody specificity in multiplex settings
Include appropriate controls for signal bleeding and non-specific binding
Use spectral unmixing for fluorescent applications to resolve overlapping signals
This approach has been successfully employed to visualize ANGPTL4 in relation to vascular structures and metabolic markers in various tissues, providing insights into its functional relationships in complex tissue environments.
Post-translational modifications (PTMs) of ANGPTL4 significantly impact its function and interactions. Advanced analytical approaches offer insights into these critical modifications:
Mass Spectrometry-Based Approaches:
Bottom-up Proteomics:
Enzymatic digestion of ANGPTL4 followed by LC-MS/MS analysis
Identification of glycosylation, phosphorylation, and other modifications
Quantification of modification stoichiometry
Top-down Proteomics:
Analysis of intact ANGPTL4 protein by high-resolution MS
Provides comprehensive view of proteoforms
Enables detection of combinatorial modifications
Site-Specific Analysis Techniques:
Glycoprofiling:
Release and analysis of N-linked and O-linked glycans
Structural characterization using exoglycosidase digestions
Linking glycan structures to functional properties
Phosphorylation Mapping:
Enrichment of phosphopeptides using titanium dioxide or IMAC
Identification of phosphorylation sites and their stoichiometry
Correlation with regulatory mechanisms
Integrated Analytical Workflow:
Initial characterization using Western blot with modification-specific antibodies
Confirmation and detailed mapping using MS-based approaches
Functional studies using site-directed mutagenesis of modified residues
Correlation of modification patterns with biological activity
These analytical approaches reveal how PTMs regulate ANGPTL4's ability to inhibit LPL and its interactions with other proteins, providing deeper insights into its role in lipid metabolism and other physiological processes.
Recent advances in ANGPTL4 antibody applications have opened new avenues for metabolic disease research. Several emerging trends demonstrate the expanding utility of these tools:
Therapeutic Antibody Development:
Single-Cell Analysis Applications:
Integration of ANGPTL4 antibodies in mass cytometry (CyTOF) panels
Single-cell proteomics to examine ANGPTL4 expression heterogeneity
Spatial transcriptomics combined with antibody detection for tissue microenvironment analysis
Methodological Innovations:
Development of proximity ligation assays to study ANGPTL4-protein interactions in situ
CRISPR-based screening with antibody readouts to identify ANGPTL4 regulators
Nanobody and aptamer alternatives to conventional antibodies for improved tissue penetration
These emerging applications are advancing our understanding of ANGPTL4's role in metabolic regulation and developing novel therapeutic strategies for metabolic disorders.
Integration of ANGPTL4 antibody-based data with multi-omics approaches provides comprehensive insights into its regulatory networks and functional implications:
Multi-Omics Integration Strategies:
Antibody-Based Proteomics with Transcriptomics:
Correlation of ANGPTL4 protein levels (detected by antibodies) with mRNA expression
Identification of post-transcriptional regulatory mechanisms
Discovery of discordant regulation between protein and transcript levels
Functional Proteomics Integration:
Immunoprecipitation with ANGPTL4 antibodies followed by mass spectrometry
Identification of protein interaction networks
Correlation with functional metabolic outcomes
Integrated Analysis Platforms:
Computational frameworks for multi-dimensional data integration
Network analysis of ANGPTL4 in lipid metabolism pathways
Systems biology approaches to model ANGPTL4 regulation
Methodological Workflow Example:
Quantify ANGPTL4 using antibody-based methods (ELISA, IHC, WB)
Perform RNA-seq or proteomics on the same samples
Integrate with metabolomics data focusing on lipid profiles
Apply computational methods to identify regulatory relationships