Ang4 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Production and Conjugation Methodology

HRP conjugation to ANGPTL4 antibodies typically involves:

  1. Antibody Preparation: Purification via Protein G chromatography to ensure >95% purity .

  2. Buffer Optimization: Removal of interfering components (e.g., BSA, Tris, sodium azide) to maximize conjugation efficiency .

  3. Conjugation Process:

    • Lightning-Link® HRP: Direct labeling with a modifier and quencher in 3 hours .

    • Sulfo-SMCC: Activation of HRP with maleimide groups, followed by thiolation of antibodies for covalent binding .

Applications in Research and Clinical Diagnostics

Ang4 HRP antibodies are primarily utilized for detecting ANGPTL4 in biological samples, with applications including:

Table 1: Key Applications and Supplier Examples

ApplicationSupplierReactivityKey Features
ELISACusabio, MyBioSourceMouse, HumanHigh specificity for serum/plasma analysis .
Western BlotAbbexaMouseLimited due to HRP's enzymatic interference .
Biomarker DetectionAssay GenieHumanValidates ANGPTL4's role in cholangiocarcinoma (CCA) metastasis .

Notable Research Findings

  • CCA Prognosis: Elevated serum ANGPTL4 levels correlate with vascular invasion (AUC = 0.751) and lymph node metastasis (AUC = 0.691) in CCA patients, detected via dot blot assays using ANGPTL4 antibodies .

  • Hypoxia Response: HRP-conjugated antibodies confirm ANGPTL4 upregulation in hypoxic endothelial cells, linked to angiogenesis inhibition .

Performance Optimization and Stability

Critical Factors for Efficiency

ParameterRecommendationImpact of Deviation
Buffer pH6.5–8.5Suboptimal conjugation or antibody denaturation .
Glycerol Content<50%Aggregation or reduced enzyme activity .
Storage-20°C in 50% glycerol/PBSLoss of HRP activity at room temperature .

Stabilizers: Products like Abcam’s LifeXtend™ HRP protect conjugates from degradation, enabling room-temperature storage .

Cross-Reactivity and Validation

Species-Specific Reactivity

SupplierHost SpeciesTarget SpeciesCross-Reactivity
CusabioRabbitMouseNo .
Assay GenieRabbitHumanLimited to human .

Validation Methods

  • Western Blot: Confirms ANGPTL4 bands at ~50 kDa in human/mouse sera .

  • ELISA: Correlates dot blot arbitrary units (AU) with ELISA concentrations (r = 0.9203, p < 0.0001) .

Clinical and Therapeutic Implications

  • Cancer Biomarker: Serum ANGPTL4 levels outperform CA 19-9 and CEA in predicting CCA metastasis .

  • Lipid Regulation: ANGPTL4 inhibition via antibodies may modulate triglyceride levels, offering therapeutic potential .

Limitations and Challenges

  • Buffer Sensitivity: Tris, sodium azide, or BSA in antibody buffers inhibit conjugation .

  • HRP Instability: Enzyme activity diminishes over time, necessitating stabilizers or cold storage .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days after receiving it. Delivery times may vary depending on the shipping method and destination. For specific delivery timeframes, please contact your local distributor.
Synonyms
Angiogenin-4 (EC 3.1.27.-), Ang4
Target Names
Ang4
Uniprot No.

Target Background

Function
Angiogenin 4 exhibits bactericidal activity against E. faecalis and L. monocytogenes, but not against L. innocua and E. coli. In vitro studies have demonstrated its ability to promote angiogenesis. Additionally, it displays low ribonuclease activity in vitro. While promoting proliferation of melanoma cells, it does not affect the proliferation of endothelial cells or fibroblasts in vitro.
Gene References Into Functions
  1. Intestinal intraepithelial lymphocyte-enterocyte crosstalk regulates the production of bactericidal angiogenin 4 by Paneth cells in response to microbial challenge. PMID: 24358364
  2. The goblet cell is identified as the cellular source of mouse Ang4 in the colon following infection with T. muris, with its expression being regulated by IL-13. PMID: 22970115
  3. The expression of Reg3 proteins and Ang4 by goblet cells highlights their crucial role in innate defense. PMID: 22723890
  4. Gene expression profiles associated with immune and defense responses, including angiogenin 4, exhibit notable differences. PMID: 19852835
  5. The protein structure of angiogenin 4 has been characterized. PMID: 15772759

Show More

Hide All

Database Links
Protein Families
Pancreatic ribonuclease family
Subcellular Location
Cytoplasmic vesicle, secretory vesicle lumen. Secreted. Nucleus, nucleolus.
Tissue Specificity
Detected in small intestine, caecum and colon, with the highest expression in Paneth cells in the intestinal epithelium.

Q&A

What is Ang4 Antibody and what are its key biological functions?

Ang4 Antibody specifically targets Angiogenin-4 (EC 3.1.27.-), a protein with multiple biological functions. Angiogenin-4 exhibits selective bactericidal activity against specific microorganisms, including Enterococcus faecalis and Listeria monocytogenes, while showing no activity against Listeria innocua and Escherichia coli . The protein promotes angiogenesis in vitro and demonstrates low ribonuclease activity. Additionally, Ang4 has been shown to stimulate proliferation specifically in melanoma cells but not in endothelial cells or fibroblasts under laboratory conditions . These properties make Ang4 a protein of interest in both microbiology and cancer research contexts, particularly for studying selective cell proliferation mechanisms.

Why is HRP conjugation commonly used with antibodies like Ang4 in research applications?

Horseradish peroxidase (HRP) conjugation is preferred for antibodies like Ang4 due to several advantageous properties for research applications. The 44kDa glycoprotein structure of HRP contains four lysine residues that facilitate effective conjugation to antibodies . When properly conjugated, HRP produces colored, fluorimetric, or luminescent derivatives that enable sensitive detection and precise quantification of target molecules . Researchers favor HRP for antibody conjugation because it is comparatively smaller, demonstrates greater stability, and is more cost-effective than alternative conjugation enzymes . Additionally, HRP's high turnover rate generates strong detectable signals within relatively short timeframes, enhancing experimental efficiency. This combination of properties makes HRP-conjugated antibodies particularly valuable for techniques requiring high sensitivity and reproducibility, such as ELISA and immunohistochemistry applications.

What detection methods are compatible with Ang4 Antibody, HRP conjugated?

Ang4 Antibody, HRP conjugated is compatible with multiple detection methodologies, with ELISA being the primary validated application . In ELISA-based detection systems, the HRP conjugate produces a measurable color change when exposed to an appropriate substrate. The enzyme-substrate reaction generates a blue color that turns yellow upon addition of the stop solution, with the optical density measured spectrophotometrically at 450 ± 2 nm . This measurement is directly proportional to the concentration of the target protein. Beyond ELISA, HRP-conjugated antibodies are generally suitable for several other detection methods including Western blotting, immunocytochemistry, and flow cytometry, though specific validation for Ang4 Antibody would be required for each application. The versatility of detection methods is enhanced by HRP's ability to catalyze reactions with various substrates to produce different types of signals (colorimetric, chemiluminescent, or fluorescent), providing researchers flexibility in experimental design based on available instrumentation and sensitivity requirements.

How should researchers optimize the working concentration of Ang4 Antibody, HRP conjugated for ELISA applications?

Optimizing the working concentration of Ang4 Antibody, HRP conjugated for ELISA requires a systematic titration approach. Begin with a broad range dilution series (e.g., 1:100, 1:500, 1:1,000, 1:5,000, and 1:10,000) of the antibody to identify the approximate optimal range. Based on the manufacturer's documentation, the recommended usage range for similar HRP-conjugated antibodies in ELISA applications is typically between 0.5-125 ng/mL . After identifying the optimal range, perform a fine-tuned titration with narrower dilution intervals to precisely determine the concentration that provides the optimal signal-to-noise ratio.

The optimization process should include both positive and negative controls to establish signal specificity. Signal-to-noise ratio evaluation involves comparing the optical density (OD) readings of samples containing Ang4 to those of negative controls. The optimal working concentration will produce distinct positive signals while maintaining minimal background. Additionally, consider performing a checkerboard titration against varying concentrations of capture antibody if using a sandwich ELISA format. This methodological approach ensures reliable and reproducible results while efficiently utilizing the antibody resource.

What are the critical steps in preparing HRP Conjugate working solution for maximum sensitivity?

Preparing HRP Conjugate working solution requires precise handling to maintain enzyme activity and ensure maximum assay sensitivity. The critical steps include:

  • Centrifugation: Begin by centrifuging the concentrated HRP Conjugate at 800×g for 1 minute to ensure all material is collected at the bottom of the tube and to remove any aggregates that could affect performance .

  • Precise dilution: Dilute the 100× Concentrated HRP Conjugate to a 1× working solution using the appropriate HRP Conjugate Diluent at precisely a 1:99 ratio (Concentrated HRP Conjugate:HRP Conjugate Diluent) . Accurate measurement is essential as improper dilution directly affects signal intensity.

  • Timing of preparation: Prepare the working solution immediately before use rather than in advance, as HRP activity can diminish over time even in optimized buffer conditions .

  • Temperature considerations: Perform all dilution steps at room temperature (20-25°C), but keep the concentrated stock at 4°C until needed to preserve enzyme activity.

  • Protection from contaminants: Use clean, enzyme-free pipettes and containers to prevent contamination with inhibitors such as sodium azide, cyanides, or sulfides that can inactivate HRP enzymatic activity .

  • Light protection: Shield the prepared solution from direct light exposure, as photosensitive components may degrade under strong illumination.

Following these methodological steps ensures optimal HRP activity and maximizes assay sensitivity by preserving the catalytic efficiency of the enzyme conjugate.

What storage conditions are recommended to maintain the activity of Ang4 Antibody, HRP conjugated?

Maintaining optimal activity of Ang4 Antibody, HRP conjugated requires adherence to specific storage protocols. For long-term storage, the lyophilized antibody should be maintained at -20°C or lower temperatures . After reconstitution, the antibody can be stored at 4°C for up to one month, but for extended preservation, aliquoting and storage at -20°C is recommended to prevent repeated freeze-thaw cycles that can compromise antibody functionality. Each freeze-thaw cycle can result in approximately 10-15% loss of activity due to protein denaturation.

It is crucial to protect HRP-conjugated antibodies from light exposure, as the HRP moiety is photosensitive and can lose activity with prolonged light exposure . When working with the antibody, minimize the time it remains at room temperature and return it to appropriate storage conditions promptly. If supplied in glycerol-containing buffer (such as 50% glycerol reported in the product specifications), ensure the antibody solution does not freeze at -20°C, as this can lead to glycerol separation and protein concentration gradients . For maximum stability preservation, some manufacturers recommend adding a protein stabilizer such as BSA (1%) if the antibody requires dilution before storage. These methodological precautions ensure the maintenance of antibody specificity and enzymatic activity throughout the research timeframe.

How can researchers validate the specificity of Ang4 Antibody, HRP conjugated in experimental systems?

Validating the specificity of Ang4 Antibody, HRP conjugated requires a multi-step approach employing several complementary techniques. Initially, perform a Western blot analysis comparing samples with known Ang4 expression against negative controls to confirm the antibody detects a single band at the expected molecular weight (approximately 16 kDa for mouse Ang4). For more stringent validation, include a competitive blocking experiment where pre-incubation of the antibody with recombinant Ang4 protein (specifically, recombinant Mouse Angiogenin-4 protein, amino acids 25-144) should substantially reduce or eliminate signal detection.

Further validation should include testing in Ang4 knockout models or following siRNA-mediated knockdown of Ang4 expression, where signal reduction should correlate with knockdown efficiency. Immunoprecipitation followed by mass spectrometry can provide definitive confirmation of antibody specificity by identifying the precise protein targets pulled down by the antibody. Additionally, cross-reactivity assessment should be performed against closely related proteins in the angiogenin family to ensure selective binding to Ang4 rather than Ang1, Ang2, or Ang3. For sandwich ELISA applications, compare results using alternative antibody pairs targeting different Ang4 epitopes to confirm consistent quantification results. This comprehensive validation approach ensures experimental findings can be confidently attributed to specific Ang4 detection.

What methodological approaches can address potential interference in ELISA using Ang4 Antibody, HRP conjugated?

Addressing potential interference in ELISA using Ang4 Antibody, HRP conjugated requires systematic troubleshooting and methodological refinements. Sample matrix effects represent a common source of interference, particularly in serum or plasma samples which may contain heterophilic antibodies or anti-animal immunoglobulins. Implement a sample pre-treatment protocol using heterophilic blocking reagents or add irrelevant animal serum (corresponding to the antibody host species) to the dilution buffer to reduce non-specific binding.

High-dose hook effect can occur when excessive antigen concentrations paradoxically produce decreased signal. Test multiple sample dilutions (including high dilutions of suspected high-concentration samples) to identify potential hook effects. According to validation data for similar ELISA systems, linearity should be maintained across serial dilutions, with samples showing recovery percentages between 85-113% across dilution ranges from 1:2 to 1:16 .

Cross-reactivity with structurally similar proteins represents another potential interference source. While the manufacturer indicates no significant cross-reactivity for this particular antibody , researchers should validate this claim in their specific experimental system, particularly when working with complex samples containing multiple angiogenin family members.

Endogenous enzyme activity that might interact with HRP substrates can be addressed by including appropriate enzyme inhibitors in sample preparation. Additionally, optimize washing steps to reduce non-specific binding while maintaining specific signals. The intra-assay and inter-assay precision reported for similar systems shows CV percentages below 6.25% , providing a benchmark for acceptable assay performance after interference mitigation strategies have been implemented.

How can Ang4 Antibody, HRP conjugated be utilized in multiplex detection systems?

Implementing Ang4 Antibody, HRP conjugated in multiplex detection systems requires strategic methodological approaches to maintain specificity and sensitivity while enabling simultaneous measurement of multiple targets. For bead-based multiplex immunoassays, the HRP-conjugated Ang4 antibody can be used alongside other conjugated detection antibodies with distinguishable reporters (e.g., phycoerythrin or alternative enzymes) that produce signals separable from HRP's output. This approach requires careful optimization of antibody ratios to prevent cross-reactivity and signal interference.

In multiplex ELISA systems using multiple capture antibodies spatially separated in defined arrays, the HRP-conjugated Ang4 antibody must be validated for compatibility with the common detection substrate and for absence of cross-reactivity with other immobilized capture antibodies. Particularly promising is the application in microfluidic immunoassay platforms, where HRP signal amplification systems can be integrated with spatial separation of capture zones.

For immunohistochemistry applications, the HRP-conjugated Ang4 antibody can be incorporated into multiplex staining protocols using sequential detection methods. This involves completely inactivating the HRP after each detection cycle through methods such as hydrogen peroxide treatment before proceeding to the next target, thus preventing signal contamination between detection cycles.

A critical validation step for multiplex applications is demonstrating that the sensitivity and dynamic range of the Ang4 antibody detection are not compromised in the multiplex format compared to singleplex detection. Researchers should establish standard curves in both formats and verify that the assay performance characteristics match the reported singleplex values, such as the detection range of 1.56-100 ng/mL demonstrated for similar ELISA systems .

What are common sources of background signal when using Ang4 Antibody, HRP conjugated, and how can they be minimized?

Background signal issues when using Ang4 Antibody, HRP conjugated can stem from multiple sources that require specific mitigation strategies. Non-specific binding of the antibody to experimental surfaces represents a primary source of background. This can be addressed by optimizing blocking protocols—using 1-5% BSA or 5% non-fat milk in TBS-T for Western blots, or commercial blocking buffers specifically formulated for ELISA applications. Extending blocking time to 2 hours at room temperature or overnight at 4°C can significantly reduce non-specific binding in challenging samples.

Insufficient washing contributes substantially to elevated background. Implement rigorous washing protocols with at least 5-6 wash cycles using fresh buffer for each cycle. For ELISA applications, include 0.05% Tween-20 in wash buffers to reduce non-specific interactions while using proper washing technique (complete well filling and emptying) to ensure removal of unbound antibodies.

Cross-reactivity with endogenous peroxidases in biological samples can generate false signals. Pre-treat samples with hydrogen peroxide (0.3% H₂O₂ for 15-30 minutes) to quench endogenous peroxidase activity before adding the HRP-conjugated antibody. For tissue samples, this treatment is particularly critical.

Improper antibody dilution often leads to excessive background. Antibody titration experiments should establish the optimal concentration that maximizes the signal-to-noise ratio. Working dilutions should be prepared fresh in appropriate diluent containing a carrier protein (0.1-0.5% BSA) to prevent non-specific binding.

Suboptimal substrate handling can also increase background. Substrate solutions should be equilibrated to room temperature before use, protected from light, and used within the recommended timeframe to prevent spontaneous oxidation that contributes to background signal. Using these methodological refinements will significantly improve signal-to-noise ratio and experimental reliability.

How should researchers interpret and troubleshoot inconsistent results in Ang4 detection assays?

Interpreting and troubleshooting inconsistent results in Ang4 detection assays requires systematic analysis of potential variables affecting assay performance. Begin by evaluating assay precision metrics across multiple experimental runs. According to validation data for similar ELISA systems, acceptable intra-assay and inter-assay precision should yield coefficient of variation (CV) values below 6.25% . Results exceeding this threshold warrant methodological investigation.

Sample quality and handling represent critical factors in result consistency. Analyze the effects of sample collection method, storage conditions, and freeze-thaw cycles on Ang4 stability. Implement standardized protocols for sample handling, including consistent time intervals between collection and processing. For plasma samples, evaluate whether different anticoagulants (EDTA, heparin, citrate) affect Ang4 measurement, as validation data shows slight variations in recovery rates between sample types (94% for serum versus 96% for EDTA plasma) .

Reagent degradation frequently underlies inconsistent performance. Document lot numbers of critical reagents, particularly antibodies and substrates, and test new lots alongside previous lots before complete transition. HRP conjugates are sensitive to storage conditions; deviations from recommended storage (typically -20°C or lower for lyophilized antibodies) can progressively reduce enzymatic activity and signal generation.

Technical variables including incubation time precision, temperature fluctuations, and washing efficiency should be standardized across experiments. Implement temperature-controlled incubation steps using calibrated equipment rather than ambient conditions subject to environmental variation. Evaluate whether automated versus manual washing procedures affect consistency, particularly for large sample sets.

When inconsistencies persist despite these controls, consider implementing standard addition methods to assess potential matrix effects. Spike known quantities of recombinant Ang4 protein into samples and measure recovery percentages, which should fall within 85-102% according to validation data for similar systems . This approach can identify sample-specific inhibitory or enhancing factors affecting antibody binding or enzymatic activity.

What statistical considerations should be applied when analyzing quantitative data from assays using Ang4 Antibody, HRP conjugated?

Analyzing quantitative data from assays using Ang4 Antibody, HRP conjugated requires specific statistical approaches to ensure scientific validity. Standard curve modeling represents the foundation for accurate quantification. Instead of linear regression, implement four-parameter logistic (4PL) or five-parameter logistic (5PL) curve fitting for ELISA data analysis, as these models better capture the sigmoidal relationship between concentration and optical density across the assay's dynamic range (1.56-100 ng/mL for similar systems) . This approach is particularly important for samples with concentrations at the extremes of the standard curve.

Outlier identification and handling must follow objective criteria rather than subjective removal. Apply Grubbs' test or the modified Z-score method to identify statistical outliers, and document all exclusion decisions with appropriate statistical justification. For replicate measurements, calculate both mean and median values, as median values are less influenced by extreme outliers that might result from technical errors.

Assay sensitivity considerations should include reliable determination of the lower limit of quantification (LLOQ). This is defined statistically as the lowest concentration where the coefficient of variation remains below 20% and recovery falls between 80-120%. For Ang4 detection, similar systems report a sensitivity of 0.94 ng/mL , but laboratory-specific validation should establish the practical LLOQ in the specific research context.

Assess dilution linearity across multiple sample dilutions to confirm that measurements follow the principle of dilutional parallelism. According to validation data, acceptable linearity would show recovery percentages between 87-113% across serial dilutions from 1:2 to 1:16 . Deviations from linearity may indicate matrix effects requiring additional sample pre-treatment steps.

For comparative studies, determine whether data follow normal distribution (using Shapiro-Wilk test) to select appropriate statistical tests (parametric vs. non-parametric). Additionally, calculate minimum sample sizes needed to detect biologically meaningful differences based on the established assay variation, typically requiring statistical power of 0.8 or higher with alpha set at 0.05. These rigorous statistical approaches ensure reliable data interpretation and reproducible research outcomes.

How can Ang4 Antibody, HRP conjugated be implemented in tissue-specific angiogenesis research?

Implementing Ang4 Antibody, HRP conjugated in tissue-specific angiogenesis research requires specialized methodological approaches to investigate the protein's role in different vascular environments. Immunohistochemical detection of Ang4 in tissue sections provides spatial information about expression patterns in relation to vascular structures. For this application, researchers should optimize antigen retrieval methods (typically citrate buffer pH 6.0 or EDTA buffer pH 9.0) to ensure accessibility of Ang4 epitopes without compromising tissue morphology. The HRP-conjugated antibody can be directly applied after blocking endogenous peroxidases and non-specific binding sites, followed by visualization using diaminobenzidine (DAB) substrate.

For quantitative assessment of angiogenesis in response to Ang4 modulation, perform systematic vessel counting across multiple microscopic fields. This should include measurements of microvessel density (MVD), vessel diameter distribution, and branching complexity to comprehensively characterize the angiogenic phenotype. The bactericidal activity of Ang4 against specific microorganisms (E. faecalis and L. monocytogenes) suggests potential roles in tissue-specific immune defense, warranting investigation of Ang4 expression in response to microbial challenges in different tissue contexts.

Laser capture microdissection combined with protein extraction protocols optimized to maintain antibody reactivity enables region-specific analysis of Ang4 levels within heterogeneous tissues. This approach is particularly valuable for understanding Ang4 distribution across tissue microenvironments. For functional studies, ex vivo tissue explant cultures allow manipulation of Ang4 levels through addition of recombinant protein or neutralizing antibodies, with subsequent analysis of angiogenic responses under controlled conditions that maintain tissue architecture. These methodological approaches collectively enable comprehensive characterization of Ang4's tissue-specific roles in both physiological and pathological angiogenesis.

What are the methodological considerations for studying the dual role of Ang4 in angiogenesis and bactericidal activity?

Studying the dual functionality of Ang4 in angiogenesis and bactericidal activity requires integrated experimental approaches that address both functions within relevant biological contexts. For bactericidal activity assessment, researchers should implement minimum inhibitory concentration (MIC) assays using purified recombinant Ang4 against the specifically susceptible bacterial strains E. faecalis and L. monocytogenes, while including L. innocua and E. coli as negative controls based on their reported resistance . Time-kill kinetics should be measured at physiologically relevant Ang4 concentrations determined through ELISA quantification in target tissues.

To investigate the molecular basis of selective bactericidal activity, structure-function analysis using truncated or mutated Ang4 variants can identify the protein domains responsible for antimicrobial activity versus those required for angiogenic functions. Testing these variants in parallel bactericidal and angiogenesis assays will reveal whether these functions can be mechanistically separated.

For angiogenesis studies, endothelial tube formation assays using HUVECs or tissue-specific endothelial cells should be conducted with controlled Ang4 concentrations, measuring multiple parameters including total tube length, number of branch points, and loop formation. The reported inhibition of endothelial cell adhesion to extracellular matrix by Ang4 should be specifically investigated through adhesion assays with different matrix components (fibronectin, laminin, collagen) to determine matrix specificity of this effect.

The potential interaction between these two functions requires examination of whether bacterial products modulate Ang4's angiogenic activity and, conversely, whether angiogenic factors affect its bactericidal properties. Co-culture systems incorporating both endothelial cells and bacteria can provide insights into such functional crosstalk. Additionally, tissue-specific expression analysis following bacterial challenge versus angiogenic stimulation will reveal whether these distinct functional roles are regulated through common or separate pathways in different physiological contexts.

How can researchers integrate Ang4 antibody detection with functional studies of lipid metabolism regulation?

Integrating Ang4 antibody detection with functional studies of lipid metabolism requires sophisticated experimental designs that connect molecular detection to physiological outcomes. Researchers should establish baseline Ang4 expression profiles in metabolically relevant tissues (liver, adipose tissue, intestine) using the HRP-conjugated antibody in Western blot and immunohistochemistry applications. These profiles should be correlated with lipid metabolism parameters including serum triglyceride levels, lipoprotein profiles, and tissue-specific lipid deposition.

To investigate the functional relationship between Ang4 and lipoprotein lipase (LPL), implement co-immunoprecipitation studies to detect potential direct interactions, followed by LPL activity assays in the presence of varying Ang4 concentrations. This approach can determine whether Ang4 modulates LPL activity through direct binding or indirect mechanisms. For in vivo studies, establish mouse models with tissue-specific Ang4 overexpression or knockdown, particularly in intestinal epithelium, followed by comprehensive lipidomic analysis to identify specific lipid species affected by Ang4 modulation.

Metabolic challenge experiments (high-fat diet, fasting/refeeding cycles) can reveal dynamic regulation of Ang4 in response to changing energetic conditions. The HRP-conjugated antibody can be used in time-course studies to detect rapid changes in Ang4 expression or secretion following metabolic perturbations. Additionally, examine the relationship between Ang4 and angiopoietin-like protein 4 (ANGPTL4), as ANGPTL4 is known to regulate triglyceride metabolism through LPL inhibition and plays roles in glucose homeostasis and insulin sensitivity .

For translational relevance, correlate Ang4 levels with metabolic syndrome parameters in human subjects, potentially identifying Ang4 as a biomarker for specific metabolic disturbances. This integrated approach connecting molecular detection to functional outcomes provides comprehensive insights into Ang4's role in metabolic regulation while establishing methodological frameworks for similar studies of multifunctional proteins.

What future directions in antibody development might enhance research applications for Ang4 detection?

Future directions in antibody development for Ang4 detection should focus on increasing specificity, sensitivity, and application versatility. Development of monoclonal antibodies targeting distinct, highly specific epitopes of Ang4 would enhance detection precision, particularly for distinguishing between full-length protein and its cleaved forms that may have different biological activities. Current polyclonal antibodies recognize multiple epitopes , potentially masking important functional differences between protein variants.

Adaptation of emerging recombinant antibody technologies, including single-chain variable fragments (scFvs) and nanobodies derived from camelid antibodies, would provide smaller detection molecules with enhanced tissue penetration for in vivo imaging applications. These smaller antibody formats could also improve detection in sterically restricted environments where conventional antibodies have limited access.

Multispecific antibody development targeting both Ang4 and its interaction partners would enable direct visualization of protein-protein interactions in situ. Bispecific antibodies simultaneously recognizing Ang4 and proteins involved in angiogenesis or lipid metabolism could provide invaluable tools for studying functional relationships in complex biological systems.

Advanced conjugation chemistries ensuring consistent antibody:HRP ratios would improve quantitative reliability across different antibody lots. Site-specific conjugation methods targeting engineered antibody sites rather than random lysine residues would preserve the antigen-binding region while providing uniform labeling. Additionally, development of pH-sensitive or environmentally responsive antibody-fluorophore conjugates could enable monitoring of Ang4 trafficking within cells and tissues under different physiological conditions.

These technological advances would significantly expand the research applications of Ang4 antibodies beyond current capabilities, facilitating deeper insights into this protein's diverse biological functions in health and disease.

How do current research findings on Ang4 contribute to our understanding of angiogenesis in pathological conditions?

Current research findings on Ang4 contribute significantly to our understanding of pathological angiogenesis through several mechanistic insights. Ang4's demonstrated ability to inhibit endothelial cell adhesion to extracellular matrix components and disrupt actin cytoskeleton reorganization suggests it may function as a contextual modulator of vascular remodeling. This modulatory role contrasts with the traditionally understood pro-angiogenic factors and suggests Ang4 may help fine-tune angiogenic responses in specific pathological microenvironments.

The selective promotion of melanoma cell proliferation by Ang4, without corresponding effects on endothelial cells or fibroblasts , indicates a potential tumor-specific growth-promoting function that may contribute to cancer progression independently of direct angiogenic effects. This finding raises important considerations for anti-angiogenic therapeutic strategies that might inadvertently affect Ang4 signaling.

Ang4's dual functionality in angiogenesis regulation and bactericidal activity points to potential mechanistic links between infection-related inflammation and pathological vascular responses. This connection is particularly relevant for conditions involving both inflammatory and angiogenic components, such as inflammatory bowel disease or wound healing complications.

The structural and functional similarities between Ang4 and other angiogenin family members suggest potential redundancy or compensatory mechanisms in pathological settings. This could explain the limited efficacy of targeted therapies against individual angiogenic factors and highlights the importance of comprehensive approaches addressing multiple factors simultaneously.

These insights collectively enhance our understanding of the complex regulatory networks governing pathological angiogenesis, moving beyond simplified models of pro- versus anti-angiogenic factors toward more nuanced understanding of context-dependent modulators like Ang4 that may have different functions depending on the specific cellular and molecular environment.

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 2025 TheBiotek. All Rights Reserved.