Ang1 is a 70-kDa secreted glycoprotein primarily expressed by pericytes and mural cells . It binds to the Tie2 receptor, triggering signaling pathways that promote vascular maturation, inhibit apoptosis, and suppress permeability . Ang1 oligomers induce Tie2 dimerization and autophosphorylation, activating downstream pathways such as PI3K/Akt, which enhance endothelial cell survival and stabilize tight junctions .
Antibody pairs are typically used in sandwich assays (e.g., ELISA) to quantify Ang1 levels or study its interactions. Two notable antibodies from the search results include:
Antibody | Source | Applications | Reactivity | Citations |
---|---|---|---|---|
ab133425 (Abcam) | Goat IgG | IHC-P, WB, ELISA | Human, Rat, Recombinant | |
27093-1-AP (Proteintech) | Rabbit IgG | WB, IHC, IF/ICC, ELISA | Human, Mouse, Rat |
Both antibodies target Ang1's functional domains, enabling precise detection in vascular tissue samples. Abcam's ab133425 reacts with the 50–100 amino acid region of human Ang1, while Proteintech's 27093-1-AP binds to a fusion protein corresponding to Ang1 .
A novel tetra-valent antibody, ASP4021, has been developed to mimic Ang1's agonistic activity on Tie2 . Unlike traditional ligands, ASP4021:
Fully activates Tie2 via receptor oligomerization, inducing Akt phosphorylation and inhibiting apoptosis .
Shows 80% inhibition of vascular permeability in rodent models, surpassing Ang1-expressing adenovirus efficacy .
Overcomes production limitations of recombinant Ang1, with stable pharmacokinetics in primates and rodents .
Diabetic retinopathy: Ang1/Tie2 signaling reduces retinal permeability and neovascularization .
Critical limb ischemia: ASP4021 enhances blood flow recovery in ischemic limbs .
Cancer: Ang1 modulates tumor angiogenesis, with potential therapeutic implications .
The ab133425/27093-1-AP pair enables:
ANG1 (Angiopoietin-1) is a secreted glycoprotein of approximately 70 kDa that plays crucial roles in the vascular system. It functions primarily by binding and activating the TEK/TIE2 receptor through inducing receptor dimerization and tyrosine phosphorylation. ANG1 regulates angiogenesis, endothelial cell survival, proliferation, migration, adhesion, and cell spreading. It also reorganizes the actin cytoskeleton and maintains vascular quiescence. During embryogenesis, ANG1 is required for normal angiogenesis and heart development. In adult tissues, ANG1 can either activate or inhibit angiogenesis depending on the context, inhibiting angiogenesis in quiescent vessels while promoting vascular stability . In quiescent vessels, ANG1 oligomers recruit TEK to cell-cell contacts, activating phosphatidylinositol 3-kinase and AKT1 signaling cascades, whereas in migrating endothelial cells, ANG1 recruits TEK to extracellular matrix contacts, leading to focal adhesion complex formation .
ANG1 antibody pairs typically consist of a capture antibody and a detection antibody that recognize different epitopes on the ANG1 protein, enabling quantitative measurement through ELISA or other immunoassay methods. For optimal detection, the capture antibody is immobilized on a solid surface, while the detection antibody is conjugated with an enzyme or fluorophore. In experimental settings, these antibody pairs allow researchers to quantify ANG1 secretion levels in conditioned medium or biological samples after appropriate sample processing. For instance, in TNBC research, supernatant collection at approximately 72 hours (when cell confluence reaches 90%) followed by centrifugation at 800g for 5 minutes at 4°C provides suitable samples for ANG1 quantification using commercial ELISA kits . The specificity of ANG1 detection relies on antibodies that do not cross-react with related proteins such as Angiopoietin-2, making epitope selection critical for assay development.
ANG1 serves as the primary physiological activator of the Tie2 receptor, initiating signaling through a complex mechanism dependent on oligomerization. When ANG1 binds Tie2, it induces receptor oligomerization, which is critical for activating Tie2 via auto-phosphorylation and initiating downstream signaling cascades . This interaction leads to vasodilation and enhanced blood flow through nitric oxide production in vascular endothelial cells . The nature of this interaction depends on cellular context: in quiescent vessels with tight endothelial cell contacts, ANG1 oligomers recruit Tie2 to cell-cell contacts forming complexes with Tie2 molecules from adjoining cells. In contrast, in migrating endothelial cells lacking cell-cell adhesions, ANG1 recruits Tie2 to contacts with the extracellular matrix, forming focal adhesion complexes and activating different downstream pathways including PTK2/FAK and MAPK1/ERK2 . This context-dependent signaling explains how ANG1 can both maintain vascular quiescence and stimulate sprouting angiogenesis under different conditions.
Researchers must implement multiple strategic approaches to differentiate between ANG1 and ANG2 functions in experimental designs. First, use highly specific antibodies that do not cross-react between ANG1 and ANG2, as demonstrated in direct ELISA studies that confirm binding specificity by comparing binding with Tie1 and other potential cross-reactive proteins . Second, employ genetic manipulation through knockdown and overexpression systems—utilize lentiviral vectors (like pSIH1) with validated target sequences (such as 5′-GGAAGAGAAAGAGAACCTT-3′ or 5′-CTATGATGATTCGACCTTT-3′ for ANG1) to create stable cell populations after puromycin selection . Third, implement rescue experiments where phenotypes from ANG1 knockdown are rescued by recombinant ANG1 but not ANG2. Fourth, assess differential downstream effects through pathway-specific readouts, as ANG1 and ANG2 often activate distinct signaling cascades despite targeting the same receptor. Finally, utilize cell type-specific experimental systems since ANG1 and ANG2 effects can vary dramatically between different cell types and vascular beds.
When validating ANG1 antibody pair specificity, researchers should implement a comprehensive set of controls. Positive controls should include recombinant human ANG1 protein at known concentrations to establish a standard curve and confirm detection sensitivity. Negative controls must include samples from ANG1 knockout cell lines or tissues and samples containing closely related proteins like ANG2, as demonstrated in Western blot analyses where wild-type endothelial cells (lane 1) can be compared with ANG1-expressing cells (lanes 2-3) and ANG2-expressing cells (lane 4) . Cross-reactivity testing should be performed against Tie1, another ANG1 receptor, using direct ELISA to confirm binding specificity . Additionally, validation should include samples from multiple species if the antibody claims cross-reactivity with mouse, human, or pig ANG1. Absorption controls where pre-incubation of the antibody with excess antigen neutralizes the signal provide further confirmation of specificity. For multiplex assays, potential interference between different antibody pairs should be assessed through spike-and-recovery experiments with mixed recombinant proteins.
To distinguish between autocrine and paracrine effects of ANG1, researchers should employ a multi-faceted experimental approach. First, conditioned medium (CM) transfer experiments are essential—collect CM from ANG1-overexpressing cells and control cells, then apply this CM to recipient cells to determine if secreted factors mediate the observed effects. As demonstrated with HCC1806 and MDA-MB-468 cells, comparing effects of direct ANG1 overexpression versus treatment with CM from ANG1-overexpressing cells can reveal whether effects are autocrine-dependent . Second, implement co-culture systems with physical separation using transwell inserts to allow secreted factors to diffuse while preventing direct cell contact. Third, use neutralizing antibodies against ANG1 or soluble Tie2 receptors in culture medium to sequester secreted ANG1, thereby blocking potential autocrine signaling. Fourth, employ genetic approaches like CRISPR/Cas9 to knockout Tie2 receptors in the same cells expressing ANG1 to eliminate autocrine signaling potential while maintaining paracrine capacity. Finally, utilize in vivo models with tissue-specific ANG1 expression and examine effects on neighboring tissues to validate findings in a physiological context.
The optimal methods for quantifying ANG1 protein vary based on sample type and research objectives. For cell culture supernatants, quantitative ELISA provides precise measurements—collect supernatant at approximately 72 hours post-seeding when cell confluence reaches 90%, centrifuge at 800g for 5 minutes at 4°C, and follow manufacturer protocols for commercial ANG1 ELISA kits . For tissue samples, preliminary homogenization in appropriate buffers containing protease inhibitors followed by centrifugation is necessary before ELISA application. Western blotting offers semi-quantitative analysis with the advantage of distinguishing different forms of ANG1 (native versus cleaved) using antibodies like ab8451 at 1:500 dilution . For simultaneous detection of multiple angiogenic factors including ANG1, multiplex bead-based immunoassays provide efficient analysis with minimal sample volume. In plasma or serum samples, ELISA remains the gold standard, but careful consideration of sample collection methods is crucial as platelets can release ANG1 during coagulation, potentially affecting measurements. For in situ analysis in tissue sections, immunohistochemistry with formalin-fixed paraffin-embedded samples using antibodies at 1:500 dilution followed by appropriate visualization systems can reveal localization patterns .
Researchers can effectively evaluate ANG1-mediated Tie2 activation through multiple complementary techniques. First, phosphorylation-specific Western blotting targeting Tie2 tyrosine residues provides direct evidence of activation—cell lysates should be collected with phosphatase inhibitors after ANG1 treatment at relevant timepoints (typically 5-30 minutes). Second, cellular viability assays using Tie2-expressing cell lines like Ba/F3 can assess functional activation by comparing responses to different ANG1 concentrations or antibody-based agonists . Third, downstream signaling evaluation through phosphorylation status of AKT and ERK1/2 offers insight into pathway-specific activation patterns, as ANG1 differentially activates PI3K/AKT in quiescent vessels versus MAPK pathways in migrating cells . Fourth, receptor clustering assays using fluorescently-tagged Tie2 and confocal microscopy can visualize receptor dimerization and oligomerization upon ANG1 binding. Fifth, in vivo models measuring vascular permeability provide functional readouts—ANG1 or agonistic antibodies like ASP4021 inhibit vascular hyper-permeability in dose-dependent manners . Finally, nitric oxide production measurement in endothelial cells offers a functional endpoint of ANG1-Tie2 signaling, as activation induces vasodilation through nitric oxide release .
For robust ANG1 gene expression analysis, researchers should employ a multi-level approach combining different methodologies. RT-qPCR represents the foundation for quantitative assessment using validated primer sets (forward: 5′-CGTGGAACCGGATTTCTCTTC-3′, reverse: 5′-CGAGAAGTTTGATTTAGTACCTGG-3′) with appropriate reference genes such as 18S rRNA . Sample preparation should involve total mRNA extraction using TRIzol® reagent followed by cDNA synthesis with commercial reverse transcription kits. For genome-wide context, RNA-sequencing provides comprehensive transcriptome analysis, allowing examination of ANG1 expression relative to related genes in angiogenesis pathways. When analyzing tissue samples, laser capture microdissection before RNA extraction ensures cell type-specific expression analysis. For spatial expression patterns, in situ hybridization with specific RNA probes complements expression data with localization information. For mechanistic studies, promoter analysis using luciferase reporter assays containing the ANG1 promoter region helps identify transcriptional regulators. When comparing across multiple samples, digital droplet PCR offers absolute quantification with higher precision than standard qPCR. Finally, single-cell RNA-sequencing provides cellular heterogeneity insights, particularly valuable when examining ANG1 expression across different cell populations within complex tissues.
Distinguishing between direct and indirect effects of ANG1 on cell proliferation requires a systematic experimental approach. First, pathway inhibition studies using selective inhibitors of known ANG1-activated signaling pathways (PI3K/AKT, MAPK) can help determine which downstream mediators are essential for the proliferative response. Second, temporal analysis of signaling events versus proliferative outcomes provides insights—rapid signaling events (minutes to hours) followed by delayed proliferation (days) may suggest indirect effects requiring transcriptional changes. Third, gene expression profiling after ANG1 treatment can identify secondary mediators upregulated by ANG1, such as CPA4 which has been shown to be upregulated by ANG1 and independently promotes TNBC cell proliferation through Cyclin D1 regulation . Fourth, conditioned medium experiments comparing direct ANG1 overexpression to treatment with medium from ANG1-overexpressing cells can reveal whether secreted factors mediate the effects, as demonstrated in HCC1806 and MDA-MB-468 cells where direct ANG1 overexpression promoted proliferation while conditioned medium had no effect . Fifth, receptor knockdown studies in target cells can confirm whether direct binding to cellular receptors is required. Finally, co-culture experiments with endothelial cells can determine if ANG1 effects on target cells require endothelial intermediaries.
To identify ANG1-regulated genes in cancer progression, researchers should implement a comprehensive analytical pipeline. First, conduct RNA-sequencing on paired samples (ANG1-knockdown/overexpression versus controls) across multiple timepoints to capture early, intermediate, and late response genes. Second, perform pathway enrichment analysis using tools like GSEA, KEGG, and Reactome to identify significantly altered biological processes. Third, validate key candidates through RT-qPCR with specific primers, as demonstrated for CPA4 (forward: 5′-CTGGACGGCAAGGAAGATTGT-3′, reverse: 5′-GACCGCGTCTTCCTCCATAA-3′) . Fourth, employ chromatin immunoprecipitation followed by sequencing (ChIP-seq) for transcription factors activated downstream of ANG1 to identify direct transcriptional targets. Fifth, integrate multi-omics data including proteomics and phosphoproteomics to connect transcriptional changes with functional protein alterations. Sixth, perform correlation analyses using clinical databases like TCGA to identify genes whose expression patterns parallel ANG1 in patient samples and correlate with survival outcomes . Seventh, conduct functional validation through siRNA/shRNA knockdown of candidate genes to determine if they phenocopy ANG1 effects, as demonstrated with CPA4 knockdown which resulted in substantial downregulation of Cyclin D1 and inhibition of TNBC cell proliferation . Finally, create in vivo models with modulated expression of both ANG1 and key target genes to validate their relationships in physiologically relevant systems.
Resolving discrepancies between ANG1 mRNA and protein expression requires systematic investigation of potential biological and technical factors. First, examine post-transcriptional regulation mechanisms—ANG1 mRNA may be subject to microRNA-mediated suppression or altered stability affecting translation efficiency. Second, consider protein half-life and turnover rates, as proteins with long half-lives may remain detectable despite decreased mRNA levels. Third, evaluate secretion dynamics—as ANG1 is a secreted glycoprotein, cellular protein levels may not reflect total production if secretion rates vary; analyze both cell lysates and conditioned media in parallel . Fourth, verify technical aspects of both assays—for mRNA detection, confirm primer specificity and efficiency using appropriate controls; for protein detection, validate antibody specificity using positive controls (ANG1-expressing cells) and negative controls (wild-type or ANG1-knockdown cells) . Fifth, examine post-translational modifications affecting antibody recognition—glycosylation patterns of ANG1 may vary between cell types, potentially altering epitope accessibility. Sixth, perform time-course analyses to identify temporal relationships between mRNA induction and subsequent protein accumulation. Finally, consider cellular heterogeneity in mixed populations—single-cell analyses may reveal subpopulations with divergent expression patterns masked in bulk analyses.
Interpreting ANG1 knockout/knockdown phenotypes requires awareness of several common pitfalls. First, compensation by related proteins—ANG2 may functionally compensate for ANG1 loss in certain contexts, potentially masking phenotypes. Second, consider incomplete knockdown efficiency—residual ANG1 expression may be sufficient to maintain partial function, particularly in dose-sensitive processes; verification of knockdown efficiency at both mRNA and protein levels is essential . Third, be aware of potential off-target effects—siRNA or shRNA approaches may affect expression of unintended genes; use multiple targeting sequences and rescue experiments with exogenous ANG1 expression to confirm specificity . Fourth, recognize context-dependency of ANG1 function—phenotypes may vary dramatically between cell types, tissues, or physiological states; ANG1 inhibits angiogenesis in quiescent vessels but promotes it in actively remodeling vasculature . Fifth, distinguish between developmental versus adult phenotypes—acute depletion in adult tissues may produce different outcomes than germline deletion affecting development. Sixth, examine cell-autonomous versus non-cell-autonomous effects—ANG1 functions in both autocrine and paracrine manners . Finally, consider compensatory pathway activation—alternative signaling mechanisms may be upregulated following chronic ANG1 depletion, potentially obscuring direct consequences of ANG1 loss.
Differentiating between specific and non-specific effects in ANG1 antibody-based therapeutic approaches requires comprehensive control experiments and validation strategies. First, implement isotype control antibodies matched to the therapeutic antibody class to identify Fc-mediated effects independent of antigen binding. Second, use multiple antibodies targeting different ANG1 epitopes—consistent phenotypes across different antibodies suggest specificity, while discrepancies may indicate epitope-specific or off-target effects. Third, perform parallel studies with genetic ANG1 manipulation (knockdown/knockout) to confirm that antibody-induced phenotypes match genetic depletion outcomes . Fourth, conduct dose-response studies to establish relationship between antibody concentration and biological effects—specific effects typically show saturable dose-response patterns while non-specific effects often increase linearly with concentration . Fifth, verify target engagement through analysis of downstream signaling (phospho-Tie2, AKT, ERK) or competitive binding assays with labeled ANG1. Sixth, examine effects in cells lacking Tie2 receptor expression—persistence of effects suggests off-target activity. Finally, for therapeutic applications, compare efficacy between multivalent formats (tetravalent antibodies like ASP4021) and bivalent or monovalent formats, as receptor clustering requirements for activation provide a specificity check—naturally oligomeric ANG1 induces Tie2 clustering for activation, which is mimicked by higher valency antibody formats but not by monovalent antibodies .
Engineered antibodies offer several advantages over natural ANG1 for therapeutic applications. First, multivalent antibody design enhances receptor activation—tetravalent antibodies like ASP4021 that fuse two Fab regions head-to-tail demonstrate potent agonistic activity comparable to natural ANG1, while monovalent antibodies show weak activity . Second, engineered antibodies provide superior stability and longer half-life—purified ANG1 and higher multimeric antibody fractions are generally unstable for therapeutic use, whereas engineered tetravalent antibodies demonstrate good pharmacokinetic properties in animal models with expected half-lives of 10-14 days in humans . Third, manufacturing scalability is improved—ASP4021 can be efficiently produced using standard CHO and 293 cell systems commonly used for mass production of therapeutic antibodies, overcoming the manufacturability limitations of ANG1 . Fourth, species cross-reactivity can be engineered—ASP4021 demonstrates binding activity to Tie2 from multiple species, allowing preclinical testing in relevant animal models, unlike previous anti-Tie2 antibodies (15B8 and 1-4h) that lacked rodent Tie2 binding . Fifth, targeted specificity minimizes off-target effects—ASP4021 shows no binding to Tie1, another ANG1 receptor, confirming binding specificity . Finally, engineered antibodies allow modular design—additional domains can be incorporated to enhance tissue targeting or provide complementary functions beyond receptor activation.
Advancing understanding of ANG1's context-dependent functions requires innovative experimental approaches. First, organ-on-chip and 3D organoid models incorporating multiple cell types (endothelial cells, pericytes, tissue-specific cells) with controlled ANG1 expression can recreate physiological microenvironments more accurately than traditional 2D cultures. Second, inducible, cell type-specific ANG1 expression systems using Cre-loxP or tetracycline-responsive elements in transgenic animal models allow temporal and spatial control of ANG1 expression to dissect tissue-specific functions. Third, single-cell multi-omics approaches combining transcriptomics, proteomics, and metabolomics at single-cell resolution can identify cell-specific responses to ANG1 within heterogeneous tissues. Fourth, CRISPR-based lineage tracing with ANG1 reporter systems can track the fate of ANG1-responsive cells during development or disease progression. Fifth, intravital microscopy with fluorescently tagged ANG1 and Tie2 enables real-time visualization of ligand-receptor interactions in living tissues. Sixth, proximity labeling techniques like BioID or APEX2 fused to ANG1 or Tie2 can identify context-specific protein interaction networks in different tissues. Finally, biomaterial-based approaches using hydrogels with controlled release of ANG1 can create defined gradients to study dose-dependent and spatial aspects of ANG1 signaling in tissue-specific contexts, particularly valuable for understanding its dual roles in maintaining vascular quiescence versus promoting angiogenesis under different conditions .
Different ANG1 antibody pair formats demonstrate varying performance characteristics that significantly impact research outcomes. The table below provides a comparative analysis:
Monoclonal/monoclonal pairs offer superior reproducibility and specificity for quantitative analysis but may have limited epitope recognition. Polyclonal-based formats provide broader epitope coverage but with potential specificity tradeoffs. For activation studies, tetravalent formats like ASP4021 demonstrate similar potent agonistic activity to natural ANG1 while monovalent formats show weak activation capacity . The format selection should be guided by experimental goals, with validation using appropriate positive controls (ANG1-expressing cells) and negative controls (wild-type endothelial cells or ANG1-knockdown cells) .
Researchers should implement comprehensive standardization strategies when comparing ANG1 expression across experimental systems. First, establish reference standards—include recombinant ANG1 protein standards of known concentration in every assay and develop stable cell lines with defined ANG1 expression levels as inter-experimental calibrators. Second, normalize with appropriate reference genes for qPCR—validate multiple candidate reference genes (such as 18S rRNA used in TNBC studies) for stability across the specific experimental conditions being compared . Third, standardize sample collection protocols—for secreted ANG1, collect conditioned medium at consistent timepoints relative to cell confluence (approximately 72 hours when confluence reaches 90%) and standardize centrifugation parameters (800g for 5 minutes at 4°C) . Fourth, implement cross-platform validation—confirm findings using multiple detection methods (ELISA, Western blot, mass spectrometry) with defined cutoffs for significant differences. Fifth, establish reporting standards—document key experimental parameters including antibody catalog numbers, dilutions, incubation conditions, and detection systems. Sixth, utilize digital PCR for absolute quantification when comparing across laboratories or platforms. Finally, consider biological context standardization—match cell densities, passage numbers, growth conditions, and tissue processing methods when comparing between different experimental systems to minimize variables that can affect ANG1 expression independent of the biological phenomenon being studied.
When designing ANG1 antibody-based experimental systems, researchers must prioritize several critical considerations. Antibody format selection significantly impacts results—tetravalent antibodies demonstrate agonistic activity similar to natural ANG1, while monovalent antibodies show weak activation capacity, making format selection crucial for functional studies . Specificity validation is essential—confirm absence of cross-reactivity with related proteins like ANG2 and Tie1 through direct binding assays . Experimental controls must be comprehensive—include isotype controls, ANG1-overexpressing positive controls, knockdown negative controls, and cross-validation with non-antibody methods . Context-dependency awareness is vital—ANG1 functions differ dramatically between quiescent and remodeling vasculature, requiring careful consideration of the cellular and tissue environment . Technical parameters require optimization—for Western blotting with ab8451, use 1:500 dilution; for ELISA, collect supernatant at ~72 hours with standardized processing . Species cross-reactivity must be verified experimentally—even antibodies with predicted cross-reactivity require validation in each species of interest . Finally, result interpretation demands integration of antibody-based findings with genetic approaches—compare antibody neutralization/activation results with knockdown/overexpression phenotypes to distinguish specific from non-specific effects and understand underlying biology rather than technical artifacts .