ANG Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Generally, we can ship your orders within 1-3 business days after receiving them. Delivery times may vary depending on the purchase method or location. Please consult your local distributors for specific delivery times.
Synonyms
ALS9 antibody; AMYOTROPHIC LATERAL SCLEROSIS antibody; ANG 1 antibody; ANG antibody; ANG I antibody; ANG1 antibody; ANGI antibody; ANGI_HUMAN antibody; Angiogenin antibody; Angiogenin ribonuclease RNase A family; 5 antibody; Epididymis luminal protein 168 antibody; HEL168 antibody; MGC22466 antibody; MGC71966 antibody; Ribonuclease 5 antibody; Ribonuclease RNase A Family 5 antibody; RNase 5 antibody; RNASE4 antibody; RNASE5 antibody
Target Names
ANG
Uniprot No.

Target Background

Function
Angiogenin binds to actin on the surface of endothelial cells. Once bound, it is endocytosed and translocated to the nucleus. Angiogenin stimulates ribosomal RNA synthesis, including the initiation site sequences of 45S rRNA. It also cleaves tRNA within anticodon loops to produce tRNA-derived stress-induced fragments (tiRNAs) that inhibit protein synthesis and trigger the assembly of stress granules (SGs). Angiogenin induces vascularization of normal and malignant tissues. Its angiogenic activity is regulated by interaction with RNH1 in vivo.
Gene References Into Functions
  • Angiogenin contributes to angiogenesis induced by Brain-derived neurotrophic factor (BDNF). PMID: 29573867
  • Phospholipase D2 (PLD2)-produced phosphatidic acid (PA) promotes cell invasion through the expression of angiogenin (ANG) in clear cell renal cell carcinoma (ccRCC) cells. PMID: 29660846
  • PCNA is predominantly localized in the cytoplasm, while hAng is distributed both in the nucleus and in the cytoplasm. hAng and PCNA colocalize in the cytoplasm, suggesting a potential interaction in this compartment. PMID: 28777577
  • RNase 5 suppresses p27 and up-regulates cyclin D1, D3, and E by activating PI3-kinase/Akt in CECs to initiate cell cycle progression. PMID: 27526633
  • When yeast genetic interaction partners held in common between human OPTN and ANG were validated in mammalian cells and zebrafish, MAP2K5 kinase emerged as a potential drug target for amyotrophic lateral sclerosis therapy. PMID: 28596290
  • There was a significant association between RNase 5 and histological differentiation in colon adenocarcinomas, but no association between RNase 5 and Necl 4 in gastric or colon adenocarcinomas. PMID: 28561015
  • ANG is established as an oncoprotein. Further research reveals that ANG contributes to oncogenesis by activating MMP2 through modulation of DNMT3b functions. PMID: 27317771
  • The RRR(33) and (50) KRSIK(54) motifs of angiogenin might play a crucial role in regulating p53-mediated apoptosis and angiogenesis in cancer cells. PMID: 29105754
  • Plexin-B2 (PLXNB2) is the functional receptor for ANG in endothelial, cancer, neuronal, and normal hematopoietic and leukemic stem and progenitor cells. PMID: 29100074
  • Delivery of the therapeutic human genes VEGF and ANG using an adenoviral vector improved functional recovery after traumatic spinal cord injury in a rat model. Immunofluorescent analysis of the post-treatment spinal cord suggests that the positive effect of Ad5-VEGF+Ad5-ANG transduction on recovery of locomotor function may be due to the action of glial cells on motor neurons. PMID: 28452633
  • Ang down-regulates the expression of Col-I, alpha-SMA and TGF-beta1/Smad2/3 and subsequently inhibits fibroblast-myofibroblast transition. PMID: 27543459
  • Ang, Tie1 and Tie2 play roles in vascular development and pathogenesis of vascular diseases. [review] PMID: 27941161
  • Results indicate that angiogenin (ANG) plays a critical role in regulating angiogenesis and follicle survival in xenografted human ovarian tissues. PMID: 28274269
  • ANG plays a significant role in EMT in squamous cell lung carcinoma and may be a valuable therapeutic target for this type of cancer. PMID: 27667357
  • There is no association between the Angiogenin (ANG) rs11701 polymorphism and amyotrophic lateral sclerosis risk. PMID: 26753798
  • ANG plays a non-cell-autonomous role in regulating hematopoiesis by simultaneously preserving hematopoietic stem/progenitor cells stemness and promoting myeloid-restricted progenitor cell proliferation. PMID: 27518564
  • ANG K17I variant is rare in Caucasian patients and controls and increases the risk for amyotrophic lateral sclerosis. PMID: 26255299
  • ANG rs11701 variant is a genetic risk factor for Parkinson's disease in the Taiwanese population. Mutations in ANG are not a common cause for idiopathic Parkinson's disease. PMID: 25386690
  • Angiogenin secretion by HCCs favors tumor development by inducing HSC activation and ECM remodeling. PMID: 25604905
  • The present study established a link between Ang and FHL3 proteins and identifies a new pathway for regulating astrocytoma progression. PMID: 25659096
  • The mechanism of ligand binding in human angiogenin was studied using the NMR chemical shift projection analysis. PMID: 25450558
  • ANG could play a pivotal role in the development of bladder cancer through regulating AKT/mTOR signaling pathway. PMID: 25564356
  • A novel regulatory pathway is suggested whereby breast tumor-derived angiogenin directly activates angiogenesis through inhibition of miR-542-3p. PMID: 26272182
  • A biological explanation is provided for the loss-of-function of D22G-Angiogenin leading to ALS, and it is suggested that the L35P-Angiogenin mutation would likely cause ALS symptoms in individuals harboring this mutation. PMID: 25372031
  • High angiogenin expression is associated with bladder cancer. PMID: 24696417
  • A novel regulatory pathway is reported whereby ANG regulates PI3K/AKT/mTOR signaling pathway via RI. PMID: 25193113
  • Up-regulated ANG expression and increased nuclear translocation were observed in cervical cancers compared to normal cervical tissues. Upregulation of ANG was positively correlated with primary tumor invasion. PMID: 25552400
  • Findings support an unrecognized interplay between ANG, ERK1/2 and MMP2 that can impact tumor growth and progression. PMID: 24561529
  • The observed pattern of angiogenin expression is compatible with a role in blood vessel formation and in cross-talk between trophoblasts and endothelial cells. PMID: 25093183
  • Serum angiogenin levels may be useful to distinguish between cancer and noncancer patients among the candidates for prostatic biopsy in regular clinical practice. PMID: 23921786
  • ANG interacts with plasminogen activation system at the leading edges of breast cancer cell surfaces and facilitates interactions of uPAR with uPA to regulate plasmin formation and cell migration. PMID: 24457100
  • Amyloidogenic ApoA-I induces cell death through attenuating the anti-stress activity of angiogenin. PMID: 24603325
  • Angiogenin was up-regulated in proliferative hemangiomas. Angiogenin serum levels correlate with the hemangioma stage. Angiogenin levels discriminate between proliferative hemangiomas and the control group and patients with venous malformations. PMID: 25068345
  • Depletion of cellular ANG expression abolished PLSCR1-enhanced rRNA transcription. PMID: 24356419
  • Human angiogenin has antimicrobial activity against C. albicans and S. pneumoniae. PMID: 12548285
  • Data show that the transcription of angiogenin (ANG) and ribonuclease 4 (RNASE4) promoter is influenced by RNA polymerase III (Pol III) elements and could be differentially regulated by an intragenic CCCTC binding factor (CTCF)-dependent chromatin loop. PMID: 24659782
  • Data show that ANG-stimulated rRNA transcription is not only an essential component for androgen-dependent growth of prostate cancer but also contributes to the transition of prostate cancer from androgen-dependent to castration-resistant growth status. PMID: 23851444
  • Nuclear ANG directly binds to the ANG-Binding Sequence within ERRgamma of ERRgamma gene and inhibits ERRgamma transcription to promote breast cancer cell proliferation. PMID: 23977052
  • These data suggest that one of the mechanisms by which ANG stimulates rRNA transcription is through an epigenetic activation of rDNA promoter. PMID: 24122807
  • Changes of angiogenin in the skin of patients with amyotrophic lateral sclerosis are related to the disease process. PMID: 23351638
  • Overexpression of ANG in pterygium body fibroblasts might be involved in active pterygium growth with thick pterygium body formation and increased risk of recurrence. PMID: 23989187
  • Our data do not support the association of angiogenin variants with Parkinson's disease in Han Chinese of mainland China. PMID: 23231972
  • Increased angiogenin may contribute to decreased T-cell zeta-chain expression in hemodialysis patients. PMID: 23379493
  • A fast molecular dynamics based method for determining the mechanisms of functional loss caused by mutations. PMID: 23665167
  • Angiogenin is up-regulated in the hypoxic environment of oral squamous cell carcinoma. PMID: 22694909
  • Dysregulated angiogenin expression may contribute to the pathogenesis of myositis as well as skin involvement via the vascular change in DM/CADM. PMID: 23229115
  • Analysis of Angiogenin in middle-aged type 1 diabetes patients. PMID: 22940420
  • Angiogenin interacts with p53 and colocalizes in the nucleus. PMID: 22266868
  • In Alzheimer disease, serum angiogenin was decreased and cognitive function was associated with correlated with angiogenin levels. PMID: 22449478
  • Serum angiogenin levels in cutaneous T cell lymphoma (CTCL) patients were significantly higher than healthy controls. Angiogenin mRNA expression levels in lesional skin were elevated in erythrodermic CTCL compared to normal skin. Results suggest enhanced angiogenin expression may be related with poor prognosis of erythrodermic CTCL. PMID: 22526325

Show More

Hide All

Database Links

HGNC: 483

OMIM: 105850

KEGG: hsa:283

STRING: 9606.ENSP00000336762

UniGene: Hs.283749

Involvement In Disease
Amyotrophic lateral sclerosis 9 (ALS9)
Protein Families
Pancreatic ribonuclease family
Subcellular Location
Cytoplasmic vesicle, secretory vesicle lumen. Secreted. Nucleus. Nucleus, nucleolus.
Tissue Specificity
Expressed predominantly in the liver. Also detected in endothelial cells and spinal cord neurons.

Q&A

What is ANG antibody and what cellular processes does it help investigate?

ANG antibody refers to antibodies that specifically recognize and bind to Angiogenin, a protein involved in blood vessel formation and RNA metabolism. These antibodies serve as critical reagents for investigating angiogenesis, tumor development, neurological disorders, and immune responses. The specificity of antibody-antigen interactions allows researchers to detect, quantify, and visualize ANG in various biological samples with high precision . ANG antibodies can be used to study how angiogenin mediates cellular responses including endothelial cell proliferation, migration, and the formation of tubular structures that eventually develop into blood vessels. Additionally, these antibodies help investigate ANG's role in stress responses and its nuclear translocation in various disease states.

How do ANG antibodies differ in terms of isotypes and how does this affect experimental design?

ANG antibodies are available in various isotypes (IgG, IgM, IgA) and subclasses (IgG1, IgG2, etc.), each with distinct properties that influence experimental applications. The isotype selection impacts:

  • Tissue penetration capabilities

  • Complement activation potential

  • Binding to Fc receptors

  • Half-life in experimental systems

  • Secondary antibody compatibility

For research applications, IgG antibodies are most commonly used due to their stability and versatility . When designing experiments, researchers must consider that different isotypes may yield varying results in techniques such as immunoprecipitation, Western blotting, or immunohistochemistry. For example, IgM antibodies with their pentameric structure provide high avidity but may cause steric hindrance in some applications, while IgG antibodies generally offer better specificity for individual epitopes. The selection should be guided by the specific requirements of the experimental technique and the biological questions being addressed.

What are the key considerations when selecting between monoclonal and polyclonal ANG antibodies?

The choice between monoclonal and polyclonal ANG antibodies significantly impacts experimental outcomes:

CharacteristicMonoclonal ANG AntibodiesPolyclonal ANG Antibodies
SpecificityHigh specificity for a single epitopeRecognize multiple epitopes on ANG
Batch consistencyHighly consistent between batchesMay vary between batches
Signal strengthMay provide lower signalOften provides stronger signal through multiple binding sites
Cross-reactivityLess prone to cross-reactivityPotentially higher cross-reactivity
ApplicationsIdeal for epitope-specific studiesBetter for detection in complex samples

Monoclonal antibodies are produced from a single B-cell clone and recognize only one epitope, making them highly specific but potentially limiting detection if the epitope is masked or altered . Polyclonal antibodies, derived from multiple B-cell clones, recognize various epitopes, increasing detection probability but potentially reducing specificity . For asymmetrical assays, researchers often use monoclonal antibodies for capture and polyclonal antibodies for detection to achieve high specificity during capture and maximum detection sensitivity .

How can ANG antibodies be effectively utilized in ELISA-based quantification of angiogenin levels?

ELISA represents one of the most robust methods for quantifying ANG in biological samples using specific antibodies. For effective implementation:

  • Selection of ELISA format: Sandwich ELISA is preferred for ANG detection, using a capture antibody immobilized on a plate to bind ANG, followed by a detection antibody conjugated to an enzyme . This format offers superior sensitivity and specificity compared to direct or indirect ELISA formats.

  • Antibody optimization: Titrate both capture and detection antibodies to determine optimal concentrations (typically 0.5–5 μg/mL) . This optimization is critical for achieving maximum sensitivity while minimizing background signal.

  • Sample preparation considerations: Biological samples should be properly processed to remove interfering substances. For serum/plasma samples, dilution in appropriate buffers helps reduce matrix effects.

  • Standard curve development: Prepare a standard curve using recombinant ANG protein at concentrations spanning the expected range in samples, typically 0-1000 pg/mL. The curve should demonstrate a clear dose-response relationship with R² values exceeding 0.98.

  • Signal development and detection: After adding the appropriate substrate, the enzymatic reaction produces a colored or luminescent product proportional to ANG concentration. For quantitative analysis, measure absorbance using a spectrophotometer at the wavelength appropriate for the substrate used .

The sandwich ELISA setup particularly benefits from using antibodies recognizing different epitopes of ANG, with capture antibodies targeting stable structural regions and detection antibodies targeting unique sequences for specificity.

What are the optimal conditions for using ANG antibodies in Western blotting applications?

Western blotting with ANG antibodies requires careful optimization of multiple parameters:

  • Sample preparation: Extract proteins using buffers containing appropriate protease inhibitors to prevent ANG degradation. For intracellular ANG, complete cell lysis is essential.

  • Gel selection: Use 12-15% SDS-PAGE gels for optimal resolution of ANG (approximately 14 kDa).

  • Transfer conditions: Employ wet transfer methods for more consistent results with small proteins like ANG. Transfer at 100V for 1 hour or 30V overnight at 4°C to PVDF or nitrocellulose membranes .

  • Blocking optimization: Use 5% non-fat dry milk or BSA in TBST (Tris-buffered saline with 0.1% Tween-20) for 1 hour at room temperature to minimize background.

  • Antibody dilution and incubation: Primary ANG antibodies typically work best at dilutions between 1:500 and 1:2000. Overnight incubation at 4°C generally yields the best signal-to-noise ratio .

  • Detection method selection: Chemiluminescence detection offers superior sensitivity for ANG compared to colorimetric methods, especially when protein expression is low .

  • Stripping and reprobing considerations: PVDF membranes allow for membrane stripping and reprobing but may generate more background compared to nitrocellulose membranes .

When troubleshooting, non-specific bands can be addressed by increasing antibody dilution or using more stringent washing conditions. For weak signals, longer primary antibody incubation or signal enhancement systems may improve results.

How can ANG antibodies be effectively employed in immunohistochemistry or immunofluorescence studies?

For successful visualization of ANG in tissue or cellular samples:

  • Fixation optimization: For ANG detection, 4% paraformaldehyde typically preserves antigen integrity better than formalin. Fixation time should be optimized to maintain epitope accessibility without compromising tissue morphology.

  • Antigen retrieval methods: Heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) often improves ANG detection in paraffin sections. For each ANG antibody, compare different retrieval methods to determine optimal conditions.

  • Antibody validation: Confirm specificity through positive and negative controls. Use tissues known to express ANG (like placenta or tumor tissues) as positive controls and antibody diluent without primary antibody as negative controls.

  • Signal amplification considerations: For weak ANG signals, employ tyramide signal amplification or higher sensitivity detection systems like polymer-based detection methods.

  • Multiplexing strategies: For co-localization studies, select ANG antibodies from different host species to allow simultaneous detection with other proteins of interest. Secondary antibodies with minimal cross-reactivity are essential for accurate results.

  • Counterstaining selection: For immunofluorescence, DAPI provides nuclear counterstaining without interfering with ANG detection. For IHC, hematoxylin concentration should be optimized to avoid masking specific ANG staining.

When interpreting results, distinguish between extracellular and intracellular ANG staining patterns, as they have different biological implications. Nuclear localization of ANG often indicates stress response activation, while extracellular staining may reflect secreted ANG involved in angiogenesis.

How can computational modeling be integrated with ANG antibody experimental data to design antibodies with customized specificity profiles?

Integrating computational approaches with experimental antibody data represents a cutting-edge strategy for designing ANG antibodies with precise specificity:

  • Binding mode identification: Computational models can identify distinct binding modes associated with ANG epitopes, allowing researchers to disentangle complex binding patterns that cannot be experimentally dissociated . This approach is particularly valuable when designing antibodies that must discriminate between closely related epitopes on ANG and similar proteins.

  • Biophysics-informed modeling: These models incorporate thermodynamic principles to describe the energetics of antibody-antigen interactions. For ANG antibodies, this allows prediction of binding properties based on sequence information . The model represents binding affinity (Δ𝐺) as the sum of individual amino acid contributions, accounting for context-dependent effects where neighboring residues influence binding.

  • Specificity optimization: Mathematical optimization of energy functions associated with different binding modes enables computational generation of novel antibody sequences with desired specificity profiles:

    • For cross-specific ANG antibodies that recognize multiple forms of ANG, minimize energy functions for desired ligands

    • For highly specific antibodies, minimize energy for desired epitopes while maximizing energy for undesired epitopes

  • Experimental validation strategies: After computational design, antibodies must be validated experimentally through:

    • Affinity measurements using surface plasmon resonance

    • Epitope mapping to confirm binding to target regions

    • Cross-reactivity testing against related proteins

    • Functional assays to verify intended biological activity

This computational-experimental pipeline has demonstrated success in designing antibodies that can discriminate between chemically similar ligands, a capability critical for advanced ANG research where distinguishing between different forms or modified versions of ANG is essential .

What approaches can be used to develop ANG antibodies capable of distinguishing between post-translationally modified forms of angiogenin?

Developing antibodies that differentiate between post-translational modifications (PTMs) of ANG requires sophisticated approaches:

  • Modification-specific immunogen design: Generate peptides containing specific PTMs (phosphorylation, glycosylation, etc.) at known modification sites on ANG. The strategic design incorporates:

    • Sufficient sequence length (typically 10-15 amino acids)

    • Central positioning of the modified residue

    • Conjugation to carrier proteins (KLH or BSA) to enhance immunogenicity

  • Differential screening strategies: Implement parallel screening against modified and unmodified ANG to identify clones with the desired selectivity profile:

    • ELISA-based screens using modified and unmodified peptides

    • Competition assays to determine relative affinities

    • Western blot validation with controlled samples

  • Epitope refinement techniques: For antibodies showing partial selectivity, affinity maturation through phage display can enhance specificity:

    • Start with modestly selective antibodies

    • Create targeted mutation libraries focused on CDR regions

    • Apply stringent selection with negative selection steps against unmodified ANG

    • Validate improved clones through comprehensive binding analysis

  • Validation in biological contexts: Confirm specificity in cellular contexts where PTMs occur naturally:

    • Induction of specific modifications through cellular stimuli

    • Treatment with modification-specific enzymes

    • Correlation with mass spectrometry data to verify detection accuracy

This approach has successfully generated antibodies that can distinguish between phosphorylated and non-phosphorylated forms of ANG, as well as antibodies specific for ANG with oxidized versus reduced disulfide bonds—distinctions critical for understanding ANG's differential functions in normal versus stress conditions.

How can ANG antibodies be employed to investigate the differential roles of ANG in normal physiology versus pathological angiogenesis?

Investigating ANG's dual roles in normal physiology and pathological conditions requires sophisticated antibody-based approaches:

  • Epitope-specific targeting: Select antibodies targeting different functional domains of ANG:

    • Nuclear localization signal (NLS) domain antibodies to block nuclear translocation

    • RNase active site antibodies to inhibit ribonucleolytic activity

    • Receptor-binding domain antibodies to prevent cellular internalization

    Each type of antibody provides insights into different functional aspects of ANG in normal versus pathological settings.

  • Temporal expression pattern analysis: Use antibodies in time-course experiments to track ANG expression, localization, and activity:

    • In development using embryonic tissues

    • During wound healing processes

    • In tumor progression models

    • Under hypoxic conditions mimicking pathological states

  • Multi-parameter flow cytometry: Employ fluorescently labeled ANG antibodies in combination with markers for:

    • Endothelial activation (CD31, VEGFR2)

    • Cell cycle progression

    • Apoptosis markers

    • Stress response indicators

    This approach allows correlation of ANG levels with cellular states in heterogeneous populations.

  • Proximity ligation assays: Identify protein-protein interactions that differ between normal and pathological conditions:

    • ANG interaction with receptors (e.g., syndecan-4)

    • Association with translation machinery components

    • Binding to stress-induced partners

  • Therapeutic intervention assessment: Evaluate ANG-targeting antibodies as potential therapeutics:

    • Neutralizing antibodies to block extracellular functions

    • Intrabodies directed against specific intracellular pools

    • Bispecific antibodies linking ANG to immune effector mechanisms

These approaches have revealed that while ANG promotes controlled angiogenesis in normal tissues, its upregulation and altered localization in tumors contribute to unregulated vessel formation. Additionally, the nuclear translocation of ANG, which can be monitored using domain-specific antibodies, has been shown to provide stress resistance to cancer cells but serves physiological roles in neuronal survival under normal conditions.

What strategies can overcome common challenges in ANG antibody specificity and cross-reactivity testing?

Ensuring ANG antibody specificity presents several challenges that can be addressed through systematic approaches:

  • Comprehensive cross-reactivity assessment: Test against related proteins in the ribonuclease superfamily:

    • RNase A (shares structural similarities)

    • Other angiogenic factors (VEGF, bFGF)

    • Species orthologs to identify conservation of epitope recognition

    Cross-reactivity testing should employ multiple techniques (ELISA, Western blot, IHC) as reactivity can differ between denatured and native forms.

  • Epitope mapping for antibody characterization: Determine the exact binding regions using:

    • Peptide arrays covering the entire ANG sequence

    • Mutational analysis of key residues

    • Hydrogen-deuterium exchange mass spectrometry

    • X-ray crystallography for definitive structural information

  • Competitive binding assays: Design assays where unlabeled antibodies compete with labeled reference antibodies for binding to ANG. The resulting competition patterns create a "fingerprint" that clusters antibodies binding similar epitopes .

  • Knockout/knockdown validation approach: Test antibodies on:

    • ANG-knockout cell lines or tissues (negative controls)

    • Cells with graded expression through siRNA knockdown

    • Overexpression systems with defined ANG variants

  • Application-specific validation: Recognize that an antibody performing well in one application may not work in others:

    • For each application (Western blot, IHC, ELISA), conduct separate validation

    • Document optimal conditions for each application

    • Create validation panels representing expected expression ranges

These strategies help establish a validation hierarchy, where antibodies are progressively tested through increasingly stringent and physiologically relevant systems. For high-confidence research applications, antibodies should demonstrate specificity across multiple validation approaches.

How can researchers troubleshoot inconsistent results when using ANG antibodies across different experimental platforms?

Inconsistent results with ANG antibodies across platforms can be methodically addressed:

  • Epitope accessibility variations: Different techniques expose different aspects of the ANG protein:

    • Western blotting detects denatured epitopes

    • ELISA may recognize both native and denatured forms

    • Immunohistochemistry requires epitopes to be accessible in fixed tissues

    Solution: Map which epitopes your antibody recognizes and whether they remain accessible in different sample preparation methods. For comprehensive analysis, use multiple antibodies targeting different regions of ANG.

  • Sample preparation standardization: Develop consistent protocols for:

    • Protein extraction buffers (detergent types, salt concentration)

    • Fixation methods and duration

    • Antigen retrieval procedures

    • Storage conditions of prepared samples

  • Antibody performance tracking: Implement quality control measures:

    • Aliquot antibodies to minimize freeze-thaw cycles

    • Include consistent positive and negative controls in every experiment

    • Maintain detailed records of antibody lot numbers and performance

    • Regularly test antibody activity against standard samples

  • Matrix effect mitigation: Address interference from sample components:

    • For serum/plasma, pre-absorb antibodies with control samples

    • Use blockers specific to the sample type (e.g., normal serum from antibody host species)

    • Optimize antibody concentration for each sample type

    • Consider sample pre-clearing for complex biological fluids

  • Instrument and detection system calibration: Ensure consistent instrument performance:

    • Regular calibration of imaging systems

    • Standardized settings for image acquisition

    • Consistent exposure times for chemiluminescence detection

    • Use of reference standards for quantitative measurements

By systematically addressing these variables, researchers can identify which factors most significantly impact their specific ANG antibody applications and develop standardized protocols that deliver consistent results across experimental platforms.

What considerations are important when analyzing contradictory data obtained from different ANG antibodies in the same experimental system?

Contradictory results from different ANG antibodies require careful analytical approaches:

  • Epitope mapping and antibody characterization: Determine precisely where each antibody binds:

    • Different antibodies may recognize distinct ANG forms (precursor vs. mature)

    • Some antibodies may detect specific conformational states

    • Certain epitopes may be masked by protein-protein interactions in vivo

    This mapping helps interpret seemingly contradictory results as potentially reflecting biologically relevant differences in ANG forms or states.

  • Validation hierarchy implementation: Establish relative reliability of conflicting results through:

    • Correlation with orthogonal detection methods (mass spectrometry, RNA expression)

    • Functional validation using knockdown/knockout systems

    • Testing in multiple cell lines or tissue types

    • Comparison with published literature using the same antibodies

  • Quantitative analysis of discrepancies: Distinguish between qualitative and quantitative differences:

    • Do antibodies show different localization patterns?

    • Are there consistent proportional differences in signal intensity?

    • Do discrepancies appear only under specific experimental conditions?

    Such analysis can reveal whether contradictions reflect technical issues or actual biological complexity.

  • Comprehensive controls design: Implement controls addressing specific contradiction sources:

    • Pre-absorption controls to verify specificity

    • Peptide competition assays to confirm epitope-specific binding

    • Isotype-matched irrelevant antibodies to assess non-specific binding

    • Sequential probing of the same samples with different antibodies

  • Integrated data interpretation: Develop frameworks that incorporate seemingly contradictory results:

    • Construct models accommodating different ANG forms or states

    • Consider temporal dynamics that might explain discrepancies

    • Evaluate whether contradictions reflect technical limitations or novel biology

This systematic approach often reveals that contradictions actually reflect biological complexity rather than technical failures. For example, antibodies recognizing different ANG epitopes have revealed distinct subcellular pools of ANG with different functions—cytoplasmic ANG involved in ribosomal RNA processing versus nuclear ANG mediating stress responses .

How might computational inference and machine learning advance the design of next-generation ANG antibodies?

The integration of computational approaches with experimental antibody development represents a revolutionary frontier for ANG antibody research:

  • Deep learning sequence-structure-function prediction: Advanced neural networks can now:

    • Predict antibody binding properties from sequence information

    • Identify optimal combinations of CDR sequences for specific ANG epitopes

    • Generate novel antibody sequences with predicted specificity profiles

    • Reduce experimental screening requirements by pre-filtering candidates in silico

  • Biophysics-informed modeling frameworks: These approaches combine:

    • Thermodynamic principles of protein-protein interactions

    • Experimental binding data from phage display libraries

    • Structural information from crystallography or cryo-EM

    • Machine learning to predict binding energetics of novel variants

    Such models can identify distinct binding modes for closely related epitopes, even those too similar to be experimentally dissociated .

  • Multi-objective optimization strategies: These computational techniques balance multiple desired properties:

    • Specificity for particular ANG forms

    • Thermal stability for robust assay performance

    • Reduced immunogenicity for in vivo applications

    • Manufacturability considerations

    By explicitly modeling these trade-offs, researchers can design antibodies optimized for specific applications rather than just maximal affinity.

  • Validation-guided refinement loops: Iterative processes where:

    • Computational models predict antibody properties

    • Key predictions are experimentally validated

    • Validation data refines subsequent computational models

    • The improved model generates next-generation candidates

This approach has already demonstrated success in designing antibodies with customized specificity profiles, either with specific high affinity for particular target epitopes or with cross-specificity for multiple defined targets . Future developments will likely enable even more precise control over binding properties, expanding our ability to create antibodies that can distinguish between subtle variations in ANG conformations, post-translational modifications, or interaction states.

What role might ANG antibodies play in developing therapeutic strategies targeting the angiogenin pathway?

ANG antibodies hold significant therapeutic potential through multiple mechanisms:

  • Targeted inhibition strategies: Different classes of ANG-targeting antibodies can:

    • Neutralize extracellular ANG to prevent receptor binding

    • Block the nuclear localization signal to inhibit nuclear translocation

    • Target the active site to inhibit ribonucleolytic activity

    • Prevent receptor-mediated internalization

    Each approach offers distinct therapeutic advantages for different disease contexts.

  • Cancer therapy applications: ANG antibodies may serve as:

    • Direct anti-angiogenic agents to restrict tumor blood supply

    • Targeting moieties for antibody-drug conjugates (ADCs)

    • Components of bispecific antibodies linking tumor cells to immune effectors

    • Biomarkers for patient stratification in precision oncology

  • Neurodegenerative disease interventions: Given ANG's neuroprotective functions:

    • Antibodies distinguishing between beneficial and pathological ANG forms

    • Intrabodies directing ANG to specific neuronal compartments

    • Antibodies enhancing ANG stability or activity in stressed neurons

    • Diagnostic tools for monitoring ANG status in CSF

  • Inflammation and autoimmunity applications: ANG antibodies can:

    • Monitor ANG as a biomarker of inflammation

    • Target ANG-producing cells in inflammatory microenvironments

    • Block specific inflammatory signaling cascades downstream of ANG

    • Provide diagnostic classification of inflammatory conditions

  • Technical considerations for therapeutic development:

    • Humanization strategies to reduce immunogenicity

    • Fc engineering to optimize effector functions or half-life

    • Format selection (IgG, Fab, scFv) based on tissue penetration requirements

    • Expression system optimization for clinical-grade production

While therapeutic ANG antibodies remain investigational, initial studies have demonstrated efficacy in animal models of cancer, where neutralizing ANG antibodies inhibited tumor angiogenesis and growth. The therapeutic development of these antibodies requires careful balancing of ANG's beneficial physiological roles against its pathological functions in disease states.

How can ANG antibodies contribute to understanding the emerging roles of ANG in stress response and RNA metabolism?

ANG antibodies provide critical tools for investigating ANG's newly recognized functions:

  • Subcellular localization analysis: Advanced imaging with domain-specific antibodies reveals:

    • Stress-induced translocation patterns

    • Association with stress granules

    • Interactions with the tRNA processing machinery

    • Recruitment to specific genomic regions

    Quantitative image analysis combined with co-localization studies maps the dynamic behavior of ANG under various stress conditions.

  • RNA-protein interaction studies: ANG antibodies enable:

    • RNA immunoprecipitation (RIP) to identify ANG-bound RNAs

    • Cross-linking immunoprecipitation (CLIP) for direct RNA binding sites

    • Proximity labeling to identify the RNA processing complexes containing ANG

    • In situ hybridization combined with immunofluorescence to visualize ANG-RNA interactions

  • Functional dissection approaches: Domain-specific antibodies help determine:

    • Which ANG domains are critical for stress response functions

    • How different cellular stressors modulate ANG activity

    • Whether disease-associated mutations affect specific functions

    • How post-translational modifications regulate ANG's distinct activities

  • Temporal dynamics investigation: Time-resolved antibody-based studies reveal:

    • The sequence of events in ANG-mediated stress responses

    • Recovery patterns after stress resolution

    • Adaptation mechanisms during chronic stress

    • Cell-type specific differences in ANG regulation

  • Integration with multi-omics datasets: ANG antibody-based findings can be contextualized with:

    • Transcriptomics data on stress-responsive genes

    • Proteomics profiles of stress-induced protein changes

    • Metabolomics analysis of cellular energy status

    • Structural biology insights on ANG conformational changes

These approaches have already yielded significant insights, revealing that ANG generates stress-induced tRNA fragments that reprogram protein translation, protects cells against stress-induced apoptosis, and participates in liquid-liquid phase separation during stress granule formation. Continuing investigations with increasingly specific antibodies promise to further elucidate ANG's multifaceted roles in cellular homeostasis and stress adaptation.

What are the recommended validation standards for ANG antibodies in research applications?

Rigorous validation is essential for reliable ANG antibody-based research:

  • Multi-technique validation approach: Verify antibody performance across:

    • Western blotting to confirm size and specificity

    • Immunoprecipitation to validate native protein recognition

    • Immunohistochemistry/immunofluorescence for localization studies

    • ELISA for quantitative applications

    Document performance characteristics for each application separately, as antibodies may perform differently across techniques .

  • Genetic validation strategies: Implement controls using:

    • ANG knockout/knockdown systems as negative controls

    • ANG overexpression systems as positive controls

    • Tagged ANG constructs for co-localization confirmation

    • Dose-response studies with graded expression levels

  • Epitope characterization requirements: Document:

    • The specific region or amino acid sequence recognized

    • Whether the antibody recognizes linear or conformational epitopes

    • Cross-reactivity with related proteins or orthologs

    • Sensitivity to post-translational modifications

  • Lot-to-lot consistency testing: Establish:

    • Reference standards for comparing new antibody lots

    • Quantitative acceptance criteria for batch variation

    • Storage and handling protocols to maintain activity

    • Regular performance monitoring throughout research projects

  • Transparent reporting standards: Include in publications:

    • Complete antibody identifiers (catalog number, lot number)

    • Detailed methods for all antibody-based procedures

    • Representative images of positive and negative controls

    • Quantification methods and reproducibility measures

These validation standards not only ensure experimental reliability but also facilitate integration of findings across different research groups working on ANG. The field is moving toward requiring multiple independent antibodies validating key findings and inclusion of appropriate genetic controls to substantiate antibody-based observations.

What experimental design considerations maximize reproducibility when working with ANG antibodies?

Ensuring reproducible results with ANG antibodies requires systematic experimental design:

  • Sample preparation standardization: Develop detailed protocols for:

    • Consistent lysis and extraction buffers

    • Standardized fixation times and temperatures

    • Controlled antigen retrieval conditions

    • Sample storage and handling procedures

    Document these parameters meticulously, as subtle variations can significantly impact antibody performance .

  • Quantitative controls implementation: Include:

    • Standard curves with recombinant ANG for quantitative applications

    • Internal loading controls for comparative studies

    • Calibrated reference samples across experimental batches

    • Serial dilution tests to confirm linearity of signal

  • Blind analysis protocols: Reduce experimenter bias through:

    • Coded sample identification during analysis

    • Automated image acquisition when possible

    • Predefined quantification parameters established before analysis

    • Independent verification of key findings by multiple researchers

  • Comprehensive metadata recording: Document:

    • Environmental conditions (temperature, humidity)

    • Reagent sources and lot numbers

    • Equipment calibration status

    • Timing of critical steps in protocols

  • Replicate design optimization: Structure experiments with:

    • Appropriate technical replicates (typically triplicate)

    • Independent biological replicates

    • Power calculations to determine sample sizes

    • Sequential replication of key findings over time

These approaches minimize variability from technical sources, allowing researchers to distinguish true biological effects from procedural artifacts. For longitudinal studies, maintaining consistent antibody lots throughout the project or performing bridging studies between lots is particularly important. Additionally, developing laboratory-specific reference samples that can be included in each experimental run provides an internal standard to normalize results across experiments.

How should researchers integrate antibody-based findings with other methodologies to build comprehensive understanding of ANG biology?

Developing robust mechanistic models of ANG function requires integrating multiple methodological approaches:

  • Orthogonal validation strategies: Confirm antibody-based findings through:

    • Mass spectrometry verification of protein identity and modifications

    • RNA-based methods (qPCR, RNA-seq) to correlate with protein data

    • Genetic approaches (CRISPR, siRNA) to confirm functional relationships

    • Live-cell imaging with fluorescent protein fusions to validate localization

  • Multi-scale investigation approaches: Connect:

    • Molecular interactions (antibody-based binding studies)

    • Cellular phenotypes (morphological and functional assays)

    • Tissue-level effects (histological analysis)

    • Systemic responses (physiological parameters)

  • Temporal dynamics integration: Combine:

    • Acute responses measured with fast-acting techniques

    • Long-term adaptations tracked through longitudinal studies

    • Developmental trajectories across model systems

    • Evolution of responses during disease progression

  • Computational model development: Incorporate:

    • Quantitative antibody-based measurements as model parameters

    • Structural information on antibody-epitope interactions

    • Systems biology approaches to place ANG in pathway contexts

    • Predictive models that generate testable hypotheses

  • Collaborative cross-validation: Establish:

    • Interlaboratory validation of key findings

    • Cross-platform confirmation of results

    • Standardized reference materials for comparative studies

    • Data sharing protocols for integrated analysis

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.