AREG Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

AREG antibodies are monoclonal or polyclonal reagents that bind specifically to amphiregulin, a 252-amino acid transmembrane glycoprotein encoded by the AREG gene . AREG functions as a ligand for the epidermal growth factor receptor (EGFR), influencing epithelial cell proliferation, tissue repair, and immune modulation . Its dual role as an extracellular signaling molecule and intracellular regulator makes it a target for studying cancer, inflammation, and developmental biology .

Key Research Applications

AREG antibodies are pivotal in studying:

  • Cancer Biology:

    • AREG promotes chemoresistance in prostate and melanoma cells by modulating IGF-1R, p21, and chromatin regulators . Neutralizing AREG antibodies restore drug sensitivity and reduce tumor growth in preclinical models .

    • Nuclear AREG correlates with low-proliferative phenotypes in melanoma, influencing histone methylation and heterochromatin dynamics .

  • Immunology:

    • Mast cell- and T cell-derived AREG suppresses inflammation and enhances regulatory T cell (Treg) activity, implicating it in immune tolerance .

    • AREG mediates tissue repair in lung and intestinal inflammation, as shown in colitis and infection models .

Clinical and Therapeutic Implications

  • Biomarker Potential: Stromal AREG levels predict chemotherapy resistance and correlate with senescence-associated secretory phenotype (SASP) in tumors .

  • Therapeutic Targeting: Anti-AREG monoclonal antibodies (e.g., AREG mAb) disrupt cancer-stroma interactions, enhancing efficacy of DNA-targeting agents like mitoxantrone .

Technical Considerations

  • Sample Preparation: Intracellular detection requires fixation/permeabilization protocols (e.g., Thermo Fisher’s Intracellular Fixation Buffer Set) .

  • Cross-Reactivity: Proteintech’s 16036-1-AP shows reactivity with human, mouse, and rat AREG .

Emerging Insights

Recent studies highlight:

  • Dual Signaling Roles: AREG’s extracellular EGFR activation and nuclear regulation of gene expression .

  • Immune Modulation: AREG from innate lymphoid cells (ILC2s) and neutrophils resolves tissue damage but may exacerbate fibrosis in chronic inflammation .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery time information.
Synonyms
AREG; AREGB; SDGF; Amphiregulin; AR; Colorectum cell-derived growth factor; CRDGF
Target Names
Uniprot No.

Target Background

Function
Amphiregulin is a ligand for the EGF receptor (EGFR). It acts as an autocrine growth factor and a mitogen for a wide range of target cells, including astrocytes, Schwann cells, and fibroblasts.
Gene References Into Functions
  1. Amphiregulin, found in non-small-cell lung carcinoma-derived exosomes, induces osteoclast differentiation through activation of the EGFR pathway. PMID: 28600504
  2. Research indicates that HIF2-alpha induces myocardial AREG expression in cardiac myocytes, increasing myocardial ischemia tolerance. PMID: 29483579
  3. AREG mediates hCG-induced StAR expression and progesterone production in human granulosa cells, providing new evidence for its role in regulating steroidogenesis. PMID: 27113901
  4. Regulatory T-cell-intrinsic amphiregulin is dispensable for its suppressive function. PMID: 27040371
  5. No significant correlations were found between YAP or AREG expression and the volume of VIII CN schwannoma. PMID: 28430338
  6. Cullin 3 regulates ADAM17-mediated ectodomain shedding of AREG. PMID: 29550478
  7. Overexpression of AREG could serve as a novel biomarker for gastric cancer (GC), and active surveillance of its expression may offer a new approach to GC diagnosis and monitoring. PMID: 27713123
  8. Sprouty2 inhibits amphiregulin-induced down-regulation of E-cadherin and cell invasion in human ovarian cancer cells. PMID: 27835572
  9. Results demonstrate that AREG expression is upregulated in gastric tumors, and its co-expression with TROP2 protein is associated with TNM stage, tumor size, lymph node metastases, and distant metastases. PMID: 28256068
  10. The secretion of IL-13 and amphiregulin suggests that intrahepatic innate lymphoid cells may be recruited to promote resolution and repair, potentially contributing to ongoing fibrogenesis in liver disease. PMID: 29261670
  11. The interplay between EGF and AREG in airway basal cell stem/progenitor cells is one of the mechanisms mediating the interconnected pathogenesis of all major smoking-induced lesions in the human airway epithelium. PMID: 27709733
  12. AREG expression might be useful for identifying CRTC1-MAML2-positive mucoepidermoid carcinomas and as a marker for favorable prognosis. PMID: 27393417
  13. Amphiregulin enhances VEGF-A production in human chondrosarcoma cells and promotes angiogenesis by inhibiting miR-206 via the FAK/c-Src/PKCdelta pathway. PMID: 27826039
  14. Amphiregulin plays a significant role in lung neoplasm resistance to amrubicinol. PMID: 28476786
  15. EREG and AREG are strongly regulated by methylation, and their expression is associated with CIMP status and primary tumor site. PMID: 27272216
  16. These findings demonstrate the posttranslational regulation of Foxp3 expression by AREG in cancer patients through the AREG/EGFR/GSK-3beta signaling pathway, which could lead to Foxp3 protein degradation in Treg cells and a potential therapeutic target for cancer treatment. PMID: 27432879
  17. Blocking soluble amphiregulin with a neutralizing antibody significantly increased apoptotic cell death of HepG2 cells due to treatment with methyl methanesulfonate, cisplatin, or a recombinant p53 adenovirus, suggesting that amphiregulin's function in inhibiting apoptosis may be a common mechanism by which hepatoma cells escape stimulus-induced apoptosis. PMID: 28351301
  18. Keratinocyte expression of hAREG elicits inflammatory epidermal hyperplasia. PMID: 26519132
  19. Low AREG expression is associated with gastric cancer. PMID: 26884344
  20. RYR2, PTDSS1, and AREG are autism susceptibility genes implicated in a Lebanese population-based study of copy number variations in this disease. PMID: 26742492
  21. High Amphiregulin enhances intercellular adhesion molecule-1 expression and promotes tumor metastasis in osteosarcoma. PMID: 26503469
  22. Results demonstrate that AREG controls G2/M progression and cytokinesis in keratinocytes through activation of a FoxM1-dependent transcriptional program, suggesting new avenues for the treatment of epithelial cancer. PMID: 26234682
  23. High expression of amphiregulin is associated with hepatocellular carcinoma. PMID: 26451607
  24. Findings show the involvement of amphiregulin and semaphorin-3A in the improvement of skin innervations and penetration of nerve fibers into the epidermis. PMID: 26201903
  25. Altered AREG expression induced by diverse luteinizing hormone receptor reactivity in granulosa cells may provide a useful marker for oocyte developmental competency. PMID: 25911599
  26. Amphiregulin enhances alpha6beta1 integrin expression and cell motility in human chondrosarcoma cells through the Ras/Raf/MEK/ERK/AP-1 pathway. PMID: 25825984
  27. Our findings implicate amphiregulin as a critical mediator of the estrogen response in ERalpha-positive breast cancer. PMID: 26527289
  28. AR induces hHSC fibrogenic activity via multiple mitogenic signaling pathways, and is upregulated in murine and human NASH, suggesting that AR antagonists may be clinically useful anti-fibrotics in NAFLD. PMID: 25744849
  29. Bradykinin (BK) stimulation of human airway smooth muscle cells increases amphiregulin secretion in a mechanism dependent on BK-induced COX-2 expression. PMID: 26047642
  30. The applied drugs showed remarkable suppression of mTOR expression, which might delay tumor progression. Interestingly, sorafenib and sunitinib increased AREG in HNSCC 11A and 14C. PMID: 25862847
  31. Expression profiling demonstrated that AREG-activated EGFR regulates gene expression differently than EGF-activated EGFR. PMID: 25454348
  32. This study shows that TGF-alpha uses common and divergent molecular mediators to regulate E-cadherin expression and cell invasion. PMID: 25869072
  33. AREG rs1615111, located in the AREG genomic region, can significantly define different prognostic cohorts in locally advanced GC. PMID: 25203737
  34. AREG induces ovarian cancer cell invasion by down-regulating E-cadherin expression. PMID: 25261255
  35. During high-pressure ventilation, Nrf2 becomes activated and induces AREG, leading to a positive feedback loop between Nrf2 and AREG, which involves the p38 MAPK and results in the expression of cytoprotective genes. PMID: 24921206
  36. AREG expression was significantly correlated with Edmondson stage and serum AFP level. PMID: 24860833
  37. AREG shedding occurs through a TNF-alpha-converting enzyme-dependent mechanism in diacetyl treated pulmonary epithelial cells. PMID: 24816162
  38. Aberrantly activated AREG-EGFR signaling is required for CRTC1-MAML2-positive MEC cell growth and survival, suggesting that EGFR-targeted therapies will benefit patients with advanced, unresectable CRTC1-MAML2-positive MEC. PMID: 23975434
  39. A self-reinforcing loop of amphiregulin and Y-box binding protein-1 contributes to poor outcomes in ovarian cancer. PMID: 23851501
  40. IL-1beta-induced amphiregulin release may be involved in the pathogenesis of rheumatoid arthritis. PMID: 24196392
  41. These findings provide mechanistic insight into the regulation of YAP and AREG by RASSF1A in human multistep hepatocarcinogenesis. PMID: 23594797
  42. Data suggest that AREG (amphiregulin), BTC (betacellulin), and EREG (epiregulin) induced prostaglandin E2 production by induction of COX-2 (prostaglandin-endoperoxide synthase 2) through MAP kinase signaling in granulosa cells. PMID: 24092824
  43. Exosome-bound WD repeat protein Monad inhibits breast cancer cell invasion by degrading amphiregulin mRNA. PMID: 23844004
  44. Promoter methylation of AREG is associated with glioblastoma. PMID: 23624749
  45. AREG plays pro-neoplastic roles in colorectal carcinogenesis. PMID: 23263765
  46. EREG-AREG and NRG1, which are members of the epidermal growth factor (EGF) family, seem to modulate Behcet's disease susceptibility through main effects and gene-gene interactions. PMID: 23625463
  47. We did not find a correlation between the presence of a K-ras mutation and the presence of Epiregulin and Amphiregulin in colon cancer tissue. PMID: 23885463
  48. Regulation of amphiregulin gene expression by beta-catenin signaling in human hepatocellular carcinoma cells. PMID: 23285165
  49. Human antigen R-mediated mRNA stabilization is required for ultraviolet B-induced autoinduction of amphiregulin in keratinocytes. PMID: 23430747
  50. Polycystin-1 regulates amphiregulin expression through CREB and AP1 signaling, which has implications in ADPKD cell proliferation. PMID: 22570239

Show More

Hide All

Database Links

HGNC: 651

OMIM: 104640

KEGG: hsa:374

STRING: 9606.ENSP00000370227

UniGene: Hs.270833

Protein Families
Amphiregulin family
Subcellular Location
Membrane; Single-pass membrane protein.

Q&A

What is AREG and why is it a significant target for antibody-based research?

AREG is a multifaceted molecule that functions both as an extracellular ligand for the EGF receptor (EGFR) and as an intracellular signaling molecule . It exists initially as a single-pass transmembrane protein that undergoes proteolytic processing by TACE/ADAM17, releasing the soluble EGFR ligand and leaving a residual transmembrane stalk that is subsequently internalized . AREG is a particularly valuable research target because it serves as an effector molecule for tissue repair and homeostasis, mediates resistance and tolerance to infection, and suppresses inflammation in the immune system context . Beyond its canonical roles, AREG has been implicated in cancer progression and drug resistance mechanisms, making it an important subject for both basic research and therapeutic development .

How do I select the most appropriate anti-AREG antibody for my specific research application?

The selection of an appropriate anti-AREG antibody should be based on:

  • Target epitope specificity: Determine whether you need antibodies that recognize:

    • The mature soluble form of AREG

    • The uncleaved transmembrane precursor

    • The cleaved transmembrane stalk (neo-epitope specific)

    • Nuclear localized AREG

  • Application compatibility: Verify validated applications for your chosen antibody:

    • Western blotting (WB): Antibodies like AF262 have been validated for detecting AREG in cell lysates and conditioned media

    • Immunocytochemistry/Immunofluorescence (ICC/IF): Consider antibodies validated for cellular localization studies

    • Flow cytometry: Antibodies like AREG559-PE are pre-tested for intracellular flow cytometry

    • Immunohistochemistry (IHC): For tissue sections, antibodies validated in FFPE samples

  • Host species and cross-reactivity: Consider the host species of the antibody and its validated reactivity profile to avoid complications in multi-species studies or when using secondary detection systems .

What are the optimal fixation and permeabilization conditions for detecting AREG in different subcellular compartments?

Detecting AREG in different subcellular compartments requires specific optimization:

  • Membrane-bound AREG:

    • For surface detection without permeabilization, use non-permeabilizing fixatives like 2-4% paraformaldehyde

    • Example protocol: Me-180 cells infected with N. gonorrhoeae showed membrane-bound AREG using polyclonal antibodies and visualization with Alexa 488-conjugated secondary antibodies

  • Intracellular/nuclear AREG:

    • Use appropriate permeabilization buffers such as the Intracellular Fixation & Permeabilization Buffer Set

    • For nuclear AREG detection critical in melanoma research, follow protocols that preserve nuclear integrity while allowing antibody penetration

    • Flow cytometry detection typically requires 5 μL (0.25 μg) of antibody per test with cell numbers ranging from 10^5 to 10^8 cells/test

  • Secreted AREG in conditioned media:

    • Detection of soluble AREG often requires concentration steps or direct analysis by ELISA or western blot

    • For western blot detection, use reducing conditions and appropriate immunoblot buffer groups

How can I validate the specificity of anti-AREG antibodies in my experimental system?

Validating anti-AREG antibody specificity is crucial and can be accomplished through several complementary approaches:

  • Genetic validation:

    • AREG knockdown/knockout controls using siRNA or CRISPR-Cas9 systems

    • Overexpression systems with tagged AREG constructs

    • Both approaches provide clear positive and negative controls for antibody validation

  • Peptide competition assays:

    • Pre-incubating the antibody with the immunizing peptide should abolish specific staining

    • Comparison of staining patterns between different anti-AREG antibodies targeting distinct epitopes

  • Stimulation experiments:

    • Treatment of cells with PMA and PHA induces AREG expression and can serve as a positive control

    • Example: T47D human breast cancer cells show increased AREG detection following PMA/PHA treatment for 3 days

  • Multiple detection methods:

    • Correlation between different techniques (IF, WB, flow cytometry) strengthens validation

    • Include secondary antibody-only controls to rule out non-specific binding

How does nuclear AREG contribute to drug resistance in melanoma, and how can this be experimentally investigated?

Nuclear AREG has been identified as a critical factor in melanoma drug resistance through several mechanisms:

  • Epigenetic regulation:

    • Nuclear AREG regulates IGF-1R, P21 (Cip1/Waf1), TP53, and JARID1B protein accumulation in the nucleus

    • It influences heterochromatin condensation through regulation of HP1beta and SETDB1

    • Nuclear AREG affects trimethylation of histones H3K9 and H3K4

  • Resistance phenotype induction:

    • SK-Mel-28-VR cells cultured under Vemurafenib (VR) selection pressure show accumulation of AREG in the nucleus

    • This nuclear accumulation correlates with JARID1B expression, a marker associated with drug-resistant slow-cycling cells

Experimental investigation approaches:

  • Nuclear fractionation and western blotting: To quantify nuclear vs. cytoplasmic AREG levels

  • Immunofluorescence microscopy: To visualize nuclear localization of AREG in resistant vs. sensitive cells

  • AREG knockdown studies: Knockdown of AREG makes previously resistant cells more sensitive to VR treatment

  • Chromatin immunoprecipitation (ChIP): To investigate AREG-associated epigenetic modifications

What role does AREG shedding play in cancer progression, and how can antibodies targeting cleaved AREG be utilized in therapeutic approaches?

AREG shedding is a critical process in cancer progression and offers unique therapeutic opportunities:

  • AREG shedding mechanism and significance:

    • AREG is a transmembrane protein proteolytically processed by TACE/ADAM17

    • Processing releases the soluble EGFR ligand, leaving a residual transmembrane stalk

    • High rates of AREG shedding are associated with cancer progression and EGFR pathway activation

  • Neo-epitope targeting approach:

    • Novel antibodies have been developed that selectively recognize the residual transmembrane stalk of cleaved AREG

    • These antibodies do not interact with uncleaved AREG, providing specificity for cells with high rates of AREG shedding

    • Example: GMF-1A3-MMAE, an antibody-drug conjugate targeting the AREG neo-epitope

  • Therapeutic applications:

    • Antibodies conjugated with cytotoxic agents (e.g., MMAE) demonstrate cytotoxicity in vitro

    • In vivo studies show these conjugates induce rapid regression of established breast tumor xenografts

    • Potential utility in breast cancer and other tumors where proteolytic AREG shedding is frequent

  • Companion diagnostics:

    • These antibodies can recognize the AREG neo-epitope in formalin-fixed, paraffin-embedded tumor tissue

    • This capability suggests their potential as companion diagnostics for patient selection in targeted therapy approaches

What immune cell populations express AREG, and how can this be accurately detected in different immunological contexts?

AREG expression has been identified in multiple immune cell populations, each with specific detection considerations:

Immune Cell TypeActivation ConditionDetection MethodReference
T cellsVarious stimuliFlow cytometry, ELISA
Group 2 innate lymphoid cellsType 2 inflammationIntracellular staining
BasophilsAllergic stimulationFlow cytometry
Mast cellsIgE-mediated activationImmunofluorescence
EosinophilsType 2 cytokine exposureFlow cytometry
NeutrophilsInflammatory stimuliIntracellular staining

For accurate detection:

  • Flow cytometric analysis:

    • Stimulated human peripheral blood cells can be analyzed using appropriate intracellular fixation and permeabilization protocols

    • Pre-diluted antibodies like AREG559 can be used at 5 μL (0.25 μg) per test

    • Excitation at 488-561 nm and emission at 578 nm are optimal for PE-conjugated anti-AREG antibodies

  • Tissue context analysis:

    • IHC/IF on tissue sections requires optimization of antigen retrieval methods

    • Dual staining with immune cell markers helps identify specific AREG-expressing populations

  • Ex vivo stimulation protocols:

    • For detecting inducible AREG expression in immune cells, appropriate stimulation protocols must be employed

    • Type 2 cytokines (IL-4, IL-13, IL-33) are effective stimulants for certain populations

How does AREG function as an effector molecule in tissue repair, and what experimental models best demonstrate this activity?

AREG serves as a pivotal effector molecule in tissue repair through several mechanisms:

  • Dual functions in immune responses:

    • Contributes to host resistance mechanisms (protective immunity)

    • Primarily functions as a key factor inducing tolerance by promoting tissue integrity restoration following inflammation

  • Tissue repair activities:

    • Promotes epithelial cell and fibroblast proliferation through EGFR activation

    • Facilitates keratinocyte migration and differentiation in skin repair

    • Mediates regulatory T cell (Treg)-dependent muscle regeneration

Optimal experimental models include:

  • Wound healing models:

    • Skin injury models with AREG knockout or overexpression

    • Analysis of re-epithelialization rates, inflammatory infiltrate, and tissue remodeling

  • Pulmonary injury models:

    • Influenza infection in AREG-deficient mice demonstrates the role in lung tissue repair

    • Analysis of epithelial integrity, lung function, and survival outcomes

  • Muscle damage models:

    • Cardiotoxin-induced muscle injury in mice with Treg-specific AREG deficiency

    • Assessment of satellite cell activation, myofiber regeneration, and functional recovery

How can AREG antibodies be optimized for targeted drug delivery in cancer therapy?

Optimization of AREG antibodies for targeted drug delivery involves several sophisticated approaches:

  • Selection of optimal antibody format:

    • Full IgG vs. antibody fragments (Fab, scFv, nanobodies)

    • Consideration of tissue penetration, half-life, and internalization rates

    • Example: The GMF-1A3-MMAE ADC targeting cleaved AREG demonstrates effective internalization and cytotoxicity

  • Conjugation chemistry optimization:

    • Selection of linker chemistry (cleavable vs. non-cleavable)

    • Drug-to-antibody ratio (DAR) optimization

    • Site-specific conjugation to preserve binding characteristics

  • Target epitope selection:

    • Neo-epitope targeting (e.g., cleaved AREG) provides specificity for cancer cells with high shedding rates

    • Epitopes that promote rapid internalization enhance drug delivery efficiency

    • For AREG, antibodies recognizing the residual transmembrane stalk showed efficient internalization in cancer cells

  • Payload selection:

    • Cytotoxic agents (MMAE, DM1, SN-38)

    • Immunomodulatory molecules

    • Radioactive isotopes for theranostic applications

  • Validation strategies:

    • In vitro internalization assays using fluorescent dye conjugation

    • Cytotoxicity assessment in relevant cell lines

    • In vivo efficacy in tumor xenograft models (as demonstrated with the AREG-targeted ADC)

How can conflicting data about AREG's role as a tumor suppressor versus oncogene be experimentally resolved?

The dual nature of AREG as both potential tumor suppressor and oncogene presents a complex research challenge that requires sophisticated experimental approaches:

  • Context-dependent analysis:

    • Studies in melanoma have shown contradictory roles for AREG

    • Systematic comparison of AREG function across different cancer types, stages, and genetic backgrounds

    • Example: In melanoma, intracellular/nuclear AREG appears to play a different role than secreted AREG

  • Subcellular localization studies:

    • Differential effects based on AREG localization (membrane-bound, secreted, cytoplasmic, nuclear)

    • Use of localization-specific antibodies and compartment-specific expression systems

    • Nuclear AREG affects IGF-1R, P21, TP53, and JARID1B accumulation, influencing cell cycle and drug sensitivity

  • Receptor-dependent vs. receptor-independent effects:

    • Distinguish EGFR-mediated from intracellular signaling functions

    • Use of EGFR inhibitors or knockdown in combination with AREG manipulation

    • Comparison of recombinant AREG treatment vs. neutralizing antibody approaches

  • Genetic manipulation approaches:

    • Generate models with selective disruption of specific AREG domains or functions

    • CRISPR-Cas9 editing to modify AREG processing, secretion, or nuclear localization signals

    • Inducible expression systems to study temporal effects

  • Multi-omics integration:

    • Combine transcriptomics, proteomics, and functional assays to develop comprehensive models

    • Network analysis to identify context-dependent interaction partners

    • Correlation with epigenetic modifications such as heterochromatin condensation (HP1beta, SETDB1) and histone trimethylation (H3K9, H3K4)

What are the most common pitfalls in AREG antibody-based experiments, and how can they be avoided?

Researchers frequently encounter several challenges when working with AREG antibodies:

  • Non-specific binding and false positives:

    • Always include proper negative controls (secondary antibody only, isotype controls)

    • Validate antibodies using AREG knockout/knockdown systems

    • For IHC/IF, include controls omitting primary antibody as demonstrated in human breast tissue staining

  • Epitope masking and detection failures:

    • AREG undergoes processing and may form complexes affecting epitope accessibility

    • Optimize antigen retrieval methods for fixed samples

    • Consider using multiple antibodies targeting different epitopes

    • For western blot, ensure appropriate reducing conditions and buffer systems (e.g., Immunoblot Buffer Group 1)

  • Inconsistent detection of processed forms:

    • AREG appears at different molecular weights depending on processing and glycosylation

    • Full-length vs. processed AREG may require different detection conditions

    • Example: In western blots, processed AREG appears at approximately 45 kDa under reducing conditions

  • Quantification challenges:

    • Standardize cell numbers (10^5 to 10^8 cells/test for flow cytometry)

    • Determine optimal antibody dilutions empirically (typically 0.3-1 μg/mL for neutralization assays)

    • Use recombinant AREG standards for quantitative assays

  • Specificity across applications:

    • An antibody validated for one application may not work in others

    • Always verify application-specific validation data

    • Example: Some antibodies are validated specifically for ICC/IF (1:50-1:200 dilution) but may require optimization for other applications

How can I validate AREG antibody functionality in neutralization assays?

Validating AREG antibody functionality in neutralization assays requires systematic approaches:

  • Proliferation neutralization assay:

    • Balb/3T3 mouse embryonic fibroblast cell line responds to recombinant Human AREG in a dose-dependent manner

    • Proliferation elicited by AREG (50 ng/mL) can be neutralized by increasing concentrations of anti-AREG antibodies

    • The ND50 (neutralizing dose, 50%) is typically 0.3-1 μg/mL for validated antibodies like Goat Anti-Human AREG Antigen Affinity-purified Polyclonal Antibody (AF262)

  • Controls and standards:

    • Include positive control (recombinant AREG stimulation alone)

    • Include negative control (neither AREG nor antibody)

    • Use irrelevant antibody control (same isotype, different specificity)

    • Test dose-response relationship with antibody titration

  • Cell system selection:

    • Choose cells with verified EGFR expression

    • Confirm AREG responsiveness before neutralization testing

    • Consider cell lines relevant to your research context (e.g., breast cancer, melanoma)

  • Endpoint measurement optimization:

    • Select appropriate proliferation assays (MTT, BrdU, cell counting)

    • Determine optimal timepoints (typically 24-72 hours)

    • Consider additional functional readouts beyond proliferation (migration, differentiation)

  • Validation across AREG sources:

    • Test neutralization against both recombinant and endogenously produced AREG

    • Compare neutralization efficiency between different cellular sources of AREG

How are AREG antibodies being utilized to study the role of AREG in infectious disease contexts?

AREG antibodies are providing valuable insights into infection biology through several innovative approaches:

  • Pathogen-induced AREG modulation:

    • N. gonorrhoeae infection increases membrane-bound AREG that co-localizes with bacterial adherence sites

    • This can be visualized using polyclonal antibodies against AREG followed by fluorophore-conjugated secondary antibodies

    • Flow cytometry reveals increased plasma membrane-bound AREG in non-permeabilized infected cells compared to controls

  • AREG in host defense and immunopathology:

    • AREG functions as an effector molecule mediating resistance and tolerance to infection

    • Antibodies enable tracking of AREG expression in immune cells responding to pathogens

    • This approach helps distinguish protective vs. pathological responses

  • Tissue repair during infection resolution:

    • AREG plays a central role in orchestrating tissue repair following infection-induced damage

    • Antibody-based detection in tissue sections and cell populations helps map the spatiotemporal dynamics of this response

    • This information is crucial for understanding resolution of inflammation and return to homeostasis

  • Therapeutic targeting considerations:

    • Understanding the dual role of AREG in infection (protective vs. pathological) informs therapeutic strategies

    • AREG blockade or enhancement might be beneficial depending on infection context and stage

    • Neutralizing antibodies serve as both research tools and potential therapeutic agents

What are the latest methodological advances in AREG antibody development for detecting novel AREG variants or post-translational modifications?

Recent technological innovations have enhanced our ability to develop and utilize AREG antibodies for detecting variants and modifications:

  • Neo-epitope specific antibodies:

    • Phage display technology has enabled identification of antibodies that selectively recognize the residual transmembrane stalk of cleaved AREG

    • These antibodies provide a novel means of targeting cells with high rates of AREG shedding

    • This approach has applications in both research and therapeutic contexts

  • Modification-specific antibodies:

    • Development of antibodies specifically recognizing phosphorylated, glycosylated, or otherwise modified AREG

    • These tools enable tracking of AREG processing and functional states

    • Application in studying the relationship between AREG modifications and functional outcomes

  • Multi-epitope recognition strategies:

    • Cocktails of antibodies recognizing different AREG epitopes improve detection sensitivity

    • Particularly valuable for detecting low-abundance AREG variants

    • Useful for comprehensive profiling of AREG expression patterns

  • Proximity ligation and multiparametric detection:

    • Combining AREG antibodies with proximity ligation assays to detect AREG-protein interactions

    • Multicolor flow cytometry and imaging using specialized AREG antibody conjugates

    • These approaches provide deeper insights into AREG signaling networks and functional states

  • Integration with mass spectrometry:

    • Immunoprecipitation with AREG antibodies followed by mass spectrometry analysis

    • Enables detailed characterization of AREG variants and post-translational modifications

    • Facilitates discovery of novel AREG forms and their functional significance

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.