Recombinant Human Amphiregulin (AREG)

Shipped with Ice Packs
In Stock

Description

Dual Roles in Homeostasis and Disease

  • Physiological Roles:

    • Promotes ductal branching in mammary glands during puberty .

    • Acts as a mitogen for hepatocytes and prevents apoptosis in the liver .

    • Enhances angiogenesis in fetal lung endothelial cells under hyperoxic stress via ERK1/2 activation .

  • Pathological Roles:

    • Cancer Promotion:

      • Drives osteosarcoma metastasis by upregulating ICAM-1 through PI3K/Akt/NF-κB signaling .

      • Mediates resistance to exemestane in breast cancer via autocrine EGFR activation .

      • Predicts poor progression-free survival in KRAS/BRAF wild-type colorectal cancer treated with cetuximab .

    • Cancer Suppression:

      • Inhibits growth in select tumor cell lines (e.g., A431 epidermoid carcinoma) .

Table 1: Key Studies on Recombinant AREG in Cancer

Cancer TypeModel/DesignKey FindingsSource
Ovarian CancerSyngeneic murine model (ID8 cells)Anti-AREG antibodies prolonged survival by 40–60% .
OsteosarcomaMG63/U2OS cell linesAREG increased migration by 2.5-fold via ICAM-1 upregulation .
Colorectal CancerPatient plasma analysisHigh baseline AREG predicted cetuximab resistance (PFS: 10.9 vs. 24.2 months) .
Lung DevelopmentHyperoxia-exposed fetal lung ECsRecombinant AREG restored angiogenesis via ERK1/2 activation .

Clinical and Therapeutic Implications

  • Biomarker Potential:

    • Plasma AREG levels correlate with cetuximab resistance in colorectal cancer, suggesting utility in treatment stratification .

  • Therapeutic Targeting:

    • Neutralizing AREG antibodies inhibit ovarian tumor growth in preclinical models .

    • Combined EGFR/PI3K inhibition may overcome AREG-driven resistance in osteosarcoma .

Future Directions

  • Investigate AREG’s role in non-cancer pathologies (e.g., bronchopulmonary dysplasia ).

  • Develop dual-targeting strategies against AREG and downstream effectors (e.g., ERK1/2, PI3K).

  • Validate AREG as a companion diagnostic for EGFR-targeted therapies .

Product Specs

Form
Lyophilized powder
Note: We prioritize shipping the format currently in stock. However, if you have specific requirements for the format, please indicate them during order placement. We will fulfill your request based on availability.
Lead Time
Delivery time may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery details.
Note: All of our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please inform us in advance as additional fees will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging this vial briefly before opening to concentrate the contents at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default glycerol concentration is 50%, which can serve as a reference.
Shelf Life
Shelf life depends on multiple factors, including storage conditions, buffer components, storage temperature, and the inherent stability of the protein itself.
Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. The shelf life of lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
The tag type will be determined during the production process. If you have a specific tag type requirement, please inform us, and we will prioritize developing the specified tag.
Synonyms
AREG; AREGB; SDGF; Amphiregulin; AR; Colorectum cell-derived growth factor; CRDGF
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
101-184
Protein Length
Full length protein
Species
Homo sapiens (Human)
Target Names
Target Protein Sequence
SVRVEQVVKPPQNKTESENTSDKPKRKKKGGKNGKNRRNRKKKNPCNAEFQNFCIHGECK YIEHLEAVTCKCQQEYFGERCGEK
Uniprot No.

Target Background

Function
Amphiregulin is a ligand of 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, contained in non-small-cell lung carcinoma-derived exosomes, induces osteoclast differentiation through the activation of the EGFR pathway. PMID: 28600504
  2. Studies indicate that HIF2-alpha induces myocardial AREG expression in cardiac myocytes, which increases myocardial ischemia tolerance. PMID: 29483579
  3. AREG mediates hCG-induced StAR expression and progesterone production in human granulosa cells, providing novel evidence for AREG's role in regulating steroidogenesis. PMID: 27113901
  4. Regulatory T-cell-intrinsic amphiregulin is dispensable for suppressive function. PMID: 27040371
  5. No significant correlations were found between YAP or AREG expression and VIII CN schwannoma volume. PMID: 28430338
  6. Cullin 3 regulates ADAM17-mediated ectodomain shedding of AREG. PMID: 29550478
  7. Over-expression of AREG could serve as a novel GC biomarker, and active surveillance of its expression could be a novel approach for 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 show that AREG expression is up-regulated in gastric tumor, and its co-expression with TROP2 protein is associated with TNM stage, tumor size, lymph node metastases, and distant metastases. PMID: 28256068
  10. 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. EGF-AREG interplay in airway basal cell stem/progenitor cells is one of the mechanisms that mediates the interconnected pathogenesis of all major smoking-induced lesions in the human airway epithelium. PMID: 27709733
  12. AREG expression may 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 an important 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 AREG/EGFR/GSK-3beta signaling, 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 also significantly increased apoptotic cell death of HepG2 cells due to treatment with methyl methanesulfonate, cisplatin, or a recombinant p53 adenovirus, suggesting that the function of amphiregulin involved in inhibiting apoptosis may be a common mechanism by which hepatoma cells escape from 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 via activation of a FoxM1-dependent transcriptional program, suggesting new avenues for treating 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 utilizes 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 may 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. 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
  42. Exosome-bound WD repeat protein Monad inhibits breast cancer cell invasion by degrading amphiregulin mRNA. PMID: 23844004
  43. Promoter methylation of AREG is associated with glioblastoma. PMID: 23624749
  44. AREG plays pro-neoplastic roles in colorectal carcinogenesis. PMID: 23263765
  45. 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
  46. 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
  47. Regulation of amphiregulin gene expression by beta-catenin signaling in human hepatocellular carcinoma cells. PMID: 23285165
  48. Human antigen R-mediated mRNA stabilization is required for ultraviolet B-induced autoinduction of amphiregulin in keratinocytes. PMID: 23430747
  49. 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 Recombinant Human Amphiregulin and what are its structural characteristics?

Recombinant Human Amphiregulin (AREG) is a member of the epidermal growth factor (EGF) family of cytokines. The protein is typically produced as a recombinant form comprising amino acids Ser101-Lys198 of the native human AREG sequence, with the full-length cDNA encoding a 252 amino acid transmembrane precursor. This particular segment represents the bioactive domain of the protein. In its native context, AREG exists in multiple forms, including variants of 78 or 84 amino acid residues with both N- and O-linked oligosaccharides . The recombinant protein is typically expressed in E. coli systems, resulting in a non-glycosylated product with a predicted molecular mass corresponding to its amino acid sequence .

The protein contains the characteristic EGF structural motif, which is essential for its receptor binding capabilities. Notably, the 98 amino acid residue long form of recombinant amphiregulin has been demonstrated to be approximately 5-10 fold more active than the 78 amino acid residue form in proliferation assays, highlighting the importance of specific structural elements for biological function .

How should Recombinant Human AREG be prepared and stored for experimental use?

Proper preparation and storage of Recombinant Human AREG are critical for maintaining its biological activity in experimental settings. The protein is typically supplied as a lyophilized powder that requires reconstitution before use. For carrier-containing formulations, reconstitution at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin is recommended. For carrier-free preparations, reconstitution at the same concentration in sterile PBS is advised .

For reconstitution protocols, the following methodology is recommended:

  • Centrifuge the tube before opening to collect all material at the bottom

  • Reconstitute to a concentration of 0.1-0.5 mg/mL in sterile distilled water

  • Avoid vortexing or vigorously pipetting the protein to prevent denaturation

  • For long-term storage, add a carrier protein or stabilizer (e.g., 0.1% BSA, 5% HSA, 10% FBS, or 5% Trehalose)

Storage recommendations indicate that the lyophilized protein should be stored at -20°C to -80°C for up to one year from the date of receipt. After reconstitution, the protein solution remains stable at -20°C for approximately 3 months or at 2-8°C for up to one week. It is essential to avoid repeated freeze-thaw cycles as these significantly diminish protein activity . Using a manual defrost freezer is recommended to maintain protein integrity over time .

What are the biological functions of AREG in normal physiological contexts?

In normal physiological contexts, AREG serves as a critical regulator of cellular proliferation and differentiation across multiple tissue types. It has been shown to stimulate the proliferation of various human and mouse keratinocytes, mammary epithelial cells, and some fibroblasts . Recent research has also revealed AREG's important role in hair follicle regeneration, where it promotes skin-derived precursor (SKP) stemness by enhancing both proliferation and hair-inducing capacity .

AREG functions as a ligand for the epidermal growth factor receptor (EGFR), activating downstream signaling cascades that regulate cellular responses. In hair regeneration specifically, AREG promotes an earlier telogen-to-anagen transition and high-efficiency hair follicle reconstitution through the phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways .

AREG mRNA expression has been detected in various human tissues including colon, stomach, breast, ovary, and kidney, suggesting widespread physiological functions across multiple organ systems . Beyond its role in normal tissue homeostasis, AREG also appears to have functions in immune response regulation, as research indicates it may be involved in protecting against post-influenza bacterial pneumonia by increasing phagocyte recruitment and reactive oxygen species (ROS) production .

How is AREG bioactivity measured in research applications?

Bioactivity assessment of Recombinant Human AREG requires specific methodological approaches to ensure reliable and reproducible results. The most commonly employed technique is the cell proliferation assay using appropriate responsive cell lines. For AREG, Balb/3T3 mouse embryonic fibroblast cells are frequently utilized as they demonstrate a dose-dependent proliferative response . In standard proliferation assays, the effective dose (ED50) typically ranges from 5-15 ng/mL, providing a quantitative measure of biological activity .

The proliferation assay methodology generally involves:

  • Seeding responsive cells (e.g., Balb/3T3) at a specified density in serum-starved conditions

  • Treating cells with serial dilutions of the recombinant AREG protein

  • Incubating for 24-72 hours (cell line dependent)

  • Measuring proliferation using techniques such as MTT assay, BrdU incorporation, or direct cell counting

  • Calculating the ED50 based on the dose-response curve

It is critical to include appropriate positive and negative controls in these assays to validate results. For advanced applications, researchers may also assess alkaline phosphatase (AP) staining as an indicator of stemness in multipotent stem cells treated with AREG, particularly when investigating its effects on cells like skin-derived precursors (SKPs) .

When comparing different forms or batches of AREG, it is important to note that the 98 amino acid residue long form has been shown to be approximately 5-10 fold more active than the 78 amino acid residue form in proliferation assays , highlighting the importance of standardization in experimental design.

What signaling pathways are activated by AREG and how are they experimentally investigated?

AREG activates multiple intracellular signaling cascades, with the PI3K and MAPK pathways being particularly well-characterized. Research has demonstrated that these pathways mediate AREG's effects on cellular proliferation and differentiation in various contexts . The experimental investigation of these signaling mechanisms requires specific methodological approaches.

To study AREG-mediated signaling pathways, researchers typically employ:

  • Pathway inhibition studies:

    • Use of specific chemical inhibitors (e.g., PI3K inhibitors like LY294002 or Wortmannin, and MAPK inhibitors like PD98059 or U0126)

    • siRNA-mediated knockdown of pathway components

    • The efficacy of siRNA transfection should be verified by fluorescence microscopy, with transfection efficiencies >70% considered acceptable

  • Protein phosphorylation analysis:

    • Western blotting for phosphorylated forms of key signaling molecules (e.g., phospho-ERK1/2, phospho-AKT)

    • Phospho-specific antibody arrays or ELISA

    • Reverse phase protein arrays (RPPA)

  • Transcriptional response analysis:

    • RT-qPCR for known pathway target genes

    • RNA-seq for comprehensive transcriptional profiling

    • Reporter gene assays for specific transcription factor activation

In hair follicle regeneration research, inhibitors of the PI3K and MAPK pathways have been shown to counteract AREG's stimulatory effects on SKP proliferation and hair-inducing capacity, confirming these pathways as critical mediators of AREG function in this context . This experimental approach provides a methodological framework for investigating AREG's signaling mechanisms in other biological systems.

What is AREG's role in cancer progression and chemoresistance?

AREG has emerged as a significant factor in cancer biology, with complex roles in tumor progression and treatment resistance. Research has revealed that AREG is a component of the senescence-associated secretory phenotype (SASP), a collection of soluble factors produced by senescent cells in the tumor microenvironment . The production of AREG is specifically triggered by DNA damage to stromal cells, which then enter senescence and release factors that influence nearby cancer cells .

The mechanisms through which AREG contributes to cancer progression are multifaceted:

  • Autocrine growth stimulation: AREG has been identified as an autocrine growth factor in certain colon carcinoma cell lines, promoting cancer cell proliferation through self-stimulation mechanisms .

  • Chemoresistance induction: AREG secretion from senescent stromal cells in the tumor microenvironment remarkably enhances cancer malignancy, including acquired resistance to therapeutic agents .

  • Immunosuppression: AREG may contribute to creating an immunosuppressive microenvironment that shields cancer cells from immune surveillance.

Experimental strategies to study AREG's role in cancer typically include:

  • Conditional media experiments: Using media from senescent stromal cells to treat cancer cells and measure changes in drug sensitivity

  • AREG neutralization: Employing anti-AREG antibodies or soluble EGFR fragments to block AREG activity

  • Genetic manipulation: Knockdown or overexpression of AREG in cancer or stromal cells to assess functional impacts

  • Combination therapy models: Testing AREG targeting agents in combination with conventional chemotherapy

Research findings indicate that targeting AREG not only minimizes chemoresistance of cancer cells but also restores immunocompetency when combined with classical chemotherapy in humanized models . This suggests potential therapeutic applications for AREG-targeting strategies in cancer treatment, particularly for overcoming resistance mechanisms.

What methodologies are used to study AREG's function in tissue regeneration?

The investigation of AREG's role in tissue regeneration, particularly in contexts like hair follicle development, employs specialized methodological approaches. Based on current research, several experimental systems have proven valuable:

  • Three-dimensional co-culture systems:
    A 3D co-culture methodology has been effectively utilized to study interactions between skin-derived precursors (SKPs) and other cell types like epidermal stem cells and adipose-derived stem cells. This system revealed that these cellular interactions enhance SKP proliferation and hair follicle regeneration capacity through AREG signaling . The co-culture approach allows for the investigation of complex cellular interactions in a controlled environment.

  • Cell isolation and culture protocols:
    For SKP isolation, specific methodologies involve:

    • Processing dermal cell suspensions through 100-μm cell strainers

    • Centrifugation at 1400 rpm at room temperature

    • Resuspension in red blood cell lysis buffer

    • Further centrifugation at 1400 rpm

    For adipose-derived stem cells (ASCs):

    • Processing through a 70-μm cell strainer

    • Centrifugation at 2000 rpm for 5 minutes

    • Culture in low-glucose DMEM containing 15% FBS, 1% L-glutamine, and 1% penicillin/streptomycin solution

  • Gene expression manipulation:
    siRNA approaches for AREG or pathway components involve:

    • Design using siRNA selection programmes

    • Transfection of cells at specific densities (e.g., 2.5 × 10^6 cells/mL)

    • Verification of transfection efficiency (>70%) using fluorescence microscopy

    • Functional assessment 48 hours post-transfection

  • Stemness assessment:
    Alkaline phosphatase (AP) staining is used to detect the reprogramming efficiency and subset of undifferentiated pluripotent stem cells with extensive self-renewal potential. This approach helps quantify SKP stemness, as AP activity decreases with differentiation .

  • Histological analysis:
    For in vivo assessment of hair follicle regeneration:

    • Skin samples are fixed with 4% PFA for 24 hours

    • Transferred to 70% ethanol

    • Embedded in paraffin and sectioned (10-μm thickness)

    • Stained using H&E staining kits according to standard procedures

These methodological approaches provide a comprehensive framework for investigating AREG's functions in tissue regeneration contexts, allowing researchers to dissect molecular mechanisms and cellular interactions in both in vitro and in vivo settings.

How should researchers design experiments to compare different forms of AREG?

When comparing different forms of AREG, researchers must carefully design experiments to account for the significant functional variations between variants. The 98 amino acid residue long form of recombinant amphiregulin has been shown to be approximately 5-10 fold more active than the 78 amino acid residue form in proliferation assays using Balb/3T3 fibroblasts . This substantial difference in potency necessitates methodological rigor in comparative studies.

Key experimental design considerations include:

  • Dose standardization:

    • Use molar concentrations rather than weight-based dosing

    • Employ wide concentration ranges (typically 0.1-100 ng/mL) to capture full dose-response relationships

    • Calculate and compare ED50 values as a quantitative measure of potency

  • Multiple readout systems:

    • Cell proliferation assays (MTT, BrdU, direct counting)

    • Receptor binding studies (competitive binding assays)

    • Downstream signaling activation (phosphorylation of EGFR, ERK, AKT)

    • Transcriptional responses (qPCR of known target genes)

  • Cell type considerations:

    • Test multiple responsive cell lines (Balb/3T3, keratinocytes, mammary epithelial cells)

    • Include both normal and cancer cell lines when relevant

    • Consider primary cells for physiological relevance

  • Time course analysis:

    • Monitor responses at multiple time points (early: 5-30 minutes; intermediate: 1-6 hours; late: 24-72 hours)

    • Assess both acute signaling responses and longer-term biological outcomes

When reporting results, researchers should clearly specify the exact form of AREG used, including amino acid sequence range, expression system, and any modifications or tags present. This information is critical for result interpretation and experimental reproducibility across different research groups.

What quality control measures should be implemented when working with Recombinant Human AREG?

Ensuring consistent quality of Recombinant Human AREG is essential for experimental reproducibility and reliable research outcomes. Comprehensive quality control measures should be implemented at multiple stages:

  • Initial protein characterization:

    • Purity assessment via SDS-PAGE (≥95-97% purity is typically required)

    • N-terminal sequence analysis to confirm identity

    • Mass spectrometry verification

    • Endotoxin testing using LAL method (<1 EU/μg is generally considered acceptable)

  • Functional validation:

    • Bioactivity testing in standardized cell proliferation assays

    • Verification of expected ED50 (5-15 ng/mL range for Balb/3T3 cells)

    • Receptor binding confirmation

    • Dose-response curve generation

  • Stability monitoring:

    • Regular testing of stored protein at defined intervals

    • Assessment of activity after reconstitution at various time points

    • Evaluation of freeze-thaw stability

    • Comparison against reference standards

  • Batch consistency verification:

    • Lot-to-lot comparison of critical parameters

    • Maintenance of internal reference standards

    • Documentation of variation between batches

  • Documentation practices:

    • Detailed record-keeping of source, lot number, and specifications

    • Documentation of reconstitution date and conditions

    • Tracking of usage and storage conditions

    • Retention of certificates of analysis

A typical quality control workflow might include initial verification upon receipt, functional validation before experimental use, and periodic stability checks throughout the usage period. For critical experiments, researchers should consider testing multiple lots or sourcing from multiple vendors to ensure result robustness and minimize batch-specific artifacts.

How can researchers effectively target AREG in disease models?

Targeting AREG in disease models requires strategic approaches tailored to specific research questions. Based on current findings, several methodological strategies have proven effective:

  • Genetic manipulation approaches:

    • siRNA-mediated knockdown: Utilizing specifically designed siRNAs to reduce AREG expression, with transfection efficiencies verified by fluorescence microscopy (>70% is considered acceptable)

    • CRISPR-Cas9 genome editing: For stable knockout models

    • Overexpression systems: To study gain-of-function effects

    • Inducible expression systems: For temporal control of manipulation

  • Protein neutralization strategies:

    • Anti-AREG neutralizing antibodies

    • Soluble EGFR fragments that compete for binding

    • Receptor-Fc fusion proteins

    • Small molecule inhibitors of AREG-EGFR interaction

  • Pathway inhibition approaches:

    • Targeting downstream signaling through PI3K inhibitors

    • MAPK pathway inhibitors to block AREG-mediated effects

    • Combination approaches targeting multiple nodes in the signaling network

  • Translational therapeutic strategies:

    • Combining AREG targeting with conventional chemotherapy

    • Assessment of immunocompetency restoration in cancer models

    • Delivery optimization for in vivo applications

In cancer research specifically, targeting AREG has shown promising results in minimizing chemoresistance and restoring immunocompetency when combined with classical chemotherapy . This suggests that AREG targeting may be particularly valuable in combination therapy approaches.

For tissue regeneration applications, modulating AREG signaling through targeted delivery or controlled release systems may enhance regenerative outcomes, as demonstrated in hair follicle regeneration models where AREG promotes SKP stemness and induces earlier telogen-to-anagen transition .

The choice of targeting strategy should be guided by specific research questions, disease context, and available model systems, with appropriate controls and validation steps to confirm targeting efficacy.

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.