Phospho-ESR1 (Ser104) Antibody

Shipped with Ice Packs
In Stock

Product Specs

Form
Supplied at 1.0mg/mL in phosphate buffered saline (without Mg2+ and Ca2+), pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
7*/654 isoform antibody; 7*/819 2 isoform antibody; 7*/822 isoform antibody; 8*/901 isoform antibody; 8*/941 isoform antibody; DKFZp686N23123 antibody; ER alpha antibody; ER antibody; ER-alpha antibody; Era antibody; ESR antibody; ESR1 antibody; ESR1_HUMAN antibody; ESRA antibody; Estradiol receptor antibody; Estrogen nuclear receptor alpha antibody; Estrogen receptor 1 antibody; Estrogen receptor alpha 3*,4,5,6,7*/822 isoform antibody; Estrogen receptor alpha antibody; Estrogen receptor alpha delta 3*,4,5,6,7*,8*/941 isoform antibody; Estrogen receptor alpha delta 3*,4,5,6,7*/819 2 isoform antibody; Estrogen receptor alpha delta 4 +49 isoform antibody; Estrogen receptor alpha delta 4*,5,6,7*/654 isoform antibody; Estrogen receptor alpha delta 4*,5,6,7,8*/901 isoform antibody; Estrogen receptor alpha E1 E2 1 2 antibody; Estrogen receptor alpha E1 N2 E2 1 2 antibody; Estrogen receptor antibody; ESTRR antibody; NR3A1 antibody; Nuclear receptor subfamily 3 group A member 1 antibody
Target Names
Uniprot No.

Target Background

Function
Estrogen receptor alpha (ERα) is a nuclear hormone receptor that plays a pivotal role in regulating eukaryotic gene expression. Steroid hormones and their receptors are involved in the regulation of cellular proliferation and differentiation in target tissues. Ligand-dependent nuclear transactivation occurs through either direct homodimer binding to a palindromic estrogen response element (ERE) sequence or by associating with other DNA-binding transcription factors, including AP-1/c-Jun, c-Fos, ATF-2, Sp1 and Sp3, to mediate ERE-independent signaling. Ligand binding induces a conformational change in ERα, enabling subsequent or combinatorial association with multiprotein coactivator complexes through LXXLL motifs of their respective components. Mutual transrepression occurs between ERα and NF-kappa-B in a cell-type specific manner. ERα decreases NF-kappa-B DNA-binding activity and inhibits NF-kappa-B-mediated transcription from the IL6 promoter. Additionally, it displaces RELA/p65 and associated coregulators from the promoter. ERα is recruited to the NF-kappa-B response element of the CCL2 and IL8 promoters and can displace CREBBP. It co-localizes with NF-kappa-B components RELA/p65 and NFKB1/p50 on ERE sequences. ERα can also act synergistically with NF-kappa-B to activate transcription involving respective recruitment adjacent response elements; this function involves CREBBP. ERα can activate the transcriptional activity of TFF1. It also mediates membrane-initiated estrogen signaling involving various kinase cascades. ERα is essential for MTA1-mediated transcriptional regulation of BRCA1 and BCAS3. It is involved in the activation of NOS3 and endothelial nitric oxide production. Isoforms lacking one or several functional domains are thought to modulate transcriptional activity by competitive ligand or DNA binding and/or heterodimerization with the full-length receptor. ERα binds to ERE and inhibits isoform 1.
Gene References Into Functions
  1. Estrogen-induced miR-191 was identified as a direct upstream regulator of DAB2 in ER-positive breast cancer cells. PMID: 29247596
  2. This research provides comprehensive genomic insights that contribute to a deeper understanding of ERα's biological roles in breast cancer. PMID: 30301189
  3. A correlation was observed between rs2046210 and rs3803662, and the risk of developing breast cancer in Vietnamese women. The A allele is associated with an increased risk for both rs2046210 (OR [95% CI] = 1.43 [1.14 - 1.78], P = 0.0015) and rs3803662 (OR [95% CI] = 1.45 [1.16 - 1.83], P = 0.001). In conclusion, two polymorphisms, rs2046210 in ESR1 and rs3803662 in TNRC9, are associated with breast cancer risk in the Vietnamese population. PMID: 30078824
  4. The study demonstrates that estrogen receptor alpha can enhance odonto/osteogenic differentiation of stem cells from apical papilla via ERK and JNK MAPK pathways. PMID: 30069950
  5. No association was found between polymorphisms in genes encoding estrogen receptors (ESR1 and ESR2) and excreted BPA levels in orthodontic patients after bracket bonding. PMID: 29961922
  6. Analysis of genome-wide ER binding sites identified mutant ER unique recruitment mediating the allele-specific transcriptional program. PMID: 29438694
  7. The study describes RNF8 as a co-activator of ERα, increasing ERα stability via a post-transcriptional pathway. This provides a new insight into the mechanisms by which RNF8 promotes cell growth in ERα-positive breast cancer. PMID: 28216286
  8. Reduced expression of ERβ1 in female ERα-negative papillary thyroid carcinoma patients is associated with greater progression of the disease. PMID: 29655286
  9. ERα and ERβ exhibit heterogeneous distribution in deep infiltrating endometriosis. PMID: 29383962
  10. ER-alpha36/EGFR signaling loop promotes growth of hepatocellular carcinoma cells. PMID: 29481815
  11. This study aimed to determine the presence and localization of estrogen receptors (ERs), progesterone receptors (PRs), and androgen receptors (ARs) in both healthy and varicose vein wall cells and their relationship with gender. PMID: 30250632
  12. Estrogen receptor-alpha was expressed only in women and showed a positive correlation with the amount of fungi in oral paracoccidioidomycosis. Progesterone receptor was observed in both genders and exhibited no correlation with estrogen receptor-alpha or fungi counting. PMID: 29796757
  13. ERα upregulates vinculin expression in breast cancer cells; Loss of vinculin promotes amoeboid features of cancer cells. PMID: 28266545
  14. Polymorphisms in the ERα gene do not predict in vitro fertilization outcome. PMID: 29916276
  15. High ESR1 expression is associated with metastasis in breast cancer. PMID: 29187405
  16. The G/G XbaI genotype of ESR1 gene is associated with breast cancer risk. PMID: 29893332
  17. miR-221 may impair the protective effect of estrogen in degenerated cartilaginous endplate cells by targeting estrogen receptor alpha. PMID: 29529124
  18. Results showed that NAT1 and ESR1 expression were increased in primary breast tumor samples compared with normal breast tissue samples, and in ER+ primary breast tumors compared with ER- tumors. Additionally, NAT1 and ESR1 expression appear to have overlapping regulation. PMID: 29901116
  19. All patients without ESR1 mutations identified by molecular barcode next-generation sequencing (MB-NGS) were confirmed to have no mutations by ddPCR. In conclusion, MB-NGS successfully detected ESR1 mutations in cfDNA with a higher sensitivity of 0.1% than conventional NGS and was considered clinically useful as ddPCR. PMID: 28905136
  20. An association was found between the presence of particular genotypes at three ESR1 polymorphisms (rs2234693, rs6902771, rs7774230) and one ESR2 polymorphism (rs3020449), and the presence of metabolic syndrome in postmenopausal women. PMID: 30049354
  21. A higher frequency of ESR1 and PIK3CA mutations was observed in plasma compared to serum in 33 MBC patients. Therefore, serum samples should not be considered the preferred source of cfDNA. PMID: 29689710
  22. These results suggest that miR-125a-3p can function as a novel tumor suppressor in ER(+) breast cancer by targeting CDK3, which may be a potential therapeutic approach for TamR breast cancer therapy. PMID: 28939591
  23. A significant finding of this study is that one out of five (20%) patients with breast cancer bone marrow metastasis (BM) had a receptor discrepancy between the primary tumor and subsequent BM. Loss of hormone receptors (ER and/or PR) expression and gain of HER2 overexpression were the most commonly observed changes. PMID: 28975433
  24. This study reports a key role of IGF-IR in the regulation of ERα-positive breast cancer cell aggressiveness and the regulation of expression levels of several extracellular matrix molecules. PMID: 28079144
  25. Associations between PvuII (T>C) and XbaI (A>G) polymorphisms of estrogen receptor alpha (ESR1) gene with type 2 diabetes mellitus (T2DM) or metabolic syndrome (MetS) are reported. PMID: 29883973
  26. The ERα gene appears to play a crucial role in stress urinary incontinence in the premenopausal period. PMID: 29769420
  27. This study reports the first discovery of naturally occurring ESR1 (Y537C) and ESR1 (Y537S) mutations in MCF7 and SUM44 ESR1-positive cell lines after acquisition of resistance to long-term-estrogen-deprivation (LTED) and subsequent resistance to fulvestrant (ICIR). These mutations became more frequent over time, impacted ESR1 binding to the genome, and altered the ESR1 interactome. PMID: 29192207
  28. Concomitant high expression of ERα36, GRP78, and GRP94 is associated with aggressive papillary thyroid cancer behavior and may be used as a predictor for extrathyroid extension, lymph node metastasis, and distant metastasis. PMID: 29368272
  29. Estrogen receptor-1 is a key regulator of HIV-1 latency that imparts gender-specific restrictions on the latent reservoir. PMID: 30061382
  30. Down-regulation of ESR1 gene expression was enhanced by the development of breast cancer. PMID: 29543921
  31. The aim of the present study was to assess whether fibrosis markers, estrogen receptor (ER)α, and the stromal derived factor (SDF)1/CXC chemokine receptor type 4 (CXCR4) axis are abnormally expressed in intrauterine adhesions endometrium. PMID: 29568895
  32. The frequency of alleles and genotypes of polymorphisms FSHR(-29G/A) and ESRI (XbaI A/G) in women with normal to poor response did not have a significant correlation. PMID: 29526845
  33. Each estrogen receptor alpha and estrogen receptor beta gene polymorphism might have a different impact on postmenopausal osteoporosis risk and bone mineral density in various ethnicities. PMID: 29458346
  34. The results suggest that the minor allele A of the ESR1 gene is associated with the development of arterial hypertension in men. PMID: 29658078
  35. The study found that tamoxifen treatment induced a decrease in PRMT2 and an increase in ER-alpha36 as well as ER-alpha36-mediated non-genomic effect in the MDA-MB-231 breast cancer cell line. PMID: 29620287
  36. ESR1 mutations are not associated with clinical resistance to fulvestrant in breast cancer patients. PMID: 27174596
  37. Overexpression of COPS5, through its isopeptidase activity, leads to ubiquitination and proteasome-mediated degradation of NCoR, a key corepressor for ERα and tamoxifen-mediated suppression of ERα target genes. PMID: 27375289
  38. ESR alpha PvuII and XbaI polymorphisms have no association with systemic lupus erythematosus. The combination of the TC/AA and CC/GG genotypes were associated with SLE susceptibility. PMID: 29356461
  39. Estrogen receptor (ER) and progesterone receptor (PR) expression in endometrial carcinoma (EC) were significantly higher than those in the paracarcinoma tissue and control. PMID: 29081408
  40. ESR1 promoter methylation was an independent risk factor and had a high value to predict 28-day mortality from acute-on-chronic hepatitis B liver failure. PMID: 29082740
  41. By analyzing different estrogen receptor-alpha(ER-a)-positive and ER-a-negative breast cancer cell lines, the role of CCN5 in the leptin-mediated regulation of growth and invasive capacity was defined. PMID: 29370782
  42. This study identified ESR1 as a direct target of miR-301a-3p. PMID: 29763890
  43. Authors report for the first time the presence of ESR1 methylation in plasma ctDNA of patients with HGSC. The agreement between ESR1 methylation in primary tumors and paired ctDNA is statistically significant. PMID: 29807696
  44. This study reports the development of a novel class of ERa AF2 inhibitors, which have the potential to effectively inhibit ERa activity by a unique mechanism and to circumvent the issue of mutation-driven resistance in breast cancer. PMID: 29462880
  45. The P2X7R rs3751143 and ER-alpha PvuII two-locus interaction confers a significantly high susceptibility to osteoporosis in Chinese postmenopausal women. PMID: 28884379
  46. Alcohol consumption may have differential effects on concordant and discordant receptor subtypes of breast cancer. PMID: 29353824
  47. ERα and ERβ mRNA expression was significantly higher (p < 0.05) in tumor tissues relative to their paired normal mucosa and correlated inversely with survival outcome. PMID: 29390981
  48. High ESR1 expression is associated with Papillary Thyroid Carcinoma. PMID: 28124274
  49. Oral administration of RAD140 substantially inhibited the growth of AR/ER(+) breast cancer patient-derived xenografts (PDX). Activation of the AR and suppression of the ER pathway, including the ESR1 gene, were observed with RAD140 treatment. PMID: 28974548
  50. Polymorphism in the ERα gene is associated with an increased risk for advanced Pelvic Organ Prolapse. However, polymorphism in the LAMC1 gene does not seem to be associated with this risk. PMID: 29241914

Show More

Hide All

Database Links

HGNC: 3467

OMIM: 133430

KEGG: hsa:2099

STRING: 9606.ENSP00000206249

UniGene: Hs.208124

Involvement In Disease
Estrogen resistance (ESTRR)
Protein Families
Nuclear hormone receptor family, NR3 subfamily
Subcellular Location
[Isoform 1]: Nucleus. Cytoplasm. Cell membrane; Peripheral membrane protein; Cytoplasmic side.; Nucleus. Golgi apparatus. Cell membrane. Note=Colocalizes with ZDHHC7 and ZDHHC21 in the Golgi apparatus where most probably palmitoylation occurs. Associated with the plasma membrane when palmitoylated.
Tissue Specificity
Widely expressed. Not expressed in the pituitary gland.; [Isoform 3]: Widely expressed, however not expressed in the pituitary gland.

Q&A

What is Phospho-ESR1 (Ser104) Antibody and what is its significance in research?

Phospho-ESR1 (Ser104) Antibody is a specialized antibody that specifically recognizes the estrogen receptor alpha (ERα) when phosphorylated at serine residue 104. This antibody is typically produced by immunizing rabbits with synthetic phosphopeptides containing the phosphorylated S104 sequence conjugated to KLH (keyhole limpet hemocyanin) . The significance of this antibody lies in its ability to detect post-translational modifications that regulate ERα activity, which is crucial for understanding estrogen signaling pathways in normal physiology and disease states.

ERα phosphorylation at specific residues in transcription activation function 1 (AF-1), including serine 104, has been shown to stimulate receptor activity in a ligand-independent manner . This phosphorylation event is particularly important in breast cancer research as it may contribute to mechanisms of endocrine therapy resistance, making phospho-specific antibodies valuable tools for investigating altered estrogen signaling in cancer progression .

What experimental applications are most suitable for Phospho-ESR1 (Ser104) Antibody?

Phospho-ESR1 (Ser104) Antibody has demonstrated utility in multiple experimental applications:

  • Western Blot (WB): Typically used at dilutions of 1:500-1:1000 for detecting phosphorylated ERα in cell or tissue lysates .

  • Immunohistochemistry (IHC): Effective at dilutions of 1:50-1:100 for examining phospho-ERα localization and expression in tissue sections .

  • ELISA: Particularly useful in cell-based ELISA formats for quantifying relative amounts of phosphorylated ERα in cultured cells .

The antibody shows reactivity with human and mouse samples, making it suitable for comparative studies across these species . For optimal results, experimental conditions should be validated and optimized for each specific application and sample type.

How can phosphorylation at Ser104 be induced or manipulated in experimental settings?

Phosphorylation of ERα at Ser104 can be experimentally induced or manipulated through several approaches:

  • MAPK pathway activation: Treatment with PMA (phorbol 12-myristate 13-acetate) activates the ERK1/2 MAPK pathway, leading to increased phosphorylation at Ser104 .

  • Estrogen treatment: Exposure to estradiol (E2) stimulates phosphorylation at Ser104, though this may involve both ligand-dependent and MAPK-mediated mechanisms .

  • Raf/Ras activation: Expression of constitutively active Raf or Ras can induce Ser104 phosphorylation through MAPK pathway stimulation .

  • Kinase inhibition studies: The MEK1/2 inhibitor U0126 can block phosphorylation at Ser104, confirming the role of the MAPK pathway and providing a negative control for experimental validation .

These experimental manipulations allow researchers to study the functional consequences of Ser104 phosphorylation in various cellular contexts.

How does phosphorylation at Ser104 interact with other ERα phosphorylation sites?

Phosphorylation at Ser104 demonstrates complex interrelationships with other ERα phosphorylation sites, particularly Ser106 and Ser118, which are also located within the AF-1 domain. Research indicates a hierarchical and potentially cooperative relationship between these sites:

  • Interdependence with Ser106 and Ser118: Phosphorylation status at Ser104 is influenced by the phosphorylation state of Ser106 and Ser118 . When Ser106 or Ser118 are substituted with alanine (preventing phosphorylation), Ser104 phosphorylation is reduced, suggesting that prior phosphorylation at these sites may facilitate Ser104 phosphorylation .

  • Sequential phosphorylation: Evidence suggests that phosphorylation at Ser118 may precede and enhance phosphorylation at Ser106, which in turn may promote phosphorylation at Ser104 . This sequential pattern indicates a coordinated regulation of multiple phosphorylation events.

  • Functional synergy: Combined phosphorylation at Ser104, Ser106, and Ser118 appears to produce synergistic effects on ERα transcriptional activity that exceed the effects of phosphorylation at individual sites .

These interactions highlight the complexity of ERα regulation through multisite phosphorylation and emphasize the importance of considering the phosphorylation status of all three sites when studying ERα function.

What role does Ser104 phosphorylation play in tamoxifen resistance mechanisms?

Phosphorylation of ERα at Ser104 contributes to tamoxifen resistance through several mechanisms:

  • Enhanced AF-1 activity: Phosphorylation at Ser104, along with Ser106, enhances the activity of the AF-1 domain, which can partially compensate for the antagonism of the AF-2 domain by tamoxifen, converting tamoxifen from an antagonist to a partial agonist .

  • 4-hydroxytamoxifen (OHT) agonist activity: Research shows that Ser104 and Ser106 are required for the agonist activity of OHT, alongside the well-established role of Ser118 . Mutation of these sites to alanine reduces the ability of OHT to stimulate ERα transcriptional activity.

  • MAPK hyperactivation: In some breast cancer cells, hyperactivation of MAPK signaling leads to increased phosphorylation at Ser104, Ser106, and Ser118, potentially contributing to tamoxifen resistance through ligand-independent activation of ERα .

  • Altered coregulator recruitment: Phosphorylation at these sites may modify interactions with transcriptional coregulators, shifting the balance from corepressor to coactivator binding in the presence of tamoxifen.

Understanding these mechanisms is crucial for developing strategies to overcome or prevent tamoxifen resistance in hormone-dependent breast cancers.

What methodological considerations are important when detecting Phospho-ERα (Ser104) in complex samples?

Detecting Phospho-ERα (Ser104) in complex biological samples requires careful methodological consideration:

  • Antibody validation: Confirm antibody specificity using appropriate controls:

    • Phosphopeptide competition assays to verify phospho-specificity

    • Comparison of wild-type ERα with S104A mutants

    • Inclusion of phosphatase-treated samples as negative controls

  • Sample preparation: Phosphorylation status can be labile:

    • Use phosphatase inhibitors (e.g., sodium fluoride, sodium orthovanadate) in all buffers

    • Process samples rapidly and maintain cold temperatures

    • Consider crosslinking agents for IHC applications to preserve phosphoepitopes

  • Signal normalization strategies:

    • Use total ERα antibodies for normalization to account for variations in total protein expression

    • Include GAPDH as an internal loading control

    • For cell-based assays, normalize to cell number using Crystal Violet staining

  • Considerations for specific techniques:

    • For Western blotting: Use 7-8% gels for optimal resolution of phosphorylated forms

    • For IHC: Optimize antigen retrieval conditions (pH, temperature, duration)

    • For ELISA: Be aware of potential cross-reactivity with other phosphorylation sites

These methodological considerations help ensure reliable and reproducible detection of Phospho-ERα (Ser104) in experimental settings.

How can I design experiments to investigate the functional consequences of Ser104 phosphorylation?

Designing experiments to investigate the functional significance of Ser104 phosphorylation requires multiple complementary approaches:

  • Site-directed mutagenesis strategies:

    • Generate S104A mutants (preventing phosphorylation) to study loss-of-function effects

    • Create S104E or S104D phosphomimetic mutants to model constitutive phosphorylation

    • Develop multiple mutants (e.g., S104A/S106A/S118A) to study combinatorial effects

  • Reporter gene assays:

    • Transfect cells with ERE-luciferase reporters alongside wild-type or mutant ERα

    • Compare transcriptional activity in response to different ligands (estradiol, SERMs)

    • Examine how MAPK pathway modulators affect wild-type versus mutant ERα activity

  • Protein-protein interaction studies:

    • Perform co-immunoprecipitation with wild-type and mutant ERα

    • Use mammalian two-hybrid assays to identify differential coregulator recruitment

    • Apply proximity ligation assays to visualize interactions in situ

  • Chromatin immunoprecipitation (ChIP):

    • Compare binding of wild-type and phospho-mutant ERα to target gene promoters

    • Investigate recruitment of coregulators in association with phosphorylated ERα

    • Perform sequential ChIP to examine multiple modifications simultaneously

These experimental approaches provide complementary data about how Ser104 phosphorylation affects ERα function at molecular, cellular, and genomic levels.

What are the common challenges and solutions when working with phospho-specific antibodies like Phospho-ESR1 (Ser104)?

Working with phospho-specific antibodies presents several challenges that require specific solutions:

ChallengeDescriptionSolution
Nonspecific bindingCross-reactivity with similar phospho-epitopesPerform peptide competition assays with phosphorylated and non-phosphorylated peptides
Low signal intensityPhosphorylation represents a small fraction of total proteinUse signal amplification methods; enrich for phosphoproteins before detection
Epitope maskingPhosphorylation site may be obscured by protein interactionsTry different fixation/extraction methods; optimize antigen retrieval
Phosphorylation labilityPhosphate groups can be lost during sample processingAdd phosphatase inhibitors to all buffers; keep samples cold; process rapidly
Batch-to-batch variabilityPolyclonal antibodies may vary between lotsValidate each new antibody lot; consider monoclonal alternatives when available
Background in IHCNonspecific tissue bindingOptimize blocking conditions; include phosphopeptide competition controls

Additional strategies to improve phospho-specific detection include using signal amplification techniques like tyramide signal amplification for IHC, or deploying proximity ligation assays to detect specific protein-protein interactions only when phosphorylation is present.

How should quantitative analysis of Phospho-ESR1 (Ser104) be performed and interpreted?

Quantitative analysis of Phospho-ESR1 (Ser104) requires rigorous methodology and careful interpretation:

  • Normalization strategies:

    • Always normalize phospho-signal to total ERα expression to account for variations in expression levels

    • Use housekeeping proteins (GAPDH) as loading controls for Western blot and cell-based assays

    • For cell-based assays, normalize to cell number using Crystal Violet staining to adjust for plating differences

  • Quantification methods:

    • For Western blots: Use digital image analysis with linear dynamic range verification

    • For IHC: Apply H-score or Allred scoring systems with blinded evaluation

    • For ELISA: Generate standard curves using recombinant proteins when possible

  • Statistical analysis:

    • Compare phospho/total ratios rather than absolute phospho-signal

    • Use appropriate statistical tests based on data distribution

    • Consider biological replicates (different samples) separately from technical replicates

  • Interpretation considerations:

    • Context-dependent phosphorylation may vary by cell type, stimulus, and time point

    • Correlation with functional outcomes requires parallel assays (transcription, proliferation)

    • Phosphorylation at S104 should be interpreted in conjunction with S106 and S118 status due to their interdependence

  • Validation approaches:

    • Confirm key findings with alternative detection methods

    • Use phosphatase treatment as negative control

    • Employ MAPK inhibitors (U0126) to demonstrate specificity of the phosphorylation mechanism

Following these guidelines ensures robust quantitative analysis of Phospho-ESR1 (Ser104) and meaningful interpretation of experimental results.

How do cell-based ELISA techniques compare with other methods for studying Phospho-ESR1 (Ser104)?

Cell-based ELISA techniques offer distinct advantages and limitations compared to other methods for studying Phospho-ESR1 (Ser104):

MethodAdvantagesLimitationsBest Applications
Cell-based ELISA- High throughput screening capability
- Quantitative results
- In situ detection in intact cells
- Normalization to cell number possible
- Parallel detection of total ERα
- Limited spatial resolution
- Cannot distinguish subcellular localization
- May detect non-specific signals
- Screening compounds that affect phosphorylation
- Dose-response studies
- siRNA screens
- Time-course experiments
Western Blot- Size verification of target protein
- Semi-quantitative
- Can detect multiple phospho-sites
- Lower throughput
- More labor-intensive
- Requires cell lysis
- Verification of antibody specificity
- Detailed analysis of phosphorylation patterns
- Detecting multiple phosphorylation sites
Immunofluorescence- Subcellular localization information
- Single-cell resolution
- Compatible with other markers
- Qualitative or semi-quantitative
- Photo-bleaching concerns
- More complex analysis
- Determining subcellular localization
- Heterogeneity studies
- Co-localization with other proteins
Mass Spectrometry- Direct identification of phospho-sites
- No antibody dependency
- Can discover novel sites
- Expensive equipment
- Lower sensitivity
- Complex sample preparation
- Comprehensive phosphorylation mapping
- Confirming antibody specificity
- Discovery research

Cell-based ELISA techniques are particularly valuable for high-throughput screening of compounds or conditions that affect ERα phosphorylation at Ser104, making them ideal for initial screens that can later be validated with more detailed but lower-throughput methods .

What emerging techniques might enhance our understanding of ERα Ser104 phosphorylation dynamics?

Several emerging technologies hold promise for advancing our understanding of ERα Ser104 phosphorylation dynamics:

  • Phospho-specific proximity ligation assays (PLA):

    • Enables visualization of phosphorylated ERα interactions with specific cofactors

    • Provides single-molecule sensitivity in intact cells

    • Can reveal cell-to-cell variability in phosphorylation status

  • CRISPR-Cas9 genome editing:

    • Generation of endogenous S104A or S104E knock-in cell lines

    • Creates physiologically relevant models with normal expression levels

    • Eliminates artifacts associated with overexpression systems

  • Phospho-proteomic approaches:

    • Tandem mass tag (TMT) labeling for quantitative comparisons across conditions

    • Phospho-enrichment strategies to enhance detection sensitivity

    • Integration with other -omics data for systems-level insights

  • Live-cell phosphorylation sensors:

    • FRET-based biosensors to monitor Ser104 phosphorylation in real-time

    • Optogenetic approaches to control kinase activity with spatial precision

    • Single-cell tracking of phosphorylation dynamics

  • Spatial transcriptomics:

    • Correlating Ser104 phosphorylation with transcriptional outputs in tissue context

    • Understanding heterogeneity of phosphorylation and its consequences

    • Linking phosphorylation to microenvironmental factors

These emerging techniques will provide unprecedented temporal and spatial resolution for studying ERα phosphorylation dynamics, potentially revealing new aspects of estrogen signaling in normal physiology and disease states.

How might Phospho-ESR1 (Ser104) research contribute to precision medicine approaches for breast cancer?

Research on Phospho-ESR1 (Ser104) has several potential applications in precision medicine for breast cancer:

  • Biomarker development:

    • Phospho-S104 levels could serve as predictive biomarkers for response to endocrine therapies

    • The ratio of phosphorylated to total ERα might identify patients at risk for resistance

    • Combined assessment of S104, S106, and S118 phosphorylation could provide a "phospho-signature" with greater predictive value than single markers

  • Therapeutic target identification:

    • Understanding the kinases responsible for S104 phosphorylation (ERK1/2 MAPK) supports the rational combination of MAPK inhibitors with endocrine therapies

    • Agents that specifically block the consequences of S104 phosphorylation without affecting beneficial ERα functions could minimize side effects

  • Resistance mechanism characterization:

    • Phosphorylation at S104 appears to contribute to tamoxifen resistance by enhancing the agonist properties of 4-hydroxytamoxifen

    • This insight can guide therapy selection, suggesting that patients with elevated S104 phosphorylation might benefit from alternative therapies

  • Patient stratification strategies:

    • Classifying tumors based on ERα phosphorylation patterns could identify distinct biological subgroups within ERα-positive breast cancers

    • This stratification could inform clinical trial design and interpretation, potentially explaining differential responses to targeted therapies

  • Monitoring treatment response:

    • Serial assessment of phosphorylation status could provide early indicators of developing resistance

    • Dynamic changes in phosphorylation might guide adaptive therapy approaches

These applications highlight how detailed understanding of ERα phosphorylation at S104 could translate into clinically relevant tools for personalizing breast cancer treatment and improving patient outcomes.

How should researchers interpret conflicting results in Phospho-ESR1 (Ser104) detection across different experimental systems?

When faced with conflicting results in Phospho-ESR1 (Ser104) detection across different experimental systems, researchers should consider several factors:

  • Antibody variables:

    • Different antibodies may have varying specificity and sensitivity for the phospho-epitope

    • Polyclonal antibodies can show batch-to-batch variation

    • Confirm results with multiple antibodies or alternative detection methods

  • Cell/tissue context considerations:

    • Phosphorylation patterns may legitimately differ between cell types due to different kinase activities

    • Primary tissues versus cell lines may show different baseline phosphorylation levels

    • Microenvironmental factors can influence phosphorylation status

  • Technical variables to evaluate:

    • Sample preparation methods (lysis buffers, fixation protocols)

    • Presence and concentration of phosphatase inhibitors

    • Time between sample collection and analysis

    • Detection method sensitivity thresholds

  • Biological complexity:

    • Interdependence between phosphorylation sites may affect epitope availability

    • Temporal dynamics of phosphorylation (rapid vs. sustained)

    • Heterogeneity within samples (single-cell methods may reveal subpopulations)

  • Validation approaches:

    • Use phosphatase treatment as a negative control

    • Include S104A mutants as specificity controls

    • Apply MAPK pathway inhibitors (U0126) to confirm pathway-specific phosphorylation

    • Consider phosphopeptide competition assays to verify antibody specificity

By systematically addressing these factors, researchers can resolve apparent contradictions and develop a more nuanced understanding of the biological variability in ERα phosphorylation.

What are the most appropriate positive and negative controls for Phospho-ESR1 (Ser104) experiments?

Establishing appropriate controls is critical for robust Phospho-ESR1 (Ser104) experiments:

Positive Controls:

  • Stimulated cell lysates:

    • Cells treated with PMA to activate MAPK signaling

    • Estradiol (E2)-treated cells expressing wild-type ERα

    • Cells expressing constitutively active Raf or Ras

  • Phosphomimetic mutants:

    • S104E or S104D ERα mutants that mimic phosphorylation

    • Can serve as controls for functional studies, though they may not perfectly replicate phosphorylation effects

  • Known positive samples:

    • MCF-7 breast cancer cells treated with growth factors

    • Certain breast cancer tissue samples with known MAPK activation

Negative Controls:

  • Phosphorylation site mutants:

    • S104A mutant ERα (prevents phosphorylation at the specific site)

    • Allows discrimination between specific and non-specific antibody binding

  • Phosphatase-treated samples:

    • Treatment with lambda phosphatase to remove phosphate groups

    • Verifies that signal depends on phosphorylation status

  • Kinase inhibition:

    • U0126 (MEK1/2 inhibitor) treatment to block MAPK-mediated phosphorylation

    • Provides pathway-specific negative control

  • Peptide competition:

    • Pre-incubation of antibody with phosphorylated peptide containing the S104 epitope

    • Should abolish specific signal while non-phosphorylated peptide should have minimal effect

  • ERα-negative cells:

    • Cell lines lacking ERα expression

    • Controls for non-specific antibody binding

Using these positive and negative controls systematically enhances the reliability and interpretability of Phospho-ESR1 (Ser104) experiments.

How can phosphorylation at Ser104 be distinguished from other post-translational modifications of ERα?

Distinguishing phosphorylation at Ser104 from other post-translational modifications (PTMs) of ERα requires specific experimental approaches:

  • Antibody-based methods with enhanced specificity:

    • Use antibodies that recognize specific phosphorylated residues and their surrounding sequence context

    • Perform peptide competition assays with phosphorylated and non-phosphorylated peptides to confirm specificity

    • Apply multiple antibodies targeting different epitopes containing the same modification

  • Mutational analyses:

    • Generate site-specific mutants (S104A) to prevent phosphorylation at the site of interest

    • Compare with wild-type ERα and other PTM site mutants

    • Create combinatorial mutants to study interactions between modifications

  • Mass spectrometry approaches:

    • Perform tandem MS (MS/MS) to identify specific modified residues

    • Use neutral loss scanning to detect phosphorylation

    • Apply multiple fragmentation methods (CID, ETD, HCD) for comprehensive PTM mapping

    • Quantify modification stoichiometry at specific sites

  • Enzymatic treatments:

    • Use phosphatase treatment to remove phosphorylation

    • Apply deubiquitinases or deacetylases to remove other specific PTMs

    • Compare modification patterns before and after treatment

  • Temporal dynamics and stimulus specificity:

    • Different PTMs may show distinct temporal patterns after stimulation

    • MAPK activators (PMA) specifically enhance Ser104 phosphorylation

    • Compare responses to different stimuli to distinguish pathway-specific modifications

By combining these approaches, researchers can confidently distinguish phosphorylation at Ser104 from other PTMs and understand their potentially independent or cooperative effects on ERα function.

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.