Phospho-ESR1 (Ser118) Antibody

Shipped with Ice Packs
In Stock

Description

Biological Significance of ESR1 Phosphorylation at Ser118

Phosphorylation of ERα at Ser118 is a hallmark of ligand-independent activation, commonly observed in hormone-resistant breast cancers . This modification:

  • Disrupts hydrophobic interactions between aromatic-rich clusters in ERα’s intrinsically disordered N-terminal transactivation domain (NTD), leading to structural expansion and enhanced transcriptional activity .

  • Enables cofactor recruitment (e.g., TIF2) and promoter-specific gene regulation (e.g., MYC, CCND1) .

  • Drives therapy resistance by maintaining ERα activity under antiestrogen treatment .

Applications in Research and Clinical Contexts

The Phospho-ESR1 (Ser118) antibody is widely used in:

ApplicationDetailsSource
Western BlottingDetects endogenous pSer118-ERα at 66 kDa; optimal dilution: 1:1,000
ImmunohistochemistryIdentifies pSer118-ERα in paraffin-embedded tissues; dilution: 1:800
Chromatin Immunoprecipitation (ChIP)Maps ERα phosphorylation-dependent promoter occupancy (e.g., TFF1, CCND1)

Gene-Specific Promoter Recruitment

  • pS2 (TFF1) promoter: ERα phosphorylated at Ser118 dissociates after estrogen treatment, contrasting with pan-ERα recruitment .

  • c-MYC and cyclin D1 promoters: S118A mutants show reduced ERα binding and transcriptional activation .

Genome-Wide Cistrome Analysis

  • ChIP-seq data reveal 14,063 high-confidence pSer118-ERα occupancy sites, 94.3% overlapping with total ERα peaks .

  • Functional enrichment: pSer118-ERα sites are associated with active enhancers (H3K27ac+) and estrogen-upregulated genes (29.9% within 100 kb of TSS) .

StudyKey InsightReference
Conformational dynamicsSer118 phosphorylation expands ERα-NTD by 5.2 Å, altering cofactor interactions
Hydrophobic disruption rescueS118A/F120A and S118A/L121A mutants restore transcriptional activity
Coregulator recruitmentpSer118 reduces ERα-NTD binding to TIF2-QRD, modulating promoter activity

Implications for Breast Cancer Therapeutics

Targeting ERα phosphorylation at Ser118 remains a promising strategy:

  • CDK7 inhibitors (e.g., SY-5609) disrupt phosphorylation-driven ERα activation in clinical trials .

  • Hydrophobic cluster disruptors could directly modulate ERα-NTD conformation, bypassing kinase dependency .

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 products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method and location. Please contact your local distributor for specific delivery information.
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
The estrogen receptor (ER) is a nuclear hormone receptor crucial for regulating eukaryotic gene expression. Steroid hormones and their receptors play vital roles in controlling cellular proliferation and differentiation in target tissues. Ligand-dependent nuclear transactivation can occur through direct homodimer binding to a palindromic estrogen response element (ERE) sequence or by associating with other DNA-binding transcription factors, such as AP-1/c-Jun, c-Fos, ATF-2, Sp1, and Sp3, to mediate ERE-independent signaling. Ligand binding triggers a conformational change, facilitating the subsequent or combinatorial association with multiprotein coactivator complexes through LXXLL motifs of their respective components. Mutual transrepression takes place between ER and NF-kappa-B in a cell-type specific manner. ER decreases NF-kappa-B DNA-binding activity, inhibits NF-kappa-B-mediated transcription from the IL6 promoter, and displaces RELA/p65 and associated coregulators from the promoter. ER is recruited to the NF-kappa-B response element of the CCL2 and IL8 promoters and can displace CREBBP. ER co-localizes with NF-kappa-B components RELA/p65 and NFKB1/p50 on ERE sequences. ER can also act synergistically with NF-kappa-B to activate transcription involving respective recruitment adjacent response elements; this function involves CREBBP. ER can activate the transcriptional activity of TFF1. Additionally, ER mediates membrane-initiated estrogen signaling involving various kinase cascades. ER is essential for MTA1-mediated transcriptional regulation of BRCA1 and BCAS3. ER 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. ER 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. The whole-genome insights carried in this work can help fully understand the biological roles of ER1 in breast cancer. PMID: 30301189
  3. There was a relationship between rs2046210 and rs3803662, and the risk of developing breast cancer in Vietnamese women. The A allele is the risk allele 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). We conclude that two polymorphisms, rs2046210 in ESR1 and rs3803662 in TNRC9, are associated with breast cancer risk in the Vietnamese population. PMID: 30078824
  4. This research demonstrates that Oestrogen receptor alpha can enhance the odonto/osteogenic differentiation of stem cells from apical papilla via ERK and JNK MAPK pathways. PMID: 30069950
  5. No association between polymorphisms in genes encoding estrogen receptors (ESR1 and ESR2) and excreted BPA levels was found in orthodontic patients after bracket bonding. PMID: 29961922
  6. Analysis of the genome-wide ER binding sites identified mutant ER unique recruitment mediating the allele-specific transcriptional program. PMID: 29438694
  7. This study describes RNF8 as a co-activator of ERalpha that increases ERalpha stability via a post-transcriptional pathway, providing a new insight into mechanisms for RNF8 to promote cell growth of ERalpha-positive breast cancer. PMID: 28216286
  8. Reduced expression of ERbeta1 in female ERalpha-negative papillary thyroid carcinoma patients is associated with greater progression of the disease. PMID: 29655286
  9. ERbeta exhibits a 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 oestrogen 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, while progesterone receptor was observed in both genders and exhibited no correlation with estrogen receptor-alpha or fungi counting. PMID: 29796757
  13. ERalpha upregulates vinculin expression in breast cancer cells; Loss of vinculin promotes amoeboid features of cancer cells. PMID: 28266545
  14. Polymorphisms 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 through 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. Furthermore, NAT1 and ESR1 expression appear to have overlapping regulation. PMID: 29901116
  19. All the patients without these mutations by molecular barcode next-generation sequencing (MB-NGS) were found to have no mutations by ddPCR. In conclusion, MB-NGS could successfully detect 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 between the presence of the particular genotypes at the three ESR1 polymorphisms (rs2234693, rs6902771, rs7774230) and one ESR2 polymorphism (rs3020449), and the presence of metabolic syndrome in postmenopausal women was found. PMID: 30049354
  21. This research suggests that serum samples should not be considered the preferred source of cfDNA, as higher frequency of ESR1 and PIK3CA mutations were observed in the plasma than in the serum in 33 MBC patients. 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 major finding of our study is that one out of five (20%) patients with breast cancer BM had a receptor discrepancy between the primary tumor and the subsequent BM, with loss of hormone receptors (ER and/or PR) expression, and gain of HER2 overexpression as the most commonly observed changes. PMID: 28975433
  24. This study reports a nodal role of IGF-IR in the regulation of ERalpha-positive breast cancer cell aggressiveness and the regulation of expression levels of several extracellular matrix molecules. PMID: 28079144
  25. The 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 ERalpha gene appears to play a key 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). Mutations were enriched with time, impacted on ESR1 binding to the genome and altered the ESR1 interactome. PMID: 29192207
  28. Concomitant high expression of ERalpha36, 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 the breast cancer. PMID: 29543921
  31. The aim of the present study was to assess whether fibrosis markers, estrogen receptor (ER)alpha 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 significant correlation. PMID: 29526845
  33. Each estrogen receptor alpha and estrogen receptor beta gene polymorphism might have different impact on postmenopausal osteoporosis risk and bone mineral density in various ethnicities. PMID: 29458346
  34. The results suggest that minor allele A of ESR1 gene is associated with the development of arterial hypertension in men. PMID: 29658078
  35. This 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 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 ERalpha and tamoxifen-mediated suppression of ERalpha 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, this research defined the role of CCN5 in the leptin-mediated regulation of growth and invasive capacity. PMID: 29370782
  42. This study identified ESR1 as a direct target of miR-301a-3p. PMID: 29763890
  43. This study reports 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. ERalpha and ERbeta 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 AR and suppression of ER pathway, including the ESR1 gene, were seen with RAD140 treatment. PMID: 28974548
  50. Polymorphism in the ERalpha 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 such 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 the biological significance of ESR1 phosphorylation at Serine 118?

ESR1 (Estrogen Receptor alpha) phosphorylation at Serine 118 plays a critical role in regulating the receptor's transcriptional activity. This specific post-translational modification occurs within the activation function-1 (AF-1) domain of the receptor. Research indicates that S118 phosphorylation significantly impacts ESR1 function, as mutation of this site substantially inhibits ESR1 activity. Mechanistically, phosphorylation at S118 directs gene-specific recruitment of ERα and various transcriptional coregulators to ERα target gene promoters . This phosphorylation event can be catalyzed by multiple kinases, including ERK1/2 mitogen-activated protein kinases (MAPK) and the cyclin-dependent protein kinase Cdk7 , suggesting integration of multiple signaling pathways in the regulation of ESR1 function.

Which kinases are responsible for phosphorylating ESR1 at Serine 118?

The phosphorylation of ESR1 at Serine 118 is mediated by several kinases as indicated in the research data. Primary kinases documented to phosphorylate this site include:

  • ERK1/2 mitogen-activated protein kinases (MAPK)

  • Cyclin-dependent protein kinase Cdk7

  • Cyclin A/CDK2

  • Casein kinase 1 (CK1)

  • LMTK3 (in vitro)

This multi-kinase regulation suggests that ESR1 S118 phosphorylation integrates signals from various cellular pathways, including cell cycle regulation, growth factor signaling, and other stimulus-dependent processes. The diversity of kinases involved indicates the importance of this phosphorylation site as a convergence point for multiple signaling networks regulating estrogen receptor function.

How does phosphorylation at S118 affect ESR1 function?

Phosphorylation at S118 has several documented effects on ESR1 function:

  • Enhancement of transcriptional activity: Research indicates that phosphorylation at this site "probably enhances transcriptional activity" .

  • Regulation of protein-protein interactions: Phosphorylation directs the recruitment of transcriptional coregulators including SRC-1, TIF-2, and AIB to ERα target gene promoters .

  • Counterregulation mechanisms: Dephosphorylation at Ser-118 by protein phosphatase PPP5C specifically inhibits ESR1's transactivation activity .

  • Self-regulation: Self-association of the receptor can induce phosphorylation .

The functional consequences of S118 phosphorylation demonstrate its central role in fine-tuning estrogen receptor signaling in response to various cellular conditions, making it a critical regulatory node in hormone-responsive tissues.

What types of Phospho-ESR1 (S118) antibodies are commercially available?

Based on the search results, there are multiple types of Phospho-ESR1 (S118) antibodies available for research use:

Antibody TypeHostApplicationsReactivityNotes
PolyclonalRabbitWB/IHC/IF/ELISAHuman/MouseAffinity-purified from rabbit antiserum
Recombinant MonoclonalRabbitELISA/IHCHumanProduced via recombinant technology
PolyclonalRabbitELISA/IHC/WBHuman/MouseLiquid in PBS with glycerol, BSA, and sodium azide

Each antibody type offers specific advantages depending on the research application, with polyclonal antibodies potentially recognizing multiple epitopes while monoclonal antibodies provide higher specificity for the phosphorylated S118 site. Researchers should select the appropriate antibody based on their specific experimental requirements and validation needs.

What are the optimal protocols for Western blot applications using Phospho-ESR1 (S118) antibody?

Based on the validated methodologies from the search results, the following protocol guidelines are recommended for Western blot applications using Phospho-ESR1 (S118) antibody:

  • Sample preparation:

    • Use appropriately treated cells (e.g., MCF7 cells treated with estradiol to induce phosphorylation)

    • Prepare lysates using standard cell lysis buffers containing phosphatase inhibitors to preserve phosphorylation status

  • Antibody dilution ranges:

    • Primary antibody: 1:500-1:2000 dilution is recommended

    • Secondary antibody: Follow manufacturer's recommendations for anti-rabbit detection systems

  • Controls:

    • Positive control: Lysates from cells known to express phosphorylated ESR1 (e.g., estradiol-treated MCF7 cells)

    • Negative control: Include a phospho-blocking peptide control to confirm specificity

  • Detection considerations:

    • The expected molecular weight for ESR1 is approximately 66 kDa

    • Phosphorylation may cause slight shifts in apparent molecular weight

For validation, one study showed specific detection of phosphorylated ESR1 in MCF7 cells treated with estradiol, with signal effectively blocked by phospho-peptide pre-treatment, confirming antibody specificity .

How should Phospho-ESR1 (S118) antibodies be validated for research applications?

Comprehensive validation of Phospho-ESR1 (S118) antibodies should include multiple approaches:

  • Specificity validation:

    • Phospho-blocking peptide competition assays: Signal should be abolished when antibody is pre-incubated with the phosphopeptide corresponding to the S118 region

    • Comparison of signal between phosphorylated and non-phosphorylated samples

    • Testing in S118A mutant cells (where serine is replaced with alanine)

  • Application-specific validation:

    • For Western blot: Demonstrate single band at expected molecular weight that disappears with phosphatase treatment

    • For IHC: Compare staining patterns with and without phospho-blocking peptide

    • For ChIP: Validate with known ERα target genes and compare with total ERα binding

  • Treatment-dependent validation:

    • Demonstrate increased signal after treatments known to induce S118 phosphorylation (e.g., estradiol, growth factors)

    • Show decreased signal after treatment with kinase inhibitors that block S118 phosphorylation

Documentation of validation results should include images of control experiments and quantification of signal differences between experimental conditions to establish robust methodology.

What are the recommended storage and handling conditions for Phospho-ESR1 (S118) antibodies?

Based on manufacturer guidelines from the search results, the following storage and handling recommendations apply to Phospho-ESR1 (S118) antibodies:

  • Long-term storage:

    • Store at -20°C for up to 1 year from the date of receipt

    • Antibodies are typically supplied in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide

  • Short-term storage:

    • For frequent use, store at 4°C for up to one month

  • Critical handling considerations:

    • Avoid repeated freeze-thaw cycles as this can degrade antibody quality

    • Aliquot antibody upon receipt if multiple uses are anticipated

    • Ensure proper centrifugation of antibody vial before opening

    • Follow manufacturer's recommendations for thawing frozen antibodies

  • Working dilution preparation:

    • Prepare working dilutions fresh on the day of use

    • Dilute in appropriate buffer according to application (e.g., blocking buffer for Western blot)

Proper storage and handling are essential for maintaining antibody specificity and sensitivity, particularly for phospho-specific antibodies which can be more vulnerable to degradation.

How can Phospho-ESR1 (S118) antibodies be used in Chromatin Immunoprecipitation (ChIP) assays?

Chromatin Immunoprecipitation using Phospho-ESR1 (S118) antibodies allows researchers to identify genomic targets specifically bound by the phosphorylated form of the receptor. Based on the methodologies described in the search results, a recommended ChIP protocol would include:

  • Sample preparation:

    • Cross-link protein-DNA complexes using formaldehyde (typically 1% for 10 minutes)

    • Isolate and sonicate chromatin to fragments of approximately 200-500 bp

    • Pre-clear chromatin using isotype-matched IgG and protein A-magnetic beads

  • Immunoprecipitation:

    • Add phospho-ERα S118 specific antibody (1-5 μg) to pre-cleared chromatin

    • Include parallel IPs with pan-ERα antibody and IgG control

    • Add protein A-magnetic beads and incubate overnight at 4°C

    • Perform stringent washing to remove non-specific interactions

  • Analysis approaches:

    • qPCR analysis of known ERα target promoters

    • ChIP-seq for genome-wide analysis of phospho-ERα binding sites

    • Compare binding profiles between phospho-ERα and total ERα

  • Validation strategies:

    • Demonstrate estrogen-dependent recruitment

    • Show differential recruitment compared to total ERα

    • Confirm association with coregulators like SRC-1, TIF-2, and AIB

This approach enables researchers to distinguish between binding events mediated by total ERα versus those specifically involving the S118-phosphorylated form, providing insights into phosphorylation-dependent gene regulation.

What are the key differences between using polyclonal versus monoclonal phospho-ESR1 (S118) antibodies?

The choice between polyclonal and monoclonal phospho-ESR1 (S118) antibodies significantly impacts experimental outcomes:

FeaturePolyclonal Antibodies Recombinant Monoclonal Antibodies
Epitope recognitionRecognize multiple epitopes around the phospho-S118 siteTarget a single epitope at the phospho-S118 site
Production methodGenerated in rabbits immunized with synthetic phosphopeptidesRecombinantly produced following cloning of antibody sequences
Batch-to-batch variabilityHigher; each immunization produces different antibody mixturesLower; recombinant production ensures consistency
Background signalPotentially higher due to multiple epitope recognitionTypically lower due to single epitope specificity
ApplicationsBroader range (WB, IHC, IF, ELISA)May be optimized for specific applications
SensitivityOften higher sensitivity due to multiple epitope bindingMay have lower sensitivity but higher specificity
Ideal use caseInitial screening or when signal amplification is neededPrecise detection of phospho-S118 with minimal cross-reactivity

When designing experiments, researchers should consider these differences and select the antibody type that best aligns with their specific research questions and technical requirements.

How does the phosphorylation status at S118 change in response to different stimuli and in different cell types?

The phosphorylation of ESR1 at S118 shows stimulus-dependent and cell type-specific regulation patterns:

Stimulus-dependent regulation:

  • Estradiol (E2) treatment: Induces rapid phosphorylation at S118, as demonstrated in MCF7 cells

  • Growth factor signaling: EGF, IGF-1, and other growth factors can induce S118 phosphorylation through MAPK pathway activation

  • Cell cycle progression: Cyclin A/CDK2 phosphorylates S118 in a cell cycle-dependent manner

Cell type-specific considerations:

  • Hormone-responsive cancer cells: Breast cancer cell lines like MCF7 show robust S118 phosphorylation in response to estradiol

  • Hormone-independent contexts: Some cells may show constitutive S118 phosphorylation through growth factor pathway activation

  • Normal versus cancer tissues: Differential regulation patterns may exist between normal breast tissue and breast carcinomas

Pathological significance:

  • Breast cancer: Altered S118 phosphorylation patterns correlate with disease progression and treatment response

  • Endocrine resistance: Increased S118 phosphorylation has been associated with tamoxifen resistance in some studies

Understanding these context-dependent phosphorylation patterns is crucial for interpreting experimental results and may have implications for therapeutic targeting in diseases where ESR1 signaling is dysregulated.

What is the relationship between ESR1 S118 phosphorylation and other post-translational modifications?

ESR1 undergoes multiple post-translational modifications that interact in complex ways with S118 phosphorylation:

  • Interconnected phosphorylation events:

    • S118 phosphorylation occurs within a cluster of phosphorylation sites in the AF-1 domain

    • Other phosphorylation sites (S104, S106, S167) may work cooperatively or antagonistically with S118

  • Relationship with ubiquitination:

    • ESR1 is subject to ubiquitination regulated by multiple E3 ligases including LATS1 via DCAF1

    • Ubiquitination leads to ESR1 proteasomal degradation

    • Deubiquitination by OTUB1 counteracts this process

    • UBR5 specifically recognizes and binds ligand-bound ESR1 for ubiquitination

    • The relationship between S118 phosphorylation and ubiquitination rate remains an active area of research

  • Glycosylation interactions:

    • ESR1 contains O-linked N-acetylglucosamine modifications

    • Potential crosstalk between glycosylation and phosphorylation at nearby sites may create complex regulatory patterns

  • Functional consequences of PTM interplay:

    • Different combinations of PTMs likely create distinct "codes" that direct ESR1 to specific gene targets

    • PTM patterns may influence protein-protein interaction profiles and subcellular localization

Understanding this complex PTM network is essential for interpreting the specific roles of S118 phosphorylation in different cellular contexts and may inform more precise therapeutic targeting strategies.

What are common issues when using Phospho-ESR1 (S118) antibodies and how can they be resolved?

Researchers frequently encounter several challenges when working with phospho-specific antibodies targeting ESR1 at S118:

  • Low or no signal issues:

    • Potential causes: Rapid dephosphorylation during sample preparation, insufficient phosphorylation induction, antibody degradation

    • Solutions: Include phosphatase inhibitors in all buffers, confirm stimulation conditions (e.g., estradiol treatment timing), verify antibody storage conditions

  • High background or non-specific binding:

    • Potential causes: Insufficient blocking, cross-reactivity with similar phospho-epitopes, too high antibody concentration

    • Solutions: Optimize blocking conditions, perform phospho-peptide competition as control , adjust antibody dilution (try 1:500-1:2000 range for WB)

  • Inconsistent results between experiments:

    • Potential causes: Variable phosphorylation levels, batch-to-batch antibody variation, inconsistent sample preparation

    • Solutions: Standardize cell treatment protocols, consider using recombinant monoclonal antibodies for greater consistency , implement rigorous positive controls

  • Discrepancies between detection methods:

    • Potential causes: Different antibody performance in various applications, epitope accessibility differences

    • Solutions: Validate antibody specifically for each application, optimize protocols for individual techniques

  • Tissue-specific detection challenges:

    • Potential causes: Epitope masking in certain fixation conditions, tissue-specific phosphatase activity

    • Solutions: Test different antigen retrieval methods for IHC, adjust fixation protocols, increase phosphatase inhibitor concentration

Creating a systematic troubleshooting workflow and maintaining detailed records of optimization attempts will help researchers overcome these common challenges.

How can I optimize immunohistochemistry protocols for Phospho-ESR1 (S118) detection in tissue samples?

Optimizing IHC protocols for phospho-specific antibodies requires careful attention to preserving phosphorylation status:

  • Tissue preparation considerations:

    • Fix tissues promptly after collection to prevent phosphatase activity

    • Consider using phosphatase inhibitor-containing fixatives

    • Standardize fixation times (typically 24-48 hours in 10% neutral buffered formalin)

  • Antigen retrieval optimization:

    • Test both heat-induced epitope retrieval (HIER) methods:

      • Citrate buffer (pH 6.0)

      • EDTA buffer (pH 9.0)

    • Optimize retrieval time (usually 10-20 minutes)

  • Blocking and antibody conditions:

    • Use 1:100-1:300 dilution range for primary antibody incubation

    • Extend primary antibody incubation to overnight at 4°C

    • Include phospho-blocking peptide controls in parallel sections

  • Signal amplification considerations:

    • Consider using polymer-based detection systems for enhanced sensitivity

    • Test tyramide signal amplification for tissues with low expression levels

  • Validation approaches:

    • Compare staining with and without phospho-blocking peptide

    • Include known positive controls (e.g., breast cancer tissues with confirmed S118 phosphorylation)

    • Compare adjacent sections stained with total ERα antibody

  • Expected staining patterns:

    • Primary nuclear localization

    • Intensity may vary between tumor regions

    • Heterogeneous expression within tissue is common

The optimal protocol will need to be determined empirically for each tissue type and fixation method, with careful documentation of all optimization steps.

How is Phospho-ESR1 (S118) being studied in relation to endocrine therapy resistance?

Phosphorylation of ESR1 at S118 has emerged as a potential biomarker and mechanism for endocrine therapy resistance:

  • Clinical correlations:

    • Increased S118 phosphorylation has been observed in some tamoxifen-resistant tumors

    • The ratio of phospho-S118 to total ESR1 may correlate with clinical outcomes

    • Growth factor receptor overexpression (e.g., HER2) can drive S118 phosphorylation independent of estrogen

  • Mechanistic insights:

    • Phosphorylation at S118 may allow ESR1 to function in a ligand-independent manner

    • S118 phosphorylation can influence the recruitment of different coregulator complexes , potentially altering the response to selective estrogen receptor modulators (SERMs)

    • Cross-talk between growth factor signaling and ESR1 phosphorylation may bypass the need for estrogen binding

  • Therapeutic implications:

    • Combining endocrine therapies with kinase inhibitors that reduce S118 phosphorylation

    • Development of biomarker assays to predict therapy response based on phosphorylation status

    • Potential for novel therapeutic agents that specifically target phosphorylated ESR1

  • Current research approaches:

    • Phosphoproteomics to map comprehensive ESR1 modification patterns in resistant versus sensitive tumors

    • Development of patient-derived xenograft models with varying S118 phosphorylation levels

    • Structure-based drug design targeting the conformation of phosphorylated ESR1

This rapidly evolving research area may lead to more personalized approaches to endocrine therapy based on ESR1 phosphorylation status.

How do genome-wide binding patterns differ between total ESR1 and Phospho-ESR1 (S118)?

Chromatin immunoprecipitation followed by sequencing (ChIP-seq) studies have revealed important differences in the genomic binding patterns of total versus phosphorylated ESR1:

  • Binding site selectivity:

    • Phospho-ESR1 (S118) appears to bind a subset of total ESR1 binding sites

    • Certain genomic regions show preferential enrichment for phospho-ESR1

    • Phosphorylation status may influence binding to non-canonical estrogen response elements

  • Coregulator recruitment patterns:

    • Phospho-ESR1 (S118) directs the recruitment of specific transcriptional coregulators including SRC-1, TIF-2, and AIB to target gene promoters

    • The coregulator complexes associated with phospho-ESR1 may differ from those bound to non-phosphorylated receptor

  • Chromatin environment associations:

    • Phospho-ESR1 binding may correlate with specific histone modifications

    • Pioneer factor requirements may differ between phospho and non-phospho ESR1

    • Chromatin accessibility patterns may influence phospho-ESR1 binding preference

  • Temporal binding dynamics:

    • Phospho-ESR1 may show distinct kinetics of recruitment to target genes

    • Early versus late estrogen-responsive genes may show differential dependence on S118 phosphorylation

    • Stimulus-specific binding patterns may emerge depending on the kinase pathway activated

  • Functional output differences:

    • Gene sets regulated by phospho-ESR1 may represent specific biological pathways

    • Phosphorylation-dependent binding correlates with specific transcriptional outcomes

    • Integration of multiple signaling inputs may occur at phospho-ESR1 binding sites

These genome-wide differences provide insight into how post-translational modifications create functional diversity in transcription factor activity and may explain context-specific effects of ESR1 signaling.

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.