Phospho-JUN (S63) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Western Blot (WB)

  • Detection Sensitivity:

    • Abcam ab32385: 1:1,000 dilution in HeLa lysates, showing specificity for S63-phosphorylated c-Jun after anisomycin treatment .

    • Bio-Techne MAB8930: Detects a ~45 kDa band in UV-treated HEK293T cells at 0.5 µg/mL .

  • Specificity Controls:

    • Loss of signal upon phosphatase treatment or use of non-phosphorylated peptide competitors .

Immunocytochemistry (ICC)/Immunofluorescence (IF)

  • Localization: Nuclear staining in HeLa cells treated with anisomycin (1 µg/mL) .

  • Protocols: Fixed cells incubated with primary antibody (1–25 µg/mL) and visualized using fluorescent secondary antibodies .

Immunohistochemistry (IHC-P)

  • Use Case: Staining of paraffin-embedded human breast carcinoma tissue at 1:50 dilution .

Simple Western

  • Bio-Techne MAB8930: Detects ~49 kDa band in UV-treated HEK293T cells at 5 µg/mL .

Biological Significance of c-Jun S63 Phosphorylation

  • Transcriptional Activation: Phosphorylation at S63 (and S73) enhances c-Jun’s ability to activate target genes like FASLG and USP28, implicated in apoptosis and colorectal cancer .

  • Disease Relevance:

    • Prolonged S63 phosphorylation observed in neurons after cerebral ischemia or nerve injury, correlating with apoptosis markers (TUNEL, Fas-ligand) .

    • Linked to KRAS-mediated oncogenic signaling in colorectal cancer .

Technical Considerations

  • Storage: Lyophilized antibodies stable at -20°C to -70°C; reconstituted solutions stored at 2–8°C for short-term use .

  • Buffer Compatibility: Performance varies by formulation (e.g., PBS/BSA vs. NFDM/TBST) .

  • Cross-Reactivity: Most antibodies show strong reactivity with human samples; some cross-react with mouse/rat (e.g., CABP1190) .

Research Findings Highlighting Utility

  • Cancer Studies:

    • Detects KRAS-mediated USP28 activation in colorectal cancer cells .

    • Used to study AP-1’s role in transcriptional regulation of oncogenes .

  • Neuroscience:

    • Persistent S63 phosphorylation observed in post-ischemic neurons, linking c-Jun to apoptosis pathways .

Limitations and Troubleshooting

  • Band Discrepancies: Observed MW (42–49 kDa) often exceeds predicted MW (35–36 kDa) due to phosphorylation or dimerization .

  • Species Specificity: Some antibodies (e.g., MAB8930) are validated only for human samples .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

CUSABIO has engineered vector clones for the expression of a recombinant JUN antibody in mammalian cells. These clones were generated by inserting the JUN antibody heavy and light chains into appropriate plasma vectors. The recombinant JUN antibody was subsequently purified from the culture medium using affinity-chromatography. This antibody is designed to detect JUN protein from human samples in various applications such as ELISA, Western Blot, Immunohistochemistry, and Immunofluorescence.

The phospho-JUN (S63) antibody is specifically designed to detect the endogenous levels of JUN phosphorylated at the Ser63 residue. Phosphorylation of Ser63 within the NH2-terminal transactivation domain of JUN is crucial for its transcriptional activity. Notably, phosphorylation of JUN has also been linked to the mediation of apoptosis when survival signals are interrupted. JUN plays a pivotal role in diverse cellular processes, encompassing proliferation, apoptosis, survival, tumorigenesis, and tissue morphogenesis.

Form
Liquid
Lead Time
Generally, we are able to dispatch products within 1-3 working days after receiving your order. The delivery timeframe may vary depending on the chosen purchasing method and location. For specific delivery details, please consult your local distributors.
Synonyms
Activator protein 1 antibody; AP 1 antibody; AP-1 antibody; AP1 antibody; cJun antibody; Enhancer Binding Protein AP1 antibody; Jun Activation Domain Binding Protein antibody; JUN antibody; Jun oncogene antibody; JUN protein antibody; Jun proto oncogene antibody; JUN_HUMAN antibody; JUNC antibody; Oncogene JUN antibody; p39 antibody; Proto oncogene c jun antibody; Proto oncogene cJun antibody; Proto-oncogene c-jun antibody; Transcription Factor AP 1 antibody; Transcription factor AP-1 antibody; Transcription Factor AP1 antibody; V jun avian sarcoma virus 17 oncogene homolog antibody; V jun sarcoma virus 17 oncogene homolog (avian) antibody; V jun sarcoma virus 17 oncogene homolog antibody; V-jun avian sarcoma virus 17 oncogene homolog antibody; vJun Avian Sarcoma Virus 17 Oncogene Homolog antibody
Target Names
JUN
Uniprot No.

Target Background

Function
JUN is a transcription factor that recognizes and binds to the enhancer heptamer motif 5'-TGA[CG]TCA-3'. When phosphorylated by HIPK3, it enhances the activity of NR5A1, leading to increased steroidogenic gene expression upon stimulation of the cAMP signaling pathway. JUN is also involved in activated KRAS-mediated transcriptional activation of USP28 in colorectal cancer (CRC) cells. It binds to the USP28 promoter in CRC cells.
Gene References Into Functions
  1. Data suggests that miR-139-5p is downregulated in the hearts of Hypertrophic cardiomyopathy patients and that it inhibits cardiac hypertrophy by targeting c-Jun expression. PMID: 29440459
  2. This study identified a critical Jun/miR-22/HuR regulatory axis in CRC (illustrated in Fig. 8). The findings highlight the crucial role of HuR and miR-22 in CRC proliferation and migration. PMID: 29351796
  3. A novel cascade mediated by AP-1 and FOXF1 that regulates oncogene-induced senescence is reported. PMID: 30119690
  4. Multivalent Interactions with Fbw7 and Pin1 Facilitate Recognition of c-Jun by the Fbw7. PMID: 29225075
  5. High AP-1 expression is associated with metastasis in colon cancer. PMID: 29305742
  6. Our findings suggest that extended AP-1 binding sites, along with adjacent binding sites for additional transcription factors, encode part of the information that governs transcription factor binding site activity in the genome. PMID: 29305491
  7. The expression of WIF-1 was low in GBC cells due to aberrant hypermethylation of its promoter region. An alternative pathogenesis of GBC was indicated where c-Jun causes hypermethylation of the WIF-1 promoter region, repressing WIF-1 expression through transcriptional regulation and interaction with DNMT1 as an early event in the tumorigenesis of GBC. PMID: 29693707
  8. Mutant cellular AP-1 proteins promote expression of a subset of Epstein-Barr virus late genes in the absence of lytic viral DNA replication. PMID: 30021895
  9. Secreted Ta9 not only has the ability to stimulate CD8+ T cells but also the potential to activate AP-1-driven transcription and contribute to T. annulata-induced leukocyte transformation PMID: 29738531
  10. MiR-216b directly targets c-Jun, thereby reducing AP-1-dependent transcription and sensitizing cells to ER stress-dependent apoptosis. PMID: 27173017
  11. Results suggest that c-Jun, p38 MAPK, PIK3CA/Akt, and GSK3 signaling are involved in the effect of miR-203 on the proliferation of hepatocellular carcinoma cells. PMID: 28887744
  12. These findings suggest that increased JUN expression and activity may contribute to gefitinib resistance in non-small cell lung cancer. PMID: 28566434
  13. The results indicated that butein has antiproliferative and proapoptotic properties through the suppression of NF-kappaB, AP-1 and Akt signaling in HTLV-1-infected T cells, both in vitro and in vivo, suggesting its therapeutic potential against HTLV-1-associated diseases including adult T-cell leukemia/lymphoma PMID: 28586006
  14. Results show that VEGFA induces c-jun expression in mediating human retinal microvascular endothelial cell migration, sprouting and tube formation, and that Pyk2-STAT3 signaling enhances cJun expression in the mediation of retinal neovascularization. PMID: 27210483
  15. Increased c-jun expression is associated with nasopharyngeal carcinoma. PMID: 28269757
  16. Thrombin binding to PAR-1 receptor activated Gi-protein/c-Src/Pyk2/EGFR/PI3K/Akt/p42/p44 MAPK cascade, which in turn elicited AP-1 activation and ultimately evoked MMP-9 expression and cell migration in SK-N-SH cells. PMID: 27181591
  17. Findings provide evidence that phospho-c-Jun activates an important regulatory mechanism to control DNMT1 expression and regulate global DNA methylation in glioblastoma. PMID: 28036297
  18. Results demonstrated for the first time the regulatory mechanism of miR-744 transcription by c-Jun, providing a potential mechanism underlying the upregulation of miR-744 in cancers PMID: 27533465
  19. Results provide evidence that NuRD represses c-Jun transcription directly which, in the absence of MBD3, activates endogenous pluripotent genes and regulates induced cancer stem cells-related genes. PMID: 27894081
  20. Taken together, these results indicated that PAR1 signalingmediated cJun activation promotes early apoptosis of HUVEC cells induced by heat stress. PMID: 28447716
  21. Cheliensisin A (Chel A)treatment led to PH domain and Leucine rich repeat Protein Phosphatases (PHLPP2) protein degradation and subsequently increased in c-Jun phosphorylation, which could be attenuated by inhibition of autophagy mediated by Beclin 1. PMID: 27556506
  22. The positive feedback regulation of OCT4 and c-JUN, resulting in the continuous expression of oncogenes such as c-JUN, seems to play a critical role in the determination of the cell fate decision from induced pluripotent stem cells to cancer stem cells in liver cancer. PMID: 27341307
  23. miR-26b plays an anti-metastatic role and is downregulated in gastric cancer tissues via the KPNA2/c-jun pathway PMID: 27078844
  24. The IL1B/AP-1/miR-30a/ADAMTS-5 axis regulates cartilage matrix degradation in osteoarthritis. PMID: 27067395
  25. TGM2 is involved in amyloid-beta (1-42)-induced pro-inflammatory activation via AP1/JNK signaling pathways in cultured monocytes. PMID: 27864692
  26. Integrative genomic analysis indicated overexpression of the AP-1 transcriptional complex suggesting experimental therapeutic rationales, including blockade of the renin-angiotensin system. This led to the repurposing of the angiotensin II receptor antagonist, irbesartan, as an anticancer therapy, resulting in the patient experiencing a dramatic and durable response. PMID: 27022066
  27. Knockdown of CD44 reduced the protein level of xCT, a cystine transporter, and increased oxidative stress. However, an increase in GSH was also observed and was associated with enhanced chemoresistance in CD44-knockdown cells. Increased GSH was mediated by the Nrf2/AP-1-induced upregulation of GCLC, a subunit of the enzyme catalyzing GSH synthesis PMID: 28185919
  28. Study highlights the role of AP1 in promoting the host gene expression profile that defines Ebola virus pathogenesis. PMID: 28931675
  29. This is the first study to show how TGF-beta regulates the expression of Claudin-4 through c-Jun signaling and how this pathway contributes to the migratory and tumorigenic phenotype of lung tumor cells. PMID: 27424491
  30. Data show that BRD4 controls RUNX2 by binding to the enhancers (ENHs) and each RUNX2 ENH is potentially controlled by a distinct set of TFs and c-JUN as the principal pivot of this regulatory platform. PMID: 28981843
  31. AP-1 likely plays a more important role in the AR cistrome in fibroblasts. PMID: 27634452
  32. Elevated levels of bile acid increase the tumorigenic potential of pancreatic cancer cells by inducing FXR/FAK/c-Jun axis to upregulate MUC4 expression. PMID: 27185392
  33. Immunohistochemistry was employed to analyze cFos, cJun and CD147 expression in 41 UCB cases and 34 noncancerous human bladder tissues. PMID: 28358415
  34. Taken together, these findings indicate that LT reduces c-Jun protein levels via two distinct mechanisms, thereby inhibiting critical cell functions, including cellular proliferation. PMID: 28893904
  35. Expression of either dominant-negative or constitutively active mutants of Nrf2, ATF4, or c-Jun confirmed that distinct transcription units are regulated by these transcription factors. PMID: 27278863
  36. Mutually exclusive transcriptional regulation by AP-1 (cjun/cfos) and non-canonical NF-kappaB (RelB/p52) downstream of MEK-ERK and NIK-IKK-alpha-NF-kappaB2 (p100) phosphorylation, respectively, was responsible for persistent Ccl20 expression in the colonic cells. PMID: 27590109
  37. Glucocorticoid receptor (GR) is recruited to activator protein-1 (AP-1) target genes in a DNA-binding-dependent manner. PMID: 28591827
  38. These results suggested that hyperphosphatemia in the patients with CKD suppresses bone resorption by inhibiting osteoclastogenesis, and this impairs the regulation of bone metabolism. PMID: 28939042
  39. These results suggest that Bacteroides fragilis enterotoxin induced accumulation of autophagosomes in endothelial cells, but activation of a signaling pathway involving JNK, AP-1, and CHOP may interfere with complete autophagy. PMID: 28694294
  40. Overall, our results suggest that miR-4632 plays an important role in regulating HPASMC proliferation and apoptosis by suppression of cJUN, providing a novel therapeutic miRNA candidate for the treatment of pulmonary vascular remodeling diseases. It also implies that serum miR-4632 has the potential to serve as a circulating biomarker for PAH diagnosis. PMID: 28701355
  41. Findings suggest that AP-1 factors are regulators of RNA polymerase III (Pol III)-driven 5S rRNA and U6 snRNA expression with a potential role in cell proliferation. PMID: 28488757
  42. Our results indicate that assessing AP1 and PEA3 transcription factor status might be a good indicator of OAC status. However, we could not detect any associations with disease stage or patient treatment regime. This suggests that the PEA3-AP1 regulatory module more likely contributes more generally to the cancer phenotype. In keeping with this observation, depletion of ETV1 and/or ETV4 causes an OAC cell growth defect PMID: 28859074
  43. shRNA-mediated inhibition of JUN decreases AML cell survival and propagation in vivo. These data uncover a previously unrecognized role of JUN as a regulator of the unfolded protein response PMID: 27840425
  44. These findings demonstrate an essential role for the ERK pathway together with c-JUN and c-FOS in the differentiation activity of LukS-PV. PMID: 27102414
  45. The present study defines the minimal TIM-3 promoter region and demonstrates its interaction with c-Jun during TIM-3 transcription in CD4(+) T cells. PMID: 27243212
  46. Taken together, our data demonstrate that JNK regulates triple-negative breast cancer (TNBC)tumorigenesis by promoting CSC phenotype through Notch1 signaling via activation of c-Jun and indicate that JNK/c-Jun/Notch1 signaling is a potential therapeutic target for TNBC PMID: 27941886
  47. Regulation of osteosarcoma cell lung metastasis by the c-Fos/AP-1 target FGFR1 PMID: 26387545
  48. c-jun promoted FOXK1-mediated proliferation and metastasis via orthotopic implantation. PMID: 27882939
  49. Data provide evidence that AP-1 is a key determinant of endocrine resistance of breast cancer cells by mediating a global shift in the estrogen receptor transcriptional program. PMID: 26965145
  50. Comparison of how AP-1 (Jun/Jun dimer) and Epstein-Barr virus Zta recognize methyl groups within their cognate response elements PMID: 28158710

Show More

Hide All

Database Links

HGNC: 6204

OMIM: 165160

KEGG: hsa:3725

STRING: 9606.ENSP00000360266

UniGene: Hs.696684

Protein Families
BZIP family, Jun subfamily
Subcellular Location
Nucleus.
Tissue Specificity
Expressed in the developing and adult prostate and prostate cancer cells.

Q&A

What is the biological significance of c-JUN phosphorylation at Ser63?

Phosphorylation of c-JUN at Ser63 represents a critical post-translational modification that directly regulates its transcriptional activity. This phosphorylation occurs in the NH2-terminal transactivation domain and is essential for c-JUN's function as a major component of the Activator Protein 1 (AP-1) complex. When phosphorylated at Ser63, c-JUN can effectively dimerize and bind to target DNA sequences, thereby influencing gene transcription. This phosphorylation event plays a crucial regulatory role in multiple cellular processes including proliferation, differentiation, malignant transformation, and apoptosis . The phosphorylation state of c-JUN at Ser63 serves as an important biomarker for JNK pathway activation and cellular stress response mechanisms.

How does c-JUN Ser63 phosphorylation differ kinetically from other c-JUN phosphorylation sites?

c-JUN phosphorylation by JNK exhibits a defined temporal order with distinctive kinetics. Research demonstrates that Ser63 phosphorylation occurs most rapidly, followed by Ser73, with threonine sites (T91 and T93) being phosphorylated significantly slower. In vitro and in vivo studies reveal the following phosphorylation order: S63 > S73 > T91 ≈ T93 . This temporal pattern has been confirmed through time-resolved NMR studies and immunoblotting with phosphorylation-specific antibodies.

Under physiological conditions with anisomycin stimulation, S63 and S73 phosphorylation occurs with half-times of approximately 12 minutes, while T91 and T93 phosphorylation proceeds more slowly with half-times of 15-17 minutes . These distinct kinetics have important implications for the temporal regulation of c-JUN activity and downstream signaling events.

What is the molecular structure of c-JUN and how does phosphorylation affect it?

c-JUN (36 kDa, 331 amino acids) belongs to the bZIP family and Jun subfamily of transcription factors. The transactivation domain (TAD) containing the phosphorylation sites is intrinsically disordered, as evidenced by 2D 1H, 15N correlation spectroscopy showing narrow chemical shift dispersion .

The protein contains four major phosphorylation sites (S63, S73, T91, T93) and a MAPK binding motif (D-motif, residues 32-50) that controls phosphorylation of these sites. Upon phosphorylation, particularly at Ser63 and Ser73, c-JUN undergoes conformational changes that facilitate dimerization and DNA binding, activating its transcriptional functions. Secondary chemical shift analyses show no significant populations of secondary structure in the unphosphorylated state, consistent with the protein's intrinsically disordered nature .

What are the optimal methods for detecting phosphorylated c-JUN at Ser63 in cell-based assays?

Several methodologies can be employed for detecting phosphorylated c-JUN at Ser63, each with distinct advantages:

  • HTRF (Homogeneous Time-Resolved Fluorescence) Assay:

    • Offers a streamlined mix-and-read no-wash protocol

    • Demonstrated 8-fold higher sensitivity compared to Western blot

    • Can be performed in single-plate or two-plate formats

    • Requires only 16 μL sample volume

    • Enables quantitative detection directly in cell lysates without electrophoresis or transfer

  • Western Blot:

    • Recommended antibody dilutions: 1:500-1:5000

    • Allows visualization of protein size (observed at 40-48 kDa)

    • Useful for comparing relative phosphorylation levels

  • Immunohistochemistry (IHC):

    • Recommended antibody dilutions: 1:50-1:200

    • Enables in situ detection in tissue sections

    • Valuable for spatial distribution analysis

  • Immunofluorescence (IF):

    • Recommended antibody dilutions: 1:20-1:200

    • Allows subcellular localization studies

    • Can be combined with other markers for co-localization analysis

For high-throughput screening applications, the HTRF assay provides significant advantages in terms of sensitivity, throughput, and reduced sample requirements. The choice of method should be determined by the specific research question, required sensitivity, and available instrumentation.

What is the recommended protocol for stimulating c-JUN phosphorylation at Ser63 in cell culture experiments?

Based on established research protocols, the following methodology is recommended for stimulating c-JUN phosphorylation at Ser63 in cell culture:

  • Cell Selection and Culture:

    • HEK293, NIH-3T3, or other relevant cell lines can be used

    • Culture cells to 70-80% confluence (approximately 50,000 cells/well in a 96-well format)

  • Stimulation Protocol:

    • Anisomycin treatment (500nM) is highly effective for inducing c-JUN phosphorylation

    • Optimal stimulation time is 30 minutes at 37°C

    • JNK activation typically begins after a 5-minute lag period and reaches maximum activity within 12 minutes

  • Cell Lysis and Detection:

    • After stimulation, remove culture medium and add appropriate lysis buffer

    • Incubate for 30 minutes to ensure complete lysis

    • Collect supernatants following centrifugation

    • Proceed with detection using HTRF, Western blot, or other methods

This protocol has been validated to produce robust and reproducible c-JUN phosphorylation that can be readily detected using phospho-specific antibodies. The stimulation conditions can be adjusted based on the specific cell type and experimental requirements.

How can I validate the specificity of phospho-JUN (S63) antibody in my experimental system?

Validation of phospho-JUN (S63) antibody specificity is crucial for ensuring reliable research outcomes. A comprehensive validation approach includes:

  • Alanine Substitution Mutants:

    • Generate S63A mutants as negative controls

    • Compare antibody reactivity between wild-type and mutant samples

    • This approach has been successfully used to verify the site specificity of phospho-antibodies

  • Peptide Competition Assay:

    • Pre-incubate antibody with synthetic phosphopeptides containing the S63 phosphorylation site

    • Test whether this competition blocks antibody binding in your assay

    • A cell-permeable c-Jun peptide has been used successfully as a competitive inhibitor

  • Phosphatase Treatment Control:

    • Treat positive samples with lambda phosphatase

    • Confirm loss of antibody reactivity after phosphate group removal

  • Stimulation/Inhibition Controls:

    • Compare samples from unstimulated cells, anisomycin-stimulated cells, and cells pre-treated with JNK inhibitors

    • A dose-dependent response should be observed with anisomycin stimulation

    • NIH-3T3 cells stimulated with increasing concentrations of anisomycin show proportional increases in S63 phosphorylation

  • Antibody Dilution Series:

    • Perform serial dilutions to establish the linear range of detection

    • Ensure your experimental conditions fall within this range

These validation steps collectively provide strong evidence for antibody specificity and help identify potential cross-reactivity issues or artifacts in your experimental system.

How does the spatial proximity of the D-motif affect c-JUN phosphorylation kinetics at different sites?

Research using engineered c-JUN TAD constructs has revealed complex relationships between D-motif positioning and phosphorylation kinetics. The D-motif (residues 32-50) serves as the MAPK binding site that controls phosphorylation of S63, S73, T91, and T93 sites.

In wild-type c-JUN, the D-motif is positioned N-terminal to the phosphorylation sites, with S63 and S73 being closer to the D-motif than T91 and T93. Experiments with transposed D-motif variants (mDock and cDock) revealed that:

  • Moving the D-motif closer to T91/T93 (mDock variant) did not accelerate their phosphorylation rates

  • Sites positioned N-terminal to the transposed D-motif showed strongly suppressed phosphorylation

  • The normally fast S63 site exhibited very slow modification in both mDock and cDock variants

These findings indicate that while spatial proximity to the D-motif contributes to phosphorylation efficiency, it is not the sole determinant. The three-dimensional structural interaction between JNK1 and c-JUN, as well as the primary amino acid sequence surrounding the phosphorylation sites, play crucial roles in determining site-specific phosphorylation kinetics.

What is the relationship between c-JUN Ser63 phosphorylation and its role in cellular pathways beyond transcriptional activation?

Beyond its well-established role in transcriptional activation, phosphorylation of c-JUN at Ser63 mediates several additional cellular functions:

  • Apoptosis Regulation:

    • c-JUN phosphorylation has been linked to apoptosis mediation when survival signals are interrupted

    • The balance between pro-survival and pro-apoptotic functions appears to be context-dependent

  • Protein Stability and Turnover:

    • Phosphorylation at Ser63 can affect c-JUN protein stability and half-life

    • This post-translational modification may protect c-JUN from ubiquitin-mediated degradation

  • Protein-Protein Interactions:

    • Phosphorylated c-JUN can recruit specific co-factors that modulate its activity

    • Phosphorylation may alter interaction patterns with other transcription factors and regulatory proteins

  • Cellular Stress Response:

    • Ser63 phosphorylation serves as a marker for cellular stress response

    • Different stressors (UV radiation, inflammatory cytokines, oxidative stress) can trigger this phosphorylation through JNK activation

  • Tissue Morphogenesis:

    • c-JUN phosphorylation contributes to developmental processes and tissue organization

    • This function appears to be regulated by both JNK-dependent and JNK-independent mechanisms

These diverse functions highlight the central role of c-JUN Ser63 phosphorylation as a regulatory node connecting multiple cellular signaling networks, extending well beyond simple transcriptional control.

How do the kinetics of c-JUN phosphorylation at Ser63 compare between different JNK isoforms?

The kinetics of c-JUN phosphorylation at Ser63 show subtle but important differences between JNK isoforms. Comparative analyses using recombinant active JNK1 and JNK2 with c-JUN TAD have revealed:

These kinetic differences may have significant implications for understanding the specific roles of different JNK isoforms in various physiological and pathological contexts, providing potential targets for isoform-selective therapeutic interventions.

What are the common technical challenges when detecting phospho-JUN (S63) and how can they be addressed?

Researchers frequently encounter several technical challenges when working with phospho-JUN (S63) detection. Here are the most common issues and recommended solutions:

  • Low Signal Intensity:

    • Cause: Insufficient stimulation, rapid dephosphorylation, or low antibody sensitivity

    • Solution: Optimize stimulation conditions (500nM anisomycin for 30 minutes works well); include phosphatase inhibitors in lysis buffer; consider using HTRF assay which offers 8-fold higher sensitivity than Western blot

  • High Background:

    • Cause: Non-specific antibody binding or insufficient blocking

    • Solution: Optimize antibody dilution (1:500-1:5000 for WB); increase blocking time/concentration; include validated negative controls (S63A mutants)

  • Inconsistent Phosphorylation:

    • Cause: Cell density variations, heterogeneous stimulation, or unstable stimulants

    • Solution: Standardize cell seeding density (50,000 cells/well recommended); ensure uniform reagent distribution; prepare fresh stimulants before each experiment

  • Rapid Dephosphorylation:

    • Cause: Phosphatase activity during sample preparation

    • Solution: Work quickly on ice; use comprehensive phosphatase inhibitor cocktails; validate inhibitor effectiveness in your system

  • Cross-Reactivity with Other Phosphorylation Sites:

    • Cause: Antibody recognizing similar epitopes around other phosphorylation sites

    • Solution: Validate antibody specificity using alanine mutants for each phosphorylation site; consider using specific blocking peptides

  • Antibody Detection Outside Linear Range:

    • Cause: Over-saturated signal or insufficient protein loading

    • Solution: Perform back-dilution series of phosphorylated samples to ensure antibody signals remain within the linear detection range

Implementing these solutions can significantly improve the reliability and reproducibility of phospho-JUN (S63) detection across different experimental platforms.

How can I quantitatively compare phospho-JUN (S63) levels across different experimental conditions?

For rigorous quantitative comparison of phospho-JUN (S63) levels across experimental conditions, consider the following methodological approaches:

  • HTRF Quantitative Assay:

    • Provides direct quantitative measurement of phosphorylated c-JUN

    • Signal intensity is directly proportional to phosphorylated protein concentration

    • Allows for high-throughput, plate-based analysis

    • Standard curves can be generated using recombinant phosphorylated standards

  • Normalization Strategies:

    • Total c-JUN Normalization: Express phospho-JUN (S63) signal relative to total c-JUN levels

    • Housekeeping Protein Normalization: Normalize to stable reference proteins (β-actin, GAPDH)

    • Loading Control Normalization: Use total protein stains (Ponceau S, REVERT)

  • Dose-Response Analysis:

    • Generate dose-response curves using varying concentrations of stimulus

    • NIH-3T3 cells stimulated with increasing anisomycin concentrations show proportional increases in S63 phosphorylation

    • Calculate EC50 values for different stimuli or inhibitors

  • Time-Course Analysis:

    • Measure phosphorylation at multiple time points (0-30 minutes recommended)

    • Calculate rates and half-times of phosphorylation

    • Allows comparison of phosphorylation kinetics under different conditions

    • In vivo studies show half-times of approximately 12 minutes for S63 phosphorylation following anisomycin treatment

  • Quantitative Image Analysis:

    • For IHC/IF applications, use software-based quantification

    • Include internal controls in each experiment

    • Consider automated high-content imaging for large-scale analysis

Each of these approaches offers distinct advantages depending on your experimental system and research questions. Combining multiple quantitative methods provides the most robust comparative analysis across experimental conditions.

What considerations are important when interpreting phospho-JUN (S63) data in the context of complex signaling networks?

Interpreting phospho-JUN (S63) data requires careful consideration of the broader signaling context. Key factors to consider include:

  • Temporal Dynamics:

    • c-JUN phosphorylation follows specific temporal patterns

    • S63 is phosphorylated rapidly (half-time ~12 min) following JNK activation

    • Transient versus sustained phosphorylation may lead to different functional outcomes

    • Consider collecting data at multiple time points to capture the complete dynamics

  • Multiple Phosphorylation Sites:

    • c-JUN contains multiple phosphorylation sites (S63, S73, T91, T93)

    • These sites are phosphorylated with different kinetics (S63 > S73 > T91 ≈ T93)

    • The combinatorial pattern of phosphorylation may determine specific functional outcomes

    • Consider analyzing multiple phosphorylation sites simultaneously

  • Pathway Crosstalk:

    • JNK is not the only kinase that can influence c-JUN activity

    • Plk3 can phosphorylate c-JUN independently of JNK

    • UV irradiation-induced phosphorylation of c-JUN and apoptosis were not significantly affected by JNK knockdown in some systems

    • Consider analyzing multiple pathway components to determine the specific contribution of each

  • Cell Type-Specific Regulation:

    • Different cell types may exhibit distinct patterns of c-JUN phosphorylation

    • Expression levels of JNK isoforms, phosphatases, and regulatory proteins vary across cell types

    • Validate findings across multiple relevant cell systems

  • Functional Correlation:

    • Phosphorylation at S63 is necessary but not always sufficient for transcriptional activation

    • Consider complementing phosphorylation data with functional readouts (AP-1 reporter assays, target gene expression)

    • Establish causality through inhibitor studies or mutation of phosphorylation sites

By integrating these considerations into your data analysis, you can develop a more comprehensive understanding of how phospho-JUN (S63) signaling contributes to specific cellular responses within complex regulatory networks.

How does the sensitivity and specificity of HTRF technology compare with traditional Western blot for phospho-JUN (S63) detection?

Comparative analysis between HTRF and Western blot technologies for phospho-JUN (S63) detection reveals significant differences in performance characteristics:

ParameterHTRF TechnologyWestern BlotPractical Implications
Sensitivity8-fold higher sensitivity demonstrated in direct comparisonLower sensitivity requires more sampleHTRF can detect lower levels of phosphorylation and requires less cellular material
ThroughputHigh (500+ assay points per kit)Low to moderateHTRF enables screening applications and larger experimental designs
Protocol ComplexitySimple mix-and-read, no-wash protocolComplex multi-step protocol with transfersHTRF reduces technical variability and hands-on time
Sample Volume16 μLTypically 20-40 μLHTRF allows for miniaturization and conservation of precious samples
QuantificationDirect quantitative readoutSemi-quantitative requiring densitometryHTRF provides more reliable quantitative comparisons
Molecular Weight InformationNo size discriminationProvides information on protein sizeWestern blot can detect unexpected bands or degradation products
SpecificityHigh (requires two antibodies in proximity)Variable (dependent on single antibody)HTRF may reduce false positives from cross-reactive species

Experimental validation with HEK293 cells stimulated with 500nM anisomycin for 30 min demonstrated that the HTRF assay could detect phospho-JUN (S63) at significantly lower concentrations than Western blot, making it particularly valuable for experiments with limited sample availability or when detecting subtle changes in phosphorylation levels .

What are the advanced experimental approaches for studying the relationship between c-JUN phosphorylation kinetics and transcriptional outcomes?

Advanced experimental strategies can elucidate the complex relationship between c-JUN phosphorylation kinetics and downstream transcriptional regulation:

  • Engineered c-JUN Variants:

    • Create "swap" variants where sequences containing fast phosphorylation sites (S63/S73) are exchanged with sequences containing slower sites (T91/T93)

    • Generate phosphomimetic mutants (S63D, S73D) to simulate constitutive phosphorylation

    • Develop phospho-deficient mutants (S63A, S73A) as negative controls

    • These engineered variants help dissect the specific contribution of each phosphorylation site to transcriptional activity

  • Time-Resolved Transcriptomics:

    • Combine time-course phosphorylation analysis with RNA-seq at multiple time points

    • Identify temporal relationships between specific phosphorylation events and transcriptional changes

    • Group target genes based on their activation kinetics relative to c-JUN phosphorylation

  • Chromatin Immunoprecipitation (ChIP) with Phospho-Specific Antibodies:

    • Use phospho-JUN (S63) antibodies for ChIP experiments

    • Determine the genomic binding sites of specifically S63-phosphorylated c-JUN

    • Compare with binding profiles of total c-JUN or other phosphorylated forms

  • Live-Cell Imaging with Phosphorylation Biosensors:

    • Develop FRET-based biosensors for real-time visualization of c-JUN phosphorylation

    • Correlate spatiotemporal patterns of phosphorylation with transcriptional reporter activity

    • Analyze single-cell dynamics to capture heterogeneity in response

  • Targeted Inhibition of Specific Kinases:

    • Compare the effects of different JNK isoform-specific inhibitors on c-JUN phosphorylation kinetics

    • Use cell-permeable c-Jun peptide inhibitors that demonstrate dose-dependent inhibition of c-Jun phosphorylation following anisomycin stimulation

These advanced approaches collectively provide a comprehensive view of how the temporal dynamics of c-JUN phosphorylation translate into specific patterns of gene expression, offering insights into the encoding of signal specificity in the JNK pathway.

How can phospho-JUN (S63) detection be integrated into multi-parameter analyses of stress response pathways?

Integration of phospho-JUN (S63) detection into multi-parameter analyses provides a more comprehensive understanding of stress response signaling networks:

  • Multiplexed Phospho-Protein Profiling:

    • Combine phospho-JUN (S63) detection with analysis of other JNK substrates

    • Include upstream activators (MKK4/7 phosphorylation) and parallel MAPK pathways (p38, ERK)

    • HTRF technology allows development of multiplexed assays using different fluorophore combinations

  • Single-Cell Multi-Parameter Analysis:

    • Utilize phospho-flow cytometry to simultaneously detect phospho-JUN (S63) and other pathway components

    • Combine with cell cycle markers or apoptosis indicators

    • Reveals cell-to-cell heterogeneity and identifies distinct cellular subpopulations

  • Pathway Activity Correlation:

    • Correlate phospho-JUN (S63) levels with functional readouts:

      • AP-1 transcriptional reporter activity

      • Expression of c-JUN target genes

      • Apoptosis markers

      • Cell proliferation indices

  • Dynamic Pathway Modeling:

    • Develop mathematical models incorporating the temporal kinetics of c-JUN phosphorylation

    • S63 is phosphorylated most rapidly, followed by S73, with T91/T93 phosphorylated more slowly

    • Model how these differential kinetics contribute to signal encoding and specificity

  • Integration with -Omics Approaches:

    • Correlate phospho-JUN (S63) levels with:

      • Transcriptomic profiles (RNA-seq)

      • Chromatin accessibility changes (ATAC-seq)

      • Global phosphoproteomic alterations (phospho-MS)

    • Identify broader network changes associated with specific phosphorylation patterns

This integrated approach provides a systems-level understanding of how phospho-JUN (S63) signaling contributes to complex cellular responses to various stressors, potentially revealing novel therapeutic targets or biomarkers for stress-related pathologies.

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 2024 Thebiotek. All Rights Reserved.