Phospho-RB1 (S788) Antibody

Shipped with Ice Packs
In Stock

Description

Introduction

The Phospho-RB1 (S788) Antibody is a highly specific immunological reagent designed to detect phosphorylated serine at position 788 (S788) of the Retinoblastoma protein 1 (RB1), a critical tumor suppressor involved in cell cycle regulation. This antibody is widely used in research to study RB1’s post-translational modifications, which are essential for its functional activity. Below, we provide a detailed analysis of its characteristics, applications, and research findings.

Antibody Characteristics

PropertyDetail
TypeRecombinant multiclonal antibody (rabbit IgG)
EpitopeSynthetic peptide containing phosphorylated serine 788 within the RB1 protein (aa 750–800)
ApplicationsImmunocytochemistry (ICC), Western blot (WB)
ReactivityHuman samples
ImmunogenPhosphorylated RB1 peptide at S788 (aa 750–800)
VendorAbcam (ab277775)

Key Features:

  • Multiclonal Design: Combines specificity of monoclonal antibodies with the broader epitope recognition of polyclonal antibodies.

  • Recombinant Origin: Ensures lot-to-lot consistency and reduced batch variability .

3.1. Immunocytochemistry (ICC)

  • Used to visualize phosphorylated RB1 in fixed cells, enabling spatial analysis of S788 modification .

  • Example: Studying RB1 localization during cell cycle progression or apoptosis.

3.2. Western Blot (WB)

  • Detects phosphorylated RB1 in lysates, aiding in quantification of S788 phosphorylation levels .

  • Ideal for analyzing RB1 activation in response to mitogenic signals or oncogenic stress.

4.1. RB1 Phosphorylation Dynamics

Phosphorylation of RB1 at specific residues (e.g., S788, S807, S608) regulates its interaction with transcription factors like E2F and its role in apoptosis . For instance:

  • S788 Phosphorylation: Emerging data suggest this site modulates RB1’s chromatin-modifying activities, including recruitment of histone methyltransferases (SUV39H1, KMT5B/C) .

  • Cross-Talk with S807/S811: Studies on S807 phosphorylation reveal its role in binding pro-apoptotic proteins like Bax . While S788-specific interactions are less characterized, antibodies targeting this site enable further exploration.

4.2. Cancer Implications

  • Elevated phosphorylation of RB1 (including S788) is observed in cancer cells, correlating with unchecked proliferation .

  • Antibodies like ab277775 facilitate mechanistic studies linking S788 phosphorylation to oncogenic pathways.

Comparative Analysis of RB1 Antibodies

AntibodyEpitopeApplicationsReactivityVendor
Phospho-RB1 (S788)S788 (phospho)ICC, WBHumanAbcam (ab277775)
Phospho-RB1 (S807)S807/S811 (phospho)WB, IP, ICCHuman, Mouse, RatAbcam (ab184796)
Phospho-RB1 (S608)S608 (phospho)WB, ELISA, IHCHuman, Mouse, RatSanta Cruz (sc-1661)
Total RB1Full-length proteinWB, IHC, IPHuman, Mouse, RatCell Signaling (9309)

Key Differences:

  • Epitope Specificity: S788 antibodies target a distinct phosphorylation site compared to S807/S811 or S608.

  • Cross-Reactivity: S788 antibodies (ab277775) are human-specific, whereas S807/S811 antibodies (ab184796) cross-react with rodent samples .

Product Specs

Buffer
Liquid in PBS containing 50% glycerol, 0.5% BSA and 0.02% sodium azide.
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery timelines may vary based on your chosen shipping method and destination. Please consult your local distributors for specific delivery information.
Synonyms
Exon 17 tumor GOS561 substitution mutation causes premature stop antibody; GOS563 exon 17 substitution mutation causes premature stop antibody; OSRC antibody; Osteosarcoma antibody; p105-Rb antibody; P105RB antibody; PP105 antibody; pp110 antibody; PPP1R130 antibody; pRb antibody; Prepro retinoblastoma associated protein antibody; Protein phosphatase 1 regulatory subunit 130 antibody; Rb antibody; RB transcriptional corepressor 1 antibody; RB_HUMAN antibody; RB1 antibody; RB1 gene antibody; Retinoblastoma 1 antibody; Retinoblastoma suspectibility protein antibody; Retinoblastoma-associated protein antibody
Target Names
RB1
Uniprot No.

Target Background

Function
The retinoblastoma protein (RB1) is a tumor suppressor that plays a crucial role in regulating the cell cycle, particularly the transition from the G1 to S phase. In its hypophosphorylated form, RB1 binds to transcription regulators of the E2F family, preventing the transcription of E2F-responsive genes. This occurs through two mechanisms: first, RB1 physically blocks the E2Fs' transactivating domain; second, it recruits chromatin-modifying enzymes that actively repress transcription. Phosphorylation of RB1 by cyclin-dependent kinases (CDKs) disrupts its interaction with E2Fs, leading to the activation of E2F-responsive genes and the initiation of S phase. RB1 also participates in the G0-G1 transition upon phosphorylation and activation by CDK3/cyclin-C.
Additionally, RB1 plays a direct role in heterochromatin formation by maintaining the overall chromatin structure, particularly that of constitutive heterochromatin. It accomplishes this by stabilizing histone methylation and recruiting histone methyltransferases such as SUV39H1, KMT5B, and KMT5C, resulting in epigenetic transcriptional repression. RB1 specifically controls histone H4 lysine 20 trimethylation. Furthermore, RB1 inhibits the intrinsic kinase activity of TAF1 and mediates transcriptional repression by SMARCA4/BRG1 by recruiting a histone deacetylase (HDAC) complex to the c-FOS promoter. In resting neurons, the transcription of the c-FOS promoter is inhibited by BRG1-dependent recruitment of a phospho-RB1-HDAC1 repressor complex. Upon calcium influx, RB1 is dephosphorylated by calcineurin, which releases the repressor complex.
In the context of viral infections, interactions with viral proteins such as SV40 large T antigen, HPV E7 protein, or adenovirus E1A protein can disrupt the RB1-E2F1 complex, thereby inhibiting RB1's activity.
Gene References Into Functions
  1. Mutations in genes like CDKN2B or RB1 have been associated with a poorer clinical outcome in lung adenocarcinoma patients with EGFR activating mutations. PMID: 29343775
  2. A study in Vietnamese patients with retinoblastoma identified three novel mutations in the germline RB1 gene. PMID: 29568217
  3. Research using phospho-defective and phospho-mimetic mutants of FoxM1b revealed a critical role for Plk1 phosphorylation sites in regulating the binding of FoxM1b to Rb and DNMT3b. PMID: 28387346
  4. The accumulation of sequence variations in the RB1 gene may influence the susceptibility of Greek patients to the progression of cervical neoplasia. PMID: 30303478
  5. Vitiligo lesions exhibit dysregulated SUMOylation and deSUMOylation in keratinocytes. Dysregulation of the cell cycle progression was observed in SUMO1 knockdown HaCaT cells. The deSUMOylation of Rb in keratinocytes may play an important role in the development of vitiligo. PMID: 30066925
  6. The Rb1 tumor suppressor gene modifies telomeric chromatin architecture by regulating TERRA expression. PMID: 28169375
  7. These findings demonstrate that developmental stage-specific, species-specific, and cell type-specific features sensitize to RB1 inactivation. Human cone precursors provide a model for retinoblastoma initiation, proliferation, premalignant arrest, and tumor growth. PMID: 30213853
  8. Low pRB expression is associated with mouth cancer. PMID: 30275188
  9. Control of the Restriction Point by Rb and p21. PMID: 30111539
  10. Studies showed that (a) alterations of the p53 and Rb pathways are associated with high proliferation of tumor cells in buccal mucosa cancer (BUC), and (b) high expression of cell-cycle proteins is associated with adverse histopathological parameters in these tumors. PMID: 29970521
  11. Results indicate that vascular smooth muscle proliferation is regulated by activation of the NF-kappaB p65/miR17/RB pathway. As NF-kappaB p65 signaling is activated in and is a master regulator of the inflammatory response, these findings may provide a mechanism for the excessive proliferation of vascular smooth muscle cells (VSMCs) under inflammation during vascular disorders, potentially identifying novel targets for the treatment of these conditions. PMID: 29115381
  12. Reduced RB expression in medullary thyroid cancer is associated with decreased patient survival in univariate and multivariable analyses, independent from patient age at surgery or advanced TNM stage. PMID: 29105562
  13. Immunohistochemistry and immunoblot analysis revealed that the expression levels of cyclin D1, cyclin E, pRb, and Ki67 in psoriasis lesions decreased after treatment and were similar to those in the normal group. PMID: 29115643
  14. Data suggest that nuclear envelope rupture in cancer cells is likely due to the loss of either the Rb or the p53 pathway. PMID: 28811362
  15. Altered pRb expression is frequently observed in gastric carcinoma and inversely correlates with tumor invasion and tumor stage, suggesting its involvement as an early event in gastric carcinogenesis. PMID: 28965621
  16. Findings show that H1.2 augments global association of pRb with chromatin, enhances transcriptional repression by pRb, and facilitates pRb-dependent cell-cycle arrest, defining a network of E2F target genes susceptible to the regulatory influence of H1.2. PMID: 28614707
  17. The increased expression of miR-503-5p significantly reduced the expression of E2F transcription factor 3 (E2F3) mRNA and retinoblastoma protein (Rb)/E2F signaling pathway mRNA in bladder cancer cells. PMID: 29169421
  18. Loss of Rb immunolabeling and KRAS mutation are promising molecular markers for the therapeutic response to platinum-based chemotherapy in pancreatic neuroendocrine neoplasm grade-3 (PanNEN-G3) and neuroendocrine tumor with G3 (NET-G3). PMID: 28455360
  19. Intensive ocular screening for retinoblastoma and neuroimaging for pineoblastoma surveillance are recommended for patients with germline RB1 mutations. Individuals with hereditary RB have an approximately 20% risk of developing second primary cancers, a higher risk among those who received radiotherapy for their primary RB tumors. PMID: 28674118
  20. The SNPs rs 216311, rs 1800383, and rs 1800386 were significantly associated with bleeding in study subjects. rs1800386 occurred in all individuals with a history of bleeding, and no ethnic variations were noted. PMID: 28091443
  21. miR-215 promoted cell migration and invasion of gastric cancer by directly targeting RB1. PMID: 28689850
  22. MiR-661 promotes metastasis of non-small cell lung cancer through RB/E2F1 signaling and epithelial-mesenchymal transition events. PMID: 28716024
  23. RB1 was identified as a direct and functional target of miR-215. RB1 expression is generally down-regulated in glioma tissues and inversely correlated with miR-215, which is up-regulated in high-grade glioma tissues. Its expression was negatively correlated with miR-215. PMID: 28573541
  24. Loss of retinoblastoma in pleomorphic fibroma: An immunohistochemical and genomic analysis. PMID: 28543636
  25. Results show that RB1 expression is regulated by cdc37, which facilitates its phosphorylation by increasing CDK4 stability. PMID: 29288563
  26. SOX2 overexpression and the loss of Rb1 protein expression might play a pivotal role in the divergent differentiation of pluripotent embryonic-like epithelial cells and the development of esophageal small-cell carcinoma. PMID: 28106103
  27. Several RB1 alterations associated with retinoblastoma in humans were also present in several non-human primates without apparent pathological effects. PMID: 28401291
  28. Results suggest that RB1 is the dominant tumor suppressor gene in Merkel cell carcinoma (MCC) and that inactivation of RB1 by MCPyV-LT is largely sufficient for its growth-supporting function in established MCPyV-positive MCC cells. PMID: 27121059
  29. The frequency and association of polymorphisms in the TP53 and RB1 genes with clinical characteristics in a group of children with retinoblastoma (RB) in northern Mexico were examined. PMID: 28210099
  30. RB underexpression is associated with tumor cell invasiveness and neuroendocrine differentiation in prostate cancer. PMID: 27015368
  31. Authors demonstrate that MYC inhibition by Omomyc, a dominant-negative MYC, suppresses the growth of small cell lung cancer (SCLC) cells with TP53 and RB1 inactivation carrying MYC, MYCL, or MYCN amplification. PMID: 27105536
  32. Data suggest that the platelet-derived growth factor receptor alpha (PDGFRalpha)/Stat3 transcription factor/Rb1 protein regulatory axis might represent a potential therapeutic target for glioblastoma (GBM) treatment. PMID: 27344175
  33. miR-590 inhibits RB1 and promotes proliferation and invasion of T-cell acute lymphoblastic leukemia cells. PMID: 27036041
  34. Causative RB1 mutations in most bilateral retinoblastoma (RB) patients and in some unilateral RB patients, including five novel mutations, were identified. PMID: 29261756
  35. Homozygous loss of RB1 is an independent prognostic marker in multiple myeloma. PMID: 28234347
  36. In certain contexts, Rb loss enables TRbeta1-dependent suppression of SKP2 as a safeguard against RB1-deficient tumorigenesis. TRbeta2 counteracts TRbeta1, thus disrupting this safeguard and promoting the development of RB1-deficient malignancies. PMID: 28972075
  37. Expression levels of miR-675-5p in glioma tissues and cells were negatively correlated with RB1 expression at both mRNA and protein levels and promoted cell proliferation and migration. PMID: 28970140
  38. Disruption of DREAM and RB-E2F complexes by oncoproteins from DNA tumor viruses leads to the upregulation of cell cycle genes and impairs growth-inhibiting pathways, including the p53-mediated downregulation of cell cycle genes. [review] PMID: 28799433
  39. A relatively stable genome in retinoblastoma tumor cells is maintained by TRb1 and TRb2-mediated PTTG1 inhibition, counteracting Rb-deficiency-related genomic instability. PMID: 28242412
  40. APC/C and pRB interact with each other via the co-activator of APC/C, FZR1, providing an alternative pathway of regulation of G1 to S transition by pRB using a post-translational mechanism. Both pRB and FZR1 have complex roles and are implicated not only in regulation of cell proliferation but also in differentiation, quiescence, apoptosis, maintenance of chromosomal integrity and metabolism. PMID: 27402801
  41. Analysis of the spectrum of RB1 variants observed in 60,706 exomes identifies 197 variants that have enough potential to disrupt splicing to warrant further consideration. PMID: 28780672
  42. AR also indirectly increases the expression of DNA replication genes through stimulatory effects on other metabolic genes with subsequent CDK activation and Rb hyperphosphorylation. PMID: 27760327
  43. Rb gene promoter methylation was more frequent in gastric cancer patients than in controls. PMID: 28319413
  44. We report the significance of genetic testing in the early detection and management of retinoblastoma from India. PMID: 26914665
  45. Results show that the functional state of protein Rb is inferred to be inactive due to its phosphorylation status in MYCN-amplified retinoblastoma without coding sequence mutations. This makes inactivation of RB1 by gene mutation or by protein phosphorylation a necessary condition for initiating retinoblastoma tumorigenesis, independent of MYCN amplification. PMID: 28211617
  46. Low RB expression is associated with osteosarcoma. PMID: 28655788
  47. Loss of RB1 is associated with papillomavirus involvement in Barrett's dysplasia and esophageal adenocarcinoma. PMID: 28722212
  48. The epigenetic interaction between Linc00441 and bidirectional transcripted neighbor RB1 may be a de novo theory cutting-point for the inactivation of RB1 in hepatocellular carcinoma (HCC). PMID: 28300839
  49. The data indicate that MAZ is essential to bypass MYB promoter repression by RB family members and to induce MYB expression. PMID: 28973440
  50. RB inactivation enhances pro-inflammatory signaling through stimulation of the interleukin-6/STAT3 pathway, which directly promotes various malignant features of cancer cells. [review] PMID: 28865172

Show More

Hide All

Database Links

HGNC: 9884

OMIM: 109800

KEGG: hsa:5925

STRING: 9606.ENSP00000267163

UniGene: Hs.408528

Involvement In Disease
Childhood cancer retinoblastoma (RB); Bladder cancer (BLC); Osteogenic sarcoma (OSRC)
Protein Families
Retinoblastoma protein (RB) family
Subcellular Location
Nucleus.
Tissue Specificity
Expressed in the retina. Expressed in foreskin keratinocytes (at protein level).

Q&A

What is the biological significance of RB1 phosphorylation at serine 788?

RB1 (Retinoblastoma 1) protein is a critical tumor suppressor that regulates cell cycle progression, particularly the G1/S transition. Phosphorylation at serine 788 (S788) is functionally significant as it directly inhibits RB's association with E2F-DP heterodimers, which are transcription factors controlling cell cycle progression. This site-specific phosphorylation contributes to a "phosphorylation code" that modulates RB1's interaction with various protein partners and consequently its tumor suppressor functions. S788 phosphorylation affects RBC (RB carboxyl terminal domain) interactions, which has distinct consequences compared to phosphorylation at other sites such as T821 or T826.

How does mono-phosphorylation at S788 differ functionally from other RB1 phosphorylation sites?

While RB1 can be phosphorylated at 14 different CDK sites, mono-phosphorylation at S788 has distinct functional consequences. Research has demonstrated that mono-phosphorylated RB1 at S788 maintains activity in arresting cells in G1-phase, with efficiency variations compared to other phosphorylation sites. In fact, studies show that S788 mono-phosphorylation is among the sites (along with T356) that provide the greatest G1 arrest increase. This indicates that mono-phosphorylation at S788 likely confers specific functional properties beyond simple inactivation of RB1, contributing to nuanced regulation of cell cycle progression.

What is the relationship between S788 phosphorylation and RB1's tumor suppressor activity?

S788 phosphorylation modulates rather than completely inactivates RB1's tumor suppressor functions. Unlike the prevailing model that only hypo-phosphorylated RB1 is active, evidence suggests that mono-phosphorylated RB1, including at S788, remains biologically active in controlling cell cycle progression. The phosphorylation at S788 specifically affects RB1's binding to E2F-DP heterodimers, thereby providing a mechanism for fine-tuning transcriptional repression of cell cycle genes rather than completely abolishing tumor suppressor activity. This challenges the traditional binary view of RB1 function (active vs. inactive) and suggests a more nuanced regulatory system.

What are the recommended experimental conditions for using Phospho-RB1 (S788) antibodies in Western blotting?

For optimal Western blotting results with Phospho-RB1 (S788) antibodies, researchers should follow these methodological guidelines:

  • Sample preparation: Use freshly prepared protein lysates from cells treated with phosphatase inhibitors to preserve phosphorylation status.

  • Dilution ratios: Initially test at 1:500-1:2000 dilution range in 5% BSA in TBST.

  • Blocking conditions: Use 5% BSA in TBST buffer for 1 hour at room temperature to reduce background.

  • Incubation parameters: Incubate with primary antibody overnight at 4°C with gentle rocking.

  • Validation controls: Include both phosphorylated and dephosphorylated samples (e.g., by treating with lambda phosphatase) to confirm specificity.

  • Detection system: Use HRP-conjugated secondary antibodies with enhanced chemiluminescence for optimal signal detection.

How can Phospho-RB1 (S788) antibodies be validated for specificity in immunohistochemistry applications?

Validating Phospho-RB1 (S788) antibodies for immunohistochemistry requires a multi-step approach:

  • Peptide competition assay: Pre-incubate antibody with the phosphorylated and non-phosphorylated peptide immunogens to confirm phospho-specificity.

  • Phosphatase treatment controls: Process serial sections with and without lambda phosphatase treatment prior to antibody incubation.

  • Knockout/knockdown controls: Use RB1-knockout or RB1-depleted tissue samples as negative controls.

  • Cross-validation: Compare staining patterns across multiple phospho-RB1 antibodies targeting different epitopes.

  • Cell cycle phase synchronization: Use samples from cells synchronized at different cell cycle stages to confirm cell cycle-dependent phosphorylation patterns.

  • Proper antigen retrieval: Optimize pH (typically using citrate buffer pH 6.0) and heat-induced epitope retrieval conditions to preserve phospho-epitope integrity.

What cell models are most appropriate for studying S788 phosphorylation dynamics?

The selection of appropriate cell models for studying S788 phosphorylation dynamics should consider:

  • Cell cycle characteristics: Primary human fibroblasts (HFFs) offer advantages for cell cycle synchronization studies through contact inhibition or serum starvation.

  • Tumor vs. normal cells: Compare retinoblastoma-derived cell lines with RPE1 cells (retinal pigment epithelial cells) to study phosphorylation differences between cancer and normal cells.

  • Tissue-specific regulation: Breast cancer cell lines like T47D, CAMA1, and MDA-MB-361 exhibit distinct patterns of RB1 phosphorylation in response to different treatments.

  • Manipulation potential: U2OS osteosarcoma cells are amenable to genetic engineering for creating isogenic cell lines expressing single phosphorylation site RB1 mutants.

  • Cell synchronization methods: Contact inhibition in RPE1 cells causes dephosphorylation of most sites but preferential phosphorylation at S780, while γ-irradiation in T47D cells generates selective phosphorylation at S807/S811, making these useful models for studying site-specific phosphorylation dynamics.

How do different cell cycle arrest mechanisms impact S788 phosphorylation status compared to other RB1 phosphorylation sites?

Different cell cycle arrest mechanisms have distinct impacts on the phosphorylation status of S788 versus other RB1 sites:

  • Contact inhibition: In RPE1 and CAMA1 cells, contact inhibition causes dephosphorylation of S608, S795, S807/S811, and T821 while leading to preferential phosphorylation at S780. S788 phosphorylation patterns differ from these sites, suggesting specific regulation mechanisms.

  • DNA damage responses: γ-irradiation or hydroxyurea treatment in T47D cells selectively maintains phosphorylation on S807/S811 while reducing phosphorylation at other sites.

  • Topoisomerase inhibition: In MDA-MB-361 cells, camptothecin treatment preferentially reduces phosphorylation of S780 and S795, while having different effects on S788.

  • CDK4/6 inhibition: Treatment with palbociclib generally reduces RB1 phosphorylation in RPE1 and T47D cells, but not all phosphorylation isoforms are equally suppressed.

These differential patterns cannot be simply attributed to differences in cell cycle position, suggesting site-specific regulatory mechanisms that may involve different kinases, phosphatases, or structural accessibility of the phosphorylation sites.

What are the methodological considerations for detecting mono-phosphorylated versus hyper-phosphorylated RB1 in complex samples?

Distinguishing mono-phosphorylated from hyper-phosphorylated RB1 species requires specialized approaches:

  • Two-dimensional isoelectric focusing (2D IEF): This technique separates RB1 isoforms based on charge differences, allowing visualization of distinct mono-phosphorylated species. Each phosphate addition shifts the protein to a more acidic position.

  • Sequential immunoprecipitation: Immunoprecipitate with one phospho-specific antibody (e.g., anti-phospho-S788) followed by immunoblotting with multiple phospho-specific antibodies targeting different sites. Mono-phosphorylated species will only be detected by the antibody matching the immunoprecipitation.

  • Phos-tag SDS-PAGE: This modified electrophoresis technique incorporates Phos-tag molecules that specifically bind phosphorylated proteins, resulting in mobility shifts proportional to phosphorylation status.

  • Mass spectrometry: Quantitative phosphoproteomics can identify and quantify site-specific phosphorylation, though this requires careful sample preparation to preserve phosphorylation status.

  • Phosphatase treatment gradients: Treating samples with increasing amounts of phosphatase can help distinguish mono- from multi-phosphorylated species based on their dephosphorylation kinetics.

How does S788 phosphorylation interact with other post-translational modifications on RB1?

S788 phosphorylation exists within a complex network of post-translational modifications (PTMs) on RB1:

  • Interdependent phosphorylation: While S807/S811 phosphorylation has been established as a priming event for other phosphorylation sites, the relationship between S788 phosphorylation and other sites requires further investigation.

  • Methylation crosstalk: K810 methylation inhibits S807/S811 phosphorylation, which indirectly affects phosphorylation throughout RB1, potentially including S788.

  • Prolyl isomerization: Phosphorylation at S608/S612 facilitates recruitment of the prolyl isomerase Pin1, which can influence further phosphorylation events on RB1.

  • Acetylation interactions: RB1 acetylation at the C-terminus may compete with or influence phosphorylation at nearby sites including S788.

  • Structural consequences: Unlike S807/S811 phosphorylation, which may not cause structural changes but rather promote intermolecular associations, S788 phosphorylation directly affects RB1's ability to bind E2F-DP complexes, suggesting distinct structural impacts.

These complex interactions highlight the importance of considering the broader PTM landscape when studying S788 phosphorylation.

How should researchers interpret seemingly contradictory results between phospho-specific antibodies and functional assays?

When faced with discrepancies between phospho-specific antibody results and functional assays:

  • Consider antibody cross-reactivity: Phospho-specific antibodies may recognize similar phospho-epitopes at different sites. Validate with peptide competition assays using the specific phospho-peptide.

  • Evaluate total vs. site-specific phosphorylation: Total RB1 phosphorylation may not correlate with specific site phosphorylation. Use multiple phospho-specific antibodies to assess the complete phosphorylation profile.

  • Assess phosphorylation stoichiometry: Low-level phosphorylation may be detected by sensitive antibodies but might not reach the threshold for functional impact.

  • Account for localization differences: Phosphorylated RB1 may be differentially distributed between nuclear and cytoplasmic compartments, affecting functional outcomes.

  • Consider temporal dynamics: Phosphorylation at S788 may be transient or occur with different kinetics than functional changes, requiring time-course analyses.

  • Evaluate other concurrent modifications: Other post-translational modifications may counteract or synergize with S788 phosphorylation, complicating interpretation of isolated phosphorylation detection.

What are the potential pitfalls in quantifying S788 phosphorylation levels across different experimental systems?

Researchers should be aware of several methodological challenges when quantifying S788 phosphorylation:

  • Antibody affinity variations: Different lots or sources of phospho-S788 antibodies may have varying affinities, making direct comparisons between studies difficult.

  • Context-dependent epitope accessibility: The accessibility of the S788 phospho-epitope may vary depending on protein conformation or complex formation.

  • Sample preparation artifacts: Phosphorylation status can be rapidly altered during sample collection and processing if phosphatase inhibitors are not properly used.

  • Normalization strategies: Proper normalization requires accounting for total RB1 levels, which may vary between samples or experimental conditions.

  • Cell synchronization differences: Cell cycle synchronization methods can themselves affect phosphorylation patterns, confounding interpretation of experimental manipulations.

  • Single-cell heterogeneity: Population-based assays may mask important cell-to-cell variability in phosphorylation status, particularly in asynchronous cultures.

  • Method-specific biases: Western blotting, immunofluorescence, and flow cytometry may yield different results due to technique-specific limitations.

How can researchers distinguish between direct and indirect effects on S788 phosphorylation in signaling pathway analyses?

To differentiate direct and indirect effects on S788 phosphorylation:

  • Kinase inhibitor specificity: Use multiple structurally distinct inhibitors of the same kinase to confirm on-target effects, or employ analog-sensitive kinase mutants for greater specificity.

  • In vitro kinase assays: Perform direct kinase assays with purified components to establish whether a kinase can directly phosphorylate S788.

  • Phosphatase identification: Use phosphatase inhibitors and genetic approaches to determine which phosphatases directly target S788.

  • Temporal resolution: Conduct high-resolution time-course experiments to establish the sequence of events in signaling cascades.

  • Substrate mutants: Create non-phosphorylatable mutants (S788A) to establish the requirement for this site in specific pathways.

  • Phospho-mimetic approaches: Compare S788D or S788E phospho-mimetic mutants with phospho-null mutants to distinguish direct phosphorylation effects from structural requirements.

  • Proximity-based approaches: Use proximity ligation assays or BioID to identify proteins that physically interact with RB1 near the S788 site under various conditions.

How does S788 phosphorylation contribute to the broader "phosphorylation code" regulating RB1 function beyond cell cycle control?

Recent research suggests that RB1 phosphorylation at S788 contributes to a sophisticated regulatory code with functions extending beyond cell cycle control:

  • Gene expression programming: S788 phosphorylation may affect RB1's regulation of specific gene sets distinct from those controlled by other phosphorylation sites, contributing to cellular identity and differentiation.

  • Protein complex assembly: While all mono-phosphorylated RB1 isoforms interact with E2F/DP proteins, they provide "different shades" of E2F regulation, with S788 mono-phosphorylation creating distinct protein complexes with unique functional outputs.

  • Metabolic regulation: Unlike S811 or T826 phosphorylation, which stimulate expression of oxidative phosphorylation genes and increase cellular oxygen consumption, S788 phosphorylation likely affects alternative metabolic pathways.

  • Developmental programming: The impact of S788 phosphorylation may vary during development or in different tissue contexts, contributing to tissue-specific functions of RB1.

  • Stress responses: The susceptibility of S788 to phosphorylation/dephosphorylation may vary under different stress conditions, providing stress-specific modulation of RB1 function.

These emerging functions highlight the need to consider S788 phosphorylation as part of an integrated regulatory system rather than in isolation.

What methodological advances are needed to better study the dynamics of S788 phosphorylation in living cells?

To advance understanding of S788 phosphorylation dynamics in living systems, several methodological developments are needed:

  • Phospho-specific biosensors: Development of FRET-based biosensors specifically responsive to S788 phosphorylation would enable real-time monitoring in living cells.

  • Engineered phospho-readers: Engineered protein domains that specifically recognize phospho-S788 could be coupled to fluorescent proteins for live imaging.

  • Site-specific incorporation of phospho-mimetics: Genetic code expansion technologies to incorporate phosphoserine directly at position 788 would facilitate functional studies.

  • Single-cell phosphoproteomics: Advances in mass spectrometry to enable single-cell resolution of RB1 phosphorylation states would reveal cell-to-cell heterogeneity.

  • Temporal control systems: Optogenetic or chemically-inducible systems to rapidly modulate kinase/phosphatase activity would help dissect the kinetics of S788 phosphorylation.

  • Intravital imaging techniques: Methods to visualize phosphorylation status in intact tissues would bridge the gap between cell culture and in vivo relevance.

  • Computational modeling: Integrative mathematical models incorporating multiple phosphorylation sites and their interdependencies would help predict system behavior.

What is the potential clinical significance of S788 phosphorylation status in cancer diagnosis or treatment strategies?

The clinical relevance of S788 phosphorylation extends to several cancer-related applications:

  • Diagnostic biomarker potential: S788 phosphorylation status may serve as a biomarker for specific cancer types or stages, particularly in retinoblastoma, bladder cancer, and osteosarcoma where RB1 mutations are common.

  • Treatment response prediction: The phosphorylation status at S788 relative to other sites might predict response to CDK4/6 inhibitors like palbociclib, which are increasingly used in cancer therapy.

  • Resistance mechanisms: Altered patterns of S788 phosphorylation could represent a mechanism of resistance to targeted therapies, offering insights for treatment adjustment.

  • Combinatorial therapy approaches: Understanding the functional consequences of S788 phosphorylation might guide rational drug combinations targeting both RB1 phosphorylation and its downstream effects.

  • Synthetic lethality opportunities: Cancer cells with specific patterns of RB1 phosphorylation, including at S788, might exhibit vulnerabilities that could be exploited through synthetic lethal approaches.

  • Patient stratification: Phosphorylation patterns at S788 and other sites could help stratify patients for clinical trials or personalized treatment approaches.

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.