Phospho-RB1 (S612) Antibody

Shipped with Ice Packs
In Stock

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 products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timelines.
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
Phospho-RB1 (S612) Antibody targets a tumor suppressor protein that plays a crucial role in regulating the G1/S transition of the cell cycle. The hypophosphorylated form of RB1 binds to transcription regulators of the E2F family, preventing the transcription of E2F-responsive genes. This inhibition occurs through two mechanisms: RB1 physically blocks the E2Fs' transactivating domain and recruits chromatin-modifying enzymes that actively repress transcription. Cyclin-dependent kinase (CDK)-mediated phosphorylation of RB1 triggers its dissociation from E2Fs, leading to the activation of E2F-responsive genes and the initiation of S phase. Moreover, RB1 promotes the G0-G1 transition upon phosphorylation and activation by CDK3/cyclin-C. RB1 is directly involved in heterochromatin formation by maintaining the overall chromatin structure, particularly stabilizing constitutive heterochromatin through the stabilization of histone methylation. It recruits and targets histone methyltransferases SUV39H1, KMT5B, and KMT5C, resulting in epigenetic transcriptional repression. RB1 controls histone H4 'Lys-20' trimethylation. Additionally, it 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, 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, leading to the release of the repressor complex. During viral infections, interactions with SV40 large T antigen, HPV E7 protein, or adenovirus E1A protein induce the disassembly of the RB1-E2F1 complex, disrupting RB1's activity.
Gene References Into Functions
  1. Concurrent mutations in genes such as CDKN2B or RB1 were associated with worse clinical outcomes in lung adenocarcinoma patients with EGFR active mutations. PMID: 29343775
  2. Mutational screening of the germline RB1 gene in Vietnamese patients with retinoblastoma revealed three novel mutations. PMID: 29568217
  3. Analyses with phospho-defective and phospho-mimetic mutants of FoxM1b identified a critical role of the 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 might influence Greek patients' susceptibility towards the progression of cervical neoplasia. PMID: 30303478
  5. Vitiligo lesions exhibited dysregulated SUMOylation and deSUMOylation in keratinocytes. Dysregulation of cell cycle progression was observed in SUMO1 knockdown HaCaT cells, suggesting that deSUMOylation of Rb in keratinocytes may play a crucial 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, as well as species- and cell type-specific features, sensitize to RB1 inactivation and reveal the human cone precursors' capacity to model 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. Results showed that: a) alterations of the p53 and Rb pathways are associated with high proliferation of tumor cells in buccal cancer (BUC), and b) high expression of cell-cycle proteins is associated with adverse histopathological parameters of these tumors. PMID: 29970521
  11. The present result indicated that vascular smooth proliferation is regulated by activation of the NF-kappaB p65/miR17/RB pathway. Since 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 and may identify novel targets for the treatment of vascular disease. 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. According to immunohistochemistry and immunoblot analysis, 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 indicate that nuclear envelope rupture in cancer cells is likely due to loss of either the Rb or the p53 pathway. PMID: 28811362
  15. Altered pRb is frequently expressed in gastric carcinoma, inversely correlates with tumor invasion and tumor stage, suggesting an early event in gastric carcinogenesis. PMID: 28965621
  16. Results define a network of E2F target genes as susceptible to the regulatory influence of H1.2, where H1.2 augments global association of pRb with chromatin, enhances transcriptional repression by pRb, and facilitates pRb-dependent cell-cycle arrest. PMID: 28614707
  17. The increased expression of miR-503-5p significantly reduced the expressions 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 of the therapeutic response to platinum-based chemotherapy for pancreatic neuroendocrine neoplasm grade-3 (PanNEN-G3), and Rb for neuroendocrine tumor with G3 (NET-G3). PMID: 28455360
  19. We recommend intensive ocular screening for patients with germline RB1 mutations for retinoblastoma as well as neuroimaging for pineoblastoma surveillance. There is an approximately 20% risk of developing second primary cancers among individuals with hereditary RB, higher among those who received radiotherapy for their primary RB tumors. PMID: 28674118
  20. The SNPs rs 216311, rs 1800383, and rs 1800386 associated significantly with bleeding in study subjects. rs1800386 occurred in all with bleeding history, 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 is generally down-regulated in glioma tissues and its expression inversely correlated with miR-215, which is up-regulated in high-grade glioma tissues, and 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 through increasing CDK4 stability. PMID: 29288563
  26. SOX2 overexpression and the loss of Rb1 protein expression might have 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 present in several non-human primates without an apparent pathological effect. PMID: 28401291
  28. Results suggest that RB1 is the dominant tumor suppressor PP 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 was examined. PMID: 28210099
  30. RB underexpression is associated with tumor cell invasiveness and neuroendocrine differentiation in prostate cancer. PMID: 27015368
  31. Authors show 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 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 the 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 S612 phosphorylation in cell cycle regulation?

The phosphorylation of RB1 at S612 is one of the key modifications that regulates its function as a cell cycle inhibitor. Located in the pocket domain, S612 phosphorylation has been mechanistically proven to strongly affect RB-E2F interactions and is therefore strongly implicated in abrogating RB's tumor suppressive capacity .

RB1 exists in different phosphorylation states throughout the cell cycle:

  • Unphosphorylated form: Predominantly in G0 phase (quiescent cells)

  • Mono-phosphorylated forms: Present in early G1 phase

  • Hyper-phosphorylated forms: Appear in late G1 and S phases

S612 can exist as one of the 14 independent mono-phosphorylated RB isoforms in early G1 phase, each potentially having unique functions . This mono-phosphorylation is primarily mediated by cyclin D:Cdk4/6 complexes .

How does mono-phosphorylation at S612 differ functionally from other RB1 phosphorylation sites?

Each mono-phosphorylated RB isoform, including S612, appears to have distinct functional properties. Research has shown that individual mono-phosphorylated RB isoforms form different protein complexes with varying compositions, suggesting site-specific functional specialization .

The functional specificity of different mono-phosphorylated RB isoforms extends beyond cell cycle regulation. For example, while S811 phosphorylation promotes association with the NuRD chromatin-remodeling complex and alters gene repression patterns, other sites like S612 may regulate different protein-protein interactions and cellular processes .

Experimental data shows that all 14 mono-phosphorylated RB isoforms are capable of arresting cells in G1-phase, confirming they remain active as cell cycle regulators, but with varying efficiencies. For instance, T356 and S788 mono-phosphorylation resulted in the greatest G1 increase .

What techniques are available for detecting RB1 S612 phosphorylation?

Several techniques can be employed to detect RB1 S612 phosphorylation:

TechniqueApplicationTypical DilutionAdvantages
Western BlotProtein quantification1:500-1:1000Quantitative assessment of phosphorylation levels
ImmunohistochemistryTissue samples1:50-1:300Spatial distribution in tissues
ELISAProtein quantification1:5000High-throughput screening
ImmunofluorescenceCellular localization1:100Visual detection of subcellular distribution

For optimal results, researchers should use phospho-specific antibodies that recognize RB1 only when phosphorylated at S612. These antibodies are typically generated using synthetic phosphopeptides corresponding to residues surrounding S612 of human RB1 .

What are the optimal conditions for using Phospho-RB1 (S612) antibody in Western blot analysis?

For optimal Western blot results with Phospho-RB1 (S612) antibody:

  • Sample preparation:

    • Use fresh cell lysates or properly stored frozen samples

    • Include phosphatase inhibitors in lysis buffer to prevent dephosphorylation

    • Normalize protein loading (30-50 μg total protein per lane recommended)

  • Antibody conditions:

    • Typical dilution: 1:1000 for Western blot applications

    • Prepare antibody solution in TBST with 5% BSA and 0.02% sodium azide

    • Incubate membrane with primary antibody overnight at 4°C

  • Controls:

    • Include both phosphorylated (e.g., S-phase arrested cells) and unphosphorylated (G0 arrested cells) controls

    • Blot for total RB1 on the same membrane after stripping or on a parallel membrane

    • Report results as the ratio of phosphorylated RB1 to total RB1 protein

  • Troubleshooting:

    • High background may indicate contaminated antibody solution; replace with fresh preparation

    • Check for cross-reactivity with other phosphorylation sites (antibodies should be validated for specificity)

How can I validate the specificity of Phospho-RB1 (S612) antibody in my experiments?

Validating specificity is crucial for phospho-specific antibodies. Several approaches include:

  • Phosphatase treatment: Treat one sample with lambda phosphatase before immunoblotting. The signal should disappear in the treated sample if the antibody is specific for the phosphorylated form.

  • Phospho-blocking peptide: Pre-incubate the antibody with the phosphopeptide used as immunogen. This should block specific binding and eliminate the signal.

  • Mutant controls: Use cell lines expressing RB1 with a serine-to-alanine mutation at position 612 (S612A) as a negative control.

  • Cross-reactivity testing: Test against other phosphorylated residues on RB1. For example, the Phospho-Rb (Ser807/811) antibody specifically doesn't cross-react with RB phosphorylated at Ser608 , and similar specificity should be confirmed for S612 antibodies.

  • Cell cycle synchronization: Compare G0-arrested cells (unphosphorylated RB) versus S-phase cells (hyperphosphorylated RB) to confirm the antibody detects the expected differences in phosphorylation patterns.

What are the best sample preparation methods for detecting RB1 S612 phosphorylation?

For optimal detection of RB1 S612 phosphorylation:

  • Cell synchronization:

    • For unphosphorylated RB1: Serum deprivation (G0 arrest)

    • For mono-phosphorylated RB1: Contact inhibition (early G1 arrest)

    • For hyper-phosphorylated RB1: S-phase arrest using thymidine block or aphidicolin

  • Lysis conditions:

    • Use fresh samples whenever possible

    • Include both phosphatase inhibitors (e.g., sodium fluoride, sodium orthovanadate) and protease inhibitors

    • Maintain cold temperature throughout sample preparation

    • Use buffers with neutral to slightly alkaline pH (7.4-8.0)

  • Storage:

    • Aliquot samples to avoid repeated freeze-thaw cycles

    • Store at -80°C for long-term preservation

    • Include glycerol (10-20%) in storage buffer

  • Normalization:

    • Always quantify total protein concentration

    • Include loading controls (e.g., β-actin, GAPDH)

    • Blot for total RB1 protein to calculate phosphorylation ratio

How can I distinguish between mono-phosphorylated and hyper-phosphorylated forms of RB1?

Distinguishing between mono-phosphorylated and hyper-phosphorylated RB1 requires specific techniques:

  • Two-dimensional isoelectric focusing (2D IEF): This technique separates proteins based on charge and can resolve different phosphorylation states. Mono-phosphorylated RB shows a distinct pattern compared to hyper-phosphorylated RB .

  • Sequential immunoprecipitation: First, immunoprecipitate with a phospho-specific antibody (e.g., S612), then immunoblot with multiple phospho-specific antibodies. For mono-phosphorylated RB, only the immunoprecipitating antibody will detect the protein, whereas hyper-phosphorylated RB will be detected by multiple phospho-specific antibodies .

  • Mobility shift: On standard SDS-PAGE, hyper-phosphorylated RB migrates more slowly than mono-phosphorylated or unphosphorylated forms.

  • Timing during cell cycle:

    • G0: Unphosphorylated RB

    • Early G1: Mono-phosphorylated RB (including S612)

    • Late G1/S: Hyper-phosphorylated RB

Research has shown that the phospho-specific immunoprecipitation of mono-phosphorylated RB from early G1 phase cells is only recognized by immunoblot with the same phospho-specific antibody and not by other phospho-specific antibodies. In contrast, immunoprecipitated hyper-phosphorylated RB from S phase arrested cells is recognized by multiple phospho-specific antibodies .

What are the implications of detecting RB1 S612 phosphorylation in cancer samples?

The detection of RB1 S612 phosphorylation in cancer samples has several important implications:

  • Diagnostic value: Phosphorylation at S612 (located in the pocket domain) strongly affects RB-E2F interactions and is therefore implicated in abrogating RB's tumor suppressive capacity. Its detection can provide valuable diagnostic information .

  • Prognostic significance: Hyperphosphorylation of RB has been associated with pathological states such as cancer. Assessing specific phosphorylation sites like S612 may provide prognostic value .

  • Therapeutic targeting: Understanding the phosphorylation pattern of RB1 in specific cancers can inform therapeutic decisions, particularly for therapies targeting cell cycle regulators like CDK4/6 inhibitors .

  • Resistance mechanisms: Changes in RB1 phosphorylation patterns may indicate development of resistance to CDK inhibitors or other targeted therapies.

  • Molecular etiology: For cancer biologists, examining S612 phosphorylation status is an important tool for understanding the molecular etiology of cancer and how RB1 function is compromised .

Research has shown that phosphorylations occurring in the pocket domain (like S612) and C-terminal domain are extremely informative regarding the oncogenic proclivity of a cell, and when conducted in human tumor samples, can provide valuable diagnostic, prognostic and therapy responsiveness information .

How do different mono-phosphorylated RB isoforms interact with other cellular proteins?

Research has revealed significant differences in protein interactions among the various mono-phosphorylated RB isoforms:

  • E2F transcription factors: Different mono-phosphorylated RB isoforms show variable binding to E2F family members (E2F-1, E2F-2, E2F-3, E2F-4). This suggests a mechanism by which RB can regulate specific E2F-responsive genes differently based on its phosphorylation state .

  • Viral oncoproteins: For example, the E1a oncoprotein from adenovirus was found to bind equally well to unphosphorylated RB and all 14 mono-phosphorylated RB isoforms, suggesting that viral proteins have evolved to overcome all forms of RB regulation .

  • Chromatin modifiers: Some specific mono-phosphorylated forms have been shown to preferentially interact with chromatin-remodeling complexes. For instance, RB phosphorylation at S811 promotes association with the NuRD chromatin-remodeling complex and alters the spectrum of genes repressed by RB .

  • Mitochondrial proteins: Proteomic studies have indicated that mitochondrial changes are a major consequence of RB inactivation, suggesting that some mono-phosphorylated forms may regulate mitochondrial function .

Research has demonstrated that the specific mono-phosphorylation event provides functional specificity that extends beyond the regulation of the cell cycle, with significant differences in the composition of protein complexes formed by individual mono-phosphorylated RB isoforms .

How should I design experiments to investigate the functional consequences of S612 phosphorylation?

To investigate the functional consequences of S612 phosphorylation:

  • Phospho-mimetic and phospho-deficient mutants:

    • Generate S612D or S612E (phospho-mimetic) and S612A (phospho-deficient) RB1 mutants

    • Express these in RB1-null cell lines to isolate the effect of this specific phosphorylation

  • Inducible systems:

    • Develop isogenic cell lines with inducible expression of wild-type or mutant RB1

    • This approach has been used successfully to study mono-phosphorylated RB isoforms

  • Proteomic analysis:

    • Perform immunoprecipitation followed by mass spectrometry to identify proteins that interact specifically with S612-phosphorylated RB1

    • Compare interaction partners between different phosphorylation states

  • Transcriptomic analysis:

    • Compare gene expression profiles in cells expressing wild-type RB1 versus S612A or S612D mutants

    • Focus on E2F-responsive genes and pathways

  • Cell cycle analysis:

    • Assess impact on G1/S transition using flow cytometry

    • Compare with other mono-phosphorylated RB isoforms, as research has shown that all 14 mono-phosphorylated RBs arrested cells in G1-phase but with varying efficiency

  • Kinase manipulation:

    • Use specific CDK4/6 inhibitors (e.g., PD0332991) or expression of p16 to prevent S612 phosphorylation

    • Assess downstream effects on cell cycle progression and gene expression

Studies have successfully used these approaches to demonstrate that different mono-phosphorylated RB isoforms have distinct activities and protein interaction partners .

What controls are essential when analyzing RB1 phosphorylation at S612?

Essential controls for RB1 S612 phosphorylation analysis include:

  • Phosphorylation state controls:

    • Unphosphorylated RB: Serum-starved G0 cells or cells treated with CDK4/6 inhibitors

    • Hyper-phosphorylated RB: S-phase synchronized cells

    • Cells expressing phospho-deficient RB (S612A mutant)

  • Antibody controls:

    • Phosphatase-treated samples to confirm phospho-specificity

    • Blocking peptide competition assays

    • Secondary antibody-only controls to assess non-specific binding

  • Total RB1 quantification:

    • Always blot for total RB protein to calculate the phosphorylation ratio

    • This is critical as the extent of RB phosphorylation should be assessed and reported as the ratio of phosphorylated RB to total RB protein

  • Cell cycle controls:

    • Synchronize cells at different cell cycle phases

    • Confirm cell cycle phase by flow cytometry or by blotting for cell cycle markers

  • Kinase manipulation:

    • CDK4/6 inhibitor (e.g., PD0332991) treatment as a negative control

    • p16 expression to specifically inhibit cyclin D:Cdk4/6 complexes

Research has shown that treatment with CDK4/6 inhibitors or expression of p16 results in the presence of unphosphorylated RB, confirming cyclin D:Cdk4/6 as the RB mono-phosphorylating kinase in vivo .

How can phospho-RB1 (S612) antibody be used in multi-parameter analyses?

Phospho-RB1 (S612) antibody can be incorporated into various multi-parameter analyses:

  • Multiplexed immunofluorescence:

    • Combine with antibodies against other phosphorylation sites on RB1

    • Co-stain with cell cycle markers (e.g., Ki67, PCNA)

    • Include markers for specific cellular compartments to assess localization

  • Flow cytometry:

    • Conjugate phospho-RB1 (S612) antibody with fluorescent dyes like Alexa Fluor 555

    • Combine with DNA content staining to correlate phosphorylation with cell cycle phase

    • Use with other intracellular markers for multi-parameter analysis

  • Proximity ligation assay (PLA):

    • Detect interactions between S612-phosphorylated RB1 and suspected binding partners

    • Provides spatial information about these interactions in situ

  • ChIP-seq analysis:

    • Use phospho-RB1 (S612) antibody for chromatin immunoprecipitation

    • Identify genomic regions bound by this specific phosphorylated form

    • Compare with binding profiles of other phosphorylated forms

  • Proteomics approaches:

    • Immunoprecipitate with phospho-RB1 (S612) antibody followed by mass spectrometry

    • Identify differential protein complexes formed specifically with S612-phosphorylated RB1

This multi-parameter approach has been used successfully to demonstrate that mono-phosphorylated RB isoforms have distinct activities and protein interaction partners. For example, RB phosphorylation at S811 promotes association with the NuRD chromatin-remodeling complex and alters gene repression patterns .

By integrating these approaches, researchers can gain comprehensive insights into the specific functions of S612 phosphorylation in various cellular contexts.

What are common issues when detecting RB1 S612 phosphorylation and how can they be resolved?

Common issues and their solutions include:

  • High background in immunoblotting:

    • Problem: Non-specific binding or contaminated antibody solutions

    • Solution: Use fresh antibody solutions, increase blocking time/concentration, optimize antibody dilution, include Tween-20 in wash buffers

  • Weak or absent signal:

    • Problem: Low phosphorylation levels or dephosphorylation during sample preparation

    • Solution: Include phosphatase inhibitors, avoid prolonged sample handling at room temperature, optimize cell synchronization to capture peak phosphorylation

  • Multiple bands or unexpected band size:

    • Problem: Degradation products or cross-reactivity

    • Solution: Use fresh samples with protease inhibitors, validate antibody specificity, include positive controls

  • Inconsistent results between experiments:

    • Problem: Variable phosphorylation levels due to cell cycle asynchrony

    • Solution: Carefully synchronize cells, standardize culture conditions, include cell cycle markers as controls

  • Poor reproducibility in phosphorylation quantification:

    • Problem: Technical variation in immunoblotting and detection

    • Solution: Use the ratio of phosphorylated RB to total RB protein rather than absolute phosphorylation levels

Research has shown that use of contaminated antibody solution usually yields high background, meaning that it is time to replace the solution with a fresh one .

How do cell cycle synchronization methods affect the detection of S612 phosphorylation?

Different synchronization methods significantly impact the detection of RB1 S612 phosphorylation:

  • Serum deprivation (G0 arrest):

    • Results in unphosphorylated RB

    • Useful as a negative control for phospho-specific antibodies

    • None of the phospho-specific RB antibodies recognize unphosphorylated RB from G0 arrested cells

  • Contact inhibition (early G1 arrest):

    • Results in mono-phosphorylated RB isoforms, including S612

    • Ideal for studying individual mono-phosphorylated forms

    • All phospho-specific RB antibodies recognize mono-phosphorylated RB isoforms from early G1 phase arrested cells

  • CDK4/6 inhibition:

    • Treatment with PD0332991 (a selective CDK4/6 inhibitor) or expression of p16 prevents RB phosphorylation

    • Results in unphosphorylated RB similar to G0 arrest

    • Confirms that cyclin D:Cdk4/6 is the RB mono-phosphorylating kinase

  • S-phase arrest (thymidine block or aphidicolin):

    • Results in hyper-phosphorylated RB

    • All phospho-specific antibodies recognize hyper-phosphorylated RB from S phase arrested cells

    • Useful as a positive control for phospho-specific antibodies

To accurately assess S612 phosphorylation across the cell cycle, researchers should combine these synchronization methods with precise timing of sample collection.

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.