Phospho-RB1 (S608) Antibody

Shipped with Ice Packs
In Stock

Description

Antibody Characteristics

Type and Reactivity

  • Abcam ab172975: A rabbit recombinant monoclonal antibody targeting phosphorylated S608 of human RB1 .

  • Assay Genie CABP0570: A rabbit polyclonal antibody validated for Western blot (WB) and ELISA .

  • Cross-reactivity: Primarily human, with some commercial variants (e.g., Biocompare products) reacting with murine and rat samples .

Applications

  • Western blot (WB): Used to detect phosphorylated RB1 in lysates (e.g., Jurkat cells treated with FBS) .

  • Immunohistochemistry (IHC): Suitable for paraffin-embedded tissues (e.g., melanoma and retina) .

  • ELISA: Quantitative measurement of phosphorylation levels .

Cancer Implications

  • Dysregulation of RB1 phosphorylation is a hallmark of cancers, including retinoblastoma and melanoma .

  • The antibody aids in studying RB1's role in tumor suppression and resistance to targeted therapies (e.g., in ovarian cancer) .

Cell Cycle Dynamics

  • S608 phosphorylation occurs early in G1 phase, preceding S780 phosphorylation .

  • Knockdown of Pin1 reduces S780 phosphorylation, underscoring the dependency of RB1 activation on S608/S612 modifications .

Clinical and Research Applications

Cancer Biology
The antibody is instrumental in:

  • Investigating RB1's role in tumor progression .

  • Validating therapeutic targets in cancers with RB1 dysregulation .

Diagnostic Potential
While not approved for clinical use, the antibody enables quantitative assessment of RB1 phosphorylation in research settings, aiding biomarker discovery .

Product Specs

Buffer
Liquid in PBS containing 50% glycerol, 0.5% BSA and 0.02% sodium azide.
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery times.
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 acts as a key regulator of the G1/S transition during the cell cycle. In its hypophosphorylated form, 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 transactivating domain of E2Fs and recruits chromatin-modifying enzymes that actively repress transcription. Cyclin and CDK-dependent phosphorylation of RB1 leads to its dissociation from E2Fs, thereby activating the transcription of E2F responsive genes and triggering entry into the S phase. Additionally, 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 that of constitutive heterochromatin. It achieves this by stabilizing histone methylation. RB1 recruits and targets histone methyltransferases SUV39H1, KMT5B and KMT5C, resulting in epigenetic transcriptional repression. It controls histone H4 'Lys-20' trimethylation. RB1 inhibits the intrinsic kinase activity of TAF1. It 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, 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 the cell cycle progression was observed in SUMO1 knockdown HaCaT cells, and 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 and reveal the capacity of human cone precursors 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 squamous cell carcinoma (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 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, the present findings may provide a mechanism for the excessive proliferation of VSMCs under inflammation during vascular disorders and may identify novel targets for the treatment of vascular d... 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 with those in the normal group. PMID: 29115643
  14. Data indicate 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 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, 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 were 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 leukaemia 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 phosphorylation at serine 608?

Phosphorylation of RB1 at serine 608 is a key regulatory event in the cell cycle. The Retinoblastoma protein functions as a tumor suppressor and negative regulator of the cell cycle. When in its hypophosphorylated state, RB1 binds to E2F family transcription factors, preventing the transcription of E2F-responsive genes necessary for cell cycle progression. Phosphorylation at S608, which depends on CDK4, contributes to the inactivation of RB1's growth-suppressive function, allowing E2F release and subsequent entry into S phase . This phosphorylation event is particularly important in understanding the mechanisms of cancer development and progression, as dysregulation of this process can lead to uncontrolled cell proliferation .

How does S608 phosphorylation differ functionally from phosphorylation at other RB1 sites?

While RB1 contains multiple phosphorylation sites, S608 phosphorylation has distinct functional implications. Unlike phosphorylation at S807/811 which has been more strongly associated with RB1's role in apoptosis and interaction with Bax , S608 phosphorylation appears more specifically involved in cell cycle regulation through E2F binding. Research indicates that various phosphorylation sites work in concert, with S608 being part of a coordinated phosphorylation program. Experimental approaches comparing the effects of phosphorylation-site mutants have revealed that S608 phosphorylation contributes to conformational changes that affect pocket domain interactions with E2F transcription factors .

What are the optimal protocols for using Phospho-RB1 (S608) antibodies in Western blotting?

For optimal Western blotting results with Phospho-RB1 (S608) antibodies:

  • Sample preparation: Use fresh cell or tissue lysates extracted with phosphatase inhibitors to preserve phosphorylation status.

  • Dilution ratios: Most Phospho-RB1 (S608) antibodies work optimally at dilutions between 1:500-1:2000 .

  • Observed molecular weight: Expect to detect bands at approximately 110 kDa .

  • Positive controls: Jurkat cell lysates are recommended as positive controls .

  • Blocking: Use 5% BSA in TBST rather than milk (which contains phosphatases).

  • Detection system: An enhanced chemiluminescence system provides sensitive detection of phosphorylated epitopes.

For recombinant monoclonal antibodies like EPR10849, a dilution of 1:1000 is typically recommended , while polyclonal antibodies may require optimization within the 1:500-1:2000 range .

What are the technical considerations for immunohistochemistry applications with Phospho-RB1 (S608) antibodies?

When performing IHC with Phospho-RB1 (S608) antibodies:

  • Fixation: Formalin-fixed, paraffin-embedded (FFPE) tissues are commonly used.

  • Antigen retrieval: Most protocols require heat-induced epitope retrieval (HIER) with citrate buffer (pH 6.0) or EDTA buffer (pH 9.0).

  • Recommended dilutions: For IHC applications, dilutions typically range from 1:100-1:300 .

  • Detection systems: Both manual and automated immunostaining systems can be used, as demonstrated in studies utilizing Leica Bond Refine polymer detection systems .

  • Controls: Include phosphatase-treated sections as negative controls to verify phospho-specificity.

  • Subcellular localization: Expect predominantly nuclear staining, consistent with RB1's cellular localization .

How can researchers validate the specificity of Phospho-RB1 (S608) antibodies in their experimental systems?

To validate antibody specificity:

  • Phosphatase treatment: Treat half of your sample with lambda phosphatase to remove phosphorylation, which should eliminate signal from a truly phospho-specific antibody.

  • Knockout/knockdown controls: Use RB1-null cell lines or RB1-knockdown samples as negative controls.

  • Peptide competition assays: Pre-incubate the antibody with phosphorylated and non-phosphorylated peptides; only the phosphorylated peptide should block specific binding.

  • Immunoprecipitation validation: Perform IP followed by Western blot with another RB1 antibody.

  • Compare multiple phospho-specific antibodies: Use antibodies from different vendors that recognize the same phosphorylation site.

  • Induction experiments: Compare samples treated with agents known to induce or reduce S608 phosphorylation (e.g., CDK4/6 inhibitors should reduce signal).

What are the relative advantages of monoclonal versus polyclonal Phospho-RB1 (S608) antibodies?

FeatureMonoclonal AntibodiesPolyclonal Antibodies
SpecificityHigher specificity for the exact phospho-epitope May recognize multiple epitopes around the phosphorylation site
Lot-to-lot consistencyExcellent reproducibility between lotsPotential variation between lots
SensitivityMay have lower sensitivity in some applicationsOften higher sensitivity due to recognition of multiple epitopes
ApplicationsExcellent for Western blot and IHC Versatile; good for WB, IHC, ELISA, IP
BackgroundTypically lower backgroundMay have higher background in some applications
Host optionsCommonly rabbit monoclonals like EPR10849 Predominantly rabbit-derived
Research useIdeal for specific mechanistic studies requiring high reproducibilityBetter for detecting low-abundance proteins or preliminary screening

Selection should be based on the specific experimental requirements, with monoclonals like EPR10849 offering high reproducibility for mechanistic studies , while polyclonals might provide advantages in detection sensitivity .

How do different phospho-specific RB1 antibodies (S608, S807/811, S795) compare in research applications?

Different phospho-specific RB1 antibodies reveal distinct aspects of RB1 function:

  • S608 phosphorylation:

    • Primary role in cell cycle regulation and E2F interaction

    • Dependent on CDK4 activity

    • Commonly used in cell cycle studies and cancer research

    • Detected at 110 kDa by Western blot in human samples

  • S807/811 phosphorylation:

    • Stronger association with RB1's role in apoptosis

    • More readily detected in Bax interaction studies

    • Required for G0-G1 transition

    • Mediated by CDK3/cyclin-C

  • S795 phosphorylation:

    • Less strongly associated with Bax interaction than S807/811

    • Can be dephosphorylated by calcineurin upon calcium stimulation

    • Important in cell cycle control mechanisms

Experimental evidence shows that while all these phosphorylation events can be detected in immunoprecipitation studies with Bax, the S807/811 phosphorylation shows the strongest signal, followed by S608 and S795, which require longer exposure times for detection .

How can Phospho-RB1 (S608) antibodies be used to investigate cancer-specific alterations in the RB pathway?

Phospho-RB1 (S608) antibodies provide valuable tools for investigating cancer-specific RB pathway alterations:

  • Alternative mechanism detection: Studies have used phospho-S608 antibodies to identify tumors that inactivate the RB pathway through hyperphosphorylation rather than genetic mutation of RB1 .

  • Multi-parameter analysis: Combine phospho-S608 detection with other markers:

    • CDK4/6 expression levels

    • Cyclin D1 overexpression

    • p16INK4a status

    • E2F target gene expression

  • Tissue microarray analysis: Screen multiple tumor types to identify patterns of RB1 phosphorylation across cancer subtypes. This has been particularly informative in studies of breast, colon, prostate, kidney, and nasopharyngeal cancers .

  • Treatment response monitoring: Monitor changes in S608 phosphorylation following treatment with CDK inhibitors or other targeted therapies.

  • Correlation with clinical outcomes: Assess whether S608 phosphorylation status correlates with patient prognosis or treatment response.

Research has shown that phosphorylation of pRb at S608 can be detected in tumors without coding alterations in RB1, suggesting this as an alternative mechanism of RB pathway inactivation in cancer development .

What experimental designs are optimal for studying the relationship between CDK4 activity and RB1 S608 phosphorylation?

To study the relationship between CDK4 activity and RB1 S608 phosphorylation:

  • CDK4 inhibition studies:

    • Treat cells with selective CDK4/6 inhibitors (palbociclib, ribociclib, abemaciclib)

    • Monitor S608 phosphorylation over time using Western blot

    • Compare with other phosphorylation sites to determine specificity

  • Genetic manipulation:

    • Express wild-type, dominant-negative, or constitutively active CDK4 constructs

    • Generate CDK4 knockout or knockdown cell lines using CRISPR-Cas9 or RNAi

    • Assess changes in S608 phosphorylation status

  • Cell synchronization experiments:

    • Synchronize cells at different cell cycle phases

    • Analyze correlation between CDK4 activity and S608 phosphorylation throughout the cell cycle

    • Use double-labeling with cell cycle markers

  • In vitro kinase assays:

    • Purify CDK4/cyclin D complexes

    • Perform in vitro kinase reactions with recombinant RB1 protein or peptides

    • Use phospho-specific antibodies to detect site-specific phosphorylation

  • Phosphoproteomic analysis:

    • Perform mass spectrometry to quantitatively assess changes in multiple RB1 phosphorylation sites

    • Compare phosphorylation patterns after CDK4 manipulation

Research has established that phosphorylation of RB1 at S608 specifically depends on CDK4 activity, distinguishing it from other phosphorylation events mediated by different CDKs .

How do post-translational modifications interact with S608 phosphorylation to regulate RB1 function?

RB1 function is regulated by a complex interplay of post-translational modifications (PTMs):

  • Phosphorylation crosstalk:

    • S608 phosphorylation occurs in coordination with other phosphorylation events

    • The sequential phosphorylation model suggests certain sites must be phosphorylated before others

    • Evidence indicates CDK3/cyclin-C-mediated phosphorylation at S807/S811 is required for G0-G1 transition, while S608 phosphorylation by CDK4 regulates G1/S transition

  • Methylation-phosphorylation interaction:

    • Monomethylation at Lys-810 by SMYD2 enhances phosphorylation at Ser-807 and Ser-811

    • N-terminal methylation by METTL11A/NTM1 may influence phosphorylation patterns

    • Monomethylation at Lys-860 by SMYD2 promotes interaction with L3MBTL1

  • Acetylation effects:

    • RB1 undergoes acetylation during keratinocyte differentiation

    • Acetylation at Lys-873 and Lys-874 occurs during cellular stress and DNA damage

    • These events may modulate the effects of phosphorylation

  • Experimental approaches:

    • Use site-specific mutants (phosphomimetic or phospho-deficient)

    • Apply specific kinase or acetyltransferase inhibitors

    • Employ phosphatase treatments alongside other PTM-specific enzymes

    • Perform sequential ChIP experiments to determine occupancy of differently modified RB1

Research has shown that these diverse modifications create a complex "RB1 code" that determines its binding partners and functional outcomes in different cellular contexts .

What are common issues when using Phospho-RB1 (S608) antibodies and how can they be resolved?

IssuePotential CausesSolutions
No signalDegraded phospho-epitopeAdd phosphatase inhibitors to all buffers; keep samples cold
Ineffective antigen retrieval (IHC)Optimize buffer pH and heating conditions
Incorrect antibody dilutionTest a dilution series (1:500, 1:1000, 1:2000)
High backgroundInsufficient blockingIncrease blocking time; use 5% BSA instead of milk
Secondary antibody cross-reactivityTest alternative secondary antibodies
Non-specific bindingPre-absorb antibody with non-phosphorylated peptide
Multiple bandsDegradation productsUse fresh samples and protease inhibitors
Cross-reactivityValidate with phospho-null mutants or phosphatase treatment
Inconsistent resultsLot-to-lot variationSwitch to monoclonal antibodies for better consistency
Variable phosphorylation levelsStandardize cell culture conditions

For optimal specificity in detecting endogenous phosphorylation, consider using rabbit-derived antibodies which demonstrate excellent reactivity with human, rat, and monkey samples when used at recommended dilutions .

How should researchers design control experiments to validate Phospho-RB1 (S608) antibody results?

Comprehensive validation controls include:

  • Phosphatase controls:

    • Treat duplicate samples with lambda phosphatase

    • Compare phosphatase-treated vs. untreated samples by Western blot

    • Signal should disappear in phosphatase-treated samples

  • Genetic controls:

    • Use RB1-null cell lines (e.g., certain SCLC lines)

    • Compare with RB1-expressing cell lines

    • No signal should be detected in RB1-null cells

  • Stimulation/inhibition controls:

    • Serum-starve cells (reduces phosphorylation)

    • Treat with CDK4/6 inhibitors (reduces S608 phosphorylation)

    • Stimulate with growth factors (increases phosphorylation)

  • Peptide competition:

    • Pre-incubate antibody with phosphorylated peptide

    • Pre-incubate with non-phosphorylated peptide

    • Only phospho-peptide should block specific signal

  • Multiple antibody validation:

    • Compare results between different antibody clones/vendors

    • Use both monoclonal and polyclonal antibodies if possible

  • Positive control samples:

    • Include Jurkat cell lysates as recommended positive controls

    • Use cell lines known to have hyperphosphorylated RB1

These controls ensure that observed signals truly represent S608 phosphorylation rather than artifacts or non-specific binding.

How is Phospho-RB1 (S608) detection being incorporated into studies of chromatin modification and gene regulation?

Emerging research applications include:

These applications are revealing how dynamic phosphorylation of RB1 at S608 contributes to epigenetic regulation across different cellular contexts.

What is the significance of RB1 S608 phosphorylation in therapeutic response to CDK4/6 inhibitors?

The phosphorylation status of RB1 at S608 has significant implications for CDK4/6 inhibitor therapy:

  • Predictive biomarker potential:

    • S608 phosphorylation levels may predict sensitivity to CDK4/6 inhibitors

    • Antibody-based detection can be used to stratify patients for clinical trials

    • Monitoring changes in phosphorylation during treatment may provide early indicators of response

  • Resistance mechanism identification:

    • Persistent S608 phosphorylation despite CDK4/6 inhibition suggests alternative kinases are active

    • This pattern may identify tumors developing resistance to CDK4/6 inhibitors

  • Combination therapy rationale:

    • Understanding the relationship between S608 phosphorylation and other signaling pathways can guide rational combination therapies

    • Targeting multiple pathways affecting RB1 phosphorylation may overcome resistance

  • Experimental approaches:

    • Time-course analysis of S608 phosphorylation after CDK4/6 inhibitor treatment

    • Correlation of phosphorylation status with clinical response

    • Comparison with other phosphorylation sites to determine site-specific effects

  • Translation to clinical practice:

    • Development of IHC-based assays for clinical samples

    • Standardization of S608 phosphorylation assessment for patient selection

Research on S608 phosphorylation provides mechanistic insights into how CDK4/6 inhibitors exert their effects and may help identify patients most likely to benefit from these targeted therapies.

What criteria should researchers use when selecting between different commercial Phospho-RB1 (S608) antibodies?

When selecting Phospho-RB1 (S608) antibodies, consider:

  • Application-specific validation:

    • Verify the antibody has been validated for your specific application (WB, IHC, IF, ELISA)

    • Review published literature using the antibody for similar applications

    • Examine validation data showing specificity in relevant experimental contexts

  • Host species and format:

    • Rabbit-derived antibodies show good reactivity across human, rat, and monkey samples

    • Consider format (unconjugated vs. conjugated) based on detection method

    • Mouse monoclonals may be advantageous for co-labeling with rabbit antibodies

  • Clonality considerations:

    • Monoclonal antibodies (e.g., EPR10849, 51B7) offer high reproducibility

    • Polyclonal antibodies may provide higher sensitivity for low abundance targets

  • Epitope specificity:

    • Review immunogen information

    • Check sequence alignment with other RB1 phosphorylation sites

    • Consider antibodies raised against longer vs. shorter peptide sequences

  • Reactivity with species of interest:

    • Verify cross-reactivity with your experimental system

    • Note that some antibodies are specifically validated for human samples only

    • Others work across multiple species including human, mouse, and rat

  • Technical support and validation data:

    • Evaluate the quality and comprehensiveness of technical documentation

    • Look for actual experimental images rather than just statements of reactivity

    • Consider the manufacturer's reputation for antibody validation

Selecting the appropriate antibody based on these criteria ensures more reliable and reproducible experimental results.

How can phospho-specific flow cytometry be optimized for measuring RB1 S608 phosphorylation in heterogeneous cell populations?

Optimizing phospho-flow for RB1 S608 analysis:

  • Cell fixation and permeabilization:

    • Test multiple fixation protocols (paraformaldehyde, methanol)

    • Compare permeabilization agents (saponin, Triton X-100)

    • Optimize timing to preserve phospho-epitopes while allowing antibody access

  • Antibody selection and validation:

    • Test both monoclonal and polyclonal antibodies for flow performance

    • Verify specificity with phosphatase-treated controls

    • Titrate antibody concentration for optimal signal-to-noise ratio

  • Multi-parameter analysis:

    • Include cell cycle markers (DNA content, Ki-67)

    • Add surface markers to identify specific cell subpopulations

    • Consider multiplexing with other phospho-proteins (pCDK4, other pRB sites)

  • Controls and standardization:

    • Use phosphatase-treated cells as negative controls

    • Include cells with known high S608 phosphorylation as positive controls

    • Implement fluorescence minus one (FMO) controls for accurate gating

  • Data analysis considerations:

    • Analyze phosphorylation relative to cell cycle phase

    • Use appropriate statistical methods for heterogeneous populations

    • Consider phosphorylation index relative to total RB1 expression

This approach enables quantitative assessment of S608 phosphorylation across different cell subpopulations, providing insights into heterogeneity of RB pathway activation within complex samples.

What are promising new applications for Phospho-RB1 (S608) antibodies in cancer research and personalized medicine?

Emerging applications include:

  • Liquid biopsy development:

    • Detection of phosphorylated RB1 in circulating tumor cells

    • Correlation with disease progression and treatment response

    • Non-invasive monitoring of RB pathway activation

  • Single-cell analysis:

    • Integration with single-cell proteomics

    • Mapping heterogeneity of RB1 phosphorylation in tumors

    • Identifying rare cell populations with altered phosphorylation

  • Therapeutic response prediction:

    • Development of companion diagnostics for CDK4/6 inhibitors

    • Standardized IHC assays for clinical decision-making

    • Integration into multi-biomarker predictive panels

  • Targeted protein degradation:

    • Monitoring phosphorylation-dependent degradation of RB1

    • Assessing effects of novel targeted protein degradation therapeutics

    • Studying phosphorylation-dependent protein-protein interactions

  • Spatial biology:

    • Implementation in multiplexed tissue imaging platforms

    • Analysis of spatial relationships between phosphorylated RB1 and other markers

    • Tumor microenvironment effects on RB1 phosphorylation

These forward-looking applications could transform how we understand and target the RB pathway in cancer, moving beyond traditional research applications toward clinically actionable insights.

How might advances in antibody engineering improve the specificity and utility of Phospho-RB1 (S608) detection tools?

Future advances may include:

  • Recombinant antibody engineering:

    • Development of single-chain variable fragments (scFvs) for improved tissue penetration

    • Bispecific antibodies targeting both phospho-S608 and total RB1

    • Engineered antibodies with reduced background and increased specificity

  • Next-generation detection systems:

    • Proximity ligation assays for studying S608 phosphorylation in situ

    • FRET-based biosensors for live-cell imaging of phosphorylation dynamics

    • Nanobody development for super-resolution microscopy applications

  • Intracellular antibodies ("intrabodies"):

    • Development of cell-permeable antibodies for live-cell applications

    • Real-time monitoring of S608 phosphorylation dynamics

    • Targeted modulation of phosphorylated RB1 function

  • Multiparametric analysis tools:

    • Mass cytometry-compatible antibodies for high-dimensional analysis

    • Oligonucleotide-conjugated antibodies for spatial transcriptomics integration

    • Multiplexed imaging antibodies for contextual analysis of RB1 phosphorylation

  • Computational approaches:

    • Machine learning algorithms to improve phosphorylation pattern recognition

    • Integrative analysis of multiple phosphorylation sites

    • Predictive modeling of phosphorylation dynamics

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.