Rb1 Antibody Pair

Shipped with Ice Packs
In Stock

Product Specs

Buffer
**Capture Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
**Detection Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. Please contact your local distributors for specific delivery times.
Notes
We recommend using the capture antibody at a concentration of 0.3 µg/mL and the detection antibody at a concentration of 0.25 µg/mL. Optimal dilutions should be determined experimentally by the researcher.
Synonyms
pRb,Rb,pp105,Rb1,Rb-1
Target Names
Rb1

Q&A

What is Rb1 and why is it significant in cancer research?

Retinoblastoma protein 1 (Rb1) is a prototypical tumor suppressor that functions as a key regulator of the G1/S transition in the cell cycle. Rb1 was the first tumor suppressor gene to be discovered in 1971 . The protein works primarily by binding to and repressing E2F transcription factors, thereby inhibiting the expression of cell cycle progression genes .

Rb1 is significant in cancer research for several reasons:

  • It serves as a convergent point for multiple cellular pathways

  • Its dysfunction is observed in most types of cancer, either through genetic alterations or disruption of upstream regulators

  • It regulates crucial cellular processes beyond cell cycle control, including:

    • Differentiation and development

    • Maintenance of genome stability

    • Immune evasion mechanisms

The canonical mechanism involves the hypophosphorylated form of Rb1 binding to E2F transcription factors, which leads to:

  • Physical blocking of E2F's transactivating domain

  • Recruitment of chromatin-modifying enzymes that actively repress transcription

Upon phosphorylation by cyclin-dependent kinases (CDKs), Rb1 releases E2F factors, allowing transcription of genes required for S-phase entry .

How do Rb1 antibody pairs function in sandwich ELISA applications?

Rb1 antibody pairs function in sandwich ELISA through a coordinated system using two antibodies that recognize different epitopes on the Rb1 protein:

ComponentFunctionSpecifications
Capture AntibodyImmobilizes Rb1 from sampleUnconjugated, typically bound to plate surface
Detection AntibodyBinds to captured Rb1 and generates signalBiotin-conjugated for signal amplification

The process works through the following steps:

  • The capture antibody (unconjugated) is immobilized on the assay plate

  • Sample containing Rb1 protein is added, allowing the protein to bind to the capture antibody

  • The biotin-conjugated detection antibody is added, forming a sandwich complex

  • Streptavidin-HRP or a similar detection system binds to the biotin

  • Substrate addition produces quantifiable signal proportional to Rb1 concentration

In commercially available Rb1 antibody pairs, polyclonal antibodies derived from rabbit hosts show good reactivity across multiple species including rat, pig, dog, rabbit, and goat samples . The detection range varies by manufacturer but typically allows for sensitive measurement of Rb1 protein in complex biological samples.

What considerations are important when designing experiments to study Rb1-E2F interactions?

When designing experiments to study Rb1-E2F interactions, researchers should consider:

Structural and functional domains:

  • Central pocket domain (residues 379-792) of Rb1 is critical for binding E2F1

  • C-terminal domain (residues 792-926) provides additional structural support

  • E2F1 binding occurs through an 18-amino acid peptide within its transactivation domain (residues 409-426)

Experimental approaches:

  • Co-immunoprecipitation studies:

    • Use anti-His-tag antibodies when working with tagged constructs

    • SV40-LT co-immunoprecipitates with all three pocket proteins, while MCPyV-LT 278 selectively binds to Rb1

  • Phosphorylation status monitoring:

    • Measure phosphorylation at key sites (Thr-821, Thr-826) which promote interaction between C-terminal and Pocket domains

    • CDK3/cyclin-C-mediated phosphorylation at Ser-807 and Ser-811 is required for G0-G1 transition

  • Chromatin modifiers recruitment:

    • Design assays to detect HDAC1, HDAC2, HDAC3, HDAC5 recruitment through the LXCXE motif

    • Include analysis of SUV39H histone methylase recruitment which leads to H3K9 trimethylation

Controls to include:

  • Viral proteins (E7, E1A) that disrupt Rb1-E2F interactions through the LXCXE motif

  • Phosphorylation-mimicking mutations to simulate active/inactive states

  • Cell synchronization to analyze cell-cycle specific interactions

Data interpretation challenges:

  • Context-dependent effects (Rb1 can both induce and repress transcription depending on gene context)

  • E2F-dependent vs. E2F-independent functions of Rb1

How should researchers validate the specificity of Rb1 antibodies?

Validating specificity of Rb1 antibodies requires a multi-faceted approach:

Essential validation methods:

  • Genetic knockout controls:

    • Include Rb1-null cells (like the LoKe cell line, which lacks Rb1 expression)

    • Use CRISPR/Cas9-generated isogenic cell lines with varying Rb1 status (wildtype, heterozygous, homozygous knockout)

  • Immunoblot analysis:

    • Verify antibody detects bands of appropriate molecular weight (~110 kDa)

    • Compare expression in multiple cell lines with known Rb1 status

    • Test detection of post-translational modifications using phospho-specific antibodies

  • Immunohistochemistry validation:

    • Utilize tissue samples with known Rb1 expression (positive/negative controls)

    • Perform parallel staining with multiple antibody clones targeting different epitopes

    • Compare with examples like MCC patient samples showing heterogeneous Rb1 expression

  • Specificity for related pocket proteins:

    • Cross-reactivity testing against p107 (RBL1) and p130 (RBL2)

    • Differential expression analysis using qPCR as complementary evidence

Experimental indicators of high-quality antibodies:

  • Detection of endogenous Rb1 levels across various cell types

  • Ability to distinguish between phosphorylated and unphosphorylated forms

  • Consistent performance across multiple application methods (IHC, IF, ELISA)

  • Limited background and non-specific binding

For monoclonal antibodies, information about the specific clone (e.g., RB1/1754, 7E4B8) should be documented along with the immunogen used for antibody production .

How can researchers use Rb1 antibodies to investigate chromatin regulation mechanisms?

Rb1 plays critical roles in chromatin regulation that can be investigated using specialized antibody-based approaches:

Chromatin immunoprecipitation (ChIP) applications:

  • Rb1 antibodies can be used to identify genomic regions where Rb1 directly regulates transcription

  • ChIP followed by sequencing (ChIP-seq) reveals genome-wide binding patterns

  • ChIP data shows decreased H3K27me3 levels at the Rb1 promoter after Rb1-P-S loop disruption

Chromatin modifier complex analysis:

  • HDAC recruitment study:

    • Rb1 recruits HDAC1-5 to E2F target promoters

    • Co-immunoprecipitation with Rb1 antibodies can identify interacting HDACs in different cellular contexts

  • Histone methyltransferase interactions:

    • Rb1 recruits SUV39H1, KMT5B, and KMT5C, leading to epigenetic transcriptional repression

    • Assess H3K9 and H4K20 trimethylation status at target regions

  • Heterochromatin formation:

    • Rb1 directly participates in heterochromatin formation and stabilizes histone methylation

    • Use antibodies to track Rb1 localization during this process

Advanced techniques combining Rb1 antibodies:

  • Proximity ligation assay (PLA) to visualize Rb1 interactions with chromatin modifiers

  • CUT&RUN or CUT&Tag approaches for higher resolution chromatin binding profiles

  • Sequential ChIP (ChIP-reChIP) to identify genomic regions where Rb1 and specific modifiers co-occupy

Specific chromatin contexts to investigate:

  • Repetitive DNA sequences (endogenous retroviruses, LINE-1 elements) where Rb1 regulates silencing through H3K27 trimethylation

  • Centromere and telomere-proximal regions showing increased UV lesion susceptibility when Rb1 is deleted

  • Cancer-related genes like telomerase reverse transcriptase (TERT) located within Rb1-regulated genomic regions

What approaches can detect aberrant chromatin looping affecting Rb1 expression in non-mutation cases?

Recent research has revealed that Rb1 dysfunction can occur without genetic mutations through aberrant chromatin looping . To detect such cases, researchers can employ:

Chromosome conformation capture (3C) techniques:

  • 3C assay can examine chromatin interactions across the entire Rb1 locus

  • The Rb1 promoter region (site E5) can interact with a suppressor region, forming an Rb1-P-S loop

  • Quantitative analysis can reveal frequency of interactions between different chromatin regions

Epigenetic modification analysis:

  • ChIP assays focusing on repressive histone marks (H3K27me3) at the Rb1 promoter

  • Analysis of EZH2 binding to the Rb1 promoter, which correlates with repression

  • Monitoring CTCF binding, which is crucial for the formation of Rb1-P-S intrachromosomal looping

Functional validation approaches:

  • CRISPR-based deletion of the Rb1 suppressor region to disrupt the Rb1-P-S loop

  • Cell proliferation assays and in vivo orthotropic xenograft experiments to assess functional consequences

  • Verification through 3C assay that the Rb1-P-S loop is abolished after deletion

Experimental workflow:

  • Identify cell lines with minimal pRB expression but no Rb1 mutations (like RB44 and IM9)

  • Design 3C assays based on known protein binding peaks (CTCF, H3K27me3, H3K4me3)

  • Analyze chromatin interactions, particularly focusing on the Rb1 promoter

  • Manipulate identified interaction regions and assess functional outcomes

  • Confirm findings through multiple techniques including ChIP, RT-PCR, and in vivo models

Data from such studies showed that disruption of the Rb1-P-S loop resulted in ~50% decrease in tumor weight in xenograft models, demonstrating the biological significance of these chromatin interactions .

How should researchers interpret discrepancies in Rb1 detection between different antibody-based methods?

When researchers encounter discrepancies in Rb1 detection across different methods, several factors should be considered:

Common sources of discrepancy:

  • Post-translational modifications:

    • Rb1 undergoes extensive phosphorylation at multiple sites (Ser-567, Thr-821, Thr-826, Ser-795, Ser-807, Ser-811)

    • Methylation at Lys-810 and Lys-860 by SMYD2 affects cell cycle progression

    • Acetylation during keratinocyte differentiation can affect antibody recognition

  • Epitope accessibility:

    • Conformation changes between hypo- and hyperphosphorylated forms

    • Protein-protein interactions may mask epitopes in complex biological samples

    • Fixation methods for IHC may affect epitope availability

  • Antibody characteristics:

    • Clone-specific recognition patterns (monoclonal vs. polyclonal)

    • Host species and isotype differences

    • Detection method (direct vs. indirect)

Systematic approach to resolve discrepancies:

MethodAdvantageLimitationWhen to Trust Results
Western BlotSize verification, semi-quantitativeDenatured proteinsWhen band size matches expected MW (~110 kDa)
IHCSpatial context, cell-specific expressionFixation artifactsWhen controls show expected patterns
ELISAQuantitative, high sensitivityNo size verificationWhen standard curve is linear and samples fall within range
IFSubcellular localizationAutofluorescence issuesWhen specific nuclear localization is observed

Best practices for reliable interpretation:

  • Use multiple antibodies targeting different epitopes

  • Include genetic controls (Rb1 null cells like LoKe)

  • Complement protein detection with mRNA analysis (qPCR)

  • Consider cell cycle phase and synchronization status

  • Document phosphorylation-dependent recognition patterns

When evaluating heterogeneous samples (like tumors), be aware that Rb1 expression can vary within the same sample, as demonstrated in MCC patient samples where some areas show Rb1 expression while others lack it entirely .

What are the consequences of Rb1 loss in cells that already have RB pathway disruption?

Research has revealed that Rb1 loss in cells with pre-existing RB pathway disruption leads to additional consequences beyond proliferative control:

DNA damage and genomic instability:

  • Even heterozygous Rb1 mutations (Rb1+/-) show increased basal levels of DNA damage

  • Elevated levels of γH2AX foci indicate spontaneous DNA damage in Rb1 mutant cells

  • Increased mitotic errors, particularly anaphase bridges, are observed

Molecular mechanisms involved:

  • Reactive oxygen species (ROS):

    • Rb1 mutant cells exhibit elevated levels of ROS, contributing to DNA damage

    • Increased sensitivity to H₂O₂ compared to wild-type cells

  • DNA repair deficiencies:

    • Impaired homologous recombination repair in Rb1 mutant cells

    • Decreased ability to repair DNA cross-links (higher sensitivity to cisplatin)

    • Lack of 53BP1 foci (a marker of non-homologous end joining) suggests altered repair pathway choice

Cancer progression implications:

  • Rb1 mutant cells show increased propensity to seed new tumors in recipient lungs in xenograft models

  • The distribution of DNA damage within common fragile sites (CFS) is altered in Rb1 mutants

Experimental approach to study these effects:

  • Generate isogenic Rb1 mutant cell lines using CRISPR/Cas9

  • Compare wildtype, heterozygous, and homozygous null genotypes

  • Assess DNA damage markers (γH2AX) and repair pathway components

  • Test chemical sensitivities to DNA-damaging agents

  • Perform in vivo xenograft experiments to evaluate metastatic potential

These findings suggest that late-arising Rb1 mutations can facilitate genome instability and cancer progression even in cells that already have RB pathway defects through other mutations .

How can researchers differentiate between the functions of Rb1 and other pocket proteins when using antibodies?

Differentiating between Rb1 and related pocket proteins (p107/RBL1 and p130/RBL2) requires careful experimental design:

Structural and functional distinctions:

Antibody-based differentiation approaches:

  • Western blot analysis:

    • Use antibodies specific to each PP (anti-Rb1, anti-p107, anti-p130)

    • Discriminate based on molecular weight differences

    • Account for different expression levels (p107 and p130 mRNA levels are typically higher than Rb1)

  • Functional differentiation:

    • Cell cycle arrest and E2F target gene repression mediated by Rb1 can be reverted by MCPyV-LT expression, unlike other PPs

    • Monitor expression of cell cycle related Rb1 target genes (CCNB1, MYB, PLK1, CDC6) after targeted knockdown

  • Experimental validation:

    • Use shRNA targeting specific PPs to analyze their individual roles

    • Combine PP-specific shRNAs with T antigen knockdown to evaluate functional differences

    • Perform cell cycle analyses to assess the impact on S and G2/M phases

Practical experimental approach:

TechniquePurposeExpected Outcome
Co-immunoprecipitationDetermine binding specificityMCPyV-LT 278 selectively binds Rb1 but not p107 or p130
Knockdown experimentsAssess functional redundancyOnly Rb1 knockdown reverses growth arrest in certain contexts
qRT-PCRMeasure expression levelsDifferent baseline expression patterns for the three PPs
ChIPIdentify genomic targetsUnique and overlapping binding sites for each PP

Key experimental finding:
In MKL-1 and WaGa cells, T antigen (TA) shRNA-induced reduction of cells in S and G2/M phase could be significantly reversed by additional knockdown of Rb1, demonstrating its dominant role compared to other pocket proteins .

What novel approaches are emerging for studying Rb1 functions in genome stability?

Recent research has revealed Rb1's involvement in genome stability beyond its classical cell cycle regulation role, suggesting several promising research directions:

UV susceptibility and carcinogen sensitivity:

  • Rb1 regulates UV susceptibility across broad genomic regions

  • Centromere and telomere-proximal regions show significant increases in UV lesion susceptibility when Rb1 is deleted

  • Cancer-related genes like TERT are located within these susceptible regions

Non-mutational Rb1 dysfunction mechanisms:

  • Aberrant chromatin looping (Rb1-P-S loop) can silence Rb1 expression without genetic mutations

  • CTCF-mediated chromatin interactions regulate Rb1 activity

  • Targeting these epigenetic mechanisms could restore Rb1 function in certain cancers

Emerging experimental approaches:

  • Genome-wide mapping techniques:

    • Combine UV damage detection with high-throughput sequencing

    • Map carcinogen susceptibility across the genome in Rb1-proficient vs. deficient cells

    • Correlate with chromatin structures and histone modifications

  • Chromosome conformation technologies:

    • 3C-based methods to understand long-range chromatin interactions affecting Rb1

    • CUT&Tag or CUT&RUN approaches for higher resolution chromatin binding profiles

    • Single-cell chromatin conformation analysis to address cellular heterogeneity

  • Therapeutic targeting strategies:

    • Develop approaches to disrupt aberrant chromatin loops silencing Rb1

    • Target synthetic lethal interactions in Rb1-deficient cells

    • Explore genome instability as a vulnerability in Rb1-mutant cancers

Interdisciplinary integration:

  • Combine structural biology with genomics to understand how Rb1 domains contribute to genome stability

  • Utilize computational approaches to predict susceptible regions and potential interventions

  • Develop patient-derived models to translate findings into clinical applications

These approaches could lead to novel therapeutic strategies for cancers with Rb1 dysfunction, focusing on restoring genomic stability rather than directly targeting cell cycle control mechanisms.

How can Rb1 antibodies be utilized to study post-translational modifications in different disease contexts?

Rb1 undergoes extensive post-translational modifications (PTMs) that regulate its function in health and disease. Advanced antibody-based approaches to study these modifications include:

Types of Rb1 PTMs to investigate:

  • Phosphorylation:

    • CDK4/6 and CDK2-mediated phosphorylation at multiple sites (Ser-567, Thr-821, Thr-826)

    • CDK3/cyclin-C-mediated phosphorylation at Ser-807 and Ser-811 required for G0-G1 transition

    • Dephosphorylation by calcineurin at Ser-795 upon calcium stimulation

  • Methylation:

    • N-terminus methylation by METTL11A/NTM1

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

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

  • Acetylation:

    • Occurs during keratinocyte differentiation

    • Affects protein-protein interactions and function

Methodological approaches:

TechniqueApplicationAdvantage
Phospho-specific antibodiesDetect specific phosphorylation sitesMonitor cell cycle-dependent changes
Mass spectrometry with IPComprehensive PTM mappingIdentifies novel modifications
Proximity ligation assayVisualize PTM-dependent interactionsSingle-cell resolution
FRET-based biosensorsReal-time PTM dynamicsLive cell applications

Disease-specific applications:

  • Cancer context:

    • Compare PTM patterns between tumor and normal tissues

    • Assess how viral oncoproteins (SV40 large T, HPV E7, adenovirus E1A) affect Rb1 PTMs

    • Evaluate PTM changes during treatment response

  • Developmental disorders:

    • Study how Rb1 PTMs influence differentiation processes

    • Investigate PTM disruptions in developmental pathologies

  • Tissue-specific regulation:

    • Compare Rb1 PTM patterns across different tissues

    • Correlate with tissue-specific transcription factors (MyoD, Runx2, C/EBP)

Emerging analytical approaches:

  • Single-cell proteomics to address cellular heterogeneity

  • PTM-specific ChIP-seq to map genomic targets of differently modified Rb1

  • Computational modeling to predict how PTM patterns affect protein function

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.