The UTP11 Antibody, HRP conjugated, combines a primary antibody targeting the UTP11 protein with horseradish peroxidase (HRP), an enzyme enabling chromogenic or chemiluminescent detection in assays. This conjugation eliminates the need for secondary antibodies, reducing cross-reactivity and simplifying workflows .
Target: UTP11, a nucleolar protein involved in 18S rRNA biosynthesis .
Detection Methods: ELISA, Western blotting, immunohistochemistry (IHC) .
HRP Function: Catalyzes reactions with substrates like diaminobenzidine (DAB) or TMB to produce visible signals .
Specificity: Monoclonal or polyclonal antibodies raised against UTP11 protein epitopes .
Conjugation Methods:
| Conjugation Method | Advantages | Time |
|---|---|---|
| Lightning-Link® HRP | Buffer-friendly, no purification needed | 2 hours |
| oYo-Link® HRP | Site-directed, hands-on time <1 minute | 2 hours |
Western Blotting: Detects UTP11 overexpression in hepatocellular carcinoma (HCC) samples, correlating with poor prognosis .
Immunofluorescence: Visualizes nucleolar localization in cancer cells .
Ribosome Biogenesis: UTP11 knockdown disrupts 18S rRNA processing, inducing nucleolar stress .
Ferroptosis: UTP11 depletion reduces SLC7A11 expression, enhancing oxidative damage in cancer cells .
Expression: TCGA and HPA databases confirm elevated UTP11 mRNA/protein levels in HCC .
Prognosis: High UTP11 correlates with reduced survival in HCC patients .
Mechanism: UTP11 stabilizes NRF2 mRNA, promoting antioxidant defense .
UTP11 (also known as UTP11L) is a protein involved in nucleolar processing of pre-18S ribosomal RNA. Recent studies have revealed that UTP11 plays an important role in cancer development through multiple mechanisms:
UTP11 is essential for the biosynthesis of 18S ribosomal RNAs by binding to the pre-rRNA processing factor MPP10
It is frequently overexpressed in human cancers and associated with poor prognoses
UTP11 deficiency inhibits cancer cell growth both in vitro and in vivo through p53-dependent and p53-independent mechanisms
Depletion of UTP11 impedes 18S rRNA biosynthesis, triggering nucleolar stress
It also represses SLC7A11 expression by promoting the decay of NRF2 mRNA, leading to enhanced ferroptosis
These findings position UTP11 as a potential oncogenic protein critical for cancer cell survival and a promising target for cancer research.
Horseradish peroxidase (HRP) conjugation serves as an enzymatic reporter system that enables highly sensitive detection of UTP11 protein. When conjugated to antibodies:
The HRP enzyme catalyzes the oxidation of substrates in the presence of hydrogen peroxide, producing a detectable signal
In chemiluminescent detection, HRP oxidizes luminol-based substrates, producing light that can be captured on film or by digital imaging systems
This enzymatic amplification significantly enhances sensitivity compared to direct labeling methods, allowing detection of low-abundance proteins like UTP11 in cancer research
The covalent linkage between antibody and HRP provides a stable conjugate that maintains both antibody specificity and enzymatic activity
This conjugation strategy is particularly valuable when studying proteins like UTP11 whose expression levels may vary significantly between normal and cancerous tissues.
UTP11 HRP-conjugated antibodies are versatile tools applicable across multiple research techniques:
Western blotting: Enables quantitative analysis of UTP11 expression in cancer cell lines and tissue lysates, with high sensitivity for detecting both overexpression and knockdown phenotypes
ELISA: Provides quantitative measurement of UTP11 in serum or cell lysates, useful for large-scale screening studies
Immunohistochemistry (IHC): Allows visualization of UTP11 localization and expression patterns in tissue sections, particularly valuable for analyzing cancer biopsies
Immunoprecipitation: When coupled with detection of binding partners like MPP10, helps elucidate UTP11's role in ribosome biogenesis
For nucleolar proteins like UTP11, immunocytochemistry with HRP-conjugated antibodies can also reveal subcellular localization patterns, confirming its nucleolar distribution and potential redistribution under stress conditions.
Direct HRP-conjugated primary antibodies provide several methodological advantages:
Reduced protocol time: Elimination of secondary antibody incubation and associated wash steps shortens experimental time by several hours
Increased specificity: Avoids potential cross-reactivity issues that can occur with secondary antibodies, particularly important when performing multi-protein analysis in complex cancer tissues
Consistent labeling ratio: Methods like site-directed conjugation ensure uniform labeling of 1-2 HRP molecules per antibody, providing reproducible signal generation
Simplified multiplexing: Enables simultaneous detection of multiple proteins without species cross-reactivity concerns
Lower background: Elimination of secondary antibody binding to endogenous immunoglobulins reduces non-specific background
These advantages make direct HRP-conjugated UTP11 antibodies particularly valuable for high-throughput cancer screening applications and complex co-localization studies.
Detecting low UTP11 expression requires methodological optimization:
Signal amplification systems:
Use enhanced chemiluminescent substrates specifically designed for low-abundance proteins
Consider tyramide signal amplification (TSA) which can increase sensitivity by 10-100 fold while maintaining specificity
Sample preparation:
Enrich for nucleolar fractions where UTP11 is concentrated
Use phosphatase inhibitors during extraction as post-translational modifications may affect antibody recognition
Antibody concentration optimization:
Perform titration experiments (1:500 to 1:5000) to determine optimal signal-to-noise ratio
Extended primary antibody incubation (overnight at 4°C) can improve detection of low-abundance targets
Background reduction strategies:
When analyzing normal tissues adjacent to tumors, these optimizations are crucial for establishing baseline UTP11 expression levels.
UTP11 deficiency triggers nucleolar stress through a complex molecular cascade that can be investigated using HRP-conjugated antibodies:
Ribosomal RNA processing disruption:
UTP11 binds to the pre-rRNA processing factor MPP10
Its depletion impedes 18S rRNA biosynthesis, disrupting ribosome assembly
HRP-conjugated antibodies against UTP11 and MPP10 can be used in co-immunoprecipitation studies to characterize this interaction
Ribosomal protein redistribution:
Nucleolar stress causes ribosomal proteins RPL5 and RPL11 to bind to MDM2
This prevents MDM2-mediated p53 ubiquitination and degradation
Multiplexed immunoblotting with HRP-conjugated antibodies can track these protein interactions
p53 stabilization and activation:
Activated p53 induces expression of growth arrest genes
HRP-conjugated antibodies against p53 and p21 can monitor this activation
Co-staining experiments can correlate UTP11 depletion with p53 accumulation
NRF2-SLC7A11 axis disruption:
These mechanistic studies benefit from the high sensitivity and specificity of HRP-conjugated antibodies, particularly in time-course experiments tracking the progression of nucleolar stress.
Rigorous validation of UTP11 HRP-conjugated antibodies should include:
Genetic validation approaches:
siRNA/shRNA knockdown: Compare signal in cells expressing UTP11 siRNA (5′-GAAGCTAAGAAAATCGAAA-3' and 5′-GGATGGAGTACATATTATT-3') versus control siRNA
CRISPR/Cas9 knockout: Generate UTP11 knockout cell lines as definitive negative controls
Overexpression: Transfect cells with Flag-tagged UTP11 expression constructs and confirm co-localization with the antibody signal
Peptide competition assays:
Pre-incubate antibody with recombinant UTP11 protein or immunogenic peptide
Valid antibodies will show reduced or eliminated signal after competition
Western blot validation:
Confirm detection of protein at expected molecular weight (~28 kDa)
Verify detection of UTP11 in known high-expressing cancers versus low-expressing normal tissues
Cross-reactivity assessment:
For UTP11 specifically, validation should include immunostaining to confirm nucleolar localization, consistent with its function in rRNA processing.
When encountering non-specific binding with UTP11 HRP-conjugated antibodies:
Optimize blocking conditions:
Use 5% BSA instead of milk for blocking phospho-epitopes
Add 0.1-0.5% Tween-20 to reduce hydrophobic interactions
Include 10% normal serum from the same species as the sample to block Fc receptors
Sample preparation refinements:
Extend fixation time for better epitope preservation
Perform antigen retrieval optimization (citrate buffer pH 6.0 vs. EDTA buffer pH 8.0)
Filter lysates to remove aggregates that cause non-specific binding
Antibody dilution and incubation adjustments:
Test higher dilutions (1:1000-1:5000) to reduce non-specific binding
Reduce incubation temperature from room temperature to 4°C
Add 0.1% non-homologous protein (like fish gelatin) to antibody diluent
Signal development modifications:
For nucleolar proteins like UTP11, additional high-salt washes (300-500 mM NaCl) can help reduce non-specific nucleic acid-mediated interactions.
Several conjugation methods are available for researchers preparing custom UTP11 HRP-conjugated antibodies:
Heterobifunctional crosslinker method:
Use Sulfo-SMCC to generate maleimide-activated HRP
Create sulfhydryl groups on UTP11 antibodies through SATA-mediated thiolation
The reaction forms stable thioether bonds between antibody and HRP
Advantages: Controlled reaction conditions, minimal antibody aggregation
Site-directed photocrosslinking:
Mix UTP11 antibody with photoactivatable HRP conjugate
Illuminate with 365nm light (6-10W) for site-specific conjugation to heavy chains
Results in uniform labeling of 1-2 HRP molecules per antibody
Advantages: Rapid protocol (under 2 hours), minimal hands-on time (30 seconds)
Rapid conjugation kits:
The choice of method depends on laboratory resources, expertise, and the specific application requirements for the UTP11 antibody conjugate.
Optimal buffer conditions for UTP11 HRP-conjugated antibodies include:
Storage buffer composition:
50% Glycerol in PBS (pH 7.4) provides cryoprotection
Addition of 0.01M PBS maintains physiological conditions
0.03% Proclin 300 prevents microbial contamination
1-5 mM EDTA chelates metal ions that could degrade the antibody
Working buffer considerations:
For Western blotting: TBS-T (20mM Tris, 150mM NaCl, 0.1% Tween-20, pH 7.6)
For ELISA: PBS-T (10mM phosphate, 150mM NaCl, 0.05% Tween-20, pH 7.4)
For IHC: TBS with 0.025% Triton X-100
Protein stabilizers:
1% BSA prevents adsorption to container surfaces
1-5 mM sodium azide inhibits microbial growth (not compatible with HRP activity assays)
0.1-1.0 mM PMSF protects against serine proteases
Temperature considerations:
For UTP11 specifically, antibody performance may be enhanced in buffers containing 1mM DTT to maintain reducing conditions that better expose the epitope.
A comprehensive control strategy for UTP11 detection should include:
Antibody controls:
Isotype control: IgG from same species as UTP11 antibody, HRP-conjugated
Secondary antibody only control (for indirect detection methods)
Peptide competition control: UTP11 antibody pre-incubated with immunizing peptide
Sample controls:
Positive control: Cell line with known UTP11 overexpression (e.g., hepatocellular carcinoma)
Negative control: UTP11 knockdown samples using validated siRNA (5′-GAAGCTAAGAAAATCGAAA-3')
Normal tissue control: Tissues with baseline UTP11 expression
Technique-specific controls:
For Western blotting: GAPDH or β-actin loading control
For IHC: Adjacent tissue sections with primary antibody omitted
For IP experiments: Non-specific IgG immunoprecipitation
Biological validation controls:
Given UTP11's role in nucleolar stress, comparing results under conditions that induce nucleolar stress (e.g., low-dose actinomycin D treatment) provides additional validation.
For comprehensive investigation of UTP11's oncogenic functions:
Expression analysis in cancer tissues:
HRP-IHC protocol:
Antigen retrieval: 10mM citrate buffer, pH 6.0, 95°C, 20 min
Antibody dilution: 1:200-1:500 in TBS + 1% BSA
Development: DAB substrate, monitor for 2-10 minutes
Counterstain: Hematoxylin for nuclei visualization
Nucleolar stress pathway analysis:
Co-immunoprecipitation protocol:
Lyse cells in buffer containing 20mM Tris-HCl, 150mM NaCl, 1% NP-40, 1mM EDTA, pH 7.4
Immunoprecipitate UTP11 with specific antibody
Probe for interactions with MPP10, RPL5, and RPL11
Detect with appropriate HRP-conjugated antibodies
p53-dependent growth arrest analysis:
Western blot protocol:
Harvest cells 48-72h after UTP11 siRNA transfection
Separate proteins on 10-12% SDS-PAGE
Transfer to PVDF membrane at 100V for 1h
Block with 5% BSA in TBS-T
Probe with HRP-conjugated antibodies against UTP11, p53, MDM2, and p21
Ferroptosis pathway analysis:
These optimized protocols leverage the sensitivity of HRP-conjugated antibodies while addressing the specific biological questions surrounding UTP11's role in cancer.
For optimal western blot detection of UTP11:
Sample preparation:
Lyse cells in RIPA buffer (50mM Tris pH 7.4, 150mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS)
Include protease inhibitors (PMSF, aprotinin, leupeptin)
Sonicate briefly to shear DNA and break nucleoli
Centrifuge at 14,000g for 15 minutes at 4°C to clear lysate
Gel electrophoresis and transfer:
Load 20-50μg protein per lane on 12% SDS-PAGE
Include molecular weight markers covering 20-30kDa range
Transfer to PVDF membrane at 100V for 60 minutes or 30V overnight
Antibody incubation:
Block membrane with 5% BSA in TBS-T for 1 hour at room temperature
Incubate with UTP11 HRP-conjugated antibody (1:1000 dilution) overnight at 4°C
Wash 4 times for 5 minutes each with TBS-T
Detection:
Apply chemiluminescent substrate and incubate for 1 minute
Expose to X-ray film or capture using digital imaging system
Expected band: ~28kDa for native UTP11
Controls to include:
Positive control: Hepatocellular carcinoma cell lysate
Negative control: UTP11 knockdown sample
For studying UTP11 interactions with MPP10 or ribosomal proteins, co-immunoprecipitation followed by western blotting provides valuable insights into complex formation.
To investigate UTP11's role in ferroptosis using HRP-conjugated antibodies:
Expression correlation analysis:
Perform western blots on cancer cell panels to correlate UTP11 expression with:
NRF2 protein levels
SLC7A11 expression
Glutathione (GSH) levels (measured biochemically)
Use HRP-conjugated antibodies for UTP11, NRF2, and SLC7A11 detection
UTP11 knockdown experiments:
Transfect cells with UTP11 siRNA (5′-GAAGCTAAGAAAATCGAAA-3')
At 48-72h post-transfection, analyze:
UTP11 protein levels by western blot
NRF2 mRNA stability using actinomycin D chase
SLC7A11 protein expression
Lipid peroxidation (using BODIPY-C11 staining)
Cell death (using annexin V/PI staining)
RNA immunoprecipitation (RIP) protocol:
Transfect cells with Flag-UTP11
Lyse in RIP buffer containing RNase inhibitors
Immunoprecipitate UTP11-RNA complexes with anti-Flag beads
Extract RNA and analyze NRF2 transcripts by RT-qPCR
Compare to input and IgG controls
Rescue experiments:
This approach combines protein detection with functional assays to establish the mechanistic link between UTP11 and ferroptosis in cancer cells.
For effective immunohistochemical detection of UTP11:
Tissue preparation:
Fix tissues in 10% neutral buffered formalin for 24-48 hours
Process and embed in paraffin
Section at 4-5μm thickness
Mount on positively charged slides
Antigen retrieval optimization:
Compare heat-induced epitope retrieval methods:
Citrate buffer (pH 6.0)
EDTA buffer (pH 8.0)
Tris-EDTA (pH 9.0)
Microwave or pressure cooker treatment for 15-20 minutes
Primary antibody protocol:
Quench endogenous peroxidase with 3% H₂O₂ for 10 minutes
Block with 5% normal serum in PBS for 1 hour
Apply UTP11 HRP-conjugated antibody at 1:50-1:200 dilution
Incubate overnight at 4°C in humidified chamber
Wash 3×5 minutes with PBS
Signal development:
Apply DAB substrate and monitor for 2-10 minutes
Counterstain with hematoxylin for 30-60 seconds
Dehydrate through graded alcohols and clear in xylene
Mount with permanent mounting medium
Special considerations for UTP11:
Include nucleolar markers (nucleolin or fibrillarin) on serial sections
Use low-dose actinomycin D-treated tissues as positive controls for nucleolar stress
Consider dual immunofluorescence with p53 to correlate UTP11 levels with p53 activation
These modifications optimize detection of UTP11's nucleolar localization and expression changes in cancer tissues.
To explore the UTP11-p53 regulatory axis:
Co-expression analysis in tissue panels:
Perform IHC on serial sections of cancer tissues for:
UTP11 (HRP-conjugated antibody)
p53 (HRP-conjugated antibody)
MDM2
p21
Quantify expression correlation using digital image analysis
UTP11 knockdown effects on p53 pathway:
Transfect cells with UTP11 siRNA
Harvest at multiple timepoints (24h, 48h, 72h)
Analyze by western blot for:
UTP11 depletion
p53 accumulation
MDM2 expression
p21 induction
Include both p53+/+ and p53-/- cell lines to distinguish p53-dependent effects
p53 ubiquitination assay:
Transfect p53-/- cells expressing shNC or shUTP11 with:
p53 expression plasmid
HA-MDM2 plasmid
His-Ub plasmid
Treat with proteasome inhibitor MG132 (4-6h)
Perform Ni-NTA pulldown of His-tagged proteins
Analyze ubiquitinated p53 by western blot
Ribosomal protein-MDM2 interaction:
This experimental design leverages the sensitivity of HRP-conjugated antibodies to detect both high-abundance (MDM2) and low-abundance proteins (ubiquitinated species) within the same pathway.
For comprehensive xenograft studies of UTP11 function:
Study design:
Animal model: 4-week-old female BALB/c nude mice
Cell lines:
CAL-51 cells stably expressing shNC or shUTP11 (6×10⁶ cells)
HCT116 p53+/+ and HCT116 p53-/- cells with shNC or shUTP11 (5×10⁶ cells)
Injection: Subcutaneous in right flank, suspended in DMEM with 50% Matrigel
Measurements: Tumor volume calculation using formula (length × width²) × 0.52
Duration: Monitor until control tumors reach ~1000mm³
Tissue analysis protocol:
Harvest and process tumors:
Fix one portion in 10% neutral buffered formalin for IHC
Snap-freeze another portion for protein/RNA extraction
Prepare single-cell suspensions for flow cytometry
Immunohistochemical analysis with HRP-conjugated antibodies:
UTP11 expression (verify knockdown efficiency)
p53 accumulation and activation
p21 expression (cell cycle arrest marker)
Ki-67 (proliferation marker)
Cleaved caspase-3 (apoptosis marker)
4-HNE staining (lipid peroxidation/ferroptosis marker)
Western blot analysis of tumor lysates:
UTP11, p53, p21, MDM2 protein levels
RPL5 and RPL11 expression
NRF2 and SLC7A11 levels
Correlation analysis:
This design allows evaluation of both p53-dependent and p53-independent mechanisms of UTP11 in tumor growth.
When facing discrepancies between UTP11 HRP-conjugated antibody results and other methods:
Epitope accessibility analysis:
Different antibodies may recognize distinct epitopes
The UTP11 epitope sequence (HIIKETKEEVTPEQLKLMRTQDVKYIEMKRVAEAKKIERLKSELHLLDFQGKQQNKHVFFFDTKKEVEQFDVATHLQTAPEL) may be differentially accessible
Test multiple antibody clones targeting different regions of UTP11
Compare native vs. denatured detection methods
Post-translational modification interference:
Some antibodies may be sensitive to phosphorylation, acetylation, or other modifications
Treat samples with phosphatases or deacetylases before analysis
Compare results from different cellular compartments (cytoplasmic vs. nuclear fractions)
Cross-reactivity assessment:
Test antibody specificity against recombinant UTP11
Perform immunoprecipitation followed by mass spectrometry
Compare results in UTP11 knockout/knockdown models
Technical validation approach:
Systematically vary protocol conditions:
Fixation methods (paraformaldehyde vs. methanol)
Detergent concentrations (0.1-1.0% Triton X-100)
Buffer compositions (phosphate vs. Tris-based)
Use orthogonal detection methods (fluorescence vs. HRP)
When properly validated, discrepancies often reveal biologically relevant information about protein conformation, interactions, or modifications in different contexts.
To dissect UTP11's dual roles in nucleolar stress and ferroptosis:
Genetic separation-of-function experiments:
Generate domain-specific UTP11 mutants:
N-terminal mutants that disrupt MPP10 binding
C-terminal mutants that affect mRNA binding
Rescue UTP11-depleted cells with these mutants
Assess nucleolar stress markers and ferroptosis independently
Pathway-specific inhibition:
Block p53 activation using pifithrin-α
Inhibit ferroptosis using ferrostatin-1 or liproxstatin-1
Supplement glutathione using N-acetylcysteine
Analyze growth inhibition and cell death in UTP11-depleted cells under each condition
Time-course experiments:
Monitor temporal sequence after UTP11 depletion:
0-24h: Nucleolar morphology changes and rRNA processing
24-48h: p53 stabilization and p21 induction
48-72h: NRF2 mRNA degradation and SLC7A11 reduction
72-96h: Lipid peroxidation and ferroptotic cell death
Selective pathway activation: