UTP18 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Constituents: 50% Glycerol, 0.01 M PBS, pH 7.4
Form
Liquid
Lead Time
Orders are typically dispatched within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
CGI 48 antibody; U3 small nucleolar RNA associated protein 18 homolog antibody; U3 small nucleolar RNA-associated protein 18 homolog antibody; UTP 18 antibody; UTP18 antibody; UTP18 small subunit (SSU) processome component homolog antibody; UTP18 small subunit processome component antibody; UTP18_HUMAN antibody; WD repeat containing protein 50 antibody; WD repeat domain 50 antibody; WD repeat-containing protein 50 antibody; WDR50 antibody
Target Names
UTP18
Uniprot No.

Target Background

Function

This antibody targets UTP18, a protein involved in the nucleolar processing of pre-18S ribosomal RNA.

Database Links

HGNC: 24274

OMIM: 612816

KEGG: hsa:51096

STRING: 9606.ENSP00000225298

UniGene: Hs.709327

Protein Families
WD repeat UTP18 family
Subcellular Location
Nucleus, nucleolus.

Q&A

What is UTP18 and why is it important in cellular research?

UTP18 (also known as WDR50, CDABP0061, CGI-48, or U3 small nucleolar RNA-associated protein 18 homolog) is part of the small subunit (SSU) processome, which serves as the first precursor of the small eukaryotic ribosomal subunit. During SSU processome assembly in the nucleolus, numerous ribosome biogenesis factors, RNA chaperones, and ribosomal proteins associate with nascent pre-rRNA to facilitate RNA folding, modifications, rearrangements, and cleavage, as well as targeted degradation of pre-ribosomal RNA by the RNA exosome. UTP18 specifically participates in nucleolar processing of pre-18S ribosomal RNA, making it a critical component in ribosome biogenesis pathways . Recent research has also implicated UTP18 in cancer progression, particularly in colorectal adenoma-carcinoma transition, highlighting its potential significance beyond basic ribosomal functions .

How does UTP18 antibody detection complement other molecular biology techniques?

UTP18 antibody detection provides unique advantages when investigating ribosome biogenesis and related cellular processes:

  • Spatial information: Unlike techniques such as RT-PCR or RNA-seq that merely quantify expression levels, immunofluorescence using FITC-conjugated UTP18 antibodies reveals subcellular localization, which is particularly valuable given UTP18's nucleocytoplasmic transport during certain cellular events like deSUMOylation .

  • Protein-protein interactions: UTP18 antibodies enable co-immunoprecipitation experiments to identify novel interaction partners within the SSU processome or other cellular complexes, providing insights into functional networks not achievable through genomic approaches.

  • Post-translational modifications: Antibodies can be used to detect specific modified states of UTP18, such as SUMOylation status, which has been shown to affect its nucleocytoplasmic transport and function in cell cycle progression .

  • Quantitative protein analysis: Western blotting with UTP18 antibodies complements transcriptomic data by confirming actual protein expression levels, which may not always correlate with mRNA abundance due to post-transcriptional regulation.

What is the molecular structure and cellular distribution of UTP18?

UTP18 is a 62 kDa protein characterized by the presence of WD repeat domains, which typically form β-propeller structures that facilitate protein-protein interactions . These structural features enable UTP18 to participate in complex formation with other processome components.

How can FITC-conjugated UTP18 antibodies be utilized to investigate the adenoma-carcinoma progression in colorectal cancer?

FITC-conjugated UTP18 antibodies offer valuable tools for investigating adenoma-carcinoma progression through several sophisticated approaches:

  • Fluorescence-based tissue analysis: FITC-conjugated UTP18 antibodies can be used to assess expression patterns across the adenoma-carcinoma continuum in tissue microarrays or patient-derived xenografts. Research has demonstrated that UTP18 is consistently upregulated during colorectal adenoma-to-carcinoma progression, making it a potential biomarker for disease advancement .

  • Organoid imaging: Patient-derived colorectal adenoma organoids can be visualized using FITC-conjugated UTP18 antibodies to monitor changes in expression and localization during experimental manipulations. Studies have shown that UTP18 overexpression promotes growth and ribosome biogenesis in adenoma organoids, suggesting a functional role in cancer progression .

  • Co-localization studies: FITC-conjugated UTP18 antibodies can be combined with antibodies against p21 or SUMOylation markers (using alternative fluorophores) to investigate the molecular mechanisms underlying UTP18's role in tumorigenesis. The deSUMOylation-induced nucleocytoplasmic transport of UTP18 has been linked to p21 mRNA instability, which drives cell cycle progression and malignant transformation .

  • Flow cytometry applications: Single-cell suspensions from adenoma and carcinoma tissues can be analyzed using FITC-conjugated UTP18 antibodies to quantify expression levels across different cell populations and correlate these with clinical outcomes or genetic signatures.

What methodological considerations are critical when designing experiments to investigate UTP18's role in ribosome biogenesis using fluorescence techniques?

When investigating UTP18's role in ribosome biogenesis using FITC-conjugated antibodies, researchers should consider:

  • Fixation protocols: UTP18's nucleolar localization necessitates optimal fixation methods to preserve nuclear architecture. Paraformaldehyde (4%) with controlled permeabilization using 0.1% Triton X-100 typically yields good results while maintaining epitope accessibility.

  • Co-staining strategies: To fully characterize UTP18's role in ribosome biogenesis, co-staining with nucleolar markers (fibrillarin, nucleolin) and other SSU processome components provides valuable context. When designing multi-color immunofluorescence panels, consider spectral overlap and employ appropriate controls.

  • Functional validation: Beyond observational studies, coupling fluorescence imaging with targeted disruption of UTP18 (via siRNA approaches similar to those described in the literature: 5′-CAGGAAGAACUAGCGGAUUtt-3′, 5′-GACGCCGGUUUCUCAUUAAtt-3′, or 5′-GAGAUUGAACGGAUCCACAtt-3′) can provide causal evidence for its role in pre-rRNA processing .

  • Ribosome biogenesis assays: Complement immunofluorescence with functional assays such as nucleolar run-on transcription or pre-rRNA processing analysis to correlate UTP18 localization patterns with actual ribosome synthesis rates.

Experimental ApproachKey ParametersExpected OutcomesControls Required
Immunofluorescence co-localization4% PFA fixation, 0.1% Triton X-100 permeabilizationNuclear/nucleolar UTP18 signal overlapping with other SSU componentsSecondary antibody only, peptide competition, siRNA knockdown
Live-cell imaging of UTP18 dynamicsTransfection with UTP18-GFP fusion constructDynamic movement between nucleolus and nucleoplasmNon-targeting control, photobleaching control
Flow cytometry quantificationSingle cell suspensions, 2% PFA fixationQuantitative expression differences between normal and cancer cellsIsotype control, unstained cells, compensation controls
BrdU incorporation with UTP18 staining16-hour BrdU pulse, dual staining with 7-AADCorrelation between UTP18 expression and S-phase entryBrdU-negative controls, cell cycle arrest controls

How can FITC-conjugated UTP18 antibodies be employed to investigate p53 pathway activation during nucleolar stress?

FITC-conjugated UTP18 antibodies provide valuable tools for investigating nucleolar stress-induced p53 activation:

  • Spatial reorganization analysis: During nucleolar stress, many nucleolar proteins exhibit altered localization patterns. FITC-conjugated UTP18 antibodies can track these changes in relation to p53 stabilization using confocal microscopy or structured illumination microscopy for enhanced resolution.

  • Stress response kinetics: Time-course experiments following induction of nucleolar stress (using actinomycin D, 5-FU, or other ribosome biogenesis inhibitors) can reveal the temporal relationship between UTP18 redistribution and p53 activation. Evidence suggests that disruption of human UTP18 can induce p53 activation, suggesting UTP18 may function as a nucleolar stress sensor .

  • Quantitative co-localization analysis: Advanced image analysis techniques can quantify the degree of co-localization between UTP18 and p53 pathway components during stress response, providing insights into molecular mechanisms.

  • Cell fate correlation: Combined with BrdU incorporation assays and 7-AAD staining for cell cycle analysis, UTP18 immunofluorescence can help determine how its expression levels correlate with cell cycle arrest and/or apoptosis following nucleolar stress .

What validation steps are essential when using a new lot of FITC-conjugated UTP18 antibody?

Thorough validation of each new FITC-conjugated UTP18 antibody lot is crucial for experimental reliability:

  • Specificity verification:

    • Western blot analysis using lysates from cells with known UTP18 expression (e.g., HeLa cells) should show a single band at the expected molecular weight of 62 kDa .

    • Peptide competition assays using the immunizing peptide (corresponding to amino acids 50-150 of human UTP18) should abolish specific staining .

    • siRNA knockdown controls using validated sequences should demonstrate reduced signal intensity proportional to knockdown efficiency .

  • Signal-to-noise optimization:

    • Titration experiments to determine optimal antibody concentration for maximum signal-to-background ratio.

    • Comparison of different fixation and permeabilization protocols to maximize epitope accessibility while preserving cellular architecture.

    • Evaluation of autofluorescence quenching methods if working with tissues known to have high background.

  • Functional validation:

    • Immunoprecipitation followed by mass spectrometry to confirm interaction with known UTP18 binding partners.

    • Correlation of staining patterns with biological responses in experimental models where UTP18 function is well-characterized (e.g., colorectal cancer models).

  • Reproducibility assessment:

    • Inter-lot comparison with previously validated antibodies using standardized samples and protocols.

    • Consistency checks across different experimental platforms (flow cytometry, microscopy, etc.).

What are the methodological differences when detecting UTP18 in cancer versus normal tissue samples?

When comparing cancer and normal tissues, several methodological adaptations are necessary:

  • Fixation considerations:

    • Cancer tissues often exhibit altered nuclear architecture requiring optimized fixation protocols to maintain UTP18 epitope accessibility.

    • Tumor samples may benefit from shorter fixation times (4-6 hours) compared to normal tissues to prevent epitope masking.

    • Antigen retrieval methods may need tissue-specific optimization, with citrate buffer (pH 6.0) typically yielding good results for UTP18 detection.

  • Signal interpretation challenges:

    • Cancer tissues often show heterogeneous UTP18 expression, necessitating quantitative analysis across multiple fields.

    • Increased background due to necrotic areas or infiltrating immune cells requires careful selection of control regions.

    • UTP18 upregulation in colorectal adenoma-carcinoma progression should be evaluated relative to matched normal tissue controls from the same patient when possible .

  • Analytical approaches:

    • For clinical samples, consider tissue microarray analysis to standardize staining conditions across multiple specimens.

    • Multiplex immunofluorescence combining UTP18 with cancer markers (Ki-67, p21) and cellular compartment markers provides contextual data for interpretation .

    • Image analysis algorithms should account for variations in cellular density and nuclear size between normal and cancer samples.

  • Validation strategies:

    • Correlation with orthogonal detection methods (RNAscope, RT-PCR) strengthens confidence in observed differences.

    • Inclusion of known positive control tissues (e.g., colorectal adenocarcinoma) with established UTP18 expression patterns .

How can cell cycle-dependent changes in UTP18 localization be accurately monitored using FITC-conjugated antibodies?

Monitoring cell cycle-dependent changes in UTP18 localization requires sophisticated experimental design:

  • Synchronization methods:

    • Double thymidine block for G1/S boundary arrest followed by release and time-course sampling.

    • Nocodazole treatment for M-phase enrichment.

    • Serum starvation for G0/G1 accumulation.

    • Each method should be validated to ensure minimal disruption to nucleolar structures.

  • Co-detection strategies:

    • Pair FITC-conjugated UTP18 antibodies with cell cycle markers in different fluorescence channels (e.g., cyclin B1 for G2/M, cyclin E for G1/S transition).

    • BrdU incorporation (16-hour pulse) with 7-AAD staining allows correlation of UTP18 expression with S-phase entry .

    • Consider using UTP18 staining in conjunction with FUCCI (fluorescent ubiquitination-based cell cycle indicator) systems for live-cell analysis.

  • Quantitative analysis:

    • Develop nucleolar-to-nucleoplasmic ratio measurements for UTP18 signal to track subcellular redistribution.

    • Single-cell analysis rather than population averages to account for cell cycle heterogeneity.

    • Time-lapse imaging with careful consideration of photobleaching and phototoxicity when using FITC conjugates.

  • Critical controls:

    • Parallel monitoring of known cell cycle-regulated nucleolar proteins (e.g., nucleophosmin) as comparative standards.

    • Validation of observed changes using cell cycle inhibitors to confirm specificity of effects.

How can researchers address nonspecific fluorescence when using FITC-conjugated UTP18 antibodies in tissue sections?

Nonspecific fluorescence is a common challenge when using FITC-conjugated antibodies in tissue sections. For UTP18 detection, consider these targeted approaches:

  • Optimized blocking protocols:

    • Extend blocking time (2-3 hours) using a combination of normal serum (5-10%) from the species of secondary antibody origin and BSA (3-5%).

    • Include 0.1-0.3% Triton X-100 in blocking solutions to reduce membrane-associated background .

    • Consider adding 0.1% glycine to quench aldehyde groups from fixation that may contribute to nonspecific binding.

  • Autofluorescence reduction:

    • Treat sections with 0.1-1% sodium borohydride (freshly prepared) for 10 minutes prior to blocking to reduce aldehyde-induced autofluorescence.

    • For tissues with high lipofuscin content (e.g., aged colorectal samples), incubate with 0.1% Sudan Black B in 70% ethanol for 20 minutes after antibody incubation.

    • Consider switching to longer wavelength fluorophores (e.g., Texas Red, Cy5) if FITC channel autofluorescence persists despite interventions.

  • Antibody optimization:

    • Titrate antibody concentration to determine the minimal effective concentration (typically 0.1-1 μg/mL based on comparable antibodies) .

    • Increase washing duration and volume (at least 3 × 15 minutes in PBS-T) following antibody incubation.

    • Consider overnight incubation at 4°C rather than shorter incubations at room temperature.

  • Controls and validation:

    • Include absorption controls using the specific immunizing peptide (amino acids 50-150 of human UTP18) .

    • Use tissue known to be negative for UTP18 expression as a background control.

    • Include secondary antibody-only controls to distinguish antibody-specific from conjugate-related background.

What analytical approaches can differentiate between nucleolar and nucleoplasmic UTP18 during deSUMOylation-induced transport?

Distinguishing between nucleolar and nucleoplasmic UTP18 localization requires sophisticated analytical approaches:

  • High-resolution imaging techniques:

    • Confocal microscopy with z-stack acquisition (0.3-0.5 μm steps) to precisely define nuclear compartments.

    • Deconvolution processing to enhance compartment boundary definition.

    • Super-resolution techniques (STED, SIM) for studying UTP18 distribution at subdiffraction resolution when more precise localization is needed.

  • Quantitative image analysis:

    • Nuclear compartment segmentation using DAPI (nuclear) and nucleolar markers (fibrillarin, nucleolin) to create masks.

    • Intensity correlation analysis between UTP18 and compartment markers before and after deSUMOylation induction.

    • Calculation of Manders' or Pearson's coefficients to quantify co-localization changes.

  • Biochemical fractionation complementation:

    • Parallel analysis using subcellular fractionation to isolate nucleolar, nucleoplasmic, and cytoplasmic fractions.

    • Western blot analysis of fractions to correlate with imaging data.

    • Consider ultracentrifugation at 200,000× g for 1 hour at 4°C to separate nucleolar components before affinity purification .

  • deSUMOylation monitoring:

    • Co-staining for SUMO1/2/3 to correlate UTP18 relocalization with SUMOylation status.

    • Use of SUMO-protease inhibitors to block deSUMOylation and observe effects on UTP18 distribution.

    • Time-course analysis following induction of deSUMOylation to track dynamic changes.

CompartmentMarker ProteinsUTP18 Expected Pattern (Normal)UTP18 Pattern (After deSUMOylation)
NucleolusFibrillarin, NucleolinStrong enrichment, punctate patternReduced signal, diffuse pattern
NucleoplasmSplicing speckles (SC35)Minimal signalIncreased signal intensity
CytoplasmCytoskeletal markersNegligible signalDetectable signal, particularly in perinuclear region
ChromatinH3K9me3, DAPI-dense regionsExcludedMinimal change, remains largely excluded

How can researchers accurately quantify changes in UTP18 expression in relation to p21 mRNA stability in experimental models?

To accurately quantify the relationship between UTP18 expression and p21 mRNA stability:

  • Integrated analytical approach:

    • Combine immunofluorescence quantification of UTP18 protein levels with RT-qPCR measurement of p21 mRNA in parallel samples.

    • Employ RNA stability assays using actinomycin D chase experiments followed by RT-qPCR at defined time points to calculate p21 mRNA half-life.

    • Use polysome profiling to assess p21 mRNA translation efficiency in relation to UTP18 expression levels.

  • Genetic manipulation strategies:

    • Implement graded UTP18 expression using doxycycline-inducible systems to establish dose-response relationships with p21 mRNA stability.

    • Utilize validated siRNA approaches (such as sequences 5′-CAGGAAGAACUAGCGGAUUtt-3′, 5′-GACGCCGGUUUCUCAUUAAtt-3′, or 5′-GAGAUUGAACGGAUCCACAtt-3′) for loss-of-function studies .

    • Create UTP18 domain mutants to identify regions specifically involved in p21 mRNA regulation.

  • Direct interaction assessment:

    • RNA immunoprecipitation (RIP) using UTP18 antibodies to detect direct binding to p21 mRNA.

    • In vitro binding assays with recombinant UTP18 and labeled p21 mRNA to characterize binding parameters.

    • Cross-linking immunoprecipitation (CLIP) to map precise UTP18 binding sites on p21 transcript.

  • Functional readouts:

    • Correlate changes in UTP18 and p21 levels with cell cycle progression using BrdU incorporation and 7-AAD staining .

    • Monitor colony formation capacity as a functional endpoint reflecting the biological significance of the UTP18-p21 axis.

    • Analyze growth curves of adenoma organoids with manipulated UTP18 expression to validate in vivo relevance .

What are the emerging applications of UTP18 as a biomarker in colorectal cancer diagnosis and treatment?

Recent studies identify UTP18 as a promising biomarker with several potential clinical applications:

  • Diagnostic and prognostic applications:

    • UTP18 upregulation correlates with adenoma-carcinoma progression, suggesting utility as a diagnostic marker for early malignant transformation .

    • Expression levels associate with adenoma recurrence and poor prognosis in colorectal cancer, offering prognostic value .

    • Development of automated image analysis algorithms for quantitative assessment of UTP18 immunofluorescence could standardize its use in clinical pathology.

  • Therapeutic targeting strategies:

    • UTP18's role in p21 mRNA destabilization suggests potential for developing inhibitors to restore p21 expression and cell cycle control.

    • The nucleocytoplasmic transport mechanism of UTP18 represents a druggable process that could be targeted by small molecules.

    • Combined targeting of UTP18 and ribosome biogenesis pathways may offer synergistic therapeutic effects.

  • Response prediction:

    • UTP18 expression levels may predict sensitivity to ribosome biogenesis inhibitors currently in clinical development.

    • Monitoring changes in UTP18 subcellular localization during treatment could provide early indicators of therapeutic response.

    • Liquid biopsy approaches detecting circulating UTP18 protein could enable non-invasive monitoring of treatment efficacy.

  • Methodological innovations:

    • Development of highly specific aptamer-based detection systems as alternatives to antibody-based methods.

    • Integration of UTP18 detection into multiplexed immunofluorescence panels for comprehensive tumor profiling.

    • Application of mass cytometry (CyTOF) for simultaneous assessment of UTP18 with dozens of other cancer-related markers at single-cell resolution.

How might the tetrameric complex of Pwp2, Utp6, Utp18, and Utp21 be studied using advanced fluorescence techniques?

The tetrameric complex comprising Pwp2, Utp6, Utp18, and Utp21 presents exciting opportunities for advanced fluorescence studies:

  • Multi-color FRET analysis:

    • Design FRET pairs using spectrally compatible fluorophores to examine proximity relationships within the complex.

    • Implement acceptor photobleaching FRET or fluorescence lifetime imaging microscopy (FLIM) for quantitative assessment of protein-protein interactions.

    • Create systematic deletion mutants to map interaction domains responsible for complex assembly.

  • Live-cell dynamics visualization:

    • Generate fluorescent protein fusions for each component to track assembly/disassembly kinetics in living cells.

    • Employ fluorescence recovery after photobleaching (FRAP) to measure residence times of individual components within the complex.

    • Use fluorescence correlation spectroscopy (FCS) to determine diffusion characteristics of intact complexes versus individual components.

  • Super-resolution approaches:

    • Apply structured illumination microscopy (SIM) or stimulated emission depletion (STED) microscopy to resolve spatial organization of the complex within nucleoli.

    • Implement single-molecule localization microscopy (PALM/STORM) for nanoscale mapping of component distribution.

    • Correlative light and electron microscopy (CLEM) to place fluorescence data in ultrastructural context.

  • Biochemical validation:

    • Recombinant reconstitution of the tetrameric complex with fluorescently labeled components to study assembly in vitro .

    • Single-molecule pull-down (SiMPull) assays to verify complex stoichiometry and assembly intermediates.

    • Native gel electrophoresis with fluorescent detection to resolve intact complexes and subcomplexes.

These advanced approaches collectively provide a comprehensive toolkit for dissecting the structure, dynamics, and function of this important macromolecular assembly in ribosome biogenesis.

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.