UMPS Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Introduction to UMPS Antibody, Biotin Conjugated

UMPS Antibody, Biotin Conjugated, is a specialized immunological reagent combining the specificity of an anti-UMPS antibody with biotin, a small molecule enabling high-affinity binding to avidin or streptavidin. UMPS (uridine monophosphate synthetase) catalyzes the conversion of orotic acid to uridine 5′ monophosphate, a critical step in pyrimidine biosynthesis. Biotin conjugation enhances the antibody’s utility in assays requiring signal amplification, such as immunoprecipitation (IP), immunohistochemistry (IHC), and enzyme-linked immunosorbent assays (ELISAs).

Application-Specific Dilutions

ApplicationRecommended Dilution
Western Blot (WB)1:500–1:2000
Immunohistochemistry (IHC)1:20–1:200
Immunoprecipitation (IP)0.5–4.0 µg per 1–3 mg lysate

Mechanism of Biotin Conjugation

Biotin is covalently attached to the antibody using NHS-ester chemistry, forming an amide bond with lysine residues. The Biotin Labeling Kit (e.g., from The Native Antigen Company) provides a streamlined protocol:

  1. Reaction: Antibody + Biotin-NHS ester → Biotinylated antibody.

  2. Purification: Optional (kit claims high efficiency without purification) .

  3. Storage: Stable at -20°C in PBS with sodium azide .

Advantages Over Unconjugated Antibodies

FeatureBiotin-Conjugated UMPS AntibodyUnconjugated UMPS Antibody
Detection SensitivityEnhanced via streptavidin-enzyme/fluorophore conjugates Limited to direct enzyme labeling
Multiplexing CapacityEnables sequential labeling (e.g., streptavidin-fluorophore + anti-biotin)Restricted to single detection systems
Signal AmplificationBiotin-SP (spacer) improves accessibility for streptavidin binding No amplification

Targeted Assays

ApplicationUse CaseExample Protocol
WB with Streptavidin-HRPDetection of UMPS in lysates (e.g., HeLa, Jurkat cells) 1:1000 dilution + streptavidin-HRP
IHC with Streptavidin-FluorophoreLocalization in tissues (e.g., colon cancer) Antigen retrieval (pH 9.0 TE buffer)
IP with Biotin-Streptavidin BeadsIsolation of UMPS complexes for mass spectrometry 2 µg antibody + streptavidin beads

Research Findings

  1. Cancer Metabolism: UMPS expression correlates with pyrimidine synthesis in pancreatic cancer, where biotinylated antibodies may enhance detection of therapeutic targets .

  2. Drug Delivery: Biotin-conjugated antibodies could enable targeted delivery via biotin receptors, though SMVT-mediated uptake requires free carboxylic acids .

  3. Live Cell Imaging: Site-specific biotinylation (via BirA ligase) and monovalent streptavidin binding enable precise localization without cross-linking .

Technical Limitations

ChallengeMitigation Strategy
Biotin InterferenceUse biotin-free buffers during conjugation .
Non-Specific BindingBlock with BSA or casein; optimize antibody dilution .
Spacer LengthBiotin-SP (6-atom spacer) improves streptavidin accessibility .

Quality Control

ParameterSpecification
Conjugation Efficiency>90% biotin incorporation (kit-dependent)
PurityAntigen affinity-purified
Stability12-month shelf life at -20°C

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchase method and location. For specific delivery times, please consult your local distributor.
Synonyms
ODC antibody; OMPdecase antibody; OPRT antibody; OPRTase antibody; Orotate phosphoribosyl transferase and orotidine 5' decarboxylase antibody; Orotate phosphoribosyltransferase antibody; Orotate phosphoribosyltransferase phosphoribosyltransferase antibody; Orotidine 5' phosphate decarboxylase antibody; Orotidine 5''-phosphate decarboxylase antibody; RP11-71H17.9 antibody; UMP synthase antibody; Umps antibody; UMPS_HUMAN antibody; Uridine 5' monophosphate synthase antibody; Uridine monophosphate synthetase antibody
Target Names
UMPS
Uniprot No.

Target Background

Gene References Into Functions
  1. Partial UMPS deficiency should be considered in the differential diagnosis of mild orotic aciduria. PMID: 28205048
  2. The UMPS 638 CC genotype may be a potential biomarker for predicting toxicity in patients undergoing preoperative chemoradiation with tegafur-uracil/leucovorin for locally advanced rectal cancer. PMID: 28347333
  3. OPRT transition state analogues are crucial components of potent inhibitors targeting OPRT enzymes. PMID: 24158442
  4. The observed mutations, aberrant splicing, and downregulation of UMPS represent novel mechanisms for acquired 5-FU resistance in colorectal cancer. PMID: 22249354
  5. [review] It is established that the type I defect in hereditary orotic aciduria is caused by a loss of uridine monophosphate (UMP) synthase activity. PMID: 19562503
  6. OPRT expression in colorectal carcinoma tissues does not correlate with the toxicity of 5-FU, but OPRT expression in normal tissues can help predict the toxicity associated with 5-FU. PMID: 22931617
  7. While no association was detected between UMPS variants and gastrointestinal cancer risk in Caucasians, polymerase chain reaction-RFLP with BsrI digestion and DHPLC set up at 59 degrees C are reliable and cost-effective methods for genotyping UMPS. PMID: 21631301
  8. Novel phosphoribosyltransferase transition states. PMID: 20527751
  9. The orotate phosphoribosyl transferase/DPD ratio has a correlation with cancer staging and survival rate. PMID: 20647710
  10. Increased orotate phosphoribosyl transferase expression is associated with bladder cancers. PMID: 20112501
  11. The expression of the OPRT gene and the OPRT/dihydropyrimidine dehydrogenase ratio may be useful as predictive parameters for the efficacy of fluoropyrimidine-based chemotherapy for metastatic colorectal cancer. PMID: 14562021
  12. Thymidylate synthase and orotate phosphoribosyl transferase, but not dihydropyrimidine dehydrogenase, are more highly expressed in prostate cancer than in benign prostatic hyperplasia. PMID: 15999119
  13. Orotate phosphoribosyl transferase plays a role in lymph node metastasis of gastric cancer. PMID: 16142362
  14. The investigated SNPs of OPRT may not have a significant influence on 5-FU sensitivity. PMID: 16328050
  15. The OPRT Gly213Ala polymorphism appears to be a useful marker for predicting toxicity to bolus 5-FU chemotherapy. PMID: 16818689
  16. Overexpression of the OPRT gene plays a significant role in the antiproliferative effect of 5-FU. PMID: 17237621
  17. High expression of OPRT is associated with response to adjuvant chemotherapy in human pancreatic cancer. PMID: 17549346
  18. Data suggest that OPRT is involved in early events of pancreatic and gallbladder carcinogenesis and invasion of hepatocellular carcinomas. PMID: 17607371
  19. Decreased sensitivities of the cultured tumor cells to 5-FU were observed. These results suggest that the OPRT expression level in tumors is an additional determinant of the efficacy of 5-FU. PMID: 17854773
  20. OPRT activity levels in tumor tissue may be a prognostic factor for survival in colorectal carcinoma with radical resection and 5-FU chemotherapy; postoperative survival was significantly better in patients with high OPRT activity. PMID: 18597678
  21. Determination of OPRT levels in gastric carcinoma tissue allows for predicting the response to S-1-based neoadjuvant/adjuvant chemotherapy. PMID: 18633253
  22. Orotate phosphoribosyl transferase is involved in the invasion and metastasis of colorectal carcinoma. PMID: 18949394
  23. Decreased activity of OPRT plays a significant role in the acquired resistance of gastric cancer cells towards 5-FU. PMID: 19020740
  24. Head & neck, gastric, colorectal, breast, lung & pancreatic cancer were examined; findings show mRNA expression & protein level of thymidylate synthase, dihydropyrimidine dehydrogenase & orotate phosphoribosyl transferase differed according to cancer type. PMID: 19020767
  25. Immunohistochemical staining for Orotate phosphoribosyl transferase (OPRT) revealed strong expression of OPRT in prostate cancer cells. There was a significant correlation between OPRT mRNA expression levels and the tumor pathological grade. PMID: 19082440
  26. High orotate phosphoribosyl transferase gene expression is associated with complete response to chemoradiotherapy in patients with squamous cell carcinoma of the esophagus. PMID: 19307741

Show More

Hide All

Database Links

HGNC: 12563

OMIM: 258900

KEGG: hsa:7372

STRING: 9606.ENSP00000232607

UniGene: Hs.2057

Involvement In Disease
Orotic aciduria 1 (ORAC1)
Protein Families
Purine/pyrimidine phosphoribosyltransferase family; OMP decarboxylase family

Q&A

What is UMPS and why is it significant in biochemical research?

UMPS (Uridine 5'-monophosphate synthase) is a bifunctional enzyme that catalyzes the final two steps of the de novo pyrimidine biosynthetic pathway. The N-terminal enzyme orotate phosphoribosyltransferase (OPRT) converts orotic acid to orotidine-5'-monophosphate, while the C-terminal enzyme OMP decarboxylase (OMPdecase) converts orotidine-5'-monophosphate to uridine monophosphate . This cytoplasmic protein plays a critical role in nucleotide metabolism, which is fundamental to DNA/RNA synthesis and cellular proliferation. Defects in the UMPS gene cause hereditary orotic aciduria, making it relevant for both basic metabolism research and clinical investigations. The enzyme's position at a critical junction in pyrimidine synthesis makes it an important target for studying metabolic regulation, cancer biology, and drug response mechanisms.

What applications is UMPS Antibody, Biotin conjugated best suited for?

UMPS Antibody, Biotin conjugated demonstrates versatility across multiple research applications, particularly Western blotting (WB), Enzyme-Linked Immunosorbent Assays (ELISA), and immunohistochemistry (both paraffin-embedded and frozen sections) . For Western blotting, the antibody performs optimally at dilutions ranging from 1:300-5000, detecting UMPS protein in cell and tissue lysates. In ELISA applications, the recommended dilution range of 1:500-1000 provides sensitive detection for quantitative analysis . The antibody excels in immunohistochemical applications (dilutions 1:200-400 for IHC-P and 1:100-500 for IHC-F) where visualization of UMPS distribution in tissues can provide insights into metabolic patterns and disease-related alterations . The antibody's reactivity with human and non-human primate samples, plus predicted cross-reactivity with multiple mammalian species, makes it valuable for comparative studies across experimental models. Research requiring sensitive detection of cytoplasmic UMPS protein in contexts like cancer metabolism, pyrimidine synthesis disorders, and drug response studies benefit significantly from this tool.

How does the structure-function relationship of UMPS influence antibody selection and experimental design?

UMPS functions as a bifunctional enzyme with distinct N-terminal OPRT and C-terminal OMPdecase domains within its 480-amino acid structure . This structural organization necessitates careful consideration when designing experiments. The specific UMPS polyclonal antibody (derived from the immunogen range 351-450/480) targets a region that may have different accessibility depending on protein conformation or post-translational modifications . When investigating specific enzyme activities, researchers should consider whether the antibody epitope might interfere with or be affected by substrate binding regions. For mechanistic studies examining either OPRT or OMPdecase functions individually, researchers may need complementary approaches beyond immunodetection. The cytoplasmic localization of UMPS informs sample preparation strategies—particularly for subcellular fractionation experiments or imaging studies where preserving native distribution is critical. Understanding the quaternary structure of UMPS in different cellular contexts may also influence experimental interpretation, as complex formation with other metabolic enzymes could potentially mask epitopes or alter apparent molecular weight in certain analytical techniques.

How should researchers validate UMPS Antibody, Biotin conjugated specificity for their experimental system?

Comprehensive validation of UMPS Antibody, Biotin conjugated requires a multi-faceted approach to ensure experimental reliability. Begin with positive and negative control samples—cell lines or tissues with well-characterized UMPS expression levels serve as critical reference points. Genetic manipulation approaches provide powerful validation tools; implementing CRISPR-Cas9 knockout or siRNA-mediated knockdown of UMPS followed by antibody testing should demonstrate corresponding signal reduction . Peptide competition assays offer another validation dimension; pre-incubating the antibody with excess immunizing peptide should effectively eliminate specific signals while non-specific binding persists. For cross-species applications, particularly with species listed under "Predicted Reactivity" rather than confirmed "Reactivity," pilot testing is essential before full-scale experiments . Western blotting validation should confirm a primary band at the expected molecular weight (~50 kDa for human UMPS), while immunofluorescence or immunohistochemistry should demonstrate the anticipated cytoplasmic localization pattern. Orthogonal validation using alternative detection methods like mass spectrometry or qPCR correlation with protein levels provides additional confidence. Document all validation steps methodically, as these strengthen the credibility of subsequent research and facilitate troubleshooting if unexpected results emerge.

What sample preparation considerations are critical for different applications using UMPS Antibody, Biotin conjugated?

Sample preparation protocols significantly impact UMPS Antibody, Biotin conjugated performance across different applications. For Western blotting, extract proteins using RIPA buffer supplemented with fresh protease inhibitors to prevent UMPS degradation. When preparing samples for SDS-PAGE, denature at 95°C for 5 minutes in Laemmli buffer containing reducing agents (DTT or β-mercaptoethanol) . For immunohistochemistry applications with formalin-fixed paraffin-embedded tissues, implementation of heat-induced epitope retrieval in citrate buffer (pH 6.0) is critical for exposing antibody binding sites masked during fixation. Frozen section protocols for IHC-F should include acetone or brief paraformaldehyde fixation followed by thorough PBS washing to remove embedding medium . For all applications involving biotinylated antibodies, researchers must address endogenous biotin concerns—implement specific blocking steps using commercial avidin/biotin blocking kits, especially for biotin-rich tissues like liver, kidney, and brain . When developing ELISA protocols, consider using synthetic blocking agents rather than milk-based blockers (which contain endogenous biotin) and optimize wash steps to reduce background. Flow cytometry applications require meticulous fixation and permeabilization optimization to maintain cellular integrity while enabling antibody access to cytoplasmic UMPS. Regardless of application, include appropriate controls processed identically to experimental samples to distinguish specific signals from technical artifacts.

What methodological approaches determine optimal antibody concentration for UMPS detection?

Determining the optimal working concentration for UMPS Antibody, Biotin conjugated requires systematic titration experiments designed for each specific application. For Western blotting, a recommended starting point is testing dilutions across the 1:300-5000 range using identical samples across all conditions . Quantitative analysis of band intensity relative to background allows identification of the dilution providing optimal signal-to-noise ratio rather than simply strongest signal. For ELISA applications, begin with the recommended 1:500-1000 range, constructing standard curves at each dilution to determine which concentration provides the best combination of sensitivity and linear dynamic range . Immunohistochemistry applications typically require more concentrated antibody solutions—test dilutions between 1:100-500 for IHC-F and 1:200-400 for IHC-P on known positive control tissues . The optimization process should evaluate not only signal intensity but also specificity factors such as background staining in negative control regions. For all applications, consider experimental variables that might necessitate concentration adjustments: sample type, target abundance, detection system sensitivity, and incubation conditions. Document optimization results comprehensively, including images of representative results at different concentrations, to establish reproducible protocols for subsequent experiments. This methodical approach ensures consistent performance while minimizing reagent waste and experimental variability.

What strategies effectively optimize Western blot protocols for UMPS Antibody, Biotin conjugated?

Optimizing Western blot protocols for UMPS Antibody, Biotin conjugated requires specific modifications to standard procedures. Begin with blocking optimization—replace milk-based blockers (which contain endogenous biotin) with 3-5% BSA in TBS-T or commercial biotin-free blocking solutions to minimize background . When preparing lysates, ensure complete protein extraction with appropriate lysis buffers containing protease inhibitors to preserve UMPS integrity. For gel separation, 10-12% polyacrylamide gels typically provide optimal resolution for UMPS (~50 kDa). During transfer, PVDF membranes often yield better results than nitrocellulose for biotin-conjugated antibody detection systems. For primary antibody incubation, start with the manufacturer's recommended dilution range (1:300-5000) and adjust based on signal intensity and background levels . Implement stringent washing steps (at least 3 × 10 minutes with TBS-T) before adding streptavidin-HRP (typically at 1:1000-1:5000 dilution). Minimize streptavidin-HRP incubation time (30-60 minutes) to reduce non-specific binding. For detection, enhanced chemiluminescence (ECL) systems provide excellent sensitivity, but exposure time must be carefully optimized to prevent background development. If high background persists despite these optimizations, implement specific avidin/biotin blocking steps before primary antibody application. For multiplex detection with other targets, consider fluorescent streptavidin conjugates combined with direct-labeled antibodies for other proteins to facilitate simultaneous imaging of multiple targets without stripping and reprobing.

How can researchers effectively manage endogenous biotin interference when using biotinylated UMPS antibodies?

Endogenous biotin presents a significant challenge when using biotinylated antibodies like UMPS Antibody, Biotin conjugated. Research has shown that certain tissues (liver, kidney, brain) contain high endogenous biotin levels, and approximately 3% of adult human serum samples contain biotin-reactive IgM antibodies that can cause false positives in biotinylation-based immunoassays . To manage this interference, implement a commercial avidin/biotin blocking kit before primary antibody application—this sequential treatment first uses avidin to bind endogenous biotin, followed by excess biotin to saturate remaining avidin binding sites. For Western blotting applications, include a control lane processed without primary antibody but with streptavidin-detection reagents to identify endogenous biotin signals. When working with human samples, consider testing for biotin-reactive antibodies using a simple ELISA approach with biotinylated proteins as capture agents . For immunohistochemistry, dual blocking is essential—block both endogenous biotin (with avidin/biotin kits) and endogenous peroxidase activity (with hydrogen peroxide treatment) when using HRP-based detection systems. In flow cytometry applications, include fluorescence-minus-one (FMO) controls with streptavidin conjugates alone to establish background thresholds. If endogenous biotin interference persists despite these measures, consider alternative detection strategies using non-biotinylated primary antibodies or detection systems that don't rely on biotin-streptavidin interaction. These comprehensive approaches minimize false positives and ensure experimental results accurately reflect UMPS expression rather than biotin-related artifacts.

What immunohistochemistry protocol modifications optimize UMPS detection in different tissue types?

Optimizing immunohistochemistry protocols for UMPS Antibody, Biotin conjugated requires tissue-specific adjustments to maximize specific signal while minimizing background. For all tissue types, begin with appropriate antigen retrieval—heat-induced epitope retrieval in citrate buffer (pH 6.0) is generally effective for UMPS detection in FFPE tissues . The critical blocking sequence involves three steps: endogenous peroxidase blocking (3% H₂O₂, 10 minutes), protein blocking (1-2% BSA or commercial blockers, 30-60 minutes), and endogenous biotin blocking (commercial avidin/biotin blocking kit). For tissues with high endogenous biotin (liver, kidney), extend the avidin incubation time in the biotin-blocking step. Apply primary antibody at optimized dilutions (1:200-400 for IHC-P, 1:100-500 for IHC-F) and incubate overnight at 4°C in a humidified chamber for best results . For detection, streptavidin-HRP systems work well at 1:500-1:1000 dilutions with 30-minute room temperature incubation. For hematoxylin counterstaining, minimize timing (15-30 seconds) to avoid obscuring cytoplasmic UMPS staining. Tissue-specific modifications include: for brain tissue—extend permeabilization time and use fluorescent detection to minimize autofluorescence interference; for highly vascularized tissues—implement additional blocking steps with normal serum matching the host species of secondary reagents; for tissues with high cell density—consider signal amplification systems like tyramide signal amplification for clear visualization of differential expression. Always process positive and negative control tissues alongside experimental samples under identical conditions for valid comparison and interpretation.

What detection systems provide optimal signal-to-noise ratios for UMPS Antibody, Biotin conjugated?

Selecting the optimal detection system for UMPS Antibody, Biotin conjugated involves balancing sensitivity requirements against background concerns. For Western blotting applications, streptavidin-HRP coupled with enhanced chemiluminescence (ECL) detection provides excellent sensitivity with manageable background when protocols are properly optimized . For applications requiring ultrasensitive detection, poly-HRP-streptavidin systems can further amplify signal through increased enzyme loading per binding event. In cases where background remains problematic despite optimization, NeutrAvidin-HRP conjugates often provide cleaner results than streptavidin-HRP due to reduced non-specific binding properties. For immunohistochemistry applications, streptavidin-HRP with DAB (3,3'-diaminobenzidine) offers reliable detection for light microscopy, while streptavidin-alkaline phosphatase with Fast Red chromogen provides an excellent alternative when endogenous peroxidase activity is difficult to block completely. Fluorescence-based detection using streptavidin conjugated to fluorophores (Alexa Fluor series, DyLight, etc.) enables greater signal precision and multiplex capability when combined with confocal microscopy. For ELISA applications, streptavidin-HRP with TMB (3,3',5,5'-tetramethylbenzidine) substrate provides a good balance of sensitivity and dynamic range. When absolute maximization of sensitivity is required, time-resolved fluorescence systems using streptavidin-europium conjugates offer exceptional signal-to-noise ratios through temporal separation of specific signal from background autofluorescence. Regardless of the chosen system, including appropriate negative controls processed identically to experimental samples is essential for accurate background assessment and signal interpretation.

How should researchers address non-specific binding issues with UMPS Antibody, Biotin conjugated?

Addressing non-specific binding requires systematic analysis and targeted interventions. First, characterize the pattern of non-specific signals—multiple unexpected bands in Western blots, diffuse staining in unexpected cellular compartments in IHC, or high background in negative controls—to guide troubleshooting. For Western blotting applications, increase antibody dilution (try 1:2000-1:5000 range) and implement more stringent washing protocols (5 x 5 minutes with 0.1% Tween-20 in TBS) . If multiple bands persist, conduct peptide competition assays to distinguish specific from non-specific signals. For immunohistochemistry applications, optimize blocking by extending incubation time (2 hours at room temperature) and using synthetic blocking agents rather than serum-based blockers. When endogenous biotin contributes to background, add an avidin/biotin blocking step before antibody application. Research has demonstrated that site-specific biotinylation methods can significantly reduce non-specific binding compared to random biotinylation approaches, improving detection limits up to 5-fold in some applications . For samples with high lipid content that can cause non-specific hydrophobic interactions, add 0.1-0.3% Triton X-100 to blocking and antibody diluent buffers. Consider substituting NeutrAvidin for streptavidin in the detection system, as it demonstrates reduced non-specific binding. For human samples where biotin IgM antibodies may cause false positives, implement additional serum pre-clearing steps or consider alternative detection methods . Always include matched isotype controls and no-primary-antibody controls processed identically to experimental samples to distinguish between true signal and background artifacts.

What analysis approaches best quantify UMPS expression in different experimental contexts?

Quantitative analysis of UMPS expression requires tailored approaches based on experimental techniques and biological questions. For Western blotting, implement digital densitometry with appropriate software (ImageJ, Image Studio, etc.) using background subtraction and normalization to validated loading controls such as β-actin or GAPDH. Include a calibration curve of recombinant UMPS protein when absolute quantification is needed. When analyzing by ELISA, construct standard curves using purified recombinant UMPS and implement four-parameter logistic regression for accurate interpolation across the assay's dynamic range. For immunohistochemistry quantification, digital pathology approaches offer the most rigorous analysis—use automated imaging systems with consistent acquisition parameters to calculate staining intensity, percent positive cells, or H-scores (combining intensity and prevalence). In flow cytometry applications, convert fluorescence intensity to standardized units using calibration beads and implement consistent gating strategies across experiments. For all quantification methods, determine technical variability through replicate analysis to establish confidence intervals for biological interpretation. When comparing UMPS expression across different experimental conditions, normalize to appropriate reference points (untreated controls, wild-type samples, etc.) and apply statistical tests suitable for the data distribution (parametric or non-parametric). For complex datasets integrating multiple parameters, consider dimension reduction techniques (PCA, t-SNE) to identify patterns not apparent in individual measurements. Regardless of the chosen quantification method, report both the analytical approach and its validation to ensure reproducibility and enable meaningful comparison with other studies.

How can researchers distinguish true UMPS expression changes from technical artifacts?

Distinguishing biological UMPS expression changes from technical artifacts requires rigorous experimental design and comprehensive controls. Implement biological replicates (minimum n=3) to account for natural variation and technical replicates to assess method reproducibility. Include calibration standards across experiments to normalize for day-to-day variations in detection sensitivity. For Western blotting, process all comparative samples simultaneously through identical conditions including lysis, electrophoresis, transfer, and detection to minimize technical variability. When analyzing by immunohistochemistry, prepare a reference tissue microarray containing known positive and negative controls to process alongside experimental samples for internal standardization. In flow cytometry studies, use quantitative beads to convert relative fluorescence to absolute values that can be compared between experiments. Implement antibody lot testing when receiving new batches—compare performance with previous lots on identical samples to detect manufacturing variations. Consider orthogonal validation of expression changes using complementary techniques—confirm protein-level changes with mRNA analysis by qPCR or transcriptomics, or validate immunodetection results with functional assays measuring UMPS enzymatic activity. For longitudinal studies, include internal reference samples processed in each experimental batch to track system performance over time. When analyzing results statistically, distinguish biological significance from statistical significance by considering effect size in addition to p-values. For complex experimental designs, consider implementing linear mixed-effects models that can account for both fixed experimental factors and random variation sources. These comprehensive approaches enable confident attribution of observed UMPS expression changes to true biological phenomena rather than methodological inconsistencies.

What strategies effectively validate UMPS localization patterns in cellular compartments?

Validating subcellular localization patterns of UMPS requires multi-technique approaches and stringent controls. Since UMPS is documented as a cytoplasmic protein, deviations from this expected pattern warrant careful validation . Begin with fluorescence microscopy using the biotinylated UMPS antibody detected with fluorescent streptavidin, combined with established markers for subcellular compartments (nucleus, mitochondria, endoplasmic reticulum, etc.). Implement super-resolution microscopy techniques (STED, STORM, or SIM) for precise localization beyond the diffraction limit of conventional microscopy. Confirm microscopy findings with biochemical fractionation—separate cellular compartments through differential centrifugation and detect UMPS in each fraction using Western blotting. For definitive validation, implement electron microscopy with immunogold labeling using the biotinylated UMPS antibody and gold-conjugated streptavidin. Control experiments are essential—verify antibody specificity through knockdown/knockout approaches and confirm compartment marker fidelity with established reagents. Use orthogonal approaches with non-biotinylated UMPS antibodies or fluorescent protein fusions to eliminate biotin-related artifacts. For unexpected localization patterns, implement proximity ligation assays to confirm protein-protein interactions that might explain the observations. When examining stress-induced or condition-specific localization changes, include appropriate time course studies and carefully matched controls. For all localization studies, quantify the distribution pattern across multiple cells (minimum 50-100 cells) to account for cell-to-cell variability and cell cycle effects. These comprehensive validation approaches ensure that observed UMPS localization patterns reflect true biological organization rather than technical artifacts.

How can researchers employ UMPS Antibody, Biotin conjugated in multiplex systems to study metabolic pathway interactions?

Integrating UMPS Antibody, Biotin conjugated into multiplex detection systems enables comprehensive analysis of metabolic pathway interactions. For fluorescence microscopy applications, combine streptavidin-fluorophore detection of biotinylated UMPS antibody with directly labeled antibodies against related enzymes in the pyrimidine synthesis pathway (CAD, DHODH) or connecting metabolic networks (mTOR signaling components, cell cycle regulators). Implement spectral unmixing for precise separation of overlapping fluorophore emissions. In tissue analyses, sequential multiplexing offers an effective approach—detect the biotinylated UMPS antibody first, followed by signal quenching and subsequent detection of additional targets with different reporting systems. For flow cytometry applications, create comprehensive panels by combining streptavidin-fluorophore detection of UMPS with antibodies against cell surface markers and other intracellular metabolic targets, allowing correlation of UMPS expression with cell phenotype and metabolic state. In protein interaction studies, implement co-immunoprecipitation using biotinylated UMPS antibody with streptavidin magnetic beads, followed by mass spectrometry analysis of binding partners. For functional metabolic studies, combine antibody-based detection with metabolite profiling using mass spectrometry—correlate UMPS protein levels with changes in pyrimidine intermediates and end products. In signaling pathway analyses, phospho-protein multiplexing with UMPS detection reveals regulatory relationships between metabolic enzymes and signaling networks. These multiplex approaches transform static single-protein detection into dynamic pathway analyses, providing mechanistic insights into how UMPS functions within broader metabolic networks, particularly in contexts like cancer metabolism where pathway rewiring is common.

What methodological approaches enable investigation of post-translational modifications of UMPS?

Investigating post-translational modifications (PTMs) of UMPS requires specialized methodological approaches beyond standard detection. Begin with immunoprecipitation using biotinylated UMPS antibody and streptavidin beads to isolate UMPS protein complexes from cell or tissue lysates. The purified protein can then be analyzed by mass spectrometry to identify specific PTMs like phosphorylation, acetylation, ubiquitination, or SUMOylation. For phosphorylation studies, complement mass spectrometry with Western blotting using phospho-specific antibodies against predicted phosphorylation sites in UMPS based on consensus sequences for known kinases. Investigate PTM dynamics by treating cells with appropriate modulators (kinase inhibitors, phosphatase inhibitors, deacetylase inhibitors, etc.) before UMPS isolation and analysis. To connect PTMs with functional outcomes, implement in vitro enzyme activity assays with purified UMPS protein containing or lacking specific modifications. For spatial regulation of PTMs, use proximity ligation assays to visualize interactions between UMPS and modifying enzymes in situ. Study PTM-dependent protein interactions using advanced techniques like BioID or APEX proximity labeling, with UMPS as the bait protein. For ubiquitination studies, treat cells with proteasome inhibitors before analysis to stabilize ubiquitinated forms of UMPS, then detect with both UMPS antibody and ubiquitin antibodies . Site-directed mutagenesis of predicted modification sites allows functional validation of specific PTMs through phenotypic analysis. These comprehensive approaches reveal how UMPS function is dynamically regulated through post-translational modifications in different cellular contexts, providing insights into the integration of pyrimidine metabolism with broader cellular signaling networks.

How can UMPS Antibody, Biotin conjugated facilitate investigation of drug response mechanisms?

Leveraging UMPS Antibody, Biotin conjugated for drug response studies provides valuable insights into therapeutic mechanisms and resistance pathways. Begin by establishing baseline UMPS expression profiles across panels of cell lines with varying drug sensitivities using quantitative Western blotting or immunohistochemistry. For chemotherapeutic agents targeting nucleotide metabolism (5-fluorouracil, gemcitabine), monitor UMPS expression changes during treatment using time-course experiments with standardized detection protocols. In patient-derived samples, correlate UMPS expression patterns with treatment outcomes to identify potential biomarker applications. Implement drug resistance modeling by creating paired sensitive/resistant cell lines and comprehensively profiling UMPS expression, localization, and post-translational modifications between matched pairs. For mechanistic insights, combine UMPS detection with broader pathway analysis—multiplex with key signaling nodes (mTOR, MAPK, p53) to identify networks regulating UMPS in response to therapy. To study functional consequences, correlate UMPS protein levels with enzymatic activity measurements and metabolomics profiling of pyrimidine intermediates before and after drug treatment. In animal models, use immunohistochemistry with UMPS antibody to track spatial and temporal expression changes during treatment courses. For high-throughput applications, develop cell-based assays using the biotinylated UMPS antibody in automated imaging platforms to screen compound libraries for modulators of UMPS expression or localization. These approaches provide mechanistic understanding of how pyrimidine metabolism adaptations contribute to therapeutic response and resistance, potentially identifying new intervention strategies targeting metabolic vulnerabilities in disease contexts.

What approaches enable integration of UMPS protein data with transcriptomics and metabolomics?

Integrating UMPS protein data with transcriptomics and metabolomics requires thoughtful experimental design and computational approaches. Begin with coordinated sample collection—harvest parallel samples from the same biological sources for protein detection, RNA sequencing, and metabolite profiling. For UMPS protein quantification, implement standardized Western blotting or ELISA protocols with recombinant protein standards to enable absolute quantification. Collect transcriptomic data using RNA-Seq or qPCR focused on pyrimidine metabolism genes, including UMPS and related pathway components. For metabolomics, employ liquid chromatography-mass spectrometry (LC-MS) optimized for pyrimidine intermediates and end products, including orotate, orotidine-5'-monophosphate, and UMP. Design time-course experiments following perturbations (drug treatment, nutrient limitation, genetic manipulation) to capture dynamic relationships between transcript levels, protein abundance, and metabolite concentrations. Implement computational integration through correlation networks, principal component analysis, and pathway enrichment analyses to identify coordinated responses across molecular levels. For causal relationship determination, apply Granger causality analysis or dynamic Bayesian networks to time-series data. Develop mathematical models incorporating enzyme kinetics to predict how changes in UMPS protein levels should affect metabolite pools, then validate predictions experimentally. Visualize integrated datasets using pathway-based representations that highlight concordant and discordant responses across biological levels. For single-cell applications, consider emerging technologies that enable parallel measurement of proteins and transcripts from the same cells. These integrative approaches reveal regulatory relationships between transcriptional control, protein abundance, and metabolic function, providing systems-level understanding of pyrimidine metabolism regulation in normal physiology and disease states.

What cutting-edge technologies can researchers combine with UMPS Antibody, Biotin conjugated for spatial metabolic profiling?

Combining UMPS Antibody, Biotin conjugated with emerging spatial technologies enables unprecedented insights into metabolic organization within tissues and cells. Implement multiplex immunofluorescence with the biotinylated UMPS antibody and additional metabolic enzymes using cyclic immunofluorescence or spectral unmixing approaches to create comprehensive metabolic maps. Integrate with spatial transcriptomics technologies like Visium or slide-seq to correlate protein detection with local gene expression profiles. For subcellular resolution, apply expansion microscopy techniques—physically expanding the sample while maintaining relative protein positions—to visualize fine-scale organization of metabolic enzyme complexes. Implement mass spectrometry imaging (MALDI-MSI or DESI-MSI) on serial sections to correlate UMPS protein distribution with spatial metabolite profiles of pyrimidine intermediates. For living systems, combine the biotinylated antibody (using cell-permeable streptavidin conjugates) with genetically encoded metabolite sensors to simultaneously monitor enzyme presence and product formation. Apply proximity labeling techniques (BioID, APEX) with UMPS as the bait protein to identify spatially restricted interaction partners in different cellular compartments. Integrate high-resolution microscopy with computational modeling to predict metabolic flux based on enzyme distribution patterns. For clinical applications, implement spatial proteomics using digital spatial profiling or imaging mass cytometry with the biotinylated UMPS antibody to characterize metabolic heterogeneity within disease tissues. These advanced spatial approaches transform our understanding of metabolism from homogeneous biochemical pathways to spatially organized processes, revealing how subcellular compartmentalization and tissue architecture influence metabolic function in both normal physiology and disease states.

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.