SPOPL Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Mechanism of SPOPL Antibody, HRP Conjugated

SPOPL Antibody, HRP conjugated refers to a horseradish peroxidase (HRP)-labeled primary antibody targeting the speckle-type POZ protein-like (SPOPL), a 45 kDa protein implicated in nuclear speckle organization and transcriptional regulation . HRP conjugation enables enzymatic detection in assays like ELISA, western blotting (WB), and immunohistochemistry (IHC) via chromogenic or chemiluminescent substrates (e.g., DAB, TMB, or ECL) .

Key Features:

PropertyDescription
TargetSPOPL protein (GenBank: BC071613; UniProt: Q6IQ16)
Host/IsotypeTypically rabbit polyclonal IgG (e.g., Proteintech 17740-1-AP)
HRP ConjugationCovalent linkage via primary amines or thiols using cross-linkers
ApplicationsWB (1:500–1:2000), ELISA, IHC
DetectionColorimetric (DAB/TMB) or chemiluminescent (ECL) signals

Applications in Research and Diagnostics

HRP-conjugated SPOPL antibodies are primarily used for detecting SPOPL expression in:

  1. Western Blotting:

    • Detects SPOPL in lysates from HeLa, PC-3, and other cell lines .

    • Optimal dilution: 1:500–1:2000 .

  2. ELISA:

    • Quantifies SPOPL in serum or lysate samples.

    • Enhanced sensitivity achieved via poly-HRP conjugation (e.g., lyophilized HRP-antibody mixtures) .

  3. Immunohistochemistry:

    • Localizes SPOPL in tissue sections (not explicitly tested in available sources but inferred from HRP compatibility) .

Conjugation Methods and Optimization

HRP conjugation methods vary in efficiency and antibody preservation:

MethodMechanismAdvantagesLimitations
Periodate OxidationOxidizes HRP carbohydrates to aldehydes; reacts with antibody amines .High conjugation efficiency, stable storageRequires lyophilization; may denature HRP .
Sulfo-SMCC/MaleimideActivates HRP with maleimide; conjugates to antibody thiols .Directional binding, retains antibody activityRequires antibody thiolation (e.g., SATA) .
Lightning-Link®Proprietary kit using pH-sensitive chemistry for rapid conjugation .No purification needed; minimal antibody lossBuffer-dependent (avoid amines/thiols) .
LYNX Rapid ConjugationLyophilized HRP mix with modifier/quencher for neutral-pH conjugation .100% antibody recovery; small-scale labelingRequires precise molar ratios (1:4–1:1 Ab:HRP) .

Critical Parameters for SPOPL HRP Conjugation:

  • Buffer: Amine-free (HEPES, MOPS) pH 6.5–8.5; avoid Tris, sodium azide .

  • Molar Ratio: 1:4–1:1 antibody-to-HRP (e.g., 100 μg HRP + 100–400 μg antibody) .

  • Storage: Lyophilized HRP conjugates stable at 4°C for extended periods .

Performance Data and Sensitivity

Studies highlight improved detection sensitivity with optimized conjugation:

Table 1: ELISA Sensitivity Comparison

Conjugation MethodAntibody DilutionSensitivity Gainp-valueSource
Classical (Periodate)1:25Baseline
Modified (Lyophilized HRP)1:5000200× higher< 0.001

Key Findings:

  • Lyophilized HRP-antibody conjugates enable higher antibody titers (1:5000 vs. 1:25), enhancing ELISA sensitivity .

  • SDS-PAGE confirms successful conjugation: no migration of HRP-antibody complexes under reducing conditions .

Research Implications and Future Directions

  • Diagnostic Potential: Enhanced sensitivity of poly-HRP conjugates may enable early detection of SPOPL-linked diseases (e.g., cancers) .

  • Limitations:

    • Cross-reactivity risks with secondary antibodies in indirect assays .

    • Buffer incompatibilities (e.g., sodium azide inhibits HRP) .

  • Recommendations: Validate conjugates via UV-Vis (peak shift at 280/430 nm) and SDS-PAGE .

References

  1. Abcam (2023): HRP Antibody Conjugation Protocol.

  2. PMC (2018): Enhanced HRP-Antibody Conjugation via Lyophilization.

  3. Proteintech (2025): SPOPL Antibody (17740-1-AP).

  4. PubMed (2015): Sulfo-SMCC-Mediated HRP Conjugation.

  5. Bio-Rad (2024): LYNX Rapid HRP Conjugation Kit.

  6. Thermo Fisher (2025): HRP-Conjugated Secondary Antibodies.

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Order fulfillment typically takes 1-3 business days. Shipping times vary depending on the order method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
SPOPL antibody; Speckle-type POZ protein-like antibody; HIB homolog 2 antibody; Roadkill homolog 2 antibody
Target Names
Uniprot No.

Target Background

Function

SPOPL is a component of cullin-RING-based BTB-CUL3-RBX1 E3 ubiquitin-protein ligase complexes. These complexes mediate the ubiquitination and subsequent proteasomal degradation of target proteins, although SPOPL's contribution is relatively low. Complexes containing SPOPL homodimers or SPOP/SPOPL heterodimers exhibit lower ubiquitin ligase activity compared to those containing only SPOP. SPOPL may function to downregulate the activity of SPOP-containing cullin-RING-based BTB-CUL3-RBX1 E3 ubiquitin-protein ligase complexes.

Gene References Into Functions
  1. Research indicates that a CUL3-SPOPL E3 ubiquitin ligase complex regulates endocytic trafficking and multivesicular body formation by ubiquitinating and degrading EPS15 at endosomes. PMID: 27008177
Database Links

HGNC: 27934

KEGG: hsa:339745

STRING: 9606.ENSP00000280098

UniGene: Hs.333297

Protein Families
Tdpoz family
Subcellular Location
Nucleus.

Q&A

What is the principle behind HRP conjugation to SPOPL antibodies and how does it enhance detection sensitivity?

HRP (Horseradish peroxidase) conjugation to SPOPL antibodies creates a detection system that significantly amplifies signal output. The principle involves covalent linking of HRP enzyme molecules to SPOPL antibodies through chemical modification of carbohydrate moieties on HRP. When sodium meta periodate oxidizes these carbohydrates, it generates aldehyde groups that react with amino groups on the antibody, forming Schiff's bases that are stabilized through reduction with sodium cyanoborohydride . This conjugation strategy preserves both the antigen-binding capability of the antibody and the enzymatic activity of HRP. The sensitivity enhancement occurs because each HRP molecule can catalyze multiple substrate conversion events, creating an amplification cascade that enables detection of low-abundance SPOPL protein in experimental samples .

What experimental applications are most suitable for SPOPL antibody, HRP conjugated?

SPOPL antibody, HRP conjugated, demonstrates optimal performance across several immunodetection techniques. For Western blotting, the conjugate enables direct detection of SPOPL protein without requiring a secondary antibody step, streamlining the protocol and reducing background interference . In ELISA applications, the conjugate allows for sensitive quantification of SPOPL proteins with detection limits in the picogram range, particularly valuable when studying SPOPL expression in different cellular contexts . Immunohistochemistry applications benefit from reduced protocol complexity and enhanced signal-to-noise ratios when visualizing SPOPL localization in tissue sections . For all applications, the direct conjugation eliminates cross-reactivity issues that might arise with two-step detection systems, particularly important when studying proteins like SPOPL that may have structural similarities with other POZ domain-containing proteins .

How do I determine the optimal working dilution for SPOPL antibody, HRP conjugated?

Determining the optimal working dilution for SPOPL antibody, HRP conjugated requires systematic titration experiments tailored to your specific application. For Western blot optimization, prepare a dilution series (typically 1:1,000 to 1:10,000) of the conjugate and test against samples with known SPOPL expression levels. Modern HRP-conjugated antibodies prepared with enhanced methodologies can maintain detection sensitivity at dilutions as high as 1:5,000, whereas traditional conjugates may require concentrations of 1:25 . For ELISA applications, perform a checkerboard titration by coating plates with various antigen concentrations and testing against a range of antibody dilutions. The optimal dilution should provide maximum specific signal with minimal background. Consider that high-quality conjugates produced using improved lyophilization techniques during the conjugation process can achieve significantly higher dilution factors than traditionally prepared conjugates while maintaining signal strength . Document the signal-to-noise ratio for each dilution to establish reproducible conditions for future experiments.

How can I troubleshoot high background when using SPOPL antibody, HRP conjugated in Western blots?

High background when using SPOPL antibody, HRP conjugated in Western blots typically stems from several methodological factors that can be systematically addressed. First, evaluate blocking efficacy—insufficient blocking allows non-specific binding to membrane proteins. Implement a more stringent blocking protocol using 5% non-fat dry milk or 3-5% BSA in TBST for at least 1-2 hours at room temperature . Second, optimize antibody concentration—HRP-conjugated antibodies prepared via enhanced methods can be used at much higher dilutions (1:5,000) than traditional conjugates (1:25) while maintaining sensitivity . Third, examine washing protocols—increase both duration (5×5 minutes) and volume of wash buffer between incubation steps to remove unbound conjugate. Fourth, adjust substrate exposure time—excessive substrate incubation amplifies background signals; implement a time-course experiment to determine optimal exposure. Fifth, consider membrane interference—certain nitrocellulose membranes may contribute to background; comparing performance with PVDF membranes might reveal improvements . Finally, integrate controls—include samples known to be negative for SPOPL expression to distinguish between specific and non-specific signals.

What strategies can enhance the specificity of SPOPL antibody, HRP conjugated for detecting post-translational modifications?

Enhancing specificity for detecting post-translational modifications (PTMs) of SPOPL protein requires sophisticated experimental design. First, implement antibody validation using PTM-null mutants where the specific modification site (e.g., phosphorylation, ubiquitination) has been altered through site-directed mutagenesis; this confirms signal specificity to the modified form . Second, employ dephosphorylation controls—treat sample aliquots with lambda phosphatase before immunodetection to verify phospho-specific signals. Third, incorporate enrichment strategies—use phosphopeptide enrichment (TiO₂, IMAC) or ubiquitin-remnant motif purification before detection to concentrate modified SPOPL forms. Fourth, utilize specialized blocking agents—phospho-blocking reagents can reduce non-specific binding to phosphorylated proteins. Fifth, optimize buffer compositions—adding phosphatase inhibitors (10 mM sodium fluoride, 1 mM sodium orthovanadate) throughout extraction and immunodetection preserves labile modifications . Finally, confirm specificity through orthogonal methods—mass spectrometry validation of detected PTM sites provides definitive evidence. This multi-faceted approach ensures that detected signals represent authentic post-translational modifications of SPOPL protein rather than non-specific binding artifacts.

How do I design experiments to investigate SPOPL protein interactions using HRP-conjugated antibodies?

Designing experiments to investigate SPOPL protein interactions requires a multi-layered approach that maximizes the advantages of HRP-conjugated antibodies. Begin with co-immunoprecipitation (Co-IP) optimization by testing various lysis conditions (RIPA, NP-40, or digitonin-based buffers) to preserve protein-protein interactions while efficiently extracting SPOPL complexes . For direct detection in Co-IP experiments, use the HRP-conjugated SPOPL antibody at optimized dilutions (1:2,000-1:5,000) to visualize pulled-down complexes without secondary antibody interference . Implement stringent controls including IgG pull-downs, SPOPL knockdown/knockout samples, and reverse Co-IPs with antibodies against suspected interaction partners. For proximity-based interaction studies, adapt HRP-conjugated SPOPL antibodies for proximity ligation assays (PLA) by conjugating additional oligonucleotide probes. To study dynamic interactions, design pulse-chase experiments with protein synthesis inhibitors, monitoring SPOPL complex formation over time. For comprehensive interactome analysis, combine antibody-based purification with mass spectrometry, using HRP activity to confirm successful enrichment before analysis. Throughout all experiments, maintain consistent buffer conditions, temperature, and incubation times to ensure reproducibility of detected interaction networks.

What factors affect the stability and shelf-life of SPOPL antibody, HRP conjugated, and how can I maximize its longevity?

The stability and shelf-life of SPOPL antibody, HRP conjugated depends on multiple storage and handling factors. HRP conjugates are susceptible to activity loss through oxidative damage, bacterial contamination, and freeze-thaw cycles. To maximize longevity, first implement proper storage conditions—store concentrated stock at -20°C in single-use aliquots (50-100 μL) containing 50% glycerol to prevent freeze-thaw damage . For working solutions, maintain at 4°C with preservatives like 0.01% thimerosal or 0.02% sodium azide, but note that high concentrations of azide can inhibit HRP activity. Second, add stabilizing proteins—supplement storage buffer with 1% BSA to prevent protein adsorption to container surfaces and provide competitive inhibition against proteases . Third, minimize light exposure—store in amber vials or wrapped in aluminum foil to prevent photo-oxidation of the heme group in HRP. Fourth, maintain optimal pH—buffers at pH 6.0-7.0 maximize HRP stability compared to more alkaline conditions. Fifth, conduct periodic quality control—test small aliquots monthly against reference standards to monitor activity retention. Properly maintained conjugates should retain >80% activity for 6-12 months at 4°C and up to 24 months at -20°C with minimal freeze-thaw cycles .

How does the periodate conjugation method affect epitope recognition of SPOPL antibodies compared to other conjugation techniques?

The periodate conjugation method's impact on SPOPL antibody epitope recognition derives from its specific chemistry and target sites. Unlike glutaraldehyde methods that predominantly target lysine residues throughout the antibody, periodate conjugation selectively modifies carbohydrate moieties on the HRP enzyme rather than the antibody itself . This selective approach preserves the antigen-binding region (Fab) structure of the SPOPL antibody, maintaining epitope recognition capacity. Comparative analysis with maleimide-based conjugation (which targets cysteine residues) shows that periodate methods result in lower binding interference, particularly important for conformational epitopes in the SPOPL protein .

The enhanced method incorporating lyophilization further preserves epitope recognition by increasing conjugation efficiency without necessitating higher concentrations of modification reagents that might affect antibody structure . Experimental validation through direct ELISA comparing native versus conjugated antibodies typically shows >95% epitope retention with optimized periodate methods compared to 70-85% with glutaraldehyde approaches. When targeting structurally complex proteins like SPOPL with multiple domains, this preservation of recognition capacity is critical for accurate experimental outcomes across applications .

What are the critical parameters to monitor when validating a new batch of SPOPL antibody, HRP conjugated for research applications?

Validating a new batch of SPOPL antibody, HRP conjugated requires comprehensive analysis of multiple parameters to ensure experimental reliability. First, assess conjugation efficiency through spectrophotometric analysis—measure absorbance at 280 nm (protein) and 403 nm (HRP) to calculate the HRP:antibody ratio, with optimal ratios typically between 2:1 and 4:1 . Second, determine enzyme activity using a kinetic assay with TMB substrate, comparing reaction rates with previous batches; activity should be within 20% of reference standards. Third, evaluate specificity through Western blotting against positive controls (cell lines with known SPOPL expression), negative controls (SPOPL-knockout lines), and potential cross-reactive proteins with similar domains. Fourth, confirm sensitivity by detecting a dilution series of recombinant SPOPL protein, establishing limits of detection and quantification .

Fifth, assess batch-to-batch consistency by overlaying titration curves from current and previous batches, with acceptable variation <15% in EC50 values. Sixth, validate application performance in your specific experimental context (e.g., cell type, tissue samples) before deploying in critical experiments. Seventh, document lot-specific optimal working dilutions for each application, as these may vary slightly between batches. This systematic validation approach ensures reliable and reproducible results when implementing new conjugate batches in SPOPL research protocols .

How can multiplexed detection systems incorporate SPOPL antibody, HRP conjugated alongside other target proteins?

Multiplexed detection incorporating SPOPL antibody, HRP conjugated requires strategic experimental design to maintain signal specificity while enabling simultaneous visualization of multiple targets. For chromogenic multiplex immunohistochemistry (IHC), implement sequential detection protocols using different substrates for HRP (DAB, AEC, TMB) that produce distinct colors, with complete HRP inactivation between rounds using hydrogen peroxide treatment . For fluorescence-based multiplex systems, convert HRP activity to fluorescent signal using tyramide signal amplification (TSA), where HRP catalyzes deposition of fluorophore-labeled tyramide at the site of antibody binding. This approach allows detection of low-abundance SPOPL alongside other proteins of interest .

When designing panels, carefully consider antibody species origin and isotypes to prevent cross-reactivity—pair SPOPL antibody, HRP conjugated with antibodies from different host species when using secondary detection systems for other targets . For automated multiplex platforms, optimize antigen retrieval conditions that accommodate all target proteins without compromising SPOPL epitope integrity. Validation of multiplexed systems should include single-stain controls, absorption controls, and spatial analysis of signal co-localization to confirm authentic multiplex detection versus artifactual overlap. This strategic implementation enables complex co-expression studies of SPOPL with interaction partners or pathway components within the same specimen .

What considerations are important when using SPOPL antibody, HRP conjugated for quantitative analysis of protein expression levels?

Quantitative analysis using SPOPL antibody, HRP conjugated demands rigorous attention to multiple variables that influence signal linearity and reproducibility. First, establish a standard curve using recombinant SPOPL protein at known concentrations (typically 0.1-100 ng/mL) to define the linear detection range; working within this range ensures proportional relationship between signal intensity and protein quantity . Second, implement loading controls—for Western blotting, normalize SPOPL signals to housekeeping proteins (β-actin, GAPDH) or total protein staining (Ponceau S, SYPRO Ruby); for ELISA, ensure consistent total protein concentrations across samples .

Third, optimize substrate kinetics—for chemiluminescent detection, conduct time-course experiments to identify the linear phase of the reaction before signal saturation. Fourth, employ image acquisition parameters that prevent pixel saturation—use exposure settings that maintain signals within the dynamic range of the detection system . Fifth, perform technical replicates (minimum triplicate) and biological replicates to establish statistical significance of observed differences. Sixth, validate quantitative findings through orthogonal methods—compare ELISA results with Western blot densitometry or mass spectrometry-based quantification. Finally, implement quality control metrics including coefficient of variation (CV) thresholds (<15% for intra-assay, <20% for inter-assay) to ensure reliable quantitative comparisons across experimental conditions .

How might emerging technologies enhance the application of SPOPL antibody, HRP conjugated in spatial biology and single-cell analysis?

Emerging technologies are revolutionizing applications of SPOPL antibody, HRP conjugated through integration with spatial biology and single-cell analytical platforms. In spatial proteomics, cyclic immunofluorescence (CycIF) techniques can incorporate HRP-conjugated SPOPL antibodies with tyramide signal amplification, followed by antibody stripping and reprobing to achieve multiplexed protein mapping with subcellular resolution . This approach allows visualization of SPOPL's spatial relationship with dozens of other proteins across tissue architectures. For digital spatial profiling platforms, HRP activity can be harnessed to release barcoded oligonucleotides from specific regions of interest, enabling quantitative analysis of SPOPL expression within precisely defined tissue microenvironments .

In the single-cell domain, integration with microfluidic technologies allows analysis of SPOPL expression in thousands of individual cells with preserved spatial context. Application in mass cytometry (CyTOF) through metal-tagged antibodies enables high-dimensional analysis of SPOPL in relation to cellular phenotypes and states . Emerging proximity-based methods like proximity extension assays (PEA) can leverage HRP-conjugated SPOPL antibodies to achieve ultrasensitive detection in limited samples through enzymatic signal amplification. Looking forward, integration with artificial intelligence-driven image analysis will enhance quantification precision, while advances in antibody engineering may produce conjugates with improved specificity and sensitivity for detecting low-abundance SPOPL variants or post-translational modifications in complex cellular environments .

What are the comparative advantages and limitations of HRP versus other enzyme conjugates for SPOPL antibody applications?

The selection between HRP and alternative enzyme conjugates for SPOPL antibody applications requires consideration of distinct performance characteristics across experimental contexts. HRP (44 kDa) offers several advantages over alkaline phosphatase (AP, 140 kDa) and β-D-galactosidase (β-Gal, 540 kDa), including smaller molecular size that facilitates tissue penetration for immunohistochemistry applications . HRP demonstrates superior catalytic efficiency with turnover rates of ~1000 molecules/second compared to AP's ~150 molecules/second, enabling more rapid signal development. Additionally, HRP conjugates exhibit excellent stability at 4°C (6-12 months) and compatibility with a diverse range of substrates for colorimetric, chemiluminescent, and fluorescent detection .

ParameterHRPAlkaline Phosphataseβ-D-Galactosidase
Molecular Weight44 kDa140 kDa540 kDa
Catalytic RateHigh (~1000/sec)Moderate (~150/sec)Low (~30/sec)
Storage Stability6-12 months (4°C)12-18 months (4°C)3-6 months (4°C)
Endogenous ActivityCommon in tissuesMainly in alkaline environmentsRare in mammalian tissues
InhibitorsAzide, cyanidePhosphate, EDTATris, high salt
Signal DevelopmentRapid (minutes)Slower (10-30 min)Slowest (30-60 min)
Signal:Noise RatioHighest for Western blotHighest for ELISAModerate

This comparative analysis guides selection of the optimal enzyme conjugate based on the specific requirements of SPOPL detection applications .

How do I effectively integrate SPOPL antibody, HRP conjugated into multi-omics research approaches?

Integrating SPOPL antibody, HRP conjugated into multi-omics research frameworks requires strategic experimental design that leverages antibody specificity while enabling complementary data generation across platforms. For proteogenomic integration, start by correlating SPOPL protein levels (detected via HRP-conjugated antibody) with corresponding mRNA expression (measured by RT-qPCR or RNA-seq) across experimental conditions to evaluate post-transcriptional regulation mechanisms . For samples with limited material, implement sequential extraction protocols that enable DNA/RNA isolation followed by protein recovery from the same specimens, preserving material for multi-omics profiling.

When combining with phosphoproteomics, use parallel sample processing where one aliquot undergoes phosphopeptide enrichment for mass spectrometry while another is analyzed using HRP-conjugated SPOPL antibody to correlate total protein levels with specific phosphorylation sites . For spatial multi-omics, implement digital spatial profiling where HRP-conjugated SPOPL antibody can be used to define regions of interest for subsequent laser capture microdissection and downstream transcriptomic or metabolomic analysis. When integrating with interactomics, use HRP-conjugated SPOPL antibody for co-immunoprecipitation followed by mass spectrometry to define the protein interaction network.

For comprehensive data integration, implement computational approaches including correlation networks, principal component analysis, and machine learning algorithms to identify patterns across multi-omics datasets. This holistic framework reveals SPOPL's functional context beyond what single-omics approaches can achieve, providing insights into regulatory mechanisms and pathway relationships .

What methodological adaptations are necessary when applying SPOPL antibody, HRP conjugated across different experimental models (cell lines, tissues, organisms)?

Methodological adaptations for SPOPL antibody, HRP conjugated across diverse experimental models require systematic optimization to account for model-specific variables. For cell line applications, adjust fixation protocols based on cellular compartmentalization of SPOPL—use 4% paraformaldehyde for preserving membrane and cytoplasmic localization versus methanol for nuclear epitopes . Cell permeabilization requires tailoring with Triton X-100 (0.1-0.5%) for adherent cells versus saponin (0.1%) for suspension cultures to maximize antibody accessibility while preserving cellular architecture.

For tissue applications, antigen retrieval methods significantly impact SPOPL detection—heat-induced epitope retrieval (HIER) with citrate buffer (pH 6.0) typically proves superior for formalin-fixed paraffin-embedded samples, while trypsin-based enzymatic retrieval may better preserve certain conformational epitopes . The table below summarizes optimal antigen retrieval conditions across tissue types:

Tissue TypeRecommended Retrieval MethodBuffer CompositionDuration
BrainHIERCitrate, pH 6.020 min
LiverHIEREDTA, pH 9.030 min
KidneyEnzymaticProteinase K10 min
LungHIERTris-EDTA, pH 9.025 min
HeartHIER + EnzymaticCitrate + Trypsin15+5 min

For organismal differences, antibody dilution requires recalibration—murine models typically require 1:500-1:1000 dilutions, while human samples often need 1:1000-1:2000 for optimal signal-to-noise ratios . Cross-species reactivity validation is essential when applied to evolutionary distant models, with Western blotting confirmation preceding immunohistochemical applications. For developmental studies, embryonic tissues often require shorter fixation times (4-8 hours) versus adult tissues (12-24 hours) to prevent epitope masking while maintaining structural integrity. These systematic adaptations ensure consistent SPOPL detection across diverse experimental models .

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.