TPP2 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Introduction to TPP2 Antibody, FITC Conjugated

TPP2 (Tripeptidyl Peptidase II) is a serine peptidase involved in processes such as antigen presentation, calcium homeostasis, and ERK signaling . The TPP2 Antibody, FITC conjugated is a fluorescence-labeled reagent designed for detecting TPP2 in techniques like flow cytometry (FCM), immunofluorescence (IF), and Western blotting (WB) . FITC (Fluorescein Isothiocyanate) conjugation enables visualization of TPP2 localization and expression dynamics in cellular and tissue samples .

Validation and Quality Control Data

  • bsm-62830r-fitc demonstrates specificity in WB (1:300–5,000 dilution), FCM (1:20–100), and IF (1:50–200) . Subcellular fractionation confirms TPP2’s enrichment in the endoplasmic reticulum (ER) .

  • ABIN7173487-FITC was validated using recombinant human TPP2 protein, with cross-reactivity confirmed in human cell lines .

Research Context and Biological Significance of TPP2

TPP2’s roles in antigen processing and calcium homeostasis make its detection critical. For example:

  • Antigen Presentation: TPP2 trims long peptide precursors for MHC class I presentation, though it is non-essential for most epitopes .

  • Calcium Signaling: TPP2 interacts with ER membrane proteins (e.g., IP3R1, SERCA1) to regulate intracellular Ca²⁺ flux .

  • ERK Phosphorylation: TPP2 inhibition reduces nuclear phosphorylated ERK1/2, impacting mitogenic signaling .

The FITC-conjugated antibody enables real-time tracking of TPP2 in these pathways via fluorescence microscopy or flow cytometry .

Limitations and Considerations

  • Species Specificity: Most FITC-conjugated TPP2 antibodies are human-reactive; cross-species validation is limited .

  • Enzymatic Activity: TPP2’s peptidase function (e.g., S449A/T mutations) may not affect antibody binding, as epitopes are often structural .

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 your order within 1-3 business days after receiving it. Delivery time may vary depending on the chosen shipping method and location. Please consult your local distributor for specific delivery timelines.
Synonyms
Tripeptidyl-peptidase 2 (TPP-2) (EC 3.4.14.10) (Tripeptidyl aminopeptidase) (Tripeptidyl-peptidase II) (TPP-II), TPP2
Target Names
TPP2
Uniprot No.

Target Background

Function
Tripeptidyl Peptidase II (TPP2) is a component of the proteolytic cascade that operates downstream of the 26S proteasome within the ubiquitin-proteasome pathway. TPP2 can partially compensate for the 26S proteasome's function when the latter is inhibited. Additionally, TPP2 stimulates adipogenesis.
Gene References Into Functions
  1. TPP2 plays a crucial role in regulating various cellular functions by controlling the phosphorylation of ERK1 and ERK2. PMID: 26041847
  2. Two consanguineous siblings have been reported to exhibit early-onset Evans syndrome, immunodeficiency, and premature immunosenescence associated with TPP2 deficiency. PMID: 25414442
  3. Research has demonstrated that autosomal recessive mutations in TPP2 cause recurrent infections, autoimmune disorders, and neurodevelopmental delays in humans. PMID: 25525876
  4. TPPII, MYBBP1A, and CDK2 form a complex protein-protein interaction network. PMID: 25303791
  5. Studies have revealed previously unknown differences between TPP II orthologues and subtilisin, along with identifying features conserved within the entire family of subtilisin-like serine peptidases. PMID: 22266401
  6. Overexpression of Tripeptidyl peptidase II (TPP2) is frequently observed during oral carcinogenesis and may be associated with the progression of Oral Squamous Cell Carcinoma (OSCC) via activation of the Spindle Assembly Checkpoint (SAC). PMID: 22986808
  7. Scientists have obtained a 3D structure of the human TPPII. PMID: 22483107
  8. Current research focuses on the structural aspects of TPPII. PMID: 21771670
  9. Research findings suggest a significant role of TPPII in maintaining viral growth, with potential implications for antiviral therapies. PMID: 21134372
  10. The promoter of TPPII has been localized to a 215 bp fragment upstream of the initiation codon. PMID: 15716107
  11. TPPII appears to promote malignant cell growth by enabling cells to exit mitosis and survive even when exhibiting severe mitotic spindle damage. PMID: 16762321
  12. TPP2 plays a specialized role in antigen processing, though it is not essential for the generation of most presented peptides. PMID: 16849449
  13. Research indicates that TPP II expression can be regulated by both positive and negative regulatory elements. PMID: 17343995
  14. Expression of mRNA for MuRF-1 increased approximately 3-fold at 10 days without changes in MAFbx or tripeptidyl peptidase II mRNA, but all decreased between 10 and 21 days of muscle disuse. PMID: 17901116
  15. Results suggest that TPPII is dispensable for the generation of proteasome-dependent HLA class I ligands, and the enzyme is not significantly involved in generating the proteasome-independent HLA-B27-bound peptide repertoire. PMID: 18286573
  16. Cross-presentation of NY-ESO-1/ISCOMATRIX cancer vaccine is proteasome independent and requires the cytosolic protease tripeptidyl peptidase II. PMID: 19155470
  17. The MHC class I-restricted LMP1 epitopes investigated are among the few known epitopes processed independently of the proteasome by tripeptidyl peptidase II. PMID: 19587004

Show More

Hide All

Database Links

HGNC: 12016

OMIM: 190470

KEGG: hsa:7174

STRING: 9606.ENSP00000365233

UniGene: Hs.432424

Protein Families
Peptidase S8 family
Subcellular Location
Cytoplasm. Nucleus. Note=Translocates to the nucleus in responce to gamma-irradiation.

Q&A

What is TPP2 and what cellular functions does it perform?

TPP2 (Tripeptidyl-peptidase 2) is a serine protease belonging to the peptidase S8 family. It plays essential roles in protein degradation pathways, cleaving tripeptides from the N-terminus of larger peptides. TPP2 is involved in multiple cellular processes including antigen processing, cell division, and stress response mechanisms. The protein has a molecular weight of approximately 138 kDa and is expressed in various human tissues including liver, colorectal, and lung tissues, as well as in multiple cell lines like Jurkat, Hela, and HepG2 . TPP2 is also found in rodent tissues, suggesting evolutionary conservation of this enzyme across mammalian species .

How does a TPP2 antibody differ from TPP peptides used in research?

While TPP2 antibodies specifically target the Tripeptidyl peptidase II protein, TPP peptides (like the Hsp70-derived 14-mer peptide TKDNNLLGRFELSG) bind to membrane-bound heat shock protein 70 (mHsp70) . TPP2 antibodies are typically generated in hosts like rabbits and recognize epitopes within the TPP2 protein structure . In contrast, TPP peptides are synthetic constructs designed to interact with specific target proteins. For immunodetection applications, TPP2 antibodies can be applied in various techniques including Western blotting, immunohistochemistry, and flow cytometry, whereas TPP peptides are often used as targeting molecules in fluorescence imaging and potential therapeutic applications .

What validation methods should be performed before using a new TPP2 antibody in experiments?

Before incorporating a new TPP2 antibody into your experimental workflow, several validation steps are essential. These include Western blot analysis to confirm target specificity and determine the correct molecular weight (approximately 138 kDa for TPP2) . Immunohistochemistry or immunofluorescence on known TPP2-expressing tissues/cells compared with negative controls helps verify tissue/cellular localization patterns . Flow cytometry validation ensures the antibody detects native protein configurations. Additionally, testing the antibody across multiple relevant cell lines/tissues (such as Jurkat, Hela, HepG2, lung and liver cancer tissues) confirms consistent recognition across sample types . Knock-down validation using TPP2-specific shRNA provides the strongest evidence for antibody specificity .

How can researchers optimize internalization kinetics experiments using FITC-labeled TPP2 antibodies?

Optimization of internalization kinetics experiments with FITC-labeled TPP2 antibodies requires systematic approach similar to methods used for other labeled antibodies. Begin by establishing optimal antibody concentration through titration experiments to determine the minimum concentration providing sufficient signal-to-noise ratio. Time-course experiments should be conducted at both 4°C (to assess surface binding without internalization) and 37°C (to measure active internalization) . Flow cytometry analysis should include propidium iodide (PI) staining to exclude non-viable cells, and appropriate isotype controls to account for non-specific binding . For quantitative analysis, calculate the internalization rate by measuring the decrease in surface fluorescence or increase in intracellular signal over time. Live-cell imaging with temperature-controlled stages can complement flow cytometry data by providing visual confirmation of antibody trafficking within cells.

What are the key differences between FITC and other fluorophores when conjugated to TPP2 antibodies?

When selecting fluorophores for TPP2 antibody conjugation, researchers should consider several distinguishing characteristics. FITC (excitation ~495nm, emission ~519nm) offers good quantum yield but suffers from relatively rapid photobleaching and pH sensitivity, with optimal fluorescence at pH 8.0 and significant reduction below pH 7.0. Compared to FITC, Alexa Fluor 488 provides enhanced photostability, brightness, and pH resistance (pH 4-10), making it superior for extended imaging sessions. IRDye800CW, used for near-infrared imaging applications, enables deeper tissue penetration and reduced autofluorescence interference, making it particularly valuable for ex vivo tissue imaging applications . Phycoerythrin (PE) provides higher brightness than FITC but has a larger size that might affect antibody penetration into tissues. Each fluorophore choice represents a trade-off between brightness, stability, size, and spectral characteristics that should align with specific experimental requirements.

How should I design control experiments when using FITC-conjugated TPP2 antibodies in flow cytometry?

Designing robust control experiments for flow cytometry with FITC-conjugated TPP2 antibodies requires multiple controls to ensure reliable data interpretation. First, include an isotype-matched FITC-conjugated control antibody (e.g., FITC-conjugated rabbit IgG if using a rabbit-derived TPP2 antibody) to establish the threshold for non-specific binding . An unstained control is essential to determine autofluorescence levels of your cell population. For validating specificity, include a TPP2-negative cell line or TPP2-knockdown cells (using shRNA targeting TPP2) as negative controls . Competitive inhibition controls where unconjugated TPP2 antibody is pre-incubated with cells before adding the FITC-conjugated version can confirm binding specificity. Temperature controls comparing staining at 4°C (surface binding only) versus 37°C (allows internalization) help distinguish between membrane and internalized antibody signals . Finally, propidium iodide (PI) staining should be used to exclude non-viable cells from analysis to prevent false-positive results .

What are the optimal fixation and permeabilization protocols for TPP2 immunofluorescence studies?

For optimal TPP2 immunofluorescence detection, fixation and permeabilization protocols should be carefully selected based on subcellular localization and epitope sensitivity. Paraformaldehyde (4%) fixation for 15-20 minutes at room temperature preserves cellular architecture while maintaining antibody accessibility. For membrane-associated TPP2, mild permeabilization with 0.1-0.2% Triton X-100 for 5-10 minutes is typically sufficient. For intracellular TPP2 detection, stronger permeabilization may be required (0.5% Triton X-100 for 10-15 minutes). If epitope masking is a concern, heat-mediated antigen retrieval in EDTA buffer (pH 8.0) can improve detection, as demonstrated in successful TPP2 immunohistochemistry protocols . When working with FITC-conjugated antibodies, minimize exposure to light throughout the protocol to prevent photobleaching. For co-localization studies with other cellular compartments, the choice between methanol (-20°C for 10 minutes) versus paraformaldehyde fixation should be determined empirically, as methanol can better preserve certain subcellular structures but may denature some epitopes.

How can TPP2 activity assays be integrated with antibody-based detection methods?

Integrating TPP2 enzymatic activity assays with antibody-based detection provides complementary data on both protein presence and functional status. The standard approach utilizes the fluorogenic substrate AAF-AMC (Ala-Ala-Phe-7-amido-4-methylcoumarin) to measure TPP2 proteolytic activity . This can be combined with immunological detection in several ways: First, perform TPP2 immunoprecipitation using the antibody, followed by activity assay of the isolated protein. Second, use specific TPP2 inhibitors (such as butabindide or B6 at 1μM concentration) to confirm the specificity of measured activity . For comprehensive analysis in cell-based systems, researchers can conduct parallel experiments where one set of samples undergoes antibody-based detection via immunoblotting, immunofluorescence, or flow cytometry, while companion samples are processed for activity assays. When evaluating TPP2 knockdown efficiency, both protein levels (by immunoblotting) and enzymatic activity should be measured to confirm functional depletion . For intact tissue samples, sections can be processed for immunohistochemistry visualization of TPP2 distribution, while adjacent sections are homogenized for biochemical activity measurements.

How should I interpret discrepancies between TPP2 protein levels detected by antibodies versus functional activity measured by enzymatic assays?

Discrepancies between TPP2 protein detection and enzymatic activity measurements require careful analysis of multiple factors. Post-translational modifications may alter TPP2 activity without changing protein levels detectable by antibodies. For instance, phosphorylation events regulated by ERK1/2 can affect TPP2 function without necessarily changing total protein abundance . Enzyme inhibition by endogenous regulators or experimental conditions (pH, temperature, ionic strength) might reduce activity despite normal protein levels. Conversely, non-specific antibody binding could lead to overestimation of TPP2 protein levels compared to activity. To resolve such discrepancies, perform Western blotting with multiple antibodies targeting different TPP2 epitopes, conduct immunoprecipitation followed by activity testing, and use specific TPP2 inhibitors like butabindide (1μM) as controls in activity assays . Time-course experiments may reveal temporal differences between protein expression and enzymatic activation. Additionally, subcellular fractionation might identify pools of TPP2 with different activity states in various cellular compartments.

What statistical approaches are most appropriate for analyzing TPP2 expression across different tissue types?

When analyzing TPP2 expression across different tissue types, several statistical approaches should be considered based on experimental design and data characteristics. For immunohistochemistry or immunofluorescence data, semi-quantitative scoring systems (0-3+ intensity combined with percentage of positive cells) can be applied, followed by non-parametric tests like Mann-Whitney U or Kruskal-Wallis for comparisons between tissue groups . For Western blot or flow cytometry quantification, normalized data can be analyzed using ANOVA with appropriate post-hoc tests (Tukey or Bonferroni) for multiple tissue comparisons. When comparing tumor versus normal tissues, paired t-tests or Wilcoxon signed-rank tests are appropriate if samples come from the same patients. Correlation analyses (Pearson's or Spearman's) can assess relationships between TPP2 expression and other variables like clinical parameters or expression of related proteins. For studies incorporating both protein detection and activity measurements, multivariate approaches like principal component analysis may reveal patterns across different tissue types. Always include adequate biological replicates (minimum n=3 for cell lines, larger for heterogeneous tissues) and report effect sizes alongside p-values.

How can multiplex imaging with FITC-conjugated TPP2 antibodies and other markers be quantitatively analyzed?

Quantitative analysis of multiplex imaging combining FITC-conjugated TPP2 antibodies with other markers requires sophisticated image processing approaches. Begin with spectral unmixing to correct for fluorophore bleed-through, particularly important when FITC emission overlaps with other green-yellow fluorophores. Single-stained controls for each fluorophore are essential for this process. For colocalization analysis, calculate Pearson's or Mander's correlation coefficients between TPP2 and other markers of interest, defining appropriate intensity thresholds to distinguish specific from background signals . Cellular segmentation using nuclear or membrane markers enables per-cell quantification of TPP2 and other markers, generating distributions rather than averages. For tissue sections, implement region-of-interest (ROI) analysis to compare TPP2 expression across different histological areas. Distance mapping can quantify spatial relationships between TPP2-positive structures and other cellular components. For high-content screening applications, machine learning algorithms can classify cells based on TPP2 expression patterns combined with other markers. When analyzing tumor-to-background ratios in tissue imaging, calculate area-under-the-curve (AUC) measurements to comprehensively assess contrast across the entire sample .

What are common sources of false positives/negatives in TPP2 antibody-based assays and how can they be prevented?

Several factors can contribute to false results in TPP2 antibody-based assays. False positives often result from non-specific antibody binding, particularly in tissues with high endogenous peroxidase activity (liver, kidney) or high protein content. This can be mitigated by thorough blocking (using 5-10% serum matched to secondary antibody species) and proper antibody titration to determine optimal concentration (typically around 0.25-2 μg/ml for TPP2 antibodies) . Cross-reactivity with related proteases can be identified by testing the antibody against purified proteins or knockout/knockdown samples . False negatives commonly occur due to epitope masking during fixation, requiring optimization of antigen retrieval methods (such as heat-mediated retrieval in EDTA buffer at pH 8.0 for TPP2) . Degraded antibodies lose specificity, so proper storage conditions and periodic validation of antibody performance using positive control samples are essential. For flow cytometry applications, high autofluorescence can mask true signals; this can be addressed through appropriate compensation settings and inclusion of unstained controls for each sample type . When using FITC-conjugated antibodies, photobleaching during sample processing can lead to signal loss, requiring minimal light exposure and efficient workflow.

How can I verify TPP2 antibody specificity in new experimental models or tissues?

Verifying TPP2 antibody specificity in new experimental models requires a multi-faceted approach. Begin with Western blot analysis to confirm detection of a single band at the expected molecular weight of 138 kDa for TPP2 . This should be conducted across multiple relevant tissues or cell types from your new model. Peptide competition assays, where the antibody is pre-incubated with the immunizing peptide before application to samples, should eliminate specific staining if the antibody is truly specific. Genetic validation through TPP2 knockdown using specific shRNA sequences targeting TPP2 (such as CCGGCCTGATCCTTTCAGGTCTGAACTCGAGTTCAGACCTGAAAGGATCAGGTTTTTG) compared with scrambled shRNA controls provides compelling evidence of specificity . For tissues with potential cross-reactivity concerns, dual-labeling with a second TPP2 antibody targeting a different epitope should show co-localization if both are specific. RNA-protein correlation analysis comparing TPP2 mRNA levels (measured by qPCR) with protein levels (detected by the antibody) across various tissues can further support specificity. Finally, functional validation by measuring TPP2 activity (using the AAF-AMC assay) in samples with high versus low antibody staining can confirm the biological relevance of the detected protein .

What quality control parameters should be monitored for long-term storage of FITC-conjugated TPP2 antibodies?

Maintaining quality of FITC-conjugated TPP2 antibodies during long-term storage requires monitoring several critical parameters. Fluorescence intensity should be measured periodically using standardized beads or consistent positive control samples, with retention of at least 80% of original signal intensity considered acceptable. Antibody concentration should be verified using protein assays to detect potential precipitation or adsorption to storage containers. The fluorophore-to-protein ratio, initially optimized during conjugation, should remain stable; significant changes suggest degradation of either component. Specificity testing using Western blot or flow cytometry with appropriate controls should be performed after extended storage periods to ensure maintained target recognition. pH stability is particularly important for FITC conjugates, as fluorescence decreases significantly below pH 7.0; buffer conditions should be monitored and maintained between pH 7.5-8.5. Sterility testing is recommended for liquid formulations to detect potential microbial contamination. For lyophilized antibodies (like the A06668-2 preparation), moisture content should be minimized during storage . Proper storage conditions include protection from light (amber vials or foil wrapping), maintenance at recommended temperature (typically -20°C for long-term storage), and avoidance of freeze-thaw cycles by preparing single-use aliquots upon reconstitution.

How can FITC-conjugated TPP2 antibodies be utilized in studying proteasome inhibition resistance mechanisms?

FITC-conjugated TPP2 antibodies offer unique advantages in investigating proteasome inhibition resistance mechanisms. Since TPP2 can partially compensate for compromised proteasome function, tracking its expression and localization changes during resistance development is critical. Flow cytometry with FITC-conjugated TPP2 antibodies enables quantitative assessment of TPP2 protein levels in single cells, revealing population heterogeneity that might be masked in bulk analyses. This approach allows identification and isolation of cell subpopulations with altered TPP2 expression for further characterization. Time-course experiments can monitor TPP2 dynamics during resistance acquisition, comparing internalization kinetics at 4°C versus 37°C to distinguish between surface binding and active internalization patterns . For spatial analysis, live-cell fluorescence microscopy with FITC-conjugated TPP2 antibodies can track TPP2 redistribution between cytoplasmic and nuclear compartments during stress responses. Co-localization studies with proteasome components can reveal potential compensatory complexes. For in-depth mechanistic studies, combining antibody-based TPP2 detection with selective inhibition using butabindide or B6 (1μM) helps distinguish between TPP2-dependent and independent resistance mechanisms . Cross-correlation with ERK1/2 phosphorylation status (using PMA stimulation) can further elucidate regulatory pathways affecting TPP2 function in resistance contexts .

What is the role of TPP2 in cancer progression and how can antibody-based detection contribute to understanding this relationship?

TPP2 plays multifaceted roles in cancer progression that can be elucidated through strategic application of antibody-based detection methods. Immunohistochemical analysis reveals elevated TPP2 expression in various cancer types including colorectal adenocarcinoma, liver cancer, and lung cancer, suggesting potential contributions to malignant phenotypes . TPP2's proteolytic activity influences antigen processing, potentially affecting immune surveillance of tumors. Multiplexed immunofluorescence combining TPP2 antibodies with markers of cancer stem cells, epithelial-mesenchymal transition, and immune infiltrates can map TPP2's relationship to these cancer-promoting processes. Correlation between TPP2 expression patterns and patient outcomes establishes prognostic significance. At the molecular level, TPP2 may regulate ERK1/2 signaling pathways, which are frequently dysregulated in cancer . This connection can be investigated using antibodies detecting both TPP2 and phosphorylated ERK1/2 forms, particularly under stimulated conditions using PMA (100 ng/ml for 15 minutes) . For functional studies, comparing TPP2 antibody detection with enzymatic activity measurements in matched normal versus tumor tissues helps distinguish between expression changes and altered activity states. In preclinical models, antibody-based imaging can track TPP2 expression during tumor progression, treatment response, and resistance development, potentially identifying patient subgroups who might benefit from therapies targeting TPP2-dependent pathways.

How can FITC-conjugated TPP2 antibodies be integrated into high-content screening approaches for drug discovery?

Integration of FITC-conjugated TPP2 antibodies into high-content screening (HCS) for drug discovery leverages multiplexed cellular analysis to identify compounds affecting TPP2 biology. The direct fluorescence detection eliminates secondary antibody requirements, streamlining workflows for large-scale screens. A typical HCS campaign would begin with optimization of antibody concentration, incubation time, and fixation/permeabilization conditions across multiple cell types relevant to the disease model. For primary screens, cells in 384- or 1536-well formats can be treated with compound libraries, then fixed and stained with FITC-conjugated TPP2 antibodies along with nuclear and cytoplasmic markers for segmentation purposes. Multi-parametric analysis can simultaneously assess: 1) TPP2 expression levels, 2) subcellular localization shifts, 3) cell morphology changes, and 4) cell viability. Hit compounds identified from primary screens should undergo validation in concentration-response experiments. Counter-screens with TPP2-knockdown cells help distinguish between on-target and off-target effects. Secondary assays should include TPP2 activity measurements using AAF-AMC substrate to determine functional consequences of expression changes . For mechanistic insights, compounds can be tested under conditions of phosphorylation stimulation using PMA (100 ng/ml) . This integrated approach not only identifies direct TPP2 modulators but also compounds affecting its regulatory pathways, potentially uncovering novel therapeutic strategies for diseases where TPP2 plays significant roles.

What are the optimal antigen retrieval methods for TPP2 detection in different tissue types?

Optimal antigen retrieval for TPP2 detection varies by tissue type, fixation method, and detection platform. For formalin-fixed, paraffin-embedded (FFPE) samples, heat-mediated antigen retrieval in EDTA buffer (pH 8.0) has proven effective across multiple tissue types including colorectal adenocarcinoma, liver cancer, and lung cancer tissues . This involves heating sections to near-boiling temperatures (95-98°C) for 15-20 minutes followed by gradual cooling. For tissues with high extracellular matrix content (like connective tissues), adding enzymatic pre-treatment with proteinase K (10-20 μg/ml for 10 minutes at 37°C) can improve antibody accessibility. Fresh frozen tissues generally require milder retrieval methods, with brief fixation in 4% paraformaldehyde followed by permeabilization with 0.1-0.2% Triton X-100 often sufficient. For immunocytochemistry applications on cultured cells (like A549), enzyme antigen retrieval reagents have shown good results . When optimizing retrieval methods, multiple conditions should be tested in parallel on serial sections, evaluating signal intensity, specificity (using appropriate controls), and tissue morphology preservation. Regardless of the method, blocking endogenous peroxidase activity (for IHC) using 0.3% H₂O₂ in methanol for 10 minutes and subsequent blocking with 10% goat serum are essential steps before primary antibody application .

How can I adapt FITC-TPP2 antibody protocols for super-resolution microscopy applications?

Adapting FITC-TPP2 antibody protocols for super-resolution microscopy requires modifications to maximize spatial resolution while maintaining signal specificity. For Structured Illumination Microscopy (SIM), standard immunofluorescence protocols can be used with additional emphasis on minimizing background fluorescence through extensive washing steps and careful blocking (5% BSA with 0.1% Triton X-100). For Stimulated Emission Depletion (STED) microscopy, FITC may not be optimal due to photobleaching under high-intensity depletion lasers; consider re-conjugating the TPP2 antibody with more photostable dyes like Abberior STAR 488 or Atto 488. For Single Molecule Localization Microscopy (SMLM) techniques like STORM or PALM, buffer optimization is critical - use oxygen-scavenging systems containing glucose oxidase, catalase, and glucose with thiols (MEA or BME) to enhance FITC blinking behavior. Cell fixation should be performed with electron microscopy-grade paraformaldehyde (4%) to preserve nanoscale structures. Sample thickness must be minimized; for tissue sections, 5-10 μm thickness is recommended. Antibody concentration should be carefully titrated, generally using lower concentrations (0.5-1 μg/ml) than conventional microscopy to reduce background and ensure single-molecule detection . For multi-color super-resolution imaging, spectral crosstalk must be eliminated through careful fluorophore selection and sequential imaging approaches.

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 2024 Thebiotek. All Rights Reserved.