MME Monoclonal Antibody,FITC Conjugated

Shipped with Ice Packs
In Stock

Description

Flow Cytometry

FITC-conjugated MME antibodies are critical for identifying CD10+ cells, including leukemia cells (e.g., pre-B ALL) and normal kidney/proximal tubule cells . For example:

  • Protocol: 20 μL antibody per 100 μL whole blood or 10610^6 cells .

  • Controls: Use mouse IgG1 isotype controls (e.g., MOPC-21 FITC) to assess nonspecific binding .

Immunofluorescence

Used to localize MME in tissue sections or cultured cells. FITC emission (green) is detected via microscopy .

Leukemia Diagnosis

CD10 (MME) is a hallmark marker for common acute lymphoblastic leukemia (ALL), present in 85% of cases . FITC-labeled MME antibodies enable rapid identification of CD10+ leukemic cells in flow cytometry .

Functional Insights

MME regulates peptide hormone activity by cleaving substrates like glucagon, enkephalins, and bradykinin . Studies using FITC-MME antibodies have mapped its expression in kidney proximal tubules and glomerular epithelium .

Therapeutic Implications

While not directly therapeutic, MME antibodies aid in characterizing cancer subtypes. Conjugated antibodies (e.g., ADCs like brentuximab vedotin) use similar targeting strategies but with cytotoxic payloads .

Comparative Analysis of MME FITC Antibodies

CloneHostConjugateReactivityApplications
MEM-78MouseFITCHumanFlow cytometry
COCLMouseFITCHumanFlow cytometry, IF
LT10MouseFITCHumanFlow cytometry, IF

Challenges and Considerations

  • Quenching Effects: High FITC concentrations may reduce fluorescence signal .

  • Cross-Reactivity: Validate specificity with isotype controls .

  • Stability: FITC is light-sensitive; store in the dark .

Product Specs

Form
Phosphate-buffered saline (PBS), pH 7.4, containing 0.09% sodium azide and 0.2% (w/v) bovine serum albumin (BSA).
Lead Time
Orders for the MME Monoclonal Antibody, FITC Conjugated, are typically shipped within 1-3 business days of order receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for precise delivery estimates.

Q&A

What is MME and why is it an important research target?

MME (Membrane Metallo-Endopeptidase), also known as CD10, NEP, or CALLA, is a zinc-dependent metalloprotease that cleaves peptide bonds on the amino side of hydrophobic amino acids. It functions as a cell surface enzyme that inactivates various peptide hormones including enkephalins, substance P, and angiotensin I. In research contexts, MME is significant as a marker for various cell types, particularly in hematological studies, where it helps identify and characterize specific leukemia subtypes. The protein is expressed on a variety of normal and neoplastic cell types, making MME monoclonal antibodies valuable tools for cellular characterization and disease diagnosis .

What is FITC conjugation and how does it enhance antibody functionality?

FITC (Fluorescein Isothiocyanate) is a reactive derivative of fluorescein that contains an isothiocyanate (-N=C=S) functional group. This group readily reacts with primary amines on proteins (including antibodies), creating a stable thiourea bond. When conjugated to antibodies, FITC provides a fluorescent tag that emits green fluorescence (peak emission ~520 nm) when excited with blue light (peak excitation ~495 nm). This conjugation transforms standard antibodies into fluorescent probes without significantly altering their binding properties, enabling direct detection in applications such as flow cytometry, immunofluorescence microscopy, and immunohistochemistry . The FITC conjugation eliminates the need for secondary detection reagents, simplifying experimental workflows while providing specific target visualization.

What are the optimal storage conditions for FITC-conjugated MME monoclonal antibodies?

FITC-conjugated antibodies require specific storage conditions to maintain their functionality and fluorescence properties. For MME monoclonal antibodies conjugated with FITC, the recommended storage conditions are:

  • Temperature: Store at 2-8°C for short-term storage (up to one month) or at -20°C for long-term storage.

  • Light exposure: Protect from prolonged exposure to light as FITC is photosensitive and can photobleach.

  • Freeze-thaw cycles: Avoid repeated freeze-thaw cycles, which can degrade both the antibody and the fluorophore.

  • Buffer composition: Most preparations are supplied in PBS with stabilizers such as BSA (0.1-1%) and preservatives like sodium azide (0.02-0.05%).

  • Aliquoting: For antibodies stored at -20°C, aliquoting is recommended to prevent repeated freeze-thaw cycles .

Following these storage guidelines will help maintain antibody performance and extend shelf life to approximately one year from the date of receipt.

How should MME-FITC antibodies be optimized for flow cytometry experiments?

Optimizing MME-FITC antibodies for flow cytometry requires systematic titration and validation steps:

Titration Protocol:

  • Prepare a single-cell suspension of known MME-expressing cells (e.g., human peripheral blood or appropriate cell lines).

  • Distribute equal cell numbers (typically 1×10⁶ cells) into separate tubes.

  • Add different concentrations of the MME-FITC antibody to each tube, starting with the manufacturer's recommended dilution (typically 20 μl per 100 μl of whole blood or 10⁶ cells in suspension) .

  • Include a range of dilutions (e.g., 1:2, 1:5, 1:10, 1:20 of the recommended concentration).

  • Incubate for 30 minutes at 2-8°C in the dark.

  • Wash cells twice with flow cytometry buffer.

  • Analyze by flow cytometry, comparing signal-to-noise ratios across different antibody concentrations.

Validation Steps:

  • Include appropriate isotype control (FITC-conjugated mouse IgG1) to establish background fluorescence.

  • Include both MME-positive and MME-negative cell populations to confirm specificity.

  • Consider compensation controls if performing multicolor flow cytometry to account for spectral overlap.

  • Evaluate the staining index (mean fluorescence intensity of positive population divided by standard deviation of negative population) to determine optimal concentration .

The optimal antibody concentration provides maximum separation between positive and negative populations while minimizing background staining.

What cell preparation techniques are recommended for MME-FITC antibody staining?

For optimal MME-FITC antibody staining, the following cell preparation techniques are recommended:

For Fresh Blood Samples:

  • Collect blood in anticoagulant tubes (EDTA or heparin).

  • For whole blood staining: Use 100 μl of blood per test.

  • For isolated cells: Perform density gradient separation using Ficoll-Paque or similar medium.

  • Wash cells twice in PBS containing 2% FBS.

  • Adjust cell concentration to 1×10⁶ cells/100 μl.

  • Add 20 μl of MME-FITC antibody per 100 μl of cell suspension .

  • Incubate for 15-30 minutes at 2-8°C in the dark.

  • For whole blood: Lyse red blood cells using commercial lysing solution.

  • Wash twice and resuspend in appropriate buffer for analysis.

For Cultured Cells:

  • Harvest adherent cells using enzyme-free cell dissociation buffer to avoid cleaving surface antigens.

  • Wash twice in cold PBS containing 2% FBS.

  • Count and adjust cell concentration to 1×10⁶ cells/100 μl.

  • Follow the staining procedure as described above.

For Fixed Cells:

  • If fixation is required, use 1-2% paraformaldehyde post-staining.

  • Note that fixation may alter MME epitope recognition; validate with fresh cells first.

  • Fixed samples can be stored at 2-8°C in the dark for up to 24 hours before analysis .

These preparation techniques ensure optimal antibody binding while preserving cell viability and antigen integrity.

How can MME-FITC antibodies be integrated into multiparameter flow cytometry panels?

Integrating MME-FITC antibodies into multiparameter flow cytometry panels requires strategic planning to maximize information while minimizing spectral overlap issues:

Panel Design Considerations:

  • FITC emits in the green spectrum (peak ~520 nm), which may overlap with PE and other fluorochromes.

  • Position the MME-FITC antibody in your panel based on expected expression levels:

    • For high-expression antigens: FITC is suitable despite its moderate brightness.

    • For low-expression antigens: Consider brighter fluorochromes or alternative MME antibody conjugates.

Recommended Compatible Fluorochromes:

FluorochromeEmission Peak (nm)Potential Spectral Overlap with FITCCompensation Requirement
Pacific Blue455MinimalLow
APC660MinimalLow
APC-Cy7785MinimalLow
PE-Cy5667ModerateMedium
PE575SignificantHigh
PE-Cy7785ModerateMedium

Implementation Protocol:

  • Perform single-color controls for each fluorochrome in your panel using the same cell type.

  • Set up a compensation matrix before analyzing multicolor samples.

  • Include an FMO (Fluorescence Minus One) control omitting the MME-FITC antibody to accurately set gates.

  • Use standardized beads to validate instrument performance before each experiment.

  • Consider tandem dye degradation when using PE-Cy7 or APC-Cy7 alongside FITC .

This strategic approach ensures accurate identification of MME-expressing populations within complex cellular phenotypes.

What approaches can be used to validate MME-FITC antibody specificity in novel tissue types?

Validating MME-FITC antibody specificity in novel tissue types requires a multi-faceted approach:

Validation Strategy:

  • Positive and Negative Controls:

    • Include known MME-positive tissues (e.g., pre-B lymphoblasts, renal proximal tubules).

    • Include known MME-negative tissues as negative controls.

    • Compare staining patterns with expectations based on literature.

  • Blocking Experiments:

    • Pre-incubate tissue sections with unlabeled anti-MME antibody.

    • Follow with MME-FITC antibody staining.

    • Significant reduction in signal indicates specific binding.

  • Antibody Clone Comparison:

    • Test multiple MME antibody clones (e.g., MEM-78 and LT10) .

    • Concordant staining patterns across different clones support specificity.

  • Molecular Validation:

    • Correlate MME protein expression with mRNA expression using qRT-PCR.

    • Compare MME-FITC staining patterns with in situ hybridization results.

  • Knockout/Knockdown Controls:

    • If available, use MME-knockout tissue or cells with MME knockdown.

    • Absence of staining in these samples confirms specificity.

  • Competitive Inhibition:

    • Pre-incubate MME-FITC antibody with purified MME protein.

    • Apply to tissue sections; reduced staining indicates specificity.

  • Western Blot Correlation:

    • Perform Western blot analysis on tissue lysates.

    • Presence of a single band at the expected molecular weight (~100 kDa for MME) supports specificity .

These complementary approaches provide robust validation of MME-FITC antibody specificity in previously untested tissue types.

What are common issues with MME-FITC antibody staining and how can they be resolved?

Researchers may encounter several challenges when working with MME-FITC antibodies. The following table outlines common issues and their solutions:

IssuePossible CausesTroubleshooting Steps
Weak or absent signal1. Insufficient antibody concentration
2. Epitope damage during sample preparation
3. Photobleaching
4. Low MME expression
1. Increase antibody concentration
2. Use gentler cell preparation methods
3. Minimize light exposure during processing
4. Verify MME expression in your sample type
5. Try alternative clones (MEM-78 vs. LT10)
High background1. Excess antibody
2. Non-specific binding
3. Insufficient washing
4. Cell autofluorescence
1. Titrate antibody to optimal concentration
2. Add 2% serum from host species to blocking buffer
3. Increase washing steps (3-4 washes)
4. Include unstained and isotype controls
5. Consider using spectral unmixing algorithms
Signal variability between experiments1. Inconsistent sample preparation
2. Antibody degradation
3. Instrument variability
1. Standardize protocols
2. Use freshly prepared antibody dilutions
3. Include calibration beads
4. Maintain consistent PMT voltages
5. Store antibody properly to prevent degradation
Non-specific staining1. Fc receptor binding
2. Dead cell binding
3. Protein aggregates
1. Block with Fc receptor blocking reagent
2. Include viability dye to exclude dead cells
3. Centrifuge antibody solution before use (10,000g, 5 min)
4. Filter samples before analysis

When these approaches fail to resolve issues, consider testing alternative MME antibody clones or consulting with the antibody manufacturer for specific recommendations.

How can researchers verify the fluorescence activity and conjugation quality of MME-FITC antibodies?

Verifying the fluorescence activity and conjugation quality of MME-FITC antibodies is essential for experimental reliability. The following methods provide comprehensive quality assessment:

Spectral Analysis:

  • Measure excitation and emission spectra using a spectrofluorometer.

  • Optimal FITC excitation peak should be at ~495 nm and emission at ~520 nm.

  • Shifts in these peaks may indicate compromised conjugation or fluorophore degradation.

Degree of Labeling (DOL) Determination:

  • Calculate the F/P (fluorophore-to-protein) ratio:

    • Measure absorbance at 280 nm (protein) and 495 nm (FITC).

    • Calculate using the formula: DOL = (A495 × ε280) ÷ [(A280 - (A495 × CF)) × εFITC]

    • Where CF is the correction factor (typically 0.35 for FITC) and ε represents molar extinction coefficients.

  • Optimal F/P ratio for MME-FITC antibodies is typically 3-7 fluorophore molecules per antibody.

Flow Cytometry Validation:

  • Compare with a reference standard or previously validated lot.

  • Calculate the staining index (SI) using the formula:
    SI = (MFIpositive - MFInegative) ÷ (2 × SDnegative)

  • Monitor SI across antibody lots to ensure consistent performance .

Antibody Binding Capacity:

  • Use quantitative flow cytometry with calibrated beads.

  • Compare the binding capacity of the FITC-conjugated antibody with unconjugated antibody.

  • Significant reduction may indicate over-conjugation affecting antigen binding.

If quality concerns are detected, researchers should contact the manufacturer for replacement or consider alternative preparations for critical experiments.

What are best practices for analyzing MME expression patterns in heterogeneous cell populations?

Analyzing MME expression patterns in heterogeneous cell populations requires sophisticated approaches to accurately identify and characterize MME-positive subsets:

Gating Strategy for Flow Cytometry:

  • Begin with standard preprocessing gates:

    • Forward/side scatter to identify viable cells

    • Single-cell discrimination using FSC-H vs. FSC-A

    • Live/dead discrimination if applicable

  • Create a specific MME gating hierarchy:

    • Set primary gates using clear MME-FITC negative and positive populations

    • Utilize FMO (Fluorescence Minus One) controls to establish accurate boundaries

    • Apply biexponential scaling for optimal visualization of FITC fluorescence

Quantification Methods:

  • Percentage-based analysis:

    • Report percentage of MME-positive cells within defined populations

    • Use consistent gating between samples

  • Expression level analysis:

    • Calculate Mean Fluorescence Intensity (MFI) or Median Fluorescence Intensity

    • Normalize to negative control or isotype control

    • Consider antibody binding capacity (ABC) calculations for absolute quantification

Advanced Analytical Approaches:

  • Dimension reduction techniques:

    • Apply tSNE or UMAP for high-dimensional visualization

    • Identify novel MME-expressing populations in complex datasets

  • Clustering algorithms:

    • Utilize FlowSOM or PhenoGraph for automated population identification

    • Validate computational findings with manual gating

  • Correlation analysis:

    • Assess relationships between MME expression and other markers

    • Generate heatmaps to visualize expression patterns across populations

These analytical frameworks enable comprehensive characterization of MME expression in heterogeneous samples, revealing biologically significant patterns that might be missed with simpler approaches.

How should researchers address potential artifacts in MME-FITC experimental data?

Common Artifacts and Mitigation Strategies:

  • Autofluorescence:

    • Manifestation: Background signal in FITC channel, especially in myeloid cells, fixed tissues, or aged samples

    • Mitigation:

      • Include unstained controls for each sample type

      • Employ spectral unmixing algorithms

      • Consider alternative fluorophores for highly autofluorescent samples

      • Use 530/30 nm bandpass filters to optimize FITC detection while minimizing autofluorescence

  • Compensation Errors:

    • Manifestation: False positive or negative MME populations due to improper fluorescence spillover correction

    • Mitigation:

      • Use single-stained controls for each fluorochrome

      • Apply automated compensation matrices with manual verification

      • Consider fluorophore combinations with minimal spectral overlap

  • Non-specific Binding:

    • Manifestation: Uniform dim staining across populations expected to be negative

    • Mitigation:

      • Include isotype controls matched to the MME-FITC antibody class and concentration

      • Implement blocking steps with serum or commercial blocking reagents

      • Validate with alternative MME detection methods

  • Dead Cell Artifacts:

    • Manifestation: Dead/dying cells showing apparent MME positivity

    • Mitigation:

      • Incorporate viability dyes in all panels

      • Perform strict viability gating before MME analysis

      • Use gentle cell preparation techniques to maximize viability

  • Photobleaching Effects:

    • Manifestation: Progressive signal reduction during acquisition or between analysis sessions

    • Mitigation:

      • Minimize sample exposure to light

      • Analyze FITC parameters early in acquisition sequences

      • Consider standardization beads to monitor instrument performance

By systematically addressing these potential artifacts, researchers can significantly improve data quality and reliability in MME-FITC experimental studies.

How does MME-FITC antibody performance compare in suspended cells versus tissue sections?

MME-FITC antibodies perform differently in suspended cells versus tissue sections due to fundamental differences in sample preparation, epitope accessibility, and detection parameters:

Comparative Performance Analysis:

ParameterSuspended CellsTissue SectionsOptimization Strategies
Signal IntensityTypically stronger with more uniform stainingOften weaker with potential regional variationsFor tissues: Increase antibody concentration by 2-3 fold; extend incubation time to overnight at 4°C
BackgroundGenerally lowerHigher due to tissue autofluorescence and non-specific bindingFor tissues: Implement additional blocking steps; include Sudan Black B (0.1%) to reduce autofluorescence
Epitope AccessibilityBetter accessibility to surface antigensMay require antigen retrieval for optimal detectionFor tissues: Test multiple antigen retrieval methods (heat-induced vs. enzymatic); optimize retrieval duration
QuantificationDirect quantification via flow cytometryTypically semi-quantitative; requires image analysisFor tissues: Use reference standards; implement computational image analysis
Clone PerformanceMEM-78 and LT10 clones perform wellClone performance may vary based on fixation methodsTest both clones on your specific tissue type; MEM-78 may perform better with certain fixatives

Protocol Adaptations for Tissue Sections:

  • Increase antibody concentration to 1:25-1:50 (versus 1:100 for suspended cells).

  • Extend incubation time (60-90 minutes at room temperature or overnight at 4°C).

  • Implement tyramide signal amplification for significantly improved sensitivity.

  • Consider confocal microscopy to improve signal-to-noise ratio in tissue sections.

Validation Approaches:
For critical studies, validate tissue section findings with complementary methods such as freshly isolated cells from the same tissue source to confirm expression patterns.

What considerations are important when using MME-FITC antibodies for rare cell detection?

Detecting rare MME-expressing cell populations requires specialized approaches to achieve sufficient sensitivity and specificity:

Optimization Strategy for Rare Cell Detection:

  • Sample Enrichment:

    • Implement magnetic bead pre-enrichment for MME-positive cells

    • Use density gradient centrifugation to remove irrelevant cell types

    • Apply negative selection to deplete abundant non-target populations

  • Staining Protocol Modifications:

    • Increase sample volume (process 10⁷-10⁸ cells for rare events)

    • Optimize antibody concentration through careful titration

    • Extend staining incubation to 45-60 minutes at 4°C

    • Include multiple MME antibody clones for improved detection sensitivity

  • Instrument Setup:

    • Adjust PMT voltages to maximize resolution of dim populations

    • Implement thresholding on FITC channel if appropriate

    • Reduce flow rate to 12-24 μl/min for improved rare event resolution

    • Collect minimum of 1-5×10⁶ events for statistically valid rare cell analysis

  • Data Analysis Approaches:

    • Use probability contour plots rather than dot plots for visualization

    • Implement sequential gating strategy with Boolean combinations

    • Apply backgating to verify population characteristics

    • Calculate background rate in negative controls to determine detection threshold:

      • Detection threshold = mean background rate + (3 × standard deviation)

Statistical Considerations:
For populations with expected frequency <0.1%, collect sufficient events to achieve a coefficient of variation (CV) <20% using the formula:
CV = 100 × √(1-p)/(n×p) where p is frequency and n is total events collected.

These specialized approaches enable reliable detection of rare MME-expressing cells with frequencies as low as 0.001% of total population.

What emerging technologies may enhance MME-FITC antibody applications in research?

Several cutting-edge technologies are poised to revolutionize MME-FITC antibody applications in research settings:

Spectral Flow Cytometry:
This technology utilizes the full emission spectrum rather than band-pass filters, enabling improved separation of FITC from autofluorescence and spectrally similar fluorophores. Spectral unmixing algorithms can distinguish subtle differences between fluorophores with overlapping emission profiles, allowing more comprehensive multiplexing while maintaining the utility of MME-FITC antibodies in complex panels .

Mass Cytometry (CyTOF):
While traditional FITC cannot be used directly in mass cytometry, new metal-tagged anti-FITC antibodies allow researchers to leverage existing FITC-conjugated antibodies in CyTOF experiments. This enables integration of MME detection into highly multiplexed panels (40+ parameters) without fluorescence spillover concerns, particularly valuable for comprehensive immune phenotyping.

Imaging Mass Cytometry:
This technology combines the high-parameter capabilities of mass cytometry with spatial resolution, allowing researchers to visualize MME expression in tissue contexts with unprecedented multiplexing capacity and spatial information retention.

Single-Cell Multiomics:
Integration of protein expression data (using MME-FITC antibodies) with transcriptomic or genomic information at single-cell resolution provides multidimensional insights into MME biology. Technologies like CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) enable simultaneous measurement of MME protein expression and whole-transcriptome profiles in the same cells.

Super-Resolution Microscopy:
Techniques such as STORM and PALM can resolve FITC-labeled structures below the diffraction limit, enabling researchers to study MME distribution at nanoscale resolution and investigate co-localization with other membrane proteins at unprecedented detail.

As these technologies mature, they will enable increasingly sophisticated analysis of MME expression and function in complex biological systems.

How might MME-FITC antibody applications evolve in clinical and translational research?

MME-FITC antibodies are positioned to play expanding roles in clinical and translational research with several promising developments on the horizon:

Liquid Biopsy Applications:
MME-FITC antibodies are increasingly being integrated into multiparameter panels for detecting circulating tumor cells (CTCs) in peripheral blood samples. This minimally invasive approach may enable real-time monitoring of disease progression and treatment response in MME-expressing malignancies, particularly in hematological disorders and certain solid tumors with MME expression.

Theranostic Approaches:
Dual-function applications combining MME-targeted diagnostics with therapeutic capabilities are emerging. Modified MME-FITC antibodies can simultaneously identify MME-expressing cells and deliver therapeutic payloads or serve as imaging agents for surgical navigation, representing a shift toward personalized medicine.

Standardized Clinical Flow Cytometry:
Efforts to standardize MME-FITC antibody use in clinical flow cytometry are advancing through initiatives like EuroFlow and ICCS. These aim to establish uniform protocols, reference standards, and reporting metrics, facilitating multi-center studies and improving diagnostic consistency across institutions .

High-Throughput Screening:
Automated platforms integrating MME-FITC antibody detection with machine learning algorithms are being developed for rapid screening and classification of patient samples. These systems promise to increase throughput while maintaining diagnostic accuracy in clinical settings.

Point-of-Care Testing: Simplified, miniaturized flow cytometry systems utilizing MME-FITC antibodies may enable rapid, bedside testing for conditions requiring MME expression analysis, particularly in resource-limited settings where traditional flow cytometry infrastructure is unavailable.

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.