SCNN1D Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

SCNN1D Antibody Basics

SCNN1D (delta-ENaC) is a nonvoltage-gated sodium channel subunit involved in epithelial sodium reabsorption, alveolar fluid clearance, and cellular migration . Antibodies targeting SCNN1D are primarily polyclonal (goat or rabbit) and unconjugated, requiring HRP-conjugated secondary antibodies for detection in techniques like Western blot (WB) and immunohistochemistry (IHC).

Biological Functions

  • Alveolar Fluid Clearance: SCNN1D regulates alveolar type 2 (AT2) cell proliferation and differentiation, critical for lung fluid homeostasis .

  • Cell Migration and Proliferation: Overexpression in glioma cells inhibits migration via MAPK signaling, while knockdown enhances metastasis in prostate cancer .

  • Tissue Expression: Widely expressed in epithelial (lung, kidney) and nonepithelial (brain, heart) tissues, with splice variants contributing to functional diversity .

Pathological Associations

  • Genetic Deficiencies: Linked to developmental disorders in the brain, heart, and respiratory systems .

  • Cancer Progression: TMEM100 regulates SCNN1D expression, modulating prostate cancer cell growth and invasion .

Western Blot and Immunohistochemistry

  • WB Data:

    • A100322: Detects SCNN1D in HT29 cell lysates (1:500–1:1000 dilution) .

    • BS74428: Validated in human, mouse, and rat cell lines (1:1000–1:2000) .

  • IHC Data:

    • A285998: Stains human uterus tissue (2.5 µg/ml), confirming epithelial expression .

Immunofluorescence (IF)

  • A99506: Localizes SCNN1D in A549 cells (1:100–1:500), with peptide-blocking controls confirming specificity .

HRP-Conjugated Detection Systems

HRP-conjugated secondary antibodies enable colorimetric or chemiluminescent detection. For example:

  1. WB Protocol:

    • Primary antibody (e.g., A100322 at 1:1000) incubated overnight.

    • Secondary antibody (e.g., donkey anti-rabbit HRP at 1:10,000) applied for 1 hour.

    • Signal detected via ECL reagents .

  2. ELISA Protocol:

    • SCNN1D-coated plates probed with primary antibody (e.g., A285998 at 1:16,000).

    • HRP-conjugated secondary antibody binds, and TMB substrate generates a measurable color change .

Table 2: SCNN1D Research Highlights

FunctionExperimental ModelKey FindingSource
Alveolar fluid clearanceHumanized transgenic miceSCNN1D regulates AT2 cell proliferation and sodium transport .
Prostate cancer progressionPCa cell lines (e.g., PC-3)TMEM100 upregulates SCNN1D, inhibiting cell migration and invasion .
Glioma cell migrationGlioma cell culturesSCNN1D overexpression reduces migration via MAPK pathway inhibition .

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Orders are typically dispatched within 1-3 business days. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
Amiloride-sensitive sodium channel subunit delta (Delta-NaCH) (Epithelial Na(+) channel subunit delta) (Delta-ENaC) (ENaCD) (Nonvoltage-gated sodium channel 1 subunit delta) (SCNED), SCNN1D, DNACH
Target Names
SCNN1D
Uniprot No.

Target Background

Function

This antibody targets the epithelial sodium channel subunit delta (ENaCδ), a non-voltage-sensitive sodium-permeable ion channel inhibited by the diuretic amiloride. ENaCδ mediates the electrodiffusion of luminal sodium (and osmotically coupled water) across the apical membrane of epithelial cells. It plays a crucial role in sodium reabsorption in the kidneys, colon, lungs, and sweat glands, and also contributes to taste perception.

Gene References Into Functions
References on ENaCδ Function and Regulation:
  1. Low delta-ENaC expression in NCI-H441, Calu-3, and ATI cells indicates minimal contribution to transepithelial sodium absorption. (PMID: 25677639)
  2. Characterization of a novel delta2 ENaC variant in lung epithelial cells, exhibiting distinct biophysical, pharmacological, regulatory, and protein lifespan properties compared to delta1betagamma ENaC. (PMID: 22505667)
  3. Delta-ENaC undergoes dynamin-independent endocytosis, unlike alphabetagamma-channels. (PMID: 22159085)
  4. COMMD1 downregulates deltaENaC activity by reducing surface expression through internalization to an intracellular recycling pool, potentially via enhanced ubiquitination. (PMID: 21741370)
  5. CPT-cAMP activates human epithelial sodium channels by relieving self-inhibition. (PMID: 21419751)
  6. CPT-cGMP stimulates human alveolar fluid clearance by releasing external Na+ self-inhibition of ENaC. (PMID: 21562313)
  7. SGK1.1 regulates delta-ENaC via a non-classical pathway, potentially integrating multiple signaling pathways converging on the channel. (PMID: 20631247)
  8. Delta ENaC is regulated by Murr1. (PMID: 14645214)
  9. ENaCδ activity is regulated by protons and may contribute to pH sensation and/or regulation in the human brain. (PMID: 14726523)
  10. ENaCδ is chemically activated by capsazepine via a proton-dependent mechanism. (PMID: 15308635)
  11. Analysis of a novel spliced variant of the ENaC delta-subunit in the human brain. (PMID: 16930535)
  12. ENaCδ isoforms are widely expressed in pyramidal cells of the human and monkey cerebral cortex and various telencephalic subcortical nuclei neuronal populations. (PMID: 17472699)
  13. Strong expression of ENaCδ transcript in human melanoma cells, confirmed via reverse transcription-polymerase chain reaction and in situ hybridization. (PMID: 18073141)
  14. ENaCδ is a potential pH sensor in the human gastrointestinal system, suggesting a novel therapeutic target for gastro-oesophageal reflux disease. (PMID: 18951889)
  15. The delta-subunit of ENaC alters proteolytic channel activation and enhances baseline channel activity. (PMID: 19717556)
  16. Delta ENaC functions as a proton sensor. (PMID: 15084585)
Database Links

HGNC: 10601

OMIM: 601328

KEGG: hsa:6339

STRING: 9606.ENSP00000368411

UniGene: Hs.512681

Protein Families
Amiloride-sensitive sodium channel (TC 1.A.6) family, SCNN1D subfamily
Subcellular Location
Cell membrane; Multi-pass membrane protein.

Q&A

What is SCNN1D and why is it important in epithelial research?

SCNN1D (Sodium Channel, Nonvoltage-Gated 1, delta) is a critical component of epithelial sodium channels that functions as a sodium permeable non-voltage-sensitive ion channel inhibited by the diuretic amiloride. The protein mediates electrodiffusion of luminal sodium (and water, which follows osmotically) through the apical membrane of epithelial cells. SCNN1D plays essential roles in controlling sodium reabsorption in various tissues including kidney, colon, lung, and sweat glands. Additionally, it contributes to taste perception mechanisms . Research on SCNN1D is particularly valuable for understanding epithelial transport physiology, sodium homeostasis, and related pathologies such as hypertension, cystic fibrosis, and certain taste disorders. When designing experiments targeting SCNN1D, researchers should consider its tissue-specific expression patterns and regulatory mechanisms to ensure physiologically relevant findings.

What applications are most suitable for HRP-conjugated SCNN1D antibodies?

HRP-conjugated SCNN1D antibodies are particularly well-suited for the following applications:

  • Enzyme-Linked Immunosorbent Assay (ELISA): These antibodies provide direct detection without requiring secondary antibodies, streamlining experimental workflows and potentially reducing background signal .

  • Immunohistochemistry on paraffin-embedded sections (IHC-P): Recommended dilutions are typically in the range of 1:100-500 for optimal staining results .

  • Western Blotting: While not explicitly mentioned for all HRP-conjugated versions, these antibodies can be used for direct detection in protein blotting applications, potentially offering greater sensitivity compared to two-step detection systems.

For specialized applications such as immunoprecipitation, researchers should consider whether the HRP conjugation might interfere with antigen binding or protein complex formation. Alternative unconjugated antibodies targeting the same epitope may be more suitable in such cases .

How should I validate a new SCNN1D antibody before incorporating it into my research protocols?

Rigorous validation of SCNN1D antibodies is essential, as research has identified issues with commercial antibody specificity. Implement the following validation protocol:

  • Positive and Negative Controls: Use tissue or cell types known to express or lack SCNN1D. Lung and kidney tissues are appropriate positive controls based on documented expression patterns .

  • Cross-Reactivity Testing: Analyze potential cross-reactivity with other ENaC subunits by examining band patterns in western blots. Be particularly vigilant about non-specific bands around 80 kDa that have been documented with some commercial antibodies .

  • Immunoprecipitation Validation: Perform immunoprecipitation followed by western blotting with different antibodies targeting the same protein to confirm specificity. This approach has been shown to eliminate off-target signals while preserving detection of legitimate target proteins .

  • Dilution Series Analysis: Create a dilution series of your lysate to establish the antibody's linear detection range and optimal working concentration, ensuring quantitative reliability .

  • Epitope Mapping: Consider the specific epitope recognized by your antibody (e.g., AA 292-519 for some SCNN1D antibodies) and how this might affect detection of processed or modified forms of the protein .

What are the recommended storage and handling protocols for maintaining HRP-conjugated SCNN1D antibody activity?

To maintain optimal activity of HRP-conjugated SCNN1D antibodies:

  • Storage Temperature: Store antibodies at -20°C for long-term storage, avoiding repeated freeze-thaw cycles which can degrade both the antibody and the HRP conjugate.

  • Working Aliquots: Prepare small working aliquots to minimize freeze-thaw cycles. When thawed, store at 4°C for short-term use (typically 1-2 weeks).

  • Buffer Considerations: Ensure storage buffers contain appropriate stabilizers for both the antibody and HRP components. Many commercial preparations include glycerol and protein stabilizers.

  • Light Sensitivity: Protect HRP-conjugated antibodies from prolonged exposure to light, as this can compromise enzymatic activity.

  • Reducing Agents: Avoid buffers containing strong reducing agents or high concentrations of sodium azide (>0.1%), as these can interfere with HRP activity.

Regular validation of antibody performance through control experiments is recommended, particularly when using antibodies that have been stored for extended periods, to ensure sensitivity and specificity are maintained.

How can I optimize immunohistochemistry protocols for SCNN1D detection in different tissue types?

Optimizing IHC protocols for SCNN1D requires tissue-specific considerations:

  • Epitope Retrieval Optimization: For SCNN1D detection in epithelial tissues, compare heat-induced epitope retrieval methods using citrate buffer (pH 6.0) versus EDTA buffer (pH 9.0) to determine which provides optimal signal-to-noise ratio for your specific tissue and fixation method.

  • Blocking Protocol Customization: For tissues with high endogenous peroxidase activity (e.g., lung), implement a two-step blocking protocol:

    • 3% hydrogen peroxide in methanol for 15 minutes to block endogenous peroxidases

    • 5% normal serum (matched to secondary antibody species) with 1% BSA for 60 minutes to minimize non-specific binding

  • Dilution Optimization Table:

Tissue TypeRecommended Starting DilutionIncubation ConditionsSpecial Considerations
Kidney1:2004°C overnightPossible high background in proximal tubules
Lung1:1004°C overnightEnhanced sensitivity with tyramide amplification
Colon1:300Room temp, 2 hoursPotential cross-reactivity with other ENaC subunits
Taste buds1:504°C, 48 hoursRequires longer incubation for adequate penetration
  • Control Sections: Always include serial sections treated with isotype-matched control antibodies at equivalent concentrations to identify non-specific binding .

What strategies can address non-specific bands when using SCNN1D antibodies in western blotting?

Non-specific bands are a documented challenge with SCNN1D antibodies. Implement these advanced troubleshooting strategies:

  • Immunoprecipitation Prior to Western Blotting: Research has demonstrated that immunoprecipitation with anti-SCNN1D antibodies prior to western blotting can effectively eliminate off-target bands (particularly the problematic 80 kDa band) while preserving detection of the authentic 95 kDa full-length SCNN1D protein .

  • Modified Blocking Protocols: Test different blocking agents (milk vs. BSA) and concentrations (3-5%) to minimize non-specific binding. For some tissues, casein-based blockers may provide superior results compared to traditional milk-based blockers.

  • Gradient Gel Electrophoresis: Employ 4-15% gradient gels to achieve better separation of proteins in the 80-100 kDa range, helping to distinguish between SCNN1D and potential cross-reactive proteins.

  • Peptide Competition Assays: Perform parallel western blots with antibody pre-incubated with immunizing peptide. Legitimate SCNN1D bands should disappear while non-specific bands typically remain.

  • Comparison Across Multiple Antibodies: Use multiple antibodies targeting different SCNN1D epitopes to confirm band identity. True SCNN1D bands should be consistently detected across antibodies targeting different regions of the protein .

How can I reliably quantify SCNN1D protein expression levels using HRP-conjugated antibodies?

For reliable quantification of SCNN1D using HRP-conjugated antibodies:

  • Establish Linear Detection Range: Determine the linear quantification range through serial dilution experiments. Research has demonstrated that SCNN1D antibodies can provide a linear detection range between 2.5-80 μg of total protein when used for western blotting of lung tissue .

  • Normalization Strategy: Use multiple housekeeping proteins for normalization rather than relying on a single reference protein. Select normalization proteins whose expression is stable under your experimental conditions.

  • Signal Acquisition Parameters:

    • For chemiluminescent detection, capture multiple exposure times to ensure measurements are made within the linear range of detection

    • For colorimetric detection, implement standardized development times and image capture settings

  • Control for Post-Translational Modifications: SCNN1D undergoes proteolytic processing, potentially yielding both full-length (~95 kDa) and cleaved forms (~30 kDa). Some antibodies may detect only the full-length form while missing cleaved products, potentially skewing quantification .

  • Statistical Validation: Perform technical replicates (minimum n=3) and calculate coefficient of variation (CV) to ensure reproducibility. Acceptable CV values should be <15% for reliable quantification.

What are the optimal immunoprecipitation protocols for SCNN1D using HRP-conjugated antibodies?

While HRP conjugation can potentially interfere with immunoprecipitation efficiency, modified protocols can be implemented:

  • Recommended IP Protocol for HRP-Conjugated Antibodies:

    a. Homogenize 20 mg of tissue in 500 μL Goldstein buffer
    b. Pre-clear with 50 μL protein G beads on ice for 45 minutes
    c. Remove beads and save 3% of lysate as input control
    d. Incubate remaining lysate with 2 μg anti-SCNN1D HRP-conjugated antibody and 40 μL protein G beads overnight at 4°C on an end-to-end rotator
    e. Perform extensive washing with PBS (minimum 5 washes)
    f. Elute proteins with 30 μL Laemmli sample buffer

  • Critical Considerations:

    • The HRP conjugate may reduce binding efficiency to protein G; if poor results are obtained, consider using a bridging antibody approach

    • Include appropriate negative controls (beads only, irrelevant HRP-conjugated antibody of same isotype)

    • For detection of immunoprecipitated material, avoid using the same antibody for detection if possible, as this can lead to detection of heavy and light chains that may interfere with visualization of target proteins

  • Alternative Approach: If direct immunoprecipitation with HRP-conjugated antibodies proves challenging, consider an unconjugated version of the same antibody clone for immunoprecipitation, followed by detection with the HRP-conjugated version in western blotting.

How does HRP conjugation affect epitope recognition and sensitivity compared to unconjugated SCNN1D antibodies?

The HRP conjugation process can impact antibody performance in several important ways:

  • Epitope Accessibility Effects: HRP (approximately 44 kDa) conjugated to antibodies can potentially cause steric hindrance, particularly affecting recognition of epitopes in structurally constrained regions of SCNN1D. This effect varies based on the specific conjugation chemistry and the epitope targeted.

  • Sensitivity Comparison Table:

ApplicationHRP-Conjugated AdvantageUnconjugated AdvantageRecommended Approach
ELISADirect detection, fewer stepsNot applicableHRP-conjugated preferred
Western BlotSingle-step detection, reduced backgroundGreater flexibility with detection systemsBoth viable; HRP-conjugated offers workflow efficiency
IHC-PSimplified protocolAmplification options with secondary systemsUnconjugated often preferred for low-abundance targets
IPNot recommendedStandard approachUnconjugated strongly preferred
  • Signal Amplification Considerations: While direct HRP conjugation eliminates the signal amplification provided by secondary antibody systems, this can be advantageous when working with highly abundant targets where oversaturation is a concern.

  • Storage Stability Differences: HRP-conjugated antibodies typically exhibit reduced shelf-life compared to unconjugated counterparts, necessitating more careful storage and handling procedures.

  • Batch-to-Batch Variation: HRP conjugation processes can introduce additional variability between manufacturing lots. Researchers conducting long-term studies should consider securing sufficient quantities from single lots when possible .

What controls should be implemented when using SCNN1D HRP-conjugated antibodies in multi-color immunofluorescence studies?

For rigorous multi-color immunofluorescence experiments with SCNN1D:

  • Essential Controls:

    • Single primary antibody controls to assess bleed-through between channels

    • Isotype-matched control antibodies at equivalent concentrations to identify non-specific binding

    • Secondary-only controls to evaluate background staining

    • Peptide competition controls to confirm specificity of SCNN1D staining

  • SCNN1D-Specific Considerations:

    • Include co-staining with markers for specific epithelial cell types to confirm expected localization patterns

    • Consider dual-staining with antibodies against other ENaC subunits (α, β) to verify co-localization patterns

    • When studying tissues with potential cross-reactivity issues, implement absorption controls with related proteins

  • Quantification Validation: For quantitative immunofluorescence studies, verify linear response range through titration experiments with tissues expressing different levels of SCNN1D.

  • Microscopy Settings: Establish consistent acquisition parameters based on brightest samples to prevent saturation while enabling detection in low-expression samples. Document all microscope settings comprehensively for reproducibility .

How can I design experiments to distinguish between total and cell-surface expressed SCNN1D?

Differentiating between total cellular SCNN1D and the functional cell-surface pool requires specialized approaches:

  • Cell Surface Biotinylation Protocol:

    • Biotinylate intact cells/tissues using membrane-impermeable sulfo-NHS-SS-biotin

    • Lyse cells and perform streptavidin pulldown to isolate surface proteins

    • Analyze SCNN1D content in total lysate versus biotinylated fraction by western blotting

    • Calculate surface expression as the ratio of biotinylated to total SCNN1D

  • Immunofluorescence Co-localization Analysis:

    • Perform dual immunofluorescence with SCNN1D antibodies and established membrane markers

    • Analyze co-localization coefficients (Pearson's or Manders') to quantify membrane association

    • Complement with Z-stack analysis to confirm apical membrane localization in polarized cells

  • Protease Protection Assays: For cultured cells, selective trypsinization of cell surface proteins before lysis can distinguish between protected intracellular pools and vulnerable surface-expressed channels.

  • Surface-Specific Labeling: When studying transfected constructs, incorporate extracellular epitope tags (e.g., FLAG, HA) that can be specifically labeled in non-permeabilized cells to quantify surface expression .

What experimental approaches can address contradictory results obtained with different SCNN1D antibodies?

Resolving discrepancies between different SCNN1D antibodies requires systematic investigation:

  • Comprehensive Antibody Comparison:

    • Map the specific epitopes recognized by each antibody

    • Test each antibody against recombinant SCNN1D fragments covering different domains

    • Compare reactivity profiles across multiple tissues and species

  • Combined Methodological Approach:

    • Implement orthogonal detection methods (e.g., mass spectrometry) to verify protein identity

    • Use genetic approaches (siRNA knockdown, CRISPR knockout) to confirm specificity

    • Perform reciprocal immunoprecipitation experiments with different antibodies to evaluate concordance

  • Processing-Dependent Recognition Analysis:

    • Investigate whether differences relate to detection of processed versus full-length forms

    • Test samples under reducing and non-reducing conditions to assess structure-dependent epitope accessibility

    • Consider deglycosylation experiments to eliminate potential differences due to post-translational modifications

  • Cross-Validation Strategy: When critical findings depend on antibody specificity, validate with at least two independent antibodies recognizing different epitopes, or complement with non-antibody-based approaches such as RT-PCR or in situ hybridization.

How can I address inconsistent SCNN1D detection across different tissue preparations?

Variations in SCNN1D detection across tissue types often stem from preparation-specific factors:

  • Fixation Optimization Matrix:

Tissue TypeOptimal FixativeFixation TimeSpecial Considerations
Kidney4% PFA24 hoursPost-fix washing critical
Lung2% PFA12-18 hoursInflation fixation improves epithelial preservation
ColonMethacarn6-12 hoursSuperior mucin preservation
Taste buds4% PFA4-6 hoursDecalcification may affect epitope
  • Antigen Retrieval Customization:

    • For formalin-fixed tissues, test both heat-mediated (citrate pH 6.0 vs. EDTA pH 9.0) and enzymatic (proteinase K) retrieval methods

    • For each tissue type, optimize retrieval duration (10-30 minutes) and temperature (95-120°C)

    • Consider dual retrieval approaches for challenging samples

  • Sample-Specific Protocol Adjustments:

    • For mucin-rich tissues, include additional washing steps with detergent-containing buffers

    • For highly vascularized tissues, implement hydrogen peroxide block before antibody incubation

    • For tissues with high lipid content, consider detergent permeabilization optimization

What strategies can optimize detection of both full-length and processed forms of SCNN1D?

SCNN1D undergoes proteolytic processing, creating detection challenges that require specialized approaches:

  • Antibody Selection Strategy:

    • Choose antibodies targeting epitopes preserved in both full-length (~95 kDa) and cleaved (~30 kDa) forms

    • Consider using multiple antibodies targeting different regions in parallel experiments

    • Verify epitope accessibility in processed forms through recombinant protein standards

  • Gel System Optimization:

    • Utilize gradient gels (4-20%) to simultaneously resolve both high and low molecular weight forms

    • For focused analysis of cleavage products, consider tricine-SDS-PAGE systems optimized for low molecular weight proteins

    • Adjust running conditions (voltage/time) to enhance separation in regions of interest

  • Immunoprecipitation Approach: Research has demonstrated that some antibodies can immunoprecipitate both full-length and cleaved SCNN1D forms but may only detect the full-length form in direct western blotting. Consider a two-antibody approach: immunoprecipitate with one antibody and detect with another that recognizes both forms .

  • Processing Control: Include samples treated with protease inhibitors to establish baseline levels of unprocessed channels for comparison.

How does sample preparation affect quantitative analysis of SCNN1D using HRP-conjugated antibodies?

Sample preparation critically influences quantitative SCNN1D analysis:

  • Optimization of Extraction Conditions:

    • Compare different lysis buffers (RIPA vs. Triton X-100 vs. Goldstein buffer) to determine optimal SCNN1D extraction efficiency

    • Evaluate the impact of detergent concentration (0.1-1%) on membrane protein solubilization

    • Test the effect of sonication parameters on SCNN1D recovery and integrity

  • Preserving Post-Translational Modifications:

    • Include appropriate phosphatase inhibitors to maintain phosphorylation status

    • Add protease inhibitor cocktails to prevent artificial processing during sample preparation

    • Consider rapid denaturation methods to "freeze" the native modification state

  • Standardization Requirements:

    • Implement consistent sample-to-buffer ratios across experimental groups

    • Standardize protein determination methods and loading amounts

    • Process all comparative samples simultaneously to minimize technical variation

  • Linear Quantification Validation: Research demonstrates that ENaC subunit antibodies provide linear detection across a specific protein concentration range (2.5-80 μg). Ensure your loading amounts fall within this verified linear range for accurate quantification .

How can SCNN1D HRP-conjugated antibodies be applied in multi-omics research approaches?

Integrating SCNN1D antibodies into multi-omics workflows offers powerful new research possibilities:

  • Chromatin Immunoprecipitation followed by Sequencing (ChIP-seq):

    • Utilize HRP-conjugated antibodies against transcription factors regulating SCNN1D expression

    • Implement sequential ChIP approaches to identify co-regulatory factor interactions

    • Correlate binding profiles with expression data to establish regulatory mechanisms

  • Proximity Labeling Proteomics:

    • Convert HRP-conjugated antibodies into proximity labeling tools using biotin-phenol substrates

    • Map the SCNN1D interactome in different cellular compartments

    • Identify novel interacting partners that may regulate channel function or trafficking

  • Single-Cell Analysis Integration:

    • Correlate antibody-based SCNN1D protein quantification with scRNA-seq data

    • Implement spatial transcriptomics approaches with SCNN1D immunohistochemistry

    • Develop computational frameworks to integrate protein and transcript data at single-cell resolution

  • Multi-Modal Imaging:

    • Combine HRP-based detection with other imaging modalities (electron microscopy, super-resolution)

    • Implement correlative light and electron microscopy to map SCNN1D distribution at ultrastructural level .

What are the current limitations in SCNN1D antibody technology and how might they be addressed?

Current limitations and potential solutions in SCNN1D antibody technology include:

  • Specificity Challenges:

    • Issue: Non-specific bands (~80 kDa) observed with some commercial antibodies

    • Solution: Development of monoclonal antibodies targeting unique SCNN1D epitopes

    • Emerging Approach: Recombinant antibody technology with enhanced epitope validation

  • Cross-Species Reactivity Limitations:

    • Issue: Many antibodies show human-specific reactivity, limiting comparative studies

    • Solution: Design of antibodies targeting evolutionarily conserved epitopes

    • Alternative: Species-specific antibody panels with matched epitopes for comparative research

  • Post-Translational Modification Detection:

    • Issue: Limited availability of modification-specific antibodies (phosphorylation, glycosylation)

    • Solution: Development of site-specific phospho-antibodies for regulatory sites

    • Future Direction: Antibodies specifically recognizing activated/cleaved channel forms

  • Quantification Standardization:

    • Issue: Variation in quantification approaches between studies limits comparability

    • Solution: Development of recombinant SCNN1D standards for absolute quantification

    • Best Practice: Implementation of digital PCR alongside protein quantification for correlation analysis .

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.