LCE3C Antibody

Shipped with Ice Packs
In Stock

Description

Applications of LCE3C Antibodies

LCE3C antibodies are utilized in diverse experimental contexts:

  • Immunodetection: ELISA, Western Blot (primary applications) .

  • Localization Studies: Immunohistochemistry (IHC) and immunofluorescence (IF) to map LCE3C expression in stratified epithelia .

  • Disease Research: Investigating psoriasis pathogenesis due to LCE3B/LCE3C deletion-associated risk .

Role in Psoriasis

  • Genetic Risk: Homozygous deletion of LCE3B and LCE3C increases psoriasis susceptibility by 1.3–1.5-fold .

  • Expression Dynamics:

    • Basal Conditions: Minimal expression in normal skin.

    • Induced States: Upregulated in lesional psoriatic epidermis and post-injury repair .

  • Functional Impact: Compromised skin barrier and impaired antimicrobial defense (e.g., against Staphylococcus aureus) .

Broader Clinical Implications

  • Autoimmune Links: Meta-analyses suggest associations with rheumatoid arthritis (OR = 1.31, P < 0.0001) .

  • Antimicrobial Activity: LCE3 proteins, including LCE3C, exhibit defensin-like properties against gram-positive/negative bacteria (MIC: 1–10 µM) .

Available Products

SupplierProduct TypeApplicationsReactivity
Biocompare Polyclonal/MonoclonalELISA, Western BlotHuman
Ximbio Monoclonal (Clone 7)ELISA, IHC, IFHuman
Protein Atlas Not specifiedIHC, IFHuman

Example Antibody Profile (Ximbio)

  • Host Species: Mouse

  • Immunogen: LCE3B fragment peptide

  • Cross-Reactivity: Pan-LCE3 (detects LCE3B, LCE3D, LCE3E)

  • Key Use: Studying epidermal differentiation in reconstructed skin models .

Mechanistic Insights from Studies

  • Epidermal Localization: LCE3C colocalizes with LCE2 proteins in the upper stratum granulosum, suggesting synergistic roles in terminal differentiation .

  • Barrier Repair: Induced by pro-inflammatory cytokines (e.g., IL-17, IL-22) during skin injury, linking it to psoriasis flare-ups .

Limitations and Future Directions

  • Specificity Challenges: High homology among LCE proteins complicates isoform-specific detection .

  • Therapeutic Potential: Modulating LCE3C expression or function could address barrier defects in psoriasis, though clinical validation is pending .

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 orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery times.
Synonyms
LCE3C antibody; LEP15 antibody; SPRL3ALate cornified envelope protein 3C antibody; Late envelope protein 15 antibody; Small proline-rich-like epidermal differentiation complex protein 3A antibody
Target Names
LCE3C
Uniprot No.

Target Background

Function
LCE3C is a structural component of the cornified envelope of the stratum corneum, playing a crucial role in innate cutaneous host defense. It exhibits defensin-like antimicrobial activity against a wide range of Gram-positive and Gram-negative bacteria, including both aerobic and anaerobic species. During inflammation, LCE3C potentially regulates skin barrier repair by influencing cutaneous microbiota composition and immune response to bacterial antigens.
Gene References Into Functions
  1. Analysis of disease variants within the LCE3 cluster revealed associations with psoriasis in patients from India. PMID: 27048876
  2. Our findings suggest that the previously identified LCE deletion in psoriasis patients is not a major contributing factor to the development of Psoriatic Arthritis in Tunisian patients. PMID: 24566688
  3. No evidence of association was found between the LCE3C_LCE3B-del and psoriasis in 34 patients from 7 multiplex Tunisian families. Additionally, no epistatic effect was observed between the deletion and the PSORS1 locus. PMID: 24485035
  4. Our meta-analysis indicates a significant association between psoriasis and the LCE3C_LCE3B-del polymorphism in European and Asian populations, but no association with psoriatic arthritis. PMID: 23631431
  5. The LCE3C_LCE3B-del might contribute to the development of familial psoriasis in the Tunisian population. PMID: 22926764
  6. Paediatric-onset psoriasis has been associated with the LCE3C_LCE3B deletion. PMID: 22512642
  7. The LCE3C_LCE3B-del is a common risk factor for autoimmune diseases. PMID: 22384135
  8. Our findings suggest that the Koebner phenomenon in psoriasis is unlikely to be influenced by the LCE3B/C genotype. PMID: 22048733
  9. The LCE3C_LCE3B-del has been identified as a significant risk factor for psoriasis. Furthermore, our research indicates that the LCE3C_LCE3B-del does not exhibit an epistatic effect with the HLA-Cw6 allele on psoriasis susceptibility in northern Chinese populations. PMID: 21711330
  10. A deletion of the LCE3B and LCE3C genes may contribute to the development of allergic contact dermatitis. PMID: 21995181
  11. Our study provides evidence for an association between the LCE3C_LCE3B-del and rheumatoid arthritis (RA) in non-Caucasian populations. Additionally, SNPs rs4112788 and rs4085613, tagging the LCE3C_LCE3B-del, have been identified as novel susceptibility factors for systemic lupus erythematosus (SLE). PMID: 21628307
  12. Our study confirms an association between the deletion of LCE3C and LCE3B and psoriasis in a Chinese population. PMID: 21509048
  13. The LCE3C_LCE3B deletion is a susceptibility factor for psoriatic arthritis, highlighting a shared risk factor involving the epidermal skin barrier in autoimmune disorders. PMID: 21400479
  14. The LCE3B/C deletion may play a role in psoriasis. PMID: 21435436
  15. The deletion of LCE3C and LCE3B is a prevalent genetic factor associated with susceptibility to psoriasis in European, Chinese, and Mongolian populations. PMID: 21107349
  16. Meta-analysis of gene-disease association. (HuGE Navigator) PMID: 20624365
  17. No genetic association was found between LCE3B and LCE3c deletions and atopic dermatitis. PMID: 20376060
  18. Observational study of gene-disease association. (HuGE Navigator) PMID: 20376060
  19. Our research has identified a pleiotropic effect of a common genetic risk factor (LCE3C_LCE3B-del) for autoimmune diseases, implicated in both psoriasis and rheumatoid arthritis. PMID: 20213803
  20. Our work suggests that homozygosity for a common LCE3C_LCE3B deletion contributes to the risk of developing chronic plaque-type psoriasis without psoriatic arthritis. PMID: 20331852
  21. Our study confirms the previously published findings indicating that the deletion of the two LCE genes is a susceptibility factor for psoriasis vulgaris with a dosage effect. PMID: 20016497
  22. Observational study of gene-disease association. (HuGE Navigator) PMID: 20213803
  23. Observational study of gene-disease association. (HuGE Navigator) PMID: 20331852
  24. The LCE3C_LCE3B-del exhibits epistatic effects with the HLA-Cw6 allele on the development of psoriasis. PMID: 19169253
  25. Observational study of gene-disease association and gene-gene interaction. (HuGE Navigator) PMID: 20016497
  26. Observational study of gene-disease association and gene-gene interaction. (HuGE Navigator) PMID: 19169253
  27. This paper describes nomenclature changes, expression in a range of tissues, and responses to UV exposure. PMID: 15854049

Show More

Hide All

Database Links

HGNC: 16612

OMIM: 612615

KEGG: hsa:353144

STRING: 9606.ENSP00000334644

UniGene: Hs.490211

Protein Families
LCE family
Tissue Specificity
Skin-specific. Expression was readily detected in adult trunk skin, adult arm skin, fetal skin, penal skin, vulva, esophagus and tongue. Not expressed in the cervix, rectum, lung, colon, or placenta.

Q&A

What is LCE3C and what is its biological significance?

LCE3C (Late Cornified Envelope 3C) is a structural protein component of the cornified envelope of the stratum corneum. It belongs to the LCE protein family, with synonyms including late envelope protein 15, small proline-rich-like epidermal differentiation complex protein 3A, and late cornified envelope protein 3C . The canonical human LCE3C protein has a reported length of 94 amino acid residues and a mass of 9.7 kDa with skin-specific expression . LCE proteins have been shown to possess antibacterial properties against both gram-positive and gram-negative bacteria, as well as aerobic and anaerobic species, suggesting a role in innate cutaneous host defense . During inflammation, LCE3 proteins are involved in skin repair processes, highlighting their importance in maintaining epidermal homeostasis .

What is the relationship between LCE3C gene deletion and disease risk?

The deletion of LCE3B and LCE3C genes (LCE3C_LCE3B-del) has been identified as a significant risk factor for psoriasis . This 32kb deletion is ancient and affects both genes simultaneously . Recent studies have also investigated its association with rheumatoid arthritis (RA). A meta-analysis combining data from Spanish, Chinese, and Dutch populations demonstrated a significant association between LCE3C_LCE3B-del and RA risk (p<0.0001, OR 1.31, 95% CI: 1.16-1.47) . This association appeared strongest in rheumatoid factor (RF) positive patients (p=0.0007, OR 1.27, 95% CI: 1.11-1.45) . The deletion may compromise barrier function, potentially allowing environmental antigens to trigger autoimmune responses in genetically susceptible individuals.

What detection methods are available for studying LCE3C expression?

Several detection methods are available for studying LCE3C expression in research settings:

  • Enzyme-Linked Immunosorbent Assay (ELISA): Commonly used for quantitative detection of LCE3C in biological samples .

  • Immunohistochemistry (IHC): Allows visualization of LCE3C protein in tissue sections, providing insights into its localization and expression patterns .

  • Immunofluorescence (IF): Enables higher sensitivity detection and co-localization studies with other epidermal markers .

  • Western Blotting: Used for semi-quantitative analysis of LCE3C protein expression and confirmation of antibody specificity .

When selecting a detection method, researchers should consider the specific research question, sample type, and required sensitivity level. For example, IF might be preferable for co-localization studies, while ELISA would be more appropriate for quantitative analysis across multiple samples.

How should researchers select the appropriate anti-LCE3C antibody for their experiments?

Selecting the appropriate anti-LCE3C antibody requires careful consideration of several factors:

  • Specificity: Determine whether you need an antibody specific to LCE3C or one that recognizes multiple LCE3 family members. For instance, the anti-pan-LCE3 monoclonal antibody (Catalog #160445) recognizes LCE3B, LCE3D, and LCE3E proteins, making it suitable for studies requiring detection of multiple LCE3 proteins .

  • Application compatibility: Verify that the antibody has been validated for your specific application (ELISA, IHC, IF, Western blot) . The documentation should include positive controls and demonstrated specificity.

  • Host species: Consider the host species (e.g., mouse for the pan-LCE3 monoclonal antibody) to avoid cross-reactivity issues when designing multi-color immunostaining protocols .

  • Monoclonal vs. polyclonal: Monoclonal antibodies offer higher specificity but may recognize only one epitope, while polyclonal antibodies might provide higher sensitivity but with potential cross-reactivity .

  • Validated applications: Review the literature and product data sheets to confirm antibody performance in applications similar to your experimental design.

What validation procedures should be performed when using a new LCE3C antibody?

When incorporating a new LCE3C antibody into your research, rigorous validation is essential to ensure reliable results:

  • Positive and negative controls: Include tissues or cells known to express high levels of LCE3C (e.g., differentiated keratinocytes) as positive controls, and tissues where LCE3C is absent as negative controls .

  • Antibody titration: Perform a dilution series to determine the optimal antibody concentration that maximizes specific signal while minimizing background.

  • Peptide competition assay: Pre-incubate the antibody with purified LCE3C peptide prior to staining to confirm binding specificity.

  • Knockout or knockdown validation: If available, utilize LCE3C knockout or knockdown samples to confirm antibody specificity.

  • Cross-reactivity assessment: Test the antibody against other LCE family members, particularly closely related proteins like LCE3B, LCE3D, and LCE3E .

  • Reproducibility testing: Perform replicate experiments to ensure consistent staining patterns across different batches of the same sample.

  • Comparison with alternative detection methods: Corroborate antibody-based detection results with mRNA expression data when possible.

What are the optimal sample preparation methods for LCE3C detection in skin tissues?

Optimal sample preparation for LCE3C detection in skin tissues depends on the specific application but generally follows these methodological guidelines:

  • For immunohistochemistry and immunofluorescence:

    • Fix tissue samples in 4% paraformaldehyde for 24 hours

    • Process and embed in paraffin or optimal cutting temperature (OCT) compound for frozen sections

    • For paraffin sections, perform antigen retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)

    • Block endogenous peroxidase activity (for IHC) and non-specific binding

    • Apply primary antibody at optimized dilution (typically 1:100 to 1:500) and incubate at 4°C overnight

    • Use appropriate detection system based on host species of primary antibody

  • For protein extraction and Western blotting:

    • Extract proteins using RIPA buffer supplemented with protease inhibitors

    • Sonicate samples to ensure complete lysis of the cornified envelope structures

    • Centrifuge at high speed to remove insoluble material

    • Quantify protein concentration using BCA or Bradford assay

    • Denature proteins with reducing agent and heat prior to loading on SDS-PAGE gels

  • For ELISA:

    • Homogenize tissue samples in PBS with protease inhibitors

    • Centrifuge to remove debris

    • Dilute samples appropriately based on expected LCE3C concentration

Proper sample handling is critical as LCE proteins are components of highly cross-linked structures in the cornified envelope, which can affect extraction efficiency and antibody accessibility.

How can researchers distinguish between different LCE3 family members in their experiments?

Distinguishing between different LCE3 family members presents a challenge due to their high sequence homology. Researchers can employ several strategies:

A combination of these approaches provides the most reliable method for distinguishing between closely related LCE3 family members.

What are the proposed molecular mechanisms by which LCE3C deletion contributes to disease pathogenesis?

Several molecular mechanisms have been proposed to explain how LCE3C deletion contributes to disease pathogenesis, particularly in psoriasis and potentially in rheumatoid arthritis:

  • Compromised epidermal barrier function: LCE3 proteins contribute to the structural integrity of the cornified envelope. Deletion of LCE3B and LCE3C may lead to impaired barrier formation, increasing susceptibility to environmental triggers and pathogens .

  • Altered antimicrobial defense: LCE3 proteins possess antibacterial properties against both gram-positive and gram-negative bacteria. Their deletion may compromise cutaneous antimicrobial defense, potentially triggering immune responses to skin microbiota .

  • Dysregulated wound healing: LCE3 proteins are involved in skin repair during inflammation. The absence of LCE3B and LCE3C might impair proper wound healing responses, contributing to chronic inflammation observed in psoriasis .

  • Enhanced antigen penetration: A defective epidermal barrier may facilitate the entry of environmental antigens, potentially triggering autoimmune responses in genetically susceptible individuals, which could explain the association with rheumatoid arthritis .

  • Genetic interaction with HLA risk alleles: Research suggests that LCE3C_LCE3B deletion may interact with HLA risk alleles, potentially amplifying disease susceptibility through synergistic effects on immune responses .

  • Evolutionary selection pressure: Higher nucleotide diversity in the LCE3BC haplotype block compared to neutral regions suggests potential balancing selection, indicating functional importance despite deletion frequency .

Understanding these mechanisms requires integrated approaches combining genetic association studies with functional analyses of barrier integrity, immune responses, and interaction with environmental factors.

How can researchers assess the functional impact of LCE3C gene deletion in experimental models?

Assessing the functional impact of LCE3C gene deletion requires multifaceted experimental approaches:

  • 3D skin equivalents and organoid models:

    • Generate skin equivalents using keratinocytes from individuals with and without LCE3C_LCE3B deletion

    • Assess barrier function using transepidermal water loss measurements

    • Challenge models with microbial pathogens to evaluate antimicrobial defense

    • Induce wounding to study repair mechanisms

  • CRISPR/Cas9-engineered cellular models:

    • Create isogenic cell lines with and without LCE3C deletion

    • Compare differentiation capacity, barrier protein expression, and response to inflammatory stimuli

    • Perform transcriptomic analyses to identify downstream pathways affected by LCE3C deletion

  • Mouse models:

    • Develop knockout models for LCE3C or the entire LCE3 gene cluster

    • Characterize skin barrier function in basal conditions and after challenges

    • Induce psoriasis-like or arthritis-like conditions to assess disease susceptibility

  • Ex vivo skin explants:

    • Collect skin samples from donors with different LCE3C genotypes

    • Perform barrier function assays and immune challenge experiments

    • Measure cytokine responses and epidermal differentiation markers

  • Functional genomic approaches:

    • Conduct chromatin immunoprecipitation sequencing (ChIP-seq) to identify transcription factors affected by LCE3C deletion

    • Perform Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) to assess chromatin accessibility changes

    • Use HiC or similar techniques to evaluate three-dimensional genomic interactions affected by the deletion

These complementary approaches provide a comprehensive understanding of how LCE3C deletion affects epidermal biology and disease susceptibility.

How should researchers interpret contradictory findings regarding LCE3C deletion association studies?

When interpreting contradictory findings in LCE3C deletion association studies, researchers should consider several methodological and biological factors:

  • Population heterogeneity: The LCE3C_LCE3B-del allele frequency varies significantly among different ethnic backgrounds. For instance, the deletion frequency is approximately 55% in Spanish and Chinese controls compared to 61% in Dutch controls . This heterogeneity may contribute to different association strengths across populations.

  • Statistical power considerations: Studies with insufficient sample sizes may fail to detect true associations. For example, a study powered to detect an odds ratio of 1.45 may be underpowered to detect a smaller effect size of 1.31, requiring approximately 1256 samples to achieve 80% power .

  • Disease heterogeneity: Different disease subtypes or endophenotypes may have variable associations with LCE3C deletion. Stratification by serological markers (e.g., anti-CCP and rheumatoid factor in RA) might reveal subgroup-specific associations .

  • Statistical model selection: The choice of genetic model (dominant, recessive, or additive) can affect results. For LCE3C_LCE3B-del, a recessive model has been found to best fit the data in psoriasis studies, which aligns with findings in rheumatoid arthritis research .

  • Environmental interactions: Gene-environment interactions may modify the effect of LCE3C deletion, potentially explaining discrepancies between studies conducted in different geographic regions or patient populations.

  • Methodological differences: Variations in genotyping techniques, quality control procedures, and statistical analysis methods can contribute to discrepant findings.

When confronted with contradictory results, researchers should consider meta-analysis approaches that combine data from multiple studies to increase statistical power and account for between-study heterogeneity .

What factors might contribute to variability in LCE3C antibody staining patterns?

Variability in LCE3C antibody staining patterns can arise from several technical and biological factors:

  • Technical factors:

    • Fixation method and duration: Overfixation can mask epitopes, while underfixation may result in tissue degradation

    • Antigen retrieval protocol: Buffer composition, pH, temperature, and duration affect epitope accessibility

    • Antibody concentration: Suboptimal dilution can lead to weak signals or high background

    • Incubation conditions: Temperature, duration, and buffer composition affect antibody binding kinetics

    • Detection system sensitivity: Various secondary antibodies and visualization reagents have different detection thresholds

    • Counterstaining intensity: Excessive counterstaining may mask specific signals

  • Biological factors:

    • Epidermal differentiation state: LCE3C expression is regulated during keratinocyte differentiation

    • Inflammatory status: Inflammation can alter LCE3 gene expression patterns

    • Genetic variations: LCE3C_LCE3B deletion or copy number variations affect expression levels

    • Tissue sampling location: Expression may vary across different body sites

    • Disease state: Pathological conditions can modify expression patterns

    • Age and sex differences: Demographic factors may influence epidermal differentiation

  • Antibody-specific factors:

    • Cross-reactivity with other LCE family members: Many antibodies recognize multiple LCE proteins, such as the pan-LCE3 antibody that detects LCE3B, LCE3D, and LCE3E

    • Lot-to-lot variability: Different production batches may have slight variations in specificity or sensitivity

    • Storage conditions: Improper storage can lead to antibody degradation and reduced performance

To minimize variability, researchers should standardize protocols, include appropriate controls, and thoroughly document all experimental conditions.

How might single-cell approaches enhance our understanding of LCE3C expression patterns?

Single-cell approaches offer unprecedented resolution for understanding LCE3C expression patterns and function:

  • Single-cell RNA sequencing (scRNA-seq):

    • Reveals cell-specific expression patterns of LCE3C and related genes within heterogeneous skin tissue

    • Identifies co-expression networks and regulatory relationships

    • Tracks dynamic changes during epidermal differentiation and wound healing

    • Compares expression profiles between normal and disease states at single-cell resolution

  • Single-cell proteomics:

    • Detects LCE3C protein levels in individual cells to assess post-transcriptional regulation

    • Evaluates co-expression with other barrier proteins at the protein level

    • Measures variability in protein expression among seemingly identical cell populations

  • Spatial transcriptomics:

    • Maps LCE3C expression within the architectural context of skin layers

    • Identifies spatial relationships between LCE3C-expressing cells and other cell types

    • Compares spatial expression patterns between normal and diseased tissues

  • CyTOF and imaging mass cytometry:

    • Simultaneously measures multiple proteins in single cells

    • Preserves spatial information while providing single-cell resolution

    • Allows correlation of LCE3C expression with cellular activation states

  • Single-cell ATAC-seq:

    • Identifies cell-specific regulatory elements controlling LCE3C expression

    • Reveals heterogeneity in chromatin accessibility among epidermal cells

    • Tracks epigenetic changes associated with differentiation and disease

  • Single-cell multi-omics approaches:

    • Integrates transcriptomic, epigenomic, and proteomic data from the same cells

    • Provides comprehensive understanding of regulatory mechanisms controlling LCE3C expression

    • Identifies causal relationships between genetic variation, epigenetic state, and gene expression

These approaches will help resolve current contradictions in the literature and provide a more nuanced understanding of how LCE3C contributes to skin barrier function and disease pathogenesis.

What are the emerging therapeutic implications of LCE3C research?

Research on LCE3C has revealed several potential therapeutic implications for skin disorders and potentially autoimmune conditions:

  • Barrier enhancement strategies:

    • Development of topical formulations containing recombinant LCE3 proteins to compensate for genetic deficiencies

    • Small molecule enhancers of endogenous LCE expression to strengthen barrier function

    • Peptide mimetics that replicate the antimicrobial properties of LCE3 proteins

  • Gene therapy approaches:

    • CRISPR/Cas9-based correction of LCE3C deletion in keratinocyte stem cells

    • Viral vector delivery of LCE3C to affected skin areas

    • RNA therapeutics to modulate expression of remaining LCE family members to compensate for deletion

  • Personalized medicine applications:

    • Genotyping for LCE3C_LCE3B-del to stratify patients for targeted therapies

    • Combinatorial approaches targeting both LCE-related barrier defects and inflammatory pathways

    • Preventive interventions for high-risk individuals carrying the deletion

  • Diagnostic applications:

    • Development of immunohistochemical panels including anti-LCE3C antibodies for improved disease classification

    • Prognostic biomarkers based on LCE expression patterns in inflammatory skin conditions

    • Non-invasive diagnostic methods to assess barrier function related to LCE status

  • Drug delivery innovations:

    • Targeting drug delivery systems to overcome compromised barrier function in patients with LCE3C deficiency

    • LCE-based vehicles for enhanced transdermal delivery of therapeutic compounds

Future research should focus on translating the growing understanding of LCE biology into clinically applicable therapeutic approaches, particularly for patients with identified LCE3C_LCE3B deletions who may benefit from targeted barrier repair strategies.

What in vitro and in vivo models are most appropriate for studying LCE3C function?

Selecting appropriate research models is crucial for advancing our understanding of LCE3C function:

In vitro models:

  • Primary human keratinocyte cultures:

    • Allow study of LCE3C expression during differentiation

    • Can be derived from donors with different LCE3C genotypes

    • Enable manipulation of expression through transfection or viral transduction

    • Most physiologically relevant cell type for studying epidermal proteins

  • 3D reconstructed human epidermis (RHE):

    • Recapitulates the stratified structure of human epidermis

    • Enables assessment of barrier function parameters

    • Allows for topical application of stimuli or therapeutic agents

    • Can incorporate keratinocytes with different LCE3C genotypes

  • Skin-on-chip models:

    • Integrate multiple skin cell types in a microfluidic environment

    • Allow for dynamic exposure to environmental factors

    • Enable real-time monitoring of barrier function

    • Facilitate high-throughput screening of compounds affecting LCE expression

In vivo models:

  • Genetically modified mouse models:

    • Knockout models for individual LCE genes or clusters

    • Humanized models expressing human LCE variants

    • Inducible models for temporal control of expression

    • Reporter models for visualization of expression patterns

  • Human skin explants:

    • Preserve native tissue architecture and cellular diversity

    • Allow for ex vivo manipulation and treatment

    • Can be obtained from patients with different LCE3C genotypes

    • Limited viability restricts long-term studies

  • Human skin xenografts in immunodeficient mice:

    • Maintain human skin characteristics in an in vivo environment

    • Allow for longer-term studies than explants

    • Enable testing of systemic treatments

    • Can utilize skin from patients with LCE3C deletions

Each model system offers distinct advantages, and researchers should select models based on their specific research questions, considering factors such as species differences in LCE gene organization, barrier properties, and immune responses.

What statistical approaches are most appropriate for analyzing LCE gene deletion association studies?

Appropriate statistical approaches for analyzing LCE gene deletion association studies require careful consideration of genetic models, population structure, and multiple testing:

Researchers should clearly report their statistical methods, adjustments for confounders, and power calculations to facilitate comparison across studies and proper interpretation of results.

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.