SNAI1 (Snail Family Transcriptional Repressor 1) antibodies are specialized immunological tools designed to detect and study the SNAI1 protein, a zinc-finger transcription factor central to epithelial-mesenchymal transition (EMT) in developmental biology and cancer metastasis . These antibodies enable researchers to investigate SNAI1's role in repressing epithelial markers (e.g., E-cadherin), promoting stemness, and driving tumor progression through techniques like western blot, immunohistochemistry (IHC), and immunofluorescence .
SNAI1 operates through multiple interconnected mechanisms:
EMT Regulation: Binds E-box motifs in promoters of epithelial genes (e.g., CDH1) to suppress E-cadherin, facilitating metastasis .
Immune Modulation: Upregulates CD73 in triple-negative breast cancer (TNBC), increasing extracellular adenosine to suppress anti-tumor immunity .
Stemness Maintenance: Represses tumor-suppressive let-7 microRNAs, enhancing cancer stem cell properties .
Commercial SNAI1 antibodies exhibit diverse properties optimized for specific experimental needs:
| Catalog No. | Host | Clonality | Applications | Reactivity |
|---|---|---|---|---|
| ABIN2854294 | Rabbit | Polyclonal | WB, IHC, IF, ICC | Human, Mouse, Rat |
| 13099-1-AP | Rabbit | Polyclonal | WB, IHC, IF, IP, ChIP | Human, Mouse, Rat |
| ABD38 | Rabbit | Polyclonal | WB (1 µg/mL) | Mouse |
| 61367 | Mouse | Monoclonal | WB, IHC | Human |
| ab167609 | Mouse | Polyclonal | WB, IF | Human, Transfected |
| Cancer Type | Stage | OS Hazard Ratio (HR) | PFS HR |
|---|---|---|---|
| STAD | 1 | 3.63* | 2.88* |
| STAD | 2 | 2.03* | 1.93* |
| CRC | All | 1.71† | 1.58† |
Phenotypic Reversal: CRISPR/Cas9-mediated SNAI1 ablation in TNBC cells (MDA-MB-231) restored epithelial markers (CDH1, ZO-1) and sensitized tumors to paclitaxel/doxorubicin .
Immune Infiltration: SNAI1 expression inversely correlates with B-cell infiltration in gastric (STAD) and colorectal (COAD) cancers .
SNAI1 is a validated prognostic marker for TNBC (HR = 2.27 for OS, P < 0.001) and gastrointestinal cancers .
Correlates with TGF-β signaling and immune subtypes (e.g., TGF-β dominant tumors) .
Nanoparticle-Delivered siRNA: Reduces SNAI1 levels in xenograft models, restoring let-7 and inhibiting tumor growth .
HDAC Inhibition: Disrupts SNAI1-HDAC1 complexes at the CDH1 promoter, reversing EMT in lens epithelial cells .
Western Blot: Use 10–30 µg lysate with 1:500–1:3000 dilution; expected band size: 29–35 kDa .
Immunohistochemistry: Optimize antigen retrieval with citrate buffer (pH 6.0); validate using FFPE tumor sections .
Immunofluorescence: Fix cells with 4% paraformaldehyde; stain with 1:100–1:1000 antibody dilution .
SNAI1, also known as Snail family transcriptional repressor 1 or SNAIL, is a zinc-finger transcription factor that plays a crucial role in regulating gene expression. It is particularly important in the process of epithelial-mesenchymal transition (EMT), which is essential for embryonic development, wound healing, and cancer metastasis. By repressing target genes, SNAI1 influences cell differentiation, migration, and survival, making it a vital protein in both normal physiology and disease progression . SNAI1 expression has been linked to cancer metastasis in epithelial cell lines through its role in directing EMT. It directs the loss of polarity in epithelial cells by down-regulating E-cadherin expression, resulting in cells becoming less adherent to their neighbors and re-engaging cell proliferation and metastasis programs .
Several types of SNAI1 antibodies are available, each with specific characteristics suited for different research applications. The most extensively characterized include:
Monoclonal antibodies: SNAI1 Antibody (G-7) is a mouse monoclonal IgG1 kappa light chain antibody designed to recognize SNAI1. This antibody targets an epitope mapped between amino acids 113-139 within an internal region of the human SNAI1 protein .
Polyclonal antibodies: Examples include SC10432 (goat polyclonal, [E-18]) from Santa Cruz Biotechnology and AF3639 (goat polyclonal) from R&D Systems . Cell Signaling also offers anti-SNAIL (#3879S) antibodies that have been used in ChIP experiments .
The selection of the appropriate antibody depends on the specific research application, required species reactivity, and the cellular localization of interest.
SNAI1 antibodies have demonstrated utility across multiple experimental applications:
Western blotting (WB): Detecting SNAI1 protein expression levels in cell or tissue lysates
Immunoprecipitation (IP): Isolating SNAI1 protein complexes
Immunofluorescence (IF): Visualizing SNAI1 localization within cells
Immunohistochemistry with paraffin-embedded sections (IHC-P): Examining SNAI1 expression in tissue samples
Enzyme-linked immunosorbent assay (ELISA): Quantifying SNAI1 in solution
Chromatin Immunoprecipitation (ChIP): Investigating SNAI1 binding to DNA sequences
The versatility of these antibodies provides researchers with robust tools for studying SNAI1's function and its implications in different biological contexts, whether investigating developmental biology or exploring therapeutic targets in oncology.
Validating SNAI1 antibody specificity is critical for ensuring reliable experimental results. Based on the literature, the following validation approaches are recommended:
Peptide blocking: Pre-incubating the antibody with its corresponding blocking peptide (e.g., SC10432P for SC10432 antibody) at a 1:10 protein ratio versus PBS as a control. Loss of signal after peptide pre-absorption confirms specificity .
siRNA knockdown: Treating cells with SNAI1-specific siRNA versus control siRNA, followed by immunoblotting or immunostaining to verify decreased signal. Studies have demonstrated a clear decrease in the ~30 kDa SNAI1 band after siRNA treatment .
Using positive and negative control samples: Well-characterized samples with known SNAI1 expression patterns, such as xenografts of cell lines (e.g., SiHa and ME180), first-trimester human placenta, or specific tumor tissues can serve as controls .
Cross-validation with multiple antibodies: Using different SNAI1 antibodies that target distinct epitopes can help confirm the specificity of observed signals .
These validation steps are essential for distinguishing true SNAI1 signals from potential non-specific binding or artifacts.
The subcellular localization detection capabilities vary significantly between SNAI1 antibodies, which can be critical for research focusing on SNAI1's function as a transcription factor:
AF3639 (goat polyclonal, R&D Systems) has demonstrated superior performance in nuclear SNAI1 detection compared to SC10432 (goat polyclonal, Santa Cruz Biotechnology). In comparative studies, AF3639 produced stronger nuclear staining with better signal-to-background ratio, while SC10432 exhibited more cytoplasmic staining . This distinction is crucial since SNAI1, as a transcription factor, exerts its primary function in the nucleus, though cytoplasmic localization may be relevant for certain regulatory mechanisms.
Interestingly, studies of preimplantation embryos reveal complex localization patterns, where SNAI1 is consistently detectable in the cytoplasm, but nuclear localization is not commonly observed despite SNAI1's role as a transcription factor . This suggests that antibody selection should be guided by the specific subcellular compartment of interest and the biological context of the study.
For experiments requiring clear visualization of nuclear SNAI1, AF3639 would be the preferred choice based on published comparisons, especially in formalin-fixed paraffin-embedded (FFPE) clinical material.
When studying rare SNAI1-positive cell populations, several methodological considerations become critical:
Signal amplification techniques: Since SNAI1 expression can be limited to small subpopulations (e.g., <5% of tumor cells in OSCC), signal amplification methods may be necessary. Consider tyramine signal amplification or multiplexed immunofluorescence techniques.
Full-section analysis: Focal nature of SNAI1 expression necessitates examination of full tissue sections rather than tissue microarrays or limited samples. This is particularly important for tumors where SNAI1-positive cells are frequently found near inflammation sites or at invasion fronts .
Cell-specific contextualization: SNAI1-positive cells should be analyzed in their microenvironmental context. For example, in tumors, SNAI1-positive cells are often located at the invasive front or near inflammatory infiltrates, suggesting functional significance of these spatial relationships .
Quantification thresholds: Establishing clear thresholds for SNAI1 positivity is essential. Studies suggest different biological significance of low-level (<5%), medium (5-10%), and high-level (>10%) SNAI1 expression, with high-level expression significantly associated with poor outcomes in cancer patients .
Combined marker analysis: Analyzing SNAI1 in conjunction with other markers (e.g., E-cadherin, FAK, p63) provides greater biological insight into the EMT process than SNAI1 analysis alone .
These considerations are essential for accurately identifying and characterizing rare SNAI1-expressing cells that may have significant biological impact despite their limited numbers.
Chromatin immunoprecipitation (ChIP) using SNAI1 antibodies is a powerful approach for investigating SNAI1's role in transcriptional regulation. Based on published methodologies, an effective ChIP protocol would include:
Antibody selection: Anti-SNAIL (#3879S) from Cell Signaling has been successfully used in ChIP experiments . Alternatively, validated SNAI1 antibodies with good nuclear localization properties (e.g., AF3639 from R&D Systems) may be suitable.
E-box motif identification: Since SNAI1 binds to E-box motifs (consensus sequence: CANNTG), in silico analysis should be performed to identify potential SNAI1 binding sites in promoters of interest. For example, the CD73 promoter contains multiple E-boxes that can be targeted in ChIP analysis .
Control regions: Include both positive controls (known SNAI1 targets such as E-cadherin promoter) and negative controls (regions lacking E-box motifs) .
Data normalization and analysis: RT-qPCR signals should be normalized to control IgG, and SNAI1 ChIP signal reported as fold enrichment over IgG control .
Cellular context considerations: SNAI1 binding activity may differ between epithelial and mesenchymal states. For example, in MDA-MB-468-iSNAI1 cells, SNAI1 binding to the CD73 promoter showed a three-fold increase specifically at E-box 5 during EMT conditions .
This methodology allows researchers to definitively establish direct regulation of target genes by SNAI1, distinguishing direct transcriptional effects from indirect consequences of SNAI1 expression.
SNAI1 displays complex, asymmetrical expression patterns during early embryonic development that require specific methodological approaches for accurate characterization:
3D confocal imaging: Since SNAI1 shows asymmetrical localization within individual blastomeres, comprehensive 3D confocal microscopy is necessary to capture the complex distribution patterns. This is especially important at the 2-cell stage, where SNAI1 can be asymmetrically localized within one blastomere and absent in the second, or show various combinations of symmetrical and asymmetrical patterns .
Quantification of expression patterns: Document the frequency of different localization patterns. For example, in 2-cell embryos, studies have observed: 16% with asymmetrical SNAI1 in one blastomere and none in the second; 9% with symmetrical distribution in one blastomere and none in the second; 16% with asymmetrical and symmetrical patterns; 25% with both asymmetrical; and 28% with symmetrical distribution in both blastomeres .
Developmental stage-specific analysis: SNAI1 localization changes dramatically throughout preimplantation development. At the 8-cell stage, SNAI1 is detected in most blastomeres. In compacted embryos, SNAI1 becomes confined to outer cells only. This pattern persists, resulting in SNAI1 detection only in the trophectoderm of the blastocyst, with progressive restriction as the blastocyst expands .
Combined protein and transcript analysis: Correlating protein localization with transcript levels (using quantitative RT-PCR) reveals distinct regulation patterns. For example, Snai1 transcripts are significantly upregulated at the 2-cell stage followed by downregulation at the 8-cell stage, while protein localization shows progressive restriction to outer cells .
These methodological considerations are essential for accurately characterizing the complex spatial and temporal regulation of SNAI1 during developmental processes.
When faced with contradictory SNAI1 antibody results across different studies or experimental conditions, researchers should consider several factors:
When contradictory results are observed, orthogonal validation approaches (combining protein detection with mRNA analysis or functional assays) can help resolve discrepancies and establish which antibody results most accurately reflect true SNAI1 biology in the specific experimental context.
Optimal fixation and permeabilization conditions for SNAI1 immunostaining vary depending on the sample type and research question:
For cell cultures and fresh tissues:
Paraformaldehyde fixation (4% PFA for 15-20 minutes at room temperature) preserves antigenicity while maintaining structural integrity.
Permeabilization with 0.1-0.5% Triton X-100 for 10-15 minutes allows antibody access to nuclear SNAI1.
For detecting both nuclear and cytoplasmic SNAI1, gentler permeabilization with 0.1% Triton X-100 or 0.5% saponin may better preserve cytoplasmic signals .
For formalin-fixed paraffin-embedded (FFPE) samples:
Heat-induced epitope retrieval using citrate buffer (pH 6.0) is generally effective for SNAI1 antibodies.
For clinical specimens, AF3639 (R&D Systems) has demonstrated superior performance with stronger nuclear staining and better signal-to-background ratio compared to SC10432 (Santa Cruz Biotechnology) .
For preimplantation embryos:
Fresh flushed embryos should be fixed immediately with 2-4% paraformaldehyde.
Gentle permeabilization conditions are crucial to preserve the unique asymmetrical distribution patterns of SNAI1 observed in early embryonic stages .
Regardless of sample type, including appropriate positive and negative controls is essential for validating staining specificity, particularly when optimizing new fixation and permeabilization protocols.
Minimizing non-specific binding when using SNAI1 antibodies is critical for accurate interpretation of results. The following strategies are recommended:
Optimized blocking protocols: Use 5-10% normal serum from the species in which the secondary antibody was raised (e.g., goat serum for anti-mouse secondary antibodies). Adding 1% BSA can further reduce background.
Antibody titration: Perform careful titration experiments to determine the optimal antibody concentration that maximizes specific signal while minimizing background. For AF3639, concentrations of 2-5 μg/ml have been successful in FFPE tissues .
Secondary antibody controls: Include controls with secondary antibody only to identify potential non-specific binding independent of the primary antibody.
Peptide competition assays: Pre-absorb the SNAI1 antibody with its specific blocking peptide (e.g., SC10432P for SC10432) at a 1:10 protein ratio versus PBS as a control. Loss of signal confirms specificity .
Cross-adsorption: For polyclonal antibodies, pre-adsorption against tissues or cell lysates from SNAI1-knockout models can reduce non-specific binding.
Wash optimization: Extended washing steps (3-5 washes of 5-10 minutes each) with PBS containing 0.1% Tween-20 can significantly reduce background.
Signal-to-noise enhancement: For challenging samples, consider signal amplification methods like tyramide signal amplification, but be aware this may amplify any remaining non-specific binding.
Implementation of these strategies, particularly peptide competition controls, has been demonstrated to significantly improve the specificity of SNAI1 detection in various experimental systems .
Quantifying SNAI1 expression in heterogeneous samples presents unique challenges due to variable expression patterns and subcellular localization. The following strategies are recommended based on published research:
Defined positivity thresholds: Establish clear criteria for SNAI1 positivity. Studies have used thresholds such as nuclear staining in ≥5% of tumor cells, with further stratification into low (5-10%) and high (>10%) expression groups, which correlated with different clinical outcomes .
Cell type-specific analysis: In heterogeneous tissues, separately analyze SNAI1 expression in different cell types and compartments. For example, in tumors, distinguish between SNAI1 expression in cancer cells versus stromal cells, as both may have biological significance .
Spatial distribution assessment: Record the spatial pattern of SNAI1-positive cells, particularly their relationship to anatomical features such as the invasive front in tumors or the outer cell layer in embryos .
Multi-parameter analysis: Combine SNAI1 quantification with assessment of related markers (e.g., E-cadherin, vimentin, FAK) to create integrated mesenchymal-epithelial profiles rather than relying on SNAI1 alone .
Digital pathology approaches: For larger sample sets, consider using digital image analysis software with machine learning algorithms to quantify nuclear versus cytoplasmic SNAI1 staining across whole tissue sections, providing more objective and comprehensive assessment.
Developmental stage consideration: In embryonic studies, quantification should account for stage-specific patterns. For example, the percentage of SNAI1-positive cells significantly decreases as embryos progress from 8-cell to blastocyst stage, with further changes as blastocysts mature and expand .
These quantification strategies enable more meaningful interpretation of SNAI1 expression patterns in complex, heterogeneous biological systems.
The choice of SNAI1 antibody can significantly impact epithelial-mesenchymal transition (EMT) research outcomes in several important ways:
To minimize antibody-dependent bias in EMT research, validation with multiple SNAI1 antibodies and correlation with functional EMT markers (e.g., E-cadherin downregulation, increased cell migration) is strongly recommended.
SNAI1 antibodies are increasingly being integrated into single-cell analysis platforms, opening new avenues for understanding SNAI1 biology at unprecedented resolution:
Single-cell immunofluorescence imaging: The observation that SNAI1 and SNAI2 display asymmetrical expression patterns in early embryonic development highlights the importance of single-cell resolution imaging. Advanced techniques like confocal microscopy combined with deconvolution algorithms allow precise quantification of SNAI1 subcellular localization in individual cells.
Mass cytometry (CyTOF): Metal-conjugated SNAI1 antibodies enable simultaneous detection of SNAI1 with dozens of other proteins in single cells. This approach is particularly valuable for analyzing rare SNAI1-positive cells in heterogeneous samples like tumors, where SNAI1-expressing cells may constitute <5% of the population .
Single-cell RNA-seq integration: Correlating single-cell SNAI1 protein expression (detected by antibodies) with transcriptomic profiles enables comprehensive characterization of SNAI1-regulated gene networks. This approach has revealed that SNAI1 directly regulates CD73 expression in human triple-negative breast cancer cells .
Spatial transcriptomics: Combining SNAI1 immunohistochemistry with spatial transcriptomics allows researchers to correlate SNAI1 protein expression with localized gene expression patterns, providing spatial context to SNAI1 function. This is particularly relevant given the observation that SNAI1-positive cells are often located at tumor invasion fronts .
Live-cell imaging: Development of non-toxic fluorescent-labeled SNAI1 antibody fragments or nanobodies enables tracking of SNAI1 dynamics in living cells, providing temporal information about SNAI1 regulation during processes like EMT.
These emerging applications promise to reveal previously unrecognized heterogeneity in SNAI1 expression and function across different cellular contexts.
SNAI1 and SNAI2 (SLUG) antibodies target distinct but related transcription factors with overlapping yet unique functions in development and disease:
SNAI1 and SNAI2 share conserved zinc-finger domains but differ in their N-terminal regions.
Most commercial antibodies target these divergent regions to ensure specificity.
Cross-reactivity testing is essential, as some antibodies may recognize both proteins due to structural similarities.
SNAI1 and SNAI2 show distinct expression patterns throughout development. Snai1 transcripts are significantly upregulated at the 2-cell stage, then downregulated at the 8-cell stage, while Snai2 transcripts are downregulated at the 2-cell stage and begin accumulating at the 8-cell stage through blastocyst stage .
Both proteins eventually become confined to outer cells in compacted embryos and trophectoderm in blastocysts, suggesting distinct temporal regulation but convergent spatial localization .
In cancer research, SNAI1 antibodies are frequently used to study primary EMT events, as SNAI1 is often an early driver of EMT.
SNAI2 antibodies are valuable for studying more sustained mesenchymal phenotypes, as SNAI2 is often expressed more stably in mesenchymal cells.
In developmental biology, combined analysis of both proteins is essential, as they display complex, sometimes asymmetrical expression patterns that may indicate functional differences .
Antibody validation approaches (peptide blocking, siRNA knockdown) are similarly effective for both SNAI1 and SNAI2 antibodies .
When studying both factors simultaneously, careful selection of compatible antibodies raised in different host species allows multiplexed detection.
Understanding these differences is crucial for designing experiments that accurately distinguish the specific contributions of SNAI1 versus SNAI2 in biological processes.
Developing SNAI1 as a clinical biomarker requires addressing several methodological considerations to ensure reliable, reproducible results that can inform patient care:
Antibody standardization: For clinical applications, standardization is critical. Among tested antibodies, AF3639 (R&D Systems) has demonstrated superior performance in FFPE clinical samples with stronger nuclear staining and better signal-background ratio . Standardizing on a single, well-validated antibody across clinical studies would improve cross-study comparability.
Scoring system development: Establish clear, reproducible scoring criteria. Evidence suggests different biological significance of SNAI1 expression levels, with high-level expression (>10% cells) significantly associated with poor outcomes . A standardized scoring system should account for:
Percentage of positive cells (e.g., <5%, 5-10%, >10%)
Staining intensity (weak, moderate, strong)
Subcellular localization (nuclear vs. cytoplasmic)
Spatial distribution (e.g., tumor core vs. invasive front)
Tissue processing protocol harmonization: SNAI1 detection can be sensitive to fixation and processing variations. Clinical biomarker development requires standardized:
Fixation time and conditions
Antigen retrieval methods
Staining protocols (automated platforms preferred)
Detection systems (e.g., polymer-based vs. avidin-biotin)
Multi-marker panels: SNAI1 is most informative when assessed as part of a panel. Studies show a mesenchymal-like immunoprofile characterized by E-cadherin loss or high cytoplasmic FAK expression together with SNAI1 correlates better with outcomes than SNAI1 alone .
Digital pathology integration: Automated image analysis improves scoring objectivity and reproducibility. Digital algorithms can quantify:
Nuclear vs. cytoplasmic SNAI1 intensity ratios
Spatial relationships to other markers
Heterogeneity across whole tumor sections
External quality assurance: Implementing proficiency testing programs for SNAI1 immunohistochemistry would ensure reliability across different laboratories, similar to established programs for ER, PR, and HER2 in breast cancer.
These methodological considerations address the challenges of translating SNAI1 from a research tool to a clinically useful biomarker.
Several promising future directions are emerging for SNAI1 antibody applications in research that will expand our understanding of SNAI1 biology and its roles in development and disease:
Super-resolution microscopy: The application of techniques like STORM (Stochastic Optical Reconstruction Microscopy) and PALM (Photo-Activated Localization Microscopy) with SNAI1 antibodies will provide nanoscale resolution of SNAI1 localization within nuclear subcompartments, potentially revealing new aspects of its transcriptional regulatory mechanisms.
Proximity labeling approaches: Combining SNAI1 antibodies with proximity labeling techniques (BioID, APEX) will map SNAI1 protein interaction networks in different cellular contexts, providing insights into how SNAI1 recruits different cofactors to regulate distinct gene sets during development versus cancer progression.
Single-molecule tracking: Development of minimally disruptive SNAI1 antibody fragments for live-cell imaging will enable real-time tracking of SNAI1 molecular dynamics, revealing the kinetics of its nuclear import/export, chromatin binding, and turnover during EMT.
Spatial multi-omics integration: Correlating SNAI1 protein localization (detected by antibodies) with spatial transcriptomics and epigenomics will generate comprehensive maps of SNAI1 activity in complex tissues, particularly important for understanding SNAI1's role at tumor invasion fronts and during embryonic development.
Therapeutic applications: SNAI1 antibodies conjugated to toxins or nanoparticles may serve as targeted therapies for cancers dependent on SNAI1-driven EMT. Early research suggests targeting cells with high SNAI1 expression might selectively eliminate metastasis-initiating cell populations.
Liquid biopsy development: Detection of SNAI1 in circulating tumor cells using highly specific antibodies may serve as a non-invasive biomarker for monitoring EMT and metastatic potential in cancer patients.
These emerging applications highlight the continuing importance of developing and validating highly specific SNAI1 antibodies with defined performance characteristics across diverse experimental conditions.
Researchers can address several current limitations in SNAI1 antibody technology through methodological innovations and rigorous validation approaches:
Improving specificity: Generate new monoclonal antibodies targeting unique SNAI1 epitopes with minimal homology to other Snail family members. Comprehensive validation should include CRISPR/Cas9 SNAI1 knockout controls and cross-reactivity testing against SNAI2 and SNAI3 .
Enhancing sensitivity for low-level expression: Develop signal amplification systems specifically optimized for SNAI1 detection. This is particularly important given that SNAI1 expression can be transient and limited to small cell subpopulations, yet biologically significant .
Standardizing quantification: Establish digital image analysis algorithms specifically calibrated for SNAI1 quantification, incorporating machine learning approaches trained on expert-annotated datasets to improve consistency in identifying positive nuclear signals versus background.
Expanding species reactivity: Current antibodies like SNAI1 Antibody (G-7) detect mouse, rat, and human SNAI1 , but broader cross-species reactivity would benefit comparative studies across model organisms. Generating antibodies against highly conserved SNAI1 regions could address this limitation.
Improving compatibility with multiplexing: Develop SNAI1 antibodies specifically designed for multiplexed immunofluorescence or mass cytometry to enable simultaneous detection of SNAI1 with numerous EMT markers in the same sample.
Creating better tools for live-cell applications: Develop non-disruptive approaches for detecting SNAI1 in living cells, such as nanobodies or antibody fragments that can be expressed intracellularly without interfering with SNAI1 function.
Establishing reference standards: Create recombinant SNAI1 protein standards and standardized positive control cell lines with defined SNAI1 expression levels to enable quantitative comparison across studies and laboratories.
Snail Family Zinc Finger 1, commonly referred to as SNAI1 or Snail, is a zinc finger transcriptional repressor. This protein plays a crucial role in the regulation of epithelial to mesenchymal transition (EMT) during embryonic development. EMT is a process where epithelial cells lose their cell polarity and cell-cell adhesion properties, gaining migratory and invasive characteristics to become mesenchymal stem cells. This transition is essential for various developmental processes, including mesoderm formation and wound healing.
The SNAI1 gene is located on chromosome 20 in humans and chromosome 2 in mice. The gene encodes a nuclear protein that is structurally similar to the Drosophila Snail protein. The protein contains several zinc finger domains, which are critical for its function as a transcriptional repressor. These zinc finger domains allow SNAI1 to bind to specific DNA sequences, known as E-boxes, in the promoter regions of target genes.
SNAI1 primarily functions by repressing the expression of E-cadherin, an adhesion molecule that maintains the epithelial phenotype. By binding to the E-cadherin promoter, SNAI1 inhibits its transcription, leading to the disassembly of adherens junctions and the subsequent loss of epithelial characteristics. This repression is facilitated by the recruitment of histone demethylase complexes, which modify chromatin structure to suppress gene expression.
In addition to E-cadherin, SNAI1 also regulates other genes involved in cell adhesion and cytoskeletal organization, such as claudin-7 (CLDN7) and keratin-8 (KRT8). The coordinated repression of these genes by SNAI1 is essential for the induction of EMT and the formation of mesenchymal cells.
During embryonic development, SNAI1 is critical for the formation and maintenance of the mesoderm, a middle layer of cells that gives rise to various tissues, including muscle, bone, and blood. The protein is also involved in the regulation of cell migration and survival, processes that are vital for proper embryogenesis.
In the context of disease, aberrant expression of SNAI1 has been implicated in cancer progression and metastasis. The ability of SNAI1 to induce EMT and promote cell migration makes it a key player in the dissemination of cancer cells from primary tumors to distant sites. Elevated levels of SNAI1 have been observed in various cancers, including breast, colorectal, and pancreatic cancers, where it is associated with poor prognosis and increased metastatic potential.
Mouse anti-human SNAI1 antibodies are commonly used in research to study the expression and function of SNAI1 in human cells. These antibodies are generated by immunizing mice with human SNAI1 protein or peptides, leading to the production of specific antibodies that can recognize and bind to human SNAI1. These antibodies are valuable tools for various applications, including Western blotting, immunohistochemistry, and immunoprecipitation, allowing researchers to investigate the role of SNAI1 in different biological contexts.