SOX2 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to SOX2 Monoclonal Antibodies

SOX2 is a transcription factor critical for embryonic development, stem cell maintenance, and tumor progression. Monoclonal antibodies targeting SOX2 enable precise detection and analysis of its expression across biological and clinical contexts. These antibodies are engineered to bind specifically to the SOX2 protein, facilitating applications in immunohistochemistry (IHC), flow cytometry, western blotting, and immunofluorescence .

Cancer Biomarker and Immunotherapy Target

SOX2 is overexpressed in multiple cancers, including gliomas, small cell lung cancer (SCLC), and breast/ovarian tumors . Key findings:

  • Glioma: SOX2 is highly expressed in malignant gliomas, with minimal expression in normal brain tissue. HLA-A*0201-restricted SOX2 peptides (e.g., TLMKKDKYTL) elicit cytotoxic T-cell responses, making SOX2 a candidate for glioma immunotherapy .

  • SCLC: 35.4% of SCLC patients show anti-SOX2 antibody reactivity, with high titers (≥1:6400) correlating with disease progression .

  • Monoclonal Gammopathy (MGUS): Anti-SOX2 T cells in MGUS patients inhibit clonogenic growth and predict favorable outcomes, highlighting its role as a precursor tumor antigen .

Stem Cell Research

SOX2 antibodies are essential for identifying pluripotent stem cells:

  • Embryonic Stem Cells: MAB2018 detects SOX2 in BG01V human embryonic stem cells, co-localizing with pluripotency markers like Oct4 .

  • Neural Differentiation: SOX2 expression persists in neural progenitor cells derived from human pluripotent stem cells (hPSCs), aiding in tracking lineage commitment .

Diagnostic Utility

  • Immunohistochemistry: Btjce and Ab171380 enable nuclear staining in FFPE tissues, such as glioblastoma and lung squamous carcinoma .

  • Flow Cytometry: MAB2018 and Btjce are optimized for intracellular staining of fixed/permeabilized cells, distinguishing SOX2+ populations (e.g., NTera-2 embryonic carcinoma cells) .

Technical Considerations

  • Antigen Retrieval: High/low pH buffers are effective for FFPE samples, as demonstrated with Btjce in human testes .

  • Cross-Reactivity: MAB2018 and SOX2/1791 exhibit minimal off-target binding, validated via western blot and immunofluorescence .

  • Purity and Stability: Antibodies like Btjce (≥90% purity) and MAB2018 (0.2 µm filtered) ensure reproducibility in high-throughput assays .

Future Directions

  • Therapeutic Antibodies: Engineered SOX2 antibodies may disrupt tumor stem cell maintenance, though challenges include targeting intracellular antigens.

  • Multiplex Assays: MAB2018’s compatibility with CyTOF and seqIF™ enables simultaneous detection of SOX2 with markers like E-cadherin and vimentin in glioblastoma .

Product Specs

Form
Purified mouse monoclonal antibody in a buffer containing 0.1M Tris-Glycine (pH 7.4, 150 mM NaCl) with 0.2% sodium azide and 50% glycerol.
Lead Time
Typically, we are able to ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the method of purchase and location. For specific delivery estimates, please contact your local distributor.
Synonyms
transcription factor SOX-2 transcription factor SOX2 SRY-related HMG-box gene 2

Q&A

What is SOX2 and why is it an important target for monoclonal antibody detection?

SOX2 is a member of the SRY-related HMG-box (SOX) family of transcription factors critically involved in embryonic development and cell fate determination. It functions as a key regulator of pluripotency in embryonic stem cells, maintaining their self-renewal capacity by controlling the expression of other transcription factors that affect Oct3/4 expression . SOX2 is essential for stem cell maintenance in the central nervous system and regulates gene expression in various tissues . The importance of SOX2 in development, pluripotency, and its dysregulation in cancer makes it a valuable target for detection using monoclonal antibodies in diverse research applications.

What are the common applications for SOX2 monoclonal antibodies in research?

SOX2 monoclonal antibodies are extensively utilized across multiple experimental platforms:

ApplicationCommon DilutionsNotes
Western Blot (WB)1:1000-1:5000Detects SOX2 at 34-40 kDa
Immunohistochemistry (IHC)1:50-1:500Often requires antigen retrieval
Immunofluorescence (IF/ICC)1:1000-1:4000For cell and tissue localization
Flow Cytometry0.5 μg per 10^6 cellsFor intracellular staining
ELISAVariableFor quantitative detection and antibody response studies

The versatility of these applications enables researchers to detect SOX2 in different experimental contexts, from protein expression levels to cellular localization studies .

What tissues and cell types typically express SOX2?

SOX2 expression has been documented in:

  • Embryonic stem cells (high expression)

  • Neural stem cells and neuroepithelium

  • Specific cancer types (particularly in small cell lung cancer, glioblastoma)

  • Adult brain tissue

  • Testis tissue

  • Esophageal tissue

Notably, SOX2 expression patterns differ between human and mouse development. While mouse primordial germ cells (PGCs) express Sox2, human PGCs during the first trimester of development do not express SOX2, highlighting important species-specific differences in developmental biology .

What antigen retrieval methods are most effective for SOX2 immunohistochemistry?

Optimal antigen retrieval methods for SOX2 IHC vary by tissue type and fixation:

  • Heat-induced epitope retrieval (HIER) using 10 mM sodium citrate buffer (pH 6.0) with microwave treatment for 8-15 minutes is commonly effective

  • For some tissues, TE buffer at pH 9.0 may provide superior results

  • Following antigen retrieval, blocking with 3% H₂O₂-methanol for 15 minutes at room temperature is often recommended before antibody incubation

The choice between high pH (pH 9.0) and low pH (pH 6.0) antigen retrieval buffers should be empirically determined for each tissue type and fixation method to optimize the signal-to-noise ratio .

How should samples be prepared for intracellular SOX2 staining in flow cytometry?

For optimal intracellular SOX2 staining in flow cytometry:

  • Fix cells using a dedicated flow cytometry fixation buffer

  • Permeabilize cells with an appropriate permeabilization/wash buffer

  • Block with appropriate serum (typically 3% BSA in PBS)

  • Incubate with SOX2 primary antibody (typically at dilutions of 1:100 to 1:200)

  • Wash thoroughly and counterstain with fluorochrome-conjugated secondary antibody

This approach has been successfully employed with multiple cell types including human embryonic stem cells, induced pluripotent stem cells, and cancer cell lines like NTera-2 .

What controls should be included when validating a new SOX2 monoclonal antibody?

Proper validation of SOX2 antibodies requires multiple controls:

  • Positive control tissues/cells known to express SOX2 (e.g., embryonic stem cells, neural stem cells, specific cancer cell lines like NTera-2)

  • Negative control tissues/cells with confirmed absence of SOX2 expression

  • Isotype control antibodies at the same concentration as the primary antibody

  • Secondary antibody-only controls to assess background staining

  • Peptide competition assays to confirm specificity

  • Comparison of results with multiple SOX2 antibody clones when possible

The combination of these controls helps distinguish between specific signal and background staining, particularly important given the nuclear localization of SOX2.

Why do different SOX2 antibodies sometimes produce conflicting results?

The discrepancy between SOX2 antibody detection results may stem from several factors:

  • Epitope recognition differences: Different antibodies target distinct regions of the SOX2 protein. For example, the R&D Systems AF2018 antibody recognizes amino acids 135-317 of SOX2, which contains the sumoylation site at amino acid 247 . This can affect detection of post-translationally modified SOX2.

  • Post-translational modifications: Modified forms of SOX2 may not be recognized by all antibodies. Research has shown that SOX2 can be sumoylated, appearing as a 50-55 kDa band instead of the expected 34-45 kDa . The Millipore AB5603 antibody detected a modified form of SOX2 in carcinoma in situ (CIS) cells that was not detected by the R&D Systems AF2018 antibody .

  • Antibody cross-reactivity: Some antibodies may cross-react with other SOX family members, although specific examples like SOX17 cross-reactivity have been ruled out in certain studies .

  • Technical differences: Variations in sample preparation, fixation methods, and antigen retrieval protocols can significantly impact results .

These factors emphasize the importance of using multiple antibody clones when investigating SOX2 expression in novel contexts.

How do SOX2 expression patterns differ between normal stem cells and cancer stem cells?

SOX2 expression shows distinct patterns in normal versus cancer stem cells:

Normal stem cells:

  • Highly regulated expression in embryonic stem cells, essential for pluripotency maintenance

  • Expression becomes restricted to specific lineages during development, particularly neuroepithelium

  • In adult tissues, expression is limited to specific stem cell niches

Cancer stem cells/tumor tissues:

  • Heterogeneous expression patterns, often with speckled nuclear localization

  • May undergo post-translational modifications (e.g., sumoylation) not typically observed in normal cells

  • Expression can be amplified in certain cancers (observed in ~20% of lung adenocarcinomas)

  • SOX2 may be strongly expressed in a small subset of tumor cells with stem-like properties

  • In small cell lung cancer, high SOX2 antibody titers are associated with limited stage disease, suggesting immune surveillance against SOX2-expressing tumor cells

Importantly, while SOX2 is often considered a cancer stem cell marker, research indicates that SOX2 expression doesn't universally guarantee cancer stem cell-like properties, particularly in lung adenocarcinoma .

What is the significance of SOX2 antibody responses in cancer patients?

Autologous antibody responses against SOX2 have significant clinical implications:

  • SOX2 antibodies are detected in approximately 10-20% of small cell lung cancer (SCLC) patients

  • The presence of anti-SOX2 antibodies correlates with limited disease stage in SCLC patients (p=0.05), potentially indicating better prognosis

  • Anti-SOX2 antibody responses are also observed in breast and ovarian cancer patients (23%), as well as non-small cell lung cancer patients (13.3%)

  • SCLC patients tend to develop higher titer antibodies compared to other cancer types

  • SOX2 antibody development correlates with the intensity of SOX2 staining in tumors (p=0.02) rather than the frequency of SOX2-expressing cells, suggesting that strong SOX2 expression, even if focal, might suffice to induce immune responses

  • These findings suggest possible active immune surveillance against SOX2-expressing tumor cells, with potential implications for immunotherapy approaches

How can contradictory data about SOX2 expression and prognosis be reconciled?

Research presents apparently conflicting data regarding SOX2 expression and clinical outcomes:

  • Tumor type specificity: SOX2 appears to have tissue-specific roles. In lung adenocarcinoma, SOX2 has been reported as an independent marker for worse outcome , while in squamous cell carcinoma of the lung, it has been associated with lower grade and better outcome .

  • Immune response as a confounding factor: The presence of anti-SOX2 antibody responses may confound analyses that only examine SOX2 protein expression. Patients with anti-SOX2 immune responses tend to have better prognosis (limited stage disease) while SOX2 protein expression alone does not predict outcomes as consistently .

  • Heterogeneity of expression patterns: SOX2 antibodies can be observed in patients whose tumors contain relatively few but strongly staining cells. This suggests that the quality (intensity) rather than quantity (frequency) of SOX2 expression may be more immunogenic and potentially relevant to prognosis .

  • Different methodologies: Studies using different antibody clones or detection methods may yield different results, as demonstrated by the varying detection of modified SOX2 forms in carcinoma in situ .

These findings highlight the need to consider both SOX2 expression and anti-SOX2 immune responses when evaluating prognostic implications.

Why might Western blot detection of SOX2 show bands at different molecular weights?

SOX2 can appear at various molecular weights in Western blot analysis due to several factors:

  • The calculated molecular weight of unmodified SOX2 is approximately 34 kDa

  • In human embryonic stem cells, SOX2 is often detected at approximately 45 kDa using standard antibodies

  • Post-translationally modified SOX2, particularly sumoylated forms, may appear at 50-55 kDa

  • Different antibodies may preferentially detect specific forms of SOX2, contributing to apparent molecular weight discrepancies

  • Variations in sample preparation, including reducing conditions and buffer systems, can affect migration patterns

When interpreting Western blot results, researchers should consider these variations and potentially employ multiple antibody clones targeting different epitopes to comprehensively analyze SOX2 expression.

What are the key optimization steps for SOX2 immunohistochemistry in different tissue types?

Successful SOX2 immunohistochemistry requires tissue-specific optimization:

  • Fixation: For most tissues, formalin fixation followed by paraffin embedding is suitable, though fixation time should be optimized to prevent epitope masking

  • Antigen retrieval:

    • For brain tissues: TE buffer at pH 9.0 often produces optimal results

    • For reproductive tissues: Citrate buffer at pH 6.0 with extended microwave treatment (8-15 minutes)

    • For tumors: Comparative testing of both high and low pH buffers is recommended

  • Antibody concentration:

    • Brain tissues typically require lower antibody concentrations (1:200 to 1:500)

    • Tumor tissues may require higher concentrations (1:50 to 1:100)

    • Embryonic tissues often show strong signal even at higher dilutions

  • Detection systems:

    • For tissues with high SOX2 expression, standard HRP-polymer systems are sufficient

    • For tissues with low or heterogeneous expression, amplification systems like tyramide signal amplification may improve detection

  • Counterstaining: Hematoxylin counterstaining should be optimized to provide nuclear detail without obscuring SOX2 nuclear staining

Each tissue type requires empirical optimization for optimal signal-to-noise ratio.

How can researchers distinguish between specific SOX2 staining and background in immunofluorescence applications?

To differentiate specific SOX2 staining from background in immunofluorescence:

  • Include proper controls:

    • Isotype-matched control antibodies at identical concentrations

    • Secondary antibody-only controls

    • Known positive and negative tissue/cell controls

  • Optimize blocking conditions:

    • Extend blocking time (1-2 hours) with appropriate serum (3-5% BSA or serum)

    • Include detergents (0.1-0.3% Triton X-100) for improved permeabilization and reduced nonspecific binding

    • Consider dual blocking with both serum and commercial blocking reagents

  • Assess staining pattern consistency:

    • Genuine SOX2 staining should be predominantly nuclear

    • In some cancer cells, a speckled nuclear pattern may be observed

    • Cytoplasmic staining should be evaluated cautiously and verified with multiple antibody clones

  • Use multi-channel fluorescence:

    • Co-stain with other lineage markers to confirm biological plausibility of SOX2 expression

    • Nuclear counterstains (DAPI or Hoechst) should be used to confirm nuclear localization

  • Titrate antibody concentrations:

    • Test a range of dilutions to identify optimal signal-to-noise ratio

    • Typical dilutions range from 1:1000 to 1:4000 for immunofluorescence

How are SOX2 monoclonal antibodies being used to study cancer stem cell populations?

SOX2 antibodies have become instrumental in cancer stem cell research:

  • Identification and isolation of stem-like cancer cells:

    • Flow cytometry with SOX2 antibodies allows identification of potential cancer stem cell populations in tumors such as glioblastoma and small cell lung cancer

    • Combination with other stem cell markers (OCT4, NANOG) provides improved specificity for cancer stem cell detection

  • Lineage tracing and fate mapping:

    • SOX2 antibodies enable tracking of cancer stem cell differentiation trajectories in experimental models

    • Sequential multiplex immunofluorescence (seqIF) allows visualization of SOX2-positive cells in relation to tumor architecture and other markers

  • Therapeutic response monitoring:

    • Changes in SOX2-positive cell populations following treatment can indicate therapeutic efficacy against cancer stem cells

    • The relationship between SOX2 expression intensity and anti-SOX2 immune responses provides insights into immune surveillance mechanisms

  • Prognostic evaluation:

    • While SOX2 expression alone shows variable association with prognosis, the presence of anti-SOX2 antibodies correlates with limited disease stage in SCLC patients (p=0.05)

    • This suggests that immune responses against SOX2-expressing cells may have prognostic significance

Recent research also indicates that SOX2 may not be universally required for cancer stem cell-like properties in all tumor types, highlighting the complexity of cancer stem cell biology .

What are the current challenges in developing standardized SOX2 detection protocols for clinical applications?

Several challenges remain in standardizing SOX2 detection for clinical applications:

  • Antibody variability:

    • Different antibody clones show variable sensitivity and specificity for different forms of SOX2

    • Post-translational modifications of SOX2 are detected inconsistently between antibody clones

  • Scoring system standardization:

    • Current studies use various scoring methods (H-score, intensity, percentage positive cells)

    • The relationship between SOX2 staining intensity versus frequency and clinical outcomes remains incompletely understood

  • Contextual interpretation:

    • SOX2 expression may have different implications depending on tumor type

    • The presence of anti-SOX2 immune responses can confound simple expression-based interpretations

  • Technical standardization:

    • Variability in fixation, antigen retrieval, and detection methods complicates cross-study comparisons

    • The need for internal and external quality controls to ensure reproducibility

  • Clinical validation:

    • Prospective studies linking standardized SOX2 detection to clinical outcomes are needed

    • Determination of clinically relevant thresholds for expression levels in different tumor types

Addressing these challenges will be essential for translating SOX2 research findings into clinically applicable diagnostic or prognostic tools.

How do SOX2 monoclonal antibodies contribute to understanding pluripotency and developmental biology?

SOX2 antibodies have made significant contributions to pluripotency and developmental research:

  • Characterization of pluripotent states:

    • SOX2 antibodies are essential for validating embryonic stem cells and induced pluripotent stem cells

    • Co-detection with other pluripotency factors (OCT4, NANOG) helps define the pluripotent state

  • Species-specific developmental differences:

    • SOX2 antibodies have revealed important differences between human and mouse development

    • While mouse primordial germ cells express Sox2, human first-trimester PGCs do not, highlighting evolutionary divergence in developmental programs

  • Lineage specification studies:

    • SOX2 antibodies enable tracking of neural lineage specification during development

    • Neural induction of human pluripotent stem cells can be monitored by SOX2 co-expression with other neuroectodermal markers (FoxG1, Pax6)

  • Reprogramming efficiency assessment:

    • SOX2 antibodies are used to validate the clearance of reprogramming factors and establishment of endogenous SOX2 expression in iPSCs

    • Flow cytometry quantification of SOX2-positive cells serves as a measure of reprogramming efficiency

  • Stem cell niche characterization:

    • In adult tissues, SOX2 antibodies help identify and characterize stem cell niches in organs such as the brain, where SOX2 maintains neural stem cell identity

These applications continue to advance our understanding of the fundamental mechanisms controlling development and cell fate decisions.

What emerging technologies are enhancing the utility of SOX2 monoclonal antibodies in research?

Recent technological advances are expanding SOX2 antibody applications:

  • Single-cell analysis:

    • Integration of SOX2 antibodies in mass cytometry (CyTOF) panels allows high-dimensional analysis of stem cell heterogeneity

    • Single-cell RNA-seq combined with SOX2 protein detection enables correlation between transcriptome and protein expression

  • Advanced imaging techniques:

    • Super-resolution microscopy provides insights into the subnuclear localization of SOX2

    • Sequential multiplexed immunofluorescence (seqIF) allows visualization of SOX2 in relation to multiple other markers in the same tissue section

  • Proximity labeling approaches:

    • BioID or APEX2 fusions with SOX2 enable identification of context-specific protein interaction networks

    • These approaches help elucidate how SOX2 functions differently across cell types

  • In vivo imaging:

    • Development of near-infrared fluorophore-conjugated SOX2 antibodies for non-invasive tracking of SOX2-positive cells in animal models

    • This enables longitudinal studies of stem cell dynamics during development and disease progression

  • Combinatorial antibody approaches:

    • Simultaneous detection of SOX2 with post-translational modification-specific antibodies to identify regulated subpopulations

    • These approaches provide insights into how SOX2 activity is modulated in different contexts

These technological advances promise to further refine our understanding of SOX2 biology across development, homeostasis, and disease.

How can researchers address contradictory findings in SOX2 expression studies?

To resolve contradictions in SOX2 research findings, investigators should:

  • Employ multiple antibody validation strategies:

    • Use at least two independent antibody clones recognizing different epitopes

    • Incorporate genetic approaches (knockdown/knockout) to confirm specificity

    • Include appropriate positive and negative controls for each application

  • Consider post-translational modifications:

    • Evaluate the potential presence of modified SOX2 forms (sumoylated, phosphorylated, etc.)

    • Use antibodies capable of detecting specific modifications when relevant

  • Report comprehensive methodological details:

    • Explicitly document antibody clones, dilutions, incubation conditions, and detection methods

    • Describe antigen retrieval protocols in detail, as these significantly impact results

  • Account for biological complexity:

    • Consider the relationship between SOX2 expression and anti-SOX2 immune responses

    • Recognize tissue-specific and context-dependent functions of SOX2

  • Integrate multi-omics approaches:

    • Correlate protein detection with transcript levels

    • Consider epigenetic regulation of SOX2 expression

By adopting these comprehensive approaches, researchers can better understand the complexities of SOX2 biology and resolve apparent contradictions in the literature.

What are the most promising future applications of SOX2 monoclonal antibodies in research and medicine?

Future applications of SOX2 antibodies hold significant promise:

  • Precision medicine approaches:

    • Development of companion diagnostics using SOX2 expression patterns to guide therapy selection

    • Integration of SOX2 antibody detection with anti-SOX2 immune response assessment for improved prognostication

  • Immunotherapy development:

    • Targeting SOX2-expressing cancer stem cells through immune checkpoint inhibition

    • Development of SOX2-targeted chimeric antigen receptor (CAR) T-cell therapies

    • Therapeutic vaccines designed to enhance anti-SOX2 immune responses in cancer patients

  • Regenerative medicine applications:

    • Quality control of stem cell-derived products for therapeutic applications

    • Monitoring of cellular reprogramming efficiency and safety

    • Tracking cell fate following transplantation in regenerative medicine approaches

  • Neurodevelopmental disorder research:

    • Investigation of SOX2 mutations associated with anophthalmia and optic nerve hypoplasia

    • Understanding the role of SOX2 in syndromic developmental disorders

  • Combined diagnostic approaches:

    • Integration of SOX2 detection with other cancer stem cell markers for improved sensitivity and specificity

    • Development of liquid biopsy approaches to detect circulating SOX2-expressing cells or anti-SOX2 antibodies

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.