SOX2 Human

SRY (sex determining region Y)-box 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Identity and Structure

The SOX2 gene is located on chromosome 3 (3q26.3-q27) and encodes a 317-amino-acid protein with three functional domains :

  • HMG Domain: Binds DNA to regulate target gene transcription.

  • Dimerization (DIM) Domain: Facilitates interaction with partner proteins (e.g., Oct4).

  • Transactivation (TAD) Domain: Activates or represses gene expression.

Table 2: SOX2 Binding Partners and Functions

Partner ProteinRoleTarget Genes
Oct4Maintain pluripotencyNANOG, SOX2, OCT4
Pax6Neural developmentNESTIN, PAX6
β-CateninWnt signaling modulationDKK1, AXIN1

Neural Development and Disease Implications

  • Neural Progenitors: SOX2 sustains NPC proliferation and inhibits premature differentiation . Silencing SOX2 in human Müller glial cells triggers apoptosis and neuronal differentiation .

  • Developmental Disorders: SOX2 mutations cause microphthalmia, anophthalmia, and optic nerve hypoplasia .

Clinical and Therapeutic Insights

  • Regenerative Medicine: SOX2, with Oct4, Klf4, and c-Myc, reprograms somatic cells into induced pluripotent stem cells (iPSCs) .

  • Anticancer Strategies: Indirect targeting via SOX2-associated pathways (e.g., Wnt, AKT) is under investigation .

Key Research Findings

  1. SOX2-DKK1 Axis: SOX2 directly activates DKK1 expression, suppressing Wnt signaling in MSCs to balance adipogenic/osteogenic differentiation .

  2. Cell-Type-Specific Binding: ChIP-seq revealed 17,992 SOX2-binding sites in hESCs vs. 67,021 in hNPCs, indicating dynamic regulatory roles .

  3. Neural Apoptosis: SOX2 knockdown in retinal Müller cells upregulated neuronal markers (βIII tubulin, rhodopsin) but induced caspase-mediated apoptosis .

Product Specs

Introduction
The transcription factor SOX2 partners with OCT4 to form a trimeric complex on DNA, which regulates the expression of genes crucial for embryonic development. These genes include YES1, FGF4, UTF1, and ZFP206. SOX4 plays a vital role in early embryogenesis and the maintenance of pluripotency in embryonic stem cells.
Description
Recombinant human SOX2 protein, expressed in E. coli, is a non-glycosylated polypeptide chain consisting of 208 amino acids (specifically, amino acids 1-200). This protein has a molecular weight of 23.1 kDa. An 8-amino acid His-Tag is fused to the C-terminus of the SOX2 protein to facilitate purification via standard chromatography techniques.
Physical Appearance
The product is a colorless solution that has undergone sterile filtration.
Formulation
The SOX2 solution is formulated with the following components: 20mM Tris-HCl at pH 8, 2mM DTT, 0.2M NaCl, and 40% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, freezing at -20°C is recommended. Adding a carrier protein such as 0.1% HSA or BSA is advisable for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the protein is determined by SDS-PAGE analysis to be greater than 90%.
Synonyms
MCOPS3, ANOP3, MGC2413, SOX2, SRY (sex determining region Y)-box 2.
Source
Escherichia Coli.
Amino Acid Sequence
MYNMMETELK PPGPQQTSGG GGGNSTAAAA GGNQKNSPDR VKRPMNAFMV WSRGQRRKMA QENPKMHNSE ISKRLGAEWK LLSETEKRPF IDEAKRLRAL HMKEHPDYKY RPRRKTKTLM KKDKYTLPGG LLAPGGNSMA SGVGVGAGLG AGVNQRMDSY AHMNGWSNGS YSMMQDQLGY PQHPGLNAHG AAQMQPMHRY LEHHHHHH.

Q&A

What is SOX2 and what are its primary functions in human cells?

SOX2 (SRY-box transcription factor 2) is a pluripotency-associated transcription factor essential for mammalian embryogenesis and adult tissue homeostasis . It functions as a master regulator that controls gene expression networks governing cell identity, differentiation, and self-renewal . In embryonic stem cells, SOX2 partners with other transcription factors like OCT4 and NANOG to maintain pluripotency . In adult tissues, SOX2 helps maintain tissue-specific stem cell populations and regulates cellular differentiation programs . Mechanistically, SOX2 binds to specific DNA sequences through its high-mobility group (HMG) box domain and recruits transcriptional machinery to modulate gene expression .

How is SOX2 expression regulated in normal human tissues?

SOX2 expression is controlled through an exquisite array of molecular mechanisms operating at multiple levels:

  • Transcriptional regulation: The SOX2 gene contains enhancer regions that are regulated by multiple transcription factors, including SOX2 itself through feedback loops .

  • microRNA regulation: Numerous microRNAs directly target SOX2 transcripts to regulate its expression post-transcriptionally .

  • Long non-coding RNA regulation: The SOX2 gene is embedded within an intron of a multi-exon lncRNA gene known as SOX2OT (SOX2 overlapping transcript), which influences SOX2 expression . SOX2OT has at least 10 exons with multiple transcription start sites and can generate at least 8 splice variants through alternative splicing .

  • Post-translational modifications: SOX2 protein function is modulated by various modifications including phosphorylation, which can affect its stability, localization, and transcriptional activity .

In embryonic stem cells, SOX2 can regulate its own expression through both positive and negative feedback loops - increasing its expression when levels are too low and decreasing it when levels are too high .

How does SOX2 expression correlate with cancer progression and patient outcomes?

SOX2 expression patterns and their correlation with patient outcomes vary significantly across cancer types:

For many cancers, increased SOX2 expression correlates with higher tumor grade and poorer patient survival . SOX2 is expressed in at least 25 different cancers, with SOX2 gene amplification observed in several cancer types . Many tumors show increased SOX2 expression during progression, suggesting its importance in advanced disease .

This contradictory data highlights the complex, context-dependent roles of SOX2 in different cancer types and stages .

What is the relationship between SOX2 and tumor-initiating cells/cancer stem cells?

SOX2 has been implicated in the tumor-initiating cell (TIC) populations (proposed cancer stem cell populations) of many cancers . The relationship between SOX2 and TICs is supported by several lines of evidence:

  • SOX2 expression in TIC-enriched populations: Cells isolated using putative cancer stem cell markers (CD133, CD44, ABCG2, and side population via Hoechst efflux) frequently show elevated SOX2 expression .

  • Functional studies:

    • Knockdown experiments: Stable SOX2 knockdown dramatically reduces tumor initiation/formation in head and neck squamous cell carcinomas and melanomas in limiting cell dilution tumor assays .

    • SOX2-positive cell isolation: When SOX2-positive cells are isolated from heterogeneous tumor populations (using GFP knocked into the endogenous SOX2 gene or GFP driven by SOX2 promoter), they exhibit higher frequency of tumor initiation compared to SOX2-negative cells .

  • Drug resistance and tumor repopulation: SOX2-positive cells are frequently members of quiescent, slowly-cycling cancer stem cell populations that survive cytotoxic drugs and repopulate tumors when treatment is withdrawn .

These findings collectively suggest that SOX2 marks and functionally contributes to tumor-initiating cell populations in multiple cancer types .

How do changes in SOX2 expression levels affect tumor growth?

SOX2 exhibits a complex, non-linear relationship with tumor growth where precise levels appear critically important:

  • Optimized expression levels: SOX2 levels in actively proliferating tumor cells appear to be optimized to maximize tumor growth - either too little or too much SOX2 dramatically alters tumor growth .

  • Conflicting effects of SOX2 overexpression:

    • Stable overexpression studies: Some report that stable SOX2 overexpression increases growth in MCF-7 (breast), DU145 (prostate), and Patu8988t (pancreatic) cancer cells .

    • Inducible overexpression studies: Controlled SOX2 elevation using inducible systems shows growth inhibition in glioblastoma (U87, U118), medulloblastoma (DAOY), breast carcinoma (MDA-MB-231), prostate carcinoma (DU145), and pancreatic cancer cell lines both in vitro and in vivo .

    • Colorectal cancer: SOX2 elevation causes growth inhibition during the initial five days, with almost complete growth arrest in HT-29 cells .

  • Context-dependent effects: The impact of SOX2 appears highly context-dependent, similar to other genes like TGFβ that can act as either tumor suppressors or oncogenes . Mutations in other genes (e.g., RB1 and p53) can permit elevated SOX2 to promote rather than inhibit tumor growth .

This complex relationship suggests that therapeutic strategies targeting SOX2 must consider the precise levels and cellular context rather than simply aiming to reduce SOX2 expression .

How does SOX2 interact with other transcription factors and chromatin regulators?

SOX2 functions within complex transcriptional networks through multiple types of interactions:

  • Protein-protein interactions: Unbiased proteomic screens of the SOX2-interactome show that SOX2 physically associates with many other master regulators, particularly OCT4 in embryonic stem cells, creating a highly integrated transcriptional network . In both embryonic stem cells and brain tumor cells, SOX2 associates with a diverse array of functionally distinct proteins involved in transcriptional regulation, signal transduction, and DNA repair/replication .

  • DNA binding specificity: SOX2 binds to specific DNA sequences through its HMG box domain, often cooperatively with partner transcription factors . This cooperative binding allows for context-specific gene regulation in different cell types.

  • Enhancer regulation: SOX2 regulates super-enhancers, which are large clusters of enhancers that drive expression of genes defining cell identity . Biochemical studies using electrophoretic mobility shift assays (EMSA) have demonstrated SOX2 binding to both naked DNA and nucleosomal DNA, suggesting its ability to access regulatory elements in various chromatin contexts .

  • Dynamic interactome: The SOX2-interactome changes when cells undergo differentiation or transformation, indicating that SOX2 functions differently depending on the cellular context . SOX2 associates with >50% of the genes that code for SOX2-associated proteins, creating a highly interconnected regulatory network .

This extensive interaction network explains why small changes in SOX2 levels can exert profound effects on cell physiology and why SOX2 function is so context-dependent .

What role do long non-coding RNAs play in SOX2 regulation?

Long non-coding RNAs (lncRNAs) play several important roles in regulating SOX2 expression:

  • SOX2OT (SOX2 overlapping transcript): The single-exon SOX2 gene is embedded within an intron of this multi-exon lncRNA gene . SOX2OT is evolutionarily conserved across vertebrates, suggesting functional importance . It possesses at least 10 exons with up to four different transcription start sites and can generate at least 8 splice variants through alternative splicing .

  • Co-expression patterns: SOX2 and SOX2OT are co-expressed in embryonic stem cells and various cancers including breast, lung, brain, and esophageal tumors . Different splice variants of SOX2OT are expressed in different cancer types .

  • Functional relationship: Evidence suggests SOX2OT contributes to SOX2 expression regulation:

    • Knockdown of SOX2OT by siRNA in lung adenocarcinoma cells (A549) reduced SOX2 expression .

    • Forced overexpression of SOX2OT in breast tumor cells (MDA-MB-231) increased SOX2 transcript and protein levels .

  • Shared microRNA regulation: Both SOX2 and SOX2OT transcripts can be targeted by the same microRNAs. For example, miR-211 targets the same sequence in both SOX2 and SOX2OT transcripts, leading to their downregulation when miR-211 is overexpressed in embryonal carcinoma cells .

This intricate relationship between SOX2 and lncRNAs adds another layer of complexity to SOX2 regulation and highlights potential new therapeutic targets for modulating SOX2 expression .

What are the most reliable methods for detecting and quantifying SOX2 expression in human samples?

Several complementary approaches are recommended for reliable SOX2 detection and quantification:

  • Transcript analysis:

    • RT-qPCR: Allows sensitive quantification of SOX2 mRNA levels but requires careful primer design to distinguish SOX2 from SOX2OT transcripts .

    • RNA sequencing: Provides comprehensive analysis of SOX2 expression alongside global transcriptomic changes and can detect alternative splice variants .

  • Protein detection:

    • Western blotting: Allows quantification of total SOX2 protein levels and can detect post-translational modifications with specific antibodies .

    • Immunohistochemistry (IHC): Essential for analyzing SOX2 expression patterns in tumor tissues and observing heterogeneity of expression within tumors .

    • Immunofluorescence: Enables co-localization studies with other proteins and subcellular localization analysis .

  • Epigenetic and binding analyses:

    • Chromatin immunoprecipitation (ChIP): Identifies SOX2 binding sites across the genome .

    • ATAC-seq: Assesses chromatin accessibility at SOX2 regulatory regions .

    • Electrophoretic mobility shift assay (EMSA): Monitors binding of SOX2 to DNA sequences, including nucleosomal DNA .

When studying SOX2 in tumors, it's critical to consider intratumoral heterogeneity, as SOX2 is often expressed heterogeneously throughout tumor cells, sometimes in only a small percentage of cells . This heterogeneity necessitates single-cell approaches or careful isolation of cell subpopulations.

What are the key considerations when manipulating SOX2 expression levels in experimental models?

When manipulating SOX2 expression in experimental models, researchers should consider several critical factors:

These considerations highlight the importance of careful experimental design when studying SOX2 function and the need to interpret results in the context of the specific experimental system used .

How can contradictory findings regarding SOX2's prognostic value in different cancers be reconciled?

The contradictory findings regarding SOX2's prognostic value across different cancers reflect its complex, context-dependent roles. Several methodological approaches can help reconcile these contradictions:

  • Integrated multi-omics analysis:

    • Combine SOX2 expression data with genomic (mutations, CNVs), epigenomic, and proteomic analyses to identify contexts where SOX2 promotes or suppresses tumor progression .

    • Investigate co-occurring molecular alterations (e.g., p53 or RB1 mutations) that may modify SOX2 function, as these have been shown to influence SOX2's effects in prostate cancer .

  • Single-cell resolution studies:

    • Given SOX2's heterogeneous expression within tumors, single-cell approaches may reveal subpopulations with distinct SOX2 expression patterns and functional states .

    • Correlating single-cell SOX2 expression with stemness markers, proliferation markers, and drug resistance can provide context for contradictory bulk tumor findings .

  • Precise quantification and standardization:

    • Standardize SOX2 detection methods and cutoff values for "high" versus "low" expression across studies .

    • Quantify not just SOX2 presence/absence but precise expression levels, since SOX2 exhibits dose-dependent effects .

  • Functional classification approaches:

    • Classify tumors based on SOX2 functional networks rather than just SOX2 expression levels .

    • Assess SOX2 in the context of its interacting partners and downstream targets, which may vary between cancer types .

  • Longitudinal and treatment-response studies:

    • Analyze SOX2 expression before and after treatment to understand its role in therapy response and resistance .

    • Study SOX2-positive cells' behavior during and after chemotherapy to examine their role in tumor recurrence .

These approaches acknowledge SOX2's context-dependent functions and may reconcile apparently contradictory findings by identifying the specific conditions under which SOX2 promotes or inhibits tumor progression .

What therapeutic strategies might effectively target SOX2-dependent cancers?

Given SOX2's properties as a transcription factor (typically challenging to target directly) and its complex dose-dependent effects, several innovative therapeutic strategies might be effective:

  • Indirect targeting approaches:

    • Target genes that enable elevated SOX2 to promote tumor growth, potentially converting SOX2 from a growth promoter to a growth inhibitor in cancer cells .

    • Identify and inhibit key components of the SOX2-dependent transcriptional network rather than SOX2 itself .

  • Synthetic lethality strategies:

    • Identify genes that are essential specifically in SOX2-high cells but dispensable in SOX2-low cells .

    • Screen for compounds that selectively kill SOX2-dependent cancer cells while sparing normal cells .

  • Regulatory RNA-based approaches:

    • Target SOX2-regulating microRNAs or long non-coding RNAs like SOX2OT, which could provide indirect ways to modulate SOX2 expression .

    • miR-211 targets both SOX2 and SOX2OT, suggesting potential for dual targeting strategies .

  • Post-translational modification modulators:

    • Develop compounds that modify SOX2's post-translational modifications to alter its stability, localization, or transcriptional activity .

    • Target enzymes responsible for SOX2 phosphorylation or other modifications .

  • Cancer stem cell-targeted approaches:

    • Develop strategies that specifically target the quiescent, SOX2-positive cancer stem cell population that survives conventional therapies .

    • Combine conventional cytotoxic therapies with drugs that force SOX2-positive cells out of quiescence to increase their susceptibility to chemotherapy .

  • Context-specific combination therapies:

    • Design treatment combinations based on molecular context (e.g., p53/Rb status) that determines SOX2's function in specific tumors .

    • In contexts where SOX2 overexpression inhibits growth, strategies to transiently increase SOX2 levels might paradoxically inhibit tumor growth .

These approaches acknowledge that direct inhibition of SOX2 may be challenging and instead focus on exploiting SOX2's biological properties and dependencies to develop effective targeted therapies .

Key SOX2 Regulatory Mechanisms and Experimental Approaches

Regulatory LevelMechanismExperimental ApproachesReference
TranscriptionalEnhancer regulation, feedback loopsChIP-seq, reporter assays, EMSA
Post-transcriptionalmicroRNA targeting (e.g., miR-211)miRNA overexpression, reporter assays
lncRNA-mediatedSOX2OT (8+ splice variants)RNA-seq, knockdown/overexpression studies
Post-translationalPhosphorylation, SUMOylationWestern blot, mass spectrometry
Protein-protein interactionsPartner TFs (e.g., OCT4)Co-IP, proteomic screens, EMSA
Chromatin-levelNucleosomal bindingEMSA with nucleosomal DNA

SOX2 Experimental Expression Systems and Their Applications

Expression SystemFeaturesApplicationsConsiderationsReference
Constitutive overexpressionStable expression from strong promotersLong-term studies, selection-based experimentsMay cause compensatory mechanisms, conflicting results
Inducible expression systemsControlled timing and level of expressionShort-term effects, dose-response studiesPreferred for growth studies, reveals immediate effects
Knockdown approachessiRNA, shRNA targeting SOX2Loss-of-function studiesMay affect SOX2OT expression, off-target effects
SOX2 reporter systemsGFP knocked into SOX2 locus or driven by SOX2 promoterIsolation of SOX2-positive cells, lineage tracingAllows study of heterogeneous expression
Mutant SOX2 expressionQuikChange PCR for site-directed mutagenesisStructure-function studiesCan identify critical domains and residues

Product Science Overview

Introduction

The SRY (Sex Determining Region Y)-Box 2, commonly referred to as SOX2, is a transcription factor that plays a crucial role in the regulation of embryonic development and the determination of cell fate. It is part of the SOX (SRY-related HMG-box) family of transcription factors, which are characterized by the presence of a conserved HMG (high mobility group) box domain that binds to DNA.

Discovery and Gene Structure

The SRY gene, located on the Y chromosome, was first identified as the key determinant of male sex development in mammals. The discovery of the SRY gene was a significant milestone in understanding the genetic mechanisms of sex determination. The SRY gene encodes a protein that acts as a transcription factor, initiating the development of male characteristics by promoting the formation of testes .

SOX2, on the other hand, is located on chromosome 3 in humans and is not directly involved in sex determination. Instead, it is essential for maintaining the pluripotency of stem cells and is involved in the regulation of genes critical for embryonic development .

Function and Mechanism

SOX2 functions as a transcription factor, meaning it binds to specific DNA sequences and regulates the expression of target genes. It is particularly important in maintaining the self-renewal and pluripotency of embryonic stem cells. Pluripotency refers to the ability of a stem cell to differentiate into any cell type in the body.

The SOX2 protein interacts with other transcription factors and co-regulators to control the expression of genes involved in cell proliferation, differentiation, and survival. It plays a pivotal role in the development of the central nervous system, sensory organs, and other tissues .

Human Recombinant SOX2

Human recombinant SOX2 refers to the SOX2 protein that has been produced using recombinant DNA technology. This involves inserting the SOX2 gene into a suitable expression system, such as bacteria or mammalian cells, to produce the protein in large quantities. Recombinant SOX2 is used in various research applications, including studies on stem cell biology, developmental biology, and regenerative medicine.

Clinical and Research Implications

SOX2 has significant implications in both clinical and research settings. Mutations or dysregulation of the SOX2 gene can lead to various developmental disorders and diseases. For example, SOX2 mutations have been associated with conditions such as anophthalmia (absence of one or both eyes), microphthalmia (abnormally small eyes), and other congenital anomalies .

In research, SOX2 is a key factor in the generation of induced pluripotent stem cells (iPSCs). iPSCs are derived from adult cells that have been reprogrammed to a pluripotent state by introducing specific transcription factors, including SOX2. These cells have the potential to differentiate into any cell type, making them valuable for studying disease mechanisms, drug screening, and regenerative therapies .

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.