SLC7A11 (solute carrier family 7 member 11), also known as xCT or CCBR1, is a transmembrane protein that forms the light chain subunit of the cystine/glutamate antiporter system xc⁻. This system mediates the exchange of intracellular glutamate for extracellular cystine, supporting glutathione synthesis and redox homeostasis . The SLC7A11 Recombinant Monoclonal Antibody targets epitopes within the N-terminal region (amino acids 1–100) of human SLC7A11, enabling precise detection in experimental models .
Recombinant monoclonal antibodies are generated using plasmid vector-based expression systems. The antibody genes are cloned into host cells (e.g., HEK293F or bacterial systems), followed by affinity chromatography purification .
Laryngeal Squamous Cell Carcinoma (LSCC): Knockdown of SLC7A11 using shRNA reduced proliferation of UMCC-5 and Hep-2 cells by 40–60% (MTT assay) and induced G1-phase cell cycle arrest (flow cytometry) .
Nasopharyngeal Carcinoma (NPC): SLC7A11 downregulation increased MHC-I membrane localization, enhancing T cell-mediated tumor cell killing (IFN-γ ELISA and colony formation assays) .
Renal Cell Carcinoma (RCC): Overexpression of SLC7A11 sensitized cells to glucose starvation-induced death, while inhibition with sulfasalazine promoted survival under low glucose .
SLC7A11 facilitates Kaposi sarcoma-associated herpesvirus (KSHV) entry into cells .
Elevated SLC7A11 in gliomas increases glutamate secretion, causing neuronal death .
Diagnostic biomarker: Overexpression correlates with poor prognosis in LSCC and NPC .
Therapeutic target: Pharmacological inhibitors (e.g., sulfasalazine) or siRNA-based knockdown disrupt SLC7A11-mediated cystine uptake, sensitizing tumors to oxidative stress .
The SLC7A11 recombinant monoclonal antibody is produced by constructing the SLC7A11 antibody genes into plasmid vectors and subsequently transfecting these vectors into suitable host cells for expression using exogenous protein expression technology. This SLC7A11 recombinant monoclonal antibody has undergone affinity-chromatography purification and has been validated for ELISA. In functional ELISA, the human SLC7A11 protein (CSB-CF892171HU(A4)) at 2 µg/mL binds to this SLC7A11 recombinant monoclonal antibody, with an EC50 of 9.452-13.79 ng/mL.
SLC7A11 is a subunit of the heterodimeric amino acid transporter, system xCT, which consists of SLC7A11 and SLC3A2. SLC7A11 acts as the light chain subunit of this transporter. The primary function of SLC7A11 is to facilitate the exchange of cystine for glutamate across the cell membrane, thereby playing a crucial role in regulating redox balance.
SLC7A11, or solute carrier family 7 member 11, is a 55.4 kilodalton membrane transport protein that functions as the light chain of the cystine/glutamate antiporter system xc-. This protein mediates the exchange of extracellular cystine for intracellular glutamate at a 1:1 ratio across plasma membranes. SLC7A11 contains multiple transmembrane domains with several extracellular and intracellular loops that serve as potential antibody binding sites. The protein plays a crucial role in maintaining cellular redox balance by importing cystine for glutathione synthesis, which protects cells against oxidative stress and programmed cell death. SLC7A11 expression is notably elevated in various cancer types, making it an important target for both basic research and therapeutic development .
SLC7A11 antibodies serve multiple experimental purposes across diverse research platforms. Western blotting represents one of the most common applications, allowing for protein detection at the expected molecular weight range of approximately 55 kDa. Immunohistochemistry applications enable visualization of SLC7A11 expression patterns in tissue sections, while immunofluorescence provides subcellular localization insights. Flow cytometry using antibodies targeting extracellular epitopes permits quantification of surface expression in live cells. Additionally, immunoprecipitation techniques facilitate protein-protein interaction studies, and ELISA methods allow for quantitative measurement of SLC7A11 levels in various sample types . The selection of application-specific antibody preparations is critical, as some formulations may perform optimally in certain techniques but not others due to differences in epitope accessibility.
The production of recombinant SLC7A11 involves several expression system options, each with distinct advantages. Bacterial systems, particularly optimized Escherichia coli strains like BL21 codon plus or Rosetta (DE3), offer cost-effective production platforms for generating sufficient protein quantities for antibody development and characterization . When expressing SLC7A11 in E. coli, researchers have successfully employed His-tag fusion strategies for subsequent Ni²⁺-chelating chromatography purification. Growth conditions significantly impact expression efficiency, with optimal results typically achieved by culturing at 28°C post-induction with IPTG concentrations between 0.05-0.4 mM . The functional activity of recombinant SLC7A11 can be verified through reconstitution in proteoliposomes and subsequent transport assays, confirming that the recombinant protein maintains native conformational characteristics essential for antibody recognition.
Most commercially available SLC7A11 recombinant monoclonal antibodies demonstrate cross-reactivity with human, mouse, and rat orthologs due to conserved sequence homology across mammalian species . This cross-reactivity facilitates translational research by allowing the same antibody to be used across multiple experimental models. When selecting antibodies for specific applications, researchers should verify the documented species reactivity through validation studies. Antibodies targeting the extracellular domains, particularly the third extracellular loop (amino acids 218-231), often show broader species reactivity profiles . For studies involving non-mammalian models or less common mammalian species, customized validation is essential as sequence divergence may impact epitope conservation and antibody binding efficiency. Researchers should carefully review available data on species reactivity when selecting antibodies for multi-species comparative studies.
Comprehensive validation of SLC7A11 recombinant monoclonal antibodies requires a multi-tiered approach to ensure specificity. The gold standard involves parallel testing in wildtype and SLC7A11 knockout models, where specific signal should be absent in knockout samples. For experimental systems where genetic knockouts are unavailable, RNA interference (siRNA or shRNA) targeting SLC7A11 can provide alternative validation by demonstrating corresponding reductions in antibody signal intensity. Pre-absorption tests, where the antibody is pre-incubated with excess antigenic peptide before application, serve as additional controls — specific binding should be significantly reduced compared to non-absorbed antibody . Multiple detection methods should be employed during validation, as antibody performance can vary considerably between applications like Western blotting and immunohistochemistry due to differences in protein conformation and epitope accessibility.
The detection of SLC7A11 across various cellular compartments requires optimization of sample preparation and imaging techniques. For plasma membrane localization studies, antibodies targeting extracellular epitopes (particularly those recognizing the third extracellular loop, amino acids 218-231) prove most effective for non-permeabilized cells . Cell surface biotinylation followed by streptavidin pull-down can enrich membrane-associated SLC7A11 prior to antibody detection. For intracellular pool assessment, permeabilization conditions require careful optimization — mild detergents like 0.1% Triton X-100 generally preserve epitope structure while allowing antibody access. Colocalization studies with established compartment markers (e.g., Na⁺/K⁺-ATPase for plasma membrane, calnexin for endoplasmic reticulum) provide valuable context for subcellular distribution patterns. Super-resolution microscopy techniques offer enhanced visualization of SLC7A11 trafficking between compartments when combined with optimized immunolabeling protocols.
The selection of target epitopes significantly influences SLC7A11 antibody performance across different applications. Antibodies directed against extracellular domains, particularly the third extracellular loop (amino acids 218-231), offer advantages for live cell applications including flow cytometry and live-cell imaging . These antibodies can detect native, non-denatured protein conformations at the cell surface. In contrast, antibodies targeting intracellular domains may perform better in applications where proteins undergo denaturation, such as Western blotting or fixed-cell immunohistochemistry. Conformational epitopes spanning multiple regions of the properly folded protein often provide greater specificity but may lose reactivity during denaturation procedures. Linear epitopes, while potentially less specific, frequently maintain reactivity across a broader range of experimental conditions and sample preparation methods.
When encountering cross-reactivity with SLC7A11 antibodies, researchers should implement systematic troubleshooting approaches. Primary strategies include optimizing antibody dilution factors — where excessive concentrations typically increase non-specific binding. Implementing more stringent blocking protocols with 5% BSA or 5% non-fat milk can reduce background signal from hydrophobic interactions. The addition of 0.1-0.3% Tween-20 to washing buffers helps minimize non-specific interactions without compromising specific binding. For Western blotting applications, extended membrane blocking (overnight at 4°C) often improves signal-to-noise ratios. When persistent cross-reactivity occurs despite optimization, epitope mapping can identify regions of sequence homology with potential cross-reactive proteins. Alternative antibody clones targeting different epitopes should be tested when cross-reactivity cannot be eliminated through protocol modifications.
Post-translational modifications (PTMs) of SLC7A11 can significantly alter antibody recognition patterns. SLC7A11 undergoes various modifications including phosphorylation, glycosylation, and oxidation that may either mask or create epitopes. Phosphorylation at specific serine residues regulates SLC7A11 activity and membrane trafficking, potentially affecting antibody accessibility to certain epitopes. For detecting phosphorylated forms, phospho-specific antibodies offer the highest sensitivity and specificity. When studying glycosylated forms, enzymatic deglycosylation treatments prior to antibody application can help determine whether glycan structures interfere with epitope recognition. Oxidative modifications, particularly relevant given SLC7A11's role in redox homeostasis, may create neo-epitopes or alter protein conformation. Researchers should carefully document experimental conditions that might induce PTMs when inconsistent antibody recognition patterns are observed across different sample preparations.
Live cell imaging with SLC7A11 antibodies requires careful consideration of several technical parameters. Antibodies targeting extracellular epitopes, particularly those recognizing the third extracellular loop (amino acids 218-231), are ideal candidates as they can bind without membrane permeabilization . Fluorophore selection significantly impacts imaging quality — smaller fluorophores like Alexa Fluor 488 or Cy3 typically cause less steric hindrance than larger ones such as phycoerythrin. Incubation temperature affects both binding kinetics and cellular metabolism; room temperature (20-25°C) often represents an optimal compromise between binding efficiency and cellular health. Minimizing exposure time and light intensity reduces phototoxicity while maintaining adequate signal-to-noise ratios. The addition of 0.1% sodium azide to imaging media prevents antibody internalization when surface localization is the primary focus. For dynamic trafficking studies, Fab fragments may be preferable to full IgG molecules due to their smaller size and reduced effect on protein function.
When encountering inconsistent results with SLC7A11 antibodies across different experimental platforms, systematic troubleshooting should address platform-specific variables. For Western blotting discrepancies, variations in sample preparation (lysis buffers, detergent types, reducing conditions) often account for result inconsistencies. Membrane protein extraction efficiency varies considerably between RIPA, NP-40, and Triton X-100 buffers, with specialized membrane protein extraction kits often yielding superior results for SLC7A11 detection. Antibody concentration optimization should be performed independently for each application rather than applying standardized dilutions across platforms. When transitioning between different detection systems (chemiluminescence, fluorescence, colorimetric), sensitivity thresholds vary substantially, necessitating adjusted exposure settings and antibody concentrations. Comprehensive documentation of all protocol variables facilitates identification of critical parameters affecting reproducibility across platforms.
The functional SLC7A11 transporter operates as a heterodimer with 4F2hc (CD98), which has significant implications for antibody-based detection methods. This dimerization can mask certain epitopes while creating unique conformational epitopes at the protein-protein interface. When studying the complete functional transporter complex, antibodies directed against regions distant from the dimerization interface generally provide more consistent results. For co-immunoprecipitation studies investigating the SLC7A11-4F2hc interaction, antibodies targeting the N-terminal region of SLC7A11 often prove most effective as this domain remains accessible in the heterodimeric complex. Size exclusion chromatography followed by Western blotting can help distinguish between monomeric SLC7A11 (~55 kDa) and the heterodimeric complex (~120 kDa), providing valuable context for interpreting antibody binding patterns. Native PAGE techniques maintain protein-protein interactions and can be preferable to SDS-PAGE when studying the intact complex.
The cellular redox environment significantly impacts SLC7A11 detection due to the protein's central role in cystine/glutamate transport and redox homeostasis. Oxidative stress conditions typically upregulate SLC7A11 expression but may simultaneously induce conformational changes through oxidation of critical cysteine residues. Sample preparation methods that maintain the native redox state (addition of N-ethylmaleimide to block free thiols or inclusion of reducing agents like DTT or β-mercaptoethanol) can preserve epitope accessibility. The timing of sample collection following oxidative stress induction critically affects both expression levels and post-translational modifications. For studying redox-dependent changes in SLC7A11 localization or function, parallel probing with antibodies targeting different epitopes provides complementary data sets that help distinguish between expression changes and conformational alterations. Correlation of antibody binding patterns with functional transport assays under varying redox conditions offers valuable mechanistic insights into structure-function relationships.
Rigorous experimental design for SLC7A11 studies requires carefully selected control samples. Positive controls should include tissues or cell lines with documented high expression levels, such as certain cancer cell lines (e.g., HT-1080 fibrosarcoma cells) that rely on SLC7A11 for redox homeostasis. Negative controls ideally include genetic knockout models or cell lines with confirmed low/absent expression. When these are unavailable, siRNA-mediated knockdown samples provide alternative negative controls. For antibody validation, peptide competition assays using the immunizing peptide confirm binding specificity. Loading controls should be selected based on sample type and fractionation method — Na⁺/K⁺-ATPase or pan-cadherin for membrane fractions, GAPDH or β-actin for total cellular protein. Internal standard curves using recombinant SLC7A11 protein at known concentrations enable quantitative analysis when absolute expression levels are required. The inclusion of multiple biologically distinct controls strengthens result interpretation and facilitates troubleshooting of unexpected findings.
SLC7A11 antibodies have become increasingly valuable tools in cancer research due to the protein's upregulation in multiple tumor types and its role in chemoresistance mechanisms. Immunohistochemical applications with these antibodies enable clinical correlation studies linking expression patterns to patient outcomes across various cancer types. Cell-based assays combining SLC7A11 antibodies with proliferation or apoptosis markers provide mechanistic insights into how this transporter contributes to tumor cell survival under oxidative stress conditions. Flow cytometry with SLC7A11 antibodies targeting extracellular epitopes allows for isolation of cancer stem cell populations that frequently exhibit elevated xCT expression. Antibody-drug conjugates targeting SLC7A11 represent an emerging therapeutic strategy, leveraging the protein's elevated expression in malignant cells as a cancer-specific delivery mechanism. Multiplexed imaging approaches combining SLC7A11 antibodies with markers of tumor metabolism offer new perspectives on redox adaptation during cancer progression and therapy resistance.
The critical role of SLC7A11 in glutamate homeostasis and oxidative stress resistance makes it particularly relevant to neurodegenerative disease research. SLC7A11 antibodies facilitate the investigation of excitotoxicity mechanisms in conditions like Alzheimer's, Parkinson's, and amyotrophic lateral sclerosis, where glutamate dysregulation contributes to pathogenesis. Immunofluorescence co-localization studies with these antibodies reveal cell type-specific expression patterns in the central nervous system, identifying which neural populations may be particularly vulnerable to oxidative stress. Brain region-specific expression analysis through immunohistochemistry helps correlate SLC7A11 levels with areas showing differential vulnerability to neurodegeneration. Studies examining alterations in SLC7A11 expression and localization during disease progression provide temporal context for potential therapeutic interventions. Antibodies recognizing specific post-translational modifications of SLC7A11 in neural tissues can help identify regulatory mechanisms that might be targeted for neuroprotective strategies.
Advanced multiplex approaches incorporating SLC7A11 antibodies enable comprehensive analysis of interrelated cellular pathways. For immunofluorescence multiplexing, careful selection of compatible antibody host species and fluorophores with minimal spectral overlap is essential. Sequential staining protocols using antibody stripping or quenching between rounds facilitate detection of multiple targets in the same sample without cross-reactivity. Mass cytometry (CyTOF) employing metal-conjugated SLC7A11 antibodies permits simultaneous detection of dozens of parameters without fluorescence spectrum limitations. Imaging mass cytometry extends this capability to tissue sections, providing spatial context for SLC7A11 expression relative to multiple markers. Proximity ligation assays combining SLC7A11 antibodies with antibodies against potential interaction partners generate fluorescent signals only when proteins are within 40nm proximity, confirming physiologically relevant associations. Single-cell proteogenomic approaches correlating SLC7A11 protein levels with transcriptomic data offer insights into regulatory mechanisms governing expression patterns across heterogeneous cell populations.