TMSB4X Antibody, HRP conjugated is a horseradish peroxidase (HRP)-linked polyclonal antibody that binds specifically to thymosin beta-4 (TMSB4X), a 5.1 kDa protein encoded by the TMSB4X gene on the X chromosome . This antibody is engineered for high-sensitivity detection in assays such as ELISA and immunohistochemistry (IHC) .
The antibody has been validated across multiple platforms:
ELISA: Demonstrates linear detection in recombinant protein dilutions .
IHC: Used to identify TMSB4X overexpression in thyroid cancer tissues, correlating with aggressive tumor characteristics (e.g., extrathyroidal extension, BRAF V600E mutation) .
Western Blot: Detects a ~8 kDa band in human cell lysates (e.g., HL-60 leukemia cells), confirming specificity .
Cross-reactivity: No significant cross-reactivity with non-target proteins reported .
TMSB4X is overexpressed in hepatocellular carcinoma (HCC) and thyroid malignancies, where it promotes tumor proliferation, migration, and resistance to ferroptosis .
The HRP-conjugated antibody has been critical in immunohistochemical studies linking high TMSB4X expression to advanced tumor stage, lymph node metastasis, and poor prognosis .
In thyroid cancer, TMSB4X expression correlates with BRAF V600E mutations and epithelial-mesenchymal transition (EMT), suggesting its role in tumor progression .
The antibody’s use in transcriptomic datasets (e.g., TCGA) has revealed associations between TMSB4X and stromal/immune scores, highlighting its microenvironmental interactions .
| Application | Dilution Range |
|---|---|
| ELISA | 1:1,000 – 1:5,000 |
| IHC | 1:50 – 1:200 |
Intended Use: Research only; not approved for diagnostic or therapeutic applications .
Limitations: Performance may vary with sample preparation and fixation methods. Optimal dilutions must be empirically determined .
Thyroid Cancer: TMSB4X immunostaining intensity correlates with papillary tumor subtype (p = 0.001), lymph node metastasis (p = 0.024), and BRAF V600E mutation (p < 0.001) .
HCC: TMSB4X regulates inflammation-associated ferroptosis and enhances tumor invasiveness in machine learning-validated prognostic models .
Current studies propose TMSB4X as a therapeutic target in cancers with dysregulated cytoskeletal dynamics or BRAF mutations. The HRP-conjugated antibody remains pivotal for translational research exploring these pathways .
TMSB4X (thymosin beta 4 X-linked) is a small protein with 44 amino acid residues and a molecular weight of approximately 5.1 kDa. It is primarily localized in the cytoplasm and belongs to the Thymosin beta protein family. The protein plays a crucial role in cytoskeletal organization by binding to G-actin and preventing its polymerization, thereby regulating actin dynamics essential for cell motility, division, and differentiation. TMSB4X is expressed in several hemopoietic cell lines and lymphoid malignant cells, making it an important target in cancer research, particularly studies involving cell migration and invasion .
HRP-conjugated TMSB4X antibodies are primarily utilized in enzyme-linked detection methods where the horseradish peroxidase enzyme catalyzes colorimetric, chemiluminescent, or fluorescent reactions. The main applications include:
Western Blot (WB): Detection of TMSB4X protein in cell or tissue lysates with enhanced sensitivity due to signal amplification by HRP
Enzyme-Linked Immunosorbent Assay (ELISA): Quantification of TMSB4X levels in biological samples
Immunohistochemistry (IHC): Visualization of TMSB4X protein localization in tissue sections
Immunocytochemistry (ICC): Detection of TMSB4X in cultured cells
Researchers should be aware that TMSB4X antibodies may exhibit cross-reactivity across different species. Based on available commercial antibodies, many TMSB4X antibodies demonstrate reactivity with human, mouse, and rat TMSB4X proteins due to the high conservation of the protein sequence across species. Some antibodies also show cross-reactivity with bovine, horse, and other mammalian species. When designing experiments, verification of species-specific reactivity is essential, especially when working with non-human models. Additionally, researchers should validate the absence of cross-reactivity with other thymosin family proteins, particularly thymosin beta 10, which shares structural similarities with TMSB4X .
For optimal Western blot detection of TMSB4X using HRP-conjugated antibodies, researchers should follow these methodological guidelines:
Sample preparation:
Use RIPA buffer with protease inhibitors for cell/tissue lysis
For this small protein (5.1 kDa), include preparation steps to prevent protein loss
Load 20-40 μg of total protein per well
Gel electrophoresis:
Use 15-20% SDS-PAGE gels or specialized Tricine-SDS gels designed for small proteins
Include a reducing agent (β-mercaptoethanol or DTT) in sample buffer
Transfer conditions:
Perform semi-dry transfer at 15V for 30 minutes or wet transfer at 30V overnight
Use 0.2 μm PVDF membrane (preferred over nitrocellulose for small proteins)
Antibody incubation:
Block with 5% non-fat milk or BSA in TBST for 1 hour at room temperature
Dilute primary antibody according to manufacturer's recommendation (typically 1:500-1:2000)
Incubate with HRP-conjugated secondary antibody if using unconjugated primary
For direct HRP-conjugated TMSB4X antibodies, a single incubation step is sufficient
Detection:
To achieve optimal immunohistochemical detection of TMSB4X in tissue sections:
Tissue preparation:
Fix tissues in 10% neutral-buffered formalin for 24-48 hours
Process and embed in paraffin following standard protocols
Cut sections at 4-5 μm thickness
Antigen retrieval:
Heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Pressure cooker method: 3 minutes at high pressure or microwave method: 20 minutes
Blocking and antibody incubation:
Block endogenous peroxidase activity with 3% H₂O₂ for 10 minutes
Block non-specific binding with 5% normal serum
For HRP-conjugated antibodies, apply at manufacturer-recommended dilution (typically 1:50-1:200)
Incubate at 4°C overnight or at room temperature for 1-2 hours
Detection and visualization:
For HRP-conjugated antibodies, apply DAB or AEC substrate directly
Counterstain with hematoxylin for nuclear visualization
Mount with appropriate mounting medium
Controls:
For quantitative analysis of TMSB4X using ELISA methodology:
Assay setup:
Sandwich ELISA: Coat plates with capture antibody (1-10 μg/ml in carbonate buffer pH 9.6)
Direct ELISA: Coat plates with sample diluted in coating buffer
Block with 1-5% BSA or suitable blocking reagent
Detection system:
For HRP-conjugated TMSB4X antibodies: Apply directly as detection antibody
Develop with TMB substrate and stop with 2N H₂SO₄
Read absorbance at 450 nm with 570 nm reference wavelength
Standard curve preparation:
Use recombinant TMSB4X protein at concentrations ranging from 0.1-1000 ng/ml
Prepare 7-8 point standard curve with 2-fold or 3-fold serial dilutions
Include blank controls
Data analysis:
Generate 4-parameter logistic curve fit for standard curve
Calculate sample concentrations using regression equation
Apply appropriate dilution factors
Perform spike-recovery tests to verify assay accuracy
Sensitivity and range:
For optimal dual immunofluorescence staining to study TMSB4X colocalization with cytoskeletal proteins:
Sample preparation:
For cultured cells: Fix with 4% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.1-0.5% Triton X-100 for 10 minutes
For tissue sections: Use freshly frozen sections or paraffin sections with appropriate antigen retrieval
Blocking and primary antibody application:
Block with 5% normal serum from species unrelated to both primary antibodies
Apply unconjugated TMSB4X antibody together with anti-cytoskeletal protein antibody (e.g., anti-actin, anti-tubulin)
Incubate overnight at 4°C in humidified chamber
Secondary antibody selection:
Select fluorophore-conjugated secondary antibodies with minimal spectral overlap
Recommended combinations: TMSB4X (green/Alexa 488) + cytoskeletal marker (red/Alexa 594)
Include proper controls for antibody cross-reactivity
Imaging parameters:
Use confocal microscopy with sequential scanning to prevent bleed-through
Optimize pinhole settings (0.7-1.0 Airy units)
Capture Z-stacks with 0.3-0.5 μm steps for 3D colocalization analysis
Colocalization analysis:
To address contradictory findings regarding TMSB4X expression across cancer types:
Multi-omics validation strategy:
Compare protein expression (Western blot, IHC, ELISA) with mRNA expression (qRT-PCR, RNA-seq)
Validate with multiple antibodies targeting different epitopes of TMSB4X
Use mass spectrometry-based proteomics as an antibody-independent method
Context-specific analysis:
Compare TMSB4X expression across different cellular compartments (cytoplasmic vs. nuclear)
Assess expression in tumor cells vs. stromal/immune cells using dual staining
Evaluate expression at different stages of cancer progression
Methodological standardization:
Standardize tissue processing protocols across studies
Use consistent scoring systems for IHC evaluation
Apply digital pathology and automated image analysis for objective quantification
Functional validation:
Perform gene knockdown and overexpression studies in multiple cell lines
Evaluate phenotypic changes in proliferation, migration, and invasion
Correlate expression with clinical outcomes in well-characterized patient cohorts
Technical considerations for hepatocellular carcinoma (HCC) studies:
To investigate TMSB4X's role in inflammation-associated ferroptosis:
Experimental design for ferroptosis detection:
Induce ferroptosis using erastin, RSL3, or sorafenib in HCC cell lines
Measure lipid peroxidation using C11-BODIPY or MDA assays
Assess cell viability using standard assays (MTT, CCK-8)
Measure GSH levels and GPX4 activity as ferroptosis markers
TMSB4X manipulation:
Perform knockdown using siRNA or CRISPR-Cas9
Overexpress TMSB4X using appropriate expression vectors
Create stable cell lines with inducible TMSB4X expression
Protein interaction studies:
Use HRP-conjugated TMSB4X antibodies for co-immunoprecipitation followed by Western blot
Perform proximity ligation assay to detect TMSB4X interaction with ferroptosis regulators
Validate interactions using recombinant proteins in vitro
Inflammatory signaling analysis:
Measure inflammatory cytokine levels (IL-1β, IL-6, TNF-α) in culture supernatants
Analyze NF-κB pathway activation using phospho-specific antibodies
Assess NLRP3 inflammasome activation in relation to TMSB4X expression
In vivo validation:
Common issues and their solutions when working with HRP-conjugated TMSB4X antibodies:
Low signal intensity:
Increase antibody concentration incrementally (1.5-2x manufacturer recommendation)
Extend incubation time to overnight at 4°C
Use signal enhancement systems (biotinyl tyramide amplification)
Ensure appropriate antigen retrieval for fixed tissues
Check HRP activity with direct enzyme assay
High background:
Increase blocking time and concentration (use 5% BSA instead of 1-3%)
Add 0.1-0.3% Triton X-100 to washing buffer
Pre-absorb antibody with tissue powder
Reduce primary antibody concentration
Include 0.05-0.1% Tween-20 in antibody diluent
Non-specific bands in Western blot:
Perform peptide competition assay to identify specific bands
Use gradient gels to better resolve proteins of similar molecular weight
Optimize transfer conditions for small proteins
Increase washing time and buffer volume
Inconsistent results across experiments:
Comprehensive validation strategies for TMSB4X antibody specificity:
Genetic validation:
Test antibody in TMSB4X knockout or knockdown models
Use overexpression systems as positive controls
Compare staining patterns in cells with known differential expression
Peptide competition assays:
Pre-incubate antibody with excess TMSB4X peptide (10-100x molar excess)
Compare results with and without competing peptide
Use unrelated peptide as negative control
Multiple antibody validation:
Compare results using antibodies targeting different epitopes
Include monoclonal and polyclonal antibodies in validation
Cross-validate with different detection methods
Orthogonal method validation:
Compare protein detection with mRNA expression (qPCR or in situ hybridization)
Correlate with mass spectrometry-based protein identification
Use recombinant TMSB4X protein as standard
Species-specific validation:
Selection considerations for direct HRP-conjugated TMSB4X antibodies versus two-step detection:
Sensitivity requirements:
| Detection Method | Sensitivity | Signal-to-Noise | Detection Limit |
|---|---|---|---|
| Direct HRP-conjugated | Moderate | Moderate | ~1-5 ng protein |
| Two-step (primary + secondary) | Higher | Higher | ~0.1-1 ng protein |
| Three-step (biotin-streptavidin) | Highest | Variable | ~0.01-0.1 ng protein |
Application-specific considerations:
Western blot: Two-step methods generally provide better sensitivity for low abundance proteins
IHC: Direct methods reduce background from endogenous biotin and non-specific binding
ELISA: Direct methods simplify workflow but may reduce detection range
Multiplexing: Direct methods allow simpler multiplex design with multiple conjugated antibodies
Experimental constraints:
Time considerations: Direct methods reduce protocol time by ~2 hours
Sample limitations: Two-step methods consume less primary antibody
Cross-reactivity concerns: Direct methods eliminate secondary antibody cross-reactivity
Signal amplification needs: Two-step methods allow for signal enhancement
Technical factors:
Recent research has established TMSB4X as a significant regulator in hepatocellular carcinoma (HCC) progression:
Experimental design to investigate TMSB4X-ferroptosis connections:
Ferroptosis induction and assessment:
Treat HCC cell lines with ferroptosis inducers (erastin, RSL3) at varying concentrations (0.1-10 μM)
Modify TMSB4X expression through overexpression and knockdown
Measure cell death via flow cytometry (Annexin V/PI staining)
Assess lipid peroxidation (C11-BODIPY, TBARS assay)
Measure cellular iron content and ROS levels
Molecular pathway analysis:
Western blot analysis of key ferroptosis pathway components:
GPX4 expression and activity
System Xc- components (SLC7A11)
Iron metabolism regulators (FTH1, TFRC)
Lipid metabolism enzymes (ACSL4, LPCAT3)
qPCR analysis of ferroptosis-related gene expression
Lipidomic analysis to identify changes in membrane phospholipid composition
Protein interaction studies:
Co-immunoprecipitation to identify TMSB4X binding partners in ferroptosis pathway
Proximity ligation assay to visualize protein interactions in situ
CRISPR-Cas9 screening to identify genetic dependencies
Transcriptomic and proteomic profiling:
RNA-seq and proteomics analysis of TMSB4X-modulated cells
Pathway enrichment analysis focusing on redox regulation
Integration with existing ferroptosis gene signatures
In vivo models:
Therapeutic implications of TMSB4X research findings:
TMSB4X as a therapeutic target:
Potential strategies for TMSB4X inhibition:
Small molecule inhibitors disrupting TMSB4X-actin interaction
Peptide-based competitive inhibitors
RNA interference approaches (siRNA, antisense oligonucleotides)
Combination approaches with existing therapies:
Ferroptosis inducers (erastin, sorafenib)
Conventional chemotherapeutics
Immune checkpoint inhibitors
Biomarker applications:
Prognostic stratification of HCC patients based on TMSB4X expression
Prediction of response to ferroptosis-inducing therapies
Monitoring treatment response through liquid biopsy approaches
Drug discovery considerations:
Screening assays for compounds that modulate TMSB4X-mediated ferroptosis resistance
Development of ferroptosis sensitizers targeting TMSB4X-dependent pathways
Repurposing of approved drugs that may influence TMSB4X activity
Translational challenges:
Tissue-specific functions of TMSB4X in different cell types
Potential off-target effects due to TMSB4X roles in normal tissue homeostasis
Delivery methods for TMSB4X-targeting therapeutics
Resistance mechanisms that might emerge during TMSB4X-targeted therapy
Future directions:
Experimental approaches to study TMSB4X post-translational modifications:
Identification of modifications:
Mass spectrometry-based proteomic analysis:
Enrichment of TMSB4X from cell lysates via immunoprecipitation
Tryptic digestion and LC-MS/MS analysis
Targeted analysis for specific modifications (phosphorylation, acetylation, etc.)
Western blotting with modification-specific antibodies
2D gel electrophoresis to separate protein isoforms
Site-directed mutagenesis studies:
Generate mutants at predicted modification sites
Create phosphomimetic mutants (Ser/Thr to Asp/Glu)
Develop non-modifiable mutants (Ser/Thr to Ala)
Assess functional consequences in cellular assays
Temporal dynamics of modifications:
Pulse-chase experiments with modification-specific detection
Time-course analysis following stimulation
Single-cell analysis using phospho-specific antibodies
Development of FRET-based biosensors for real-time monitoring
Enzyme identification:
Inhibitor screens to identify responsible kinases/phosphatases
Enzyme-substrate validation assays
Co-immunoprecipitation studies to detect enzyme-TMSB4X complexes
In vitro kinase/phosphatase assays with recombinant proteins
Functional consequences:
Applications of single-cell technologies for TMSB4X research:
Single-cell RNA sequencing (scRNA-seq):
Characterize cell-type specific expression patterns of TMSB4X in tumor tissues
Identify rare cell populations with distinct TMSB4X expression
Construct pseudo-time trajectories to understand TMSB4X expression during cellular differentiation
Integration with spatial transcriptomics data
Single-cell proteomics approaches:
Mass cytometry (CyTOF) with TMSB4X antibodies
Multiplexed ion beam imaging (MIBI) for spatial resolution
Single-cell Western blotting for protein isoform detection
Microfluidic-based single-cell secretomics
Spatial transcriptomics and proteomics:
In situ hybridization techniques (RNAscope, MERFISH)
Digital spatial profiling
Spatial mapping of TMSB4X in relation to vascular structures and immune infiltrates
Multiplexed immunofluorescence with machine learning-based image analysis
Functional single-cell assays:
Live cell imaging with TMSB4X reporters
Single-cell secretion assays for inflammation markers
Cell migration tracking in relation to TMSB4X expression
Single-cell ferroptosis sensitivity assays
Computational integration:
Methodological advances needed for TMSB4X research in disease contexts:
Improved detection systems:
Development of high-affinity, highly specific monoclonal antibodies
Creation of nanobody-based detection reagents for live imaging
Improved TMSB4X activity assays beyond simple binding measurements
Biosensors to detect TMSB4X-actin interaction dynamics in real-time
Advanced disease models:
Patient-derived organoids with defined genetic backgrounds
Humanized mouse models for studying immune interactions
CRISPR-engineered models with endogenous TMSB4X tagging
Tissue-specific conditional knockout models
Multi-parametric ferroptosis assessment:
Simultaneous measurement of multiple ferroptosis markers
Live-cell imaging methods for ferroptosis progression
Correlative light and electron microscopy for ultrastructural analysis
In vivo ferroptosis detection methods
Inflammation monitoring:
Multiplexed cytokine profiling in relation to TMSB4X expression
Single-cell inflammasome activation assays
Live tracking of inflammatory cell recruitment
Non-invasive imaging of inflammation in animal models
Translational methodologies: