IRX4 antibodies are immunochemical tools targeting the IRX4 protein, encoded by the IRX4 gene (UniProt ID: P78413). These antibodies enable the detection, quantification, and functional analysis of IRX4 in experimental models and clinical samples. IRX4 plays roles in cardiac development, prostate cancer (PCa) progression, and stem cell regulation, making its study critical for both basic and translational research.
IRX4 antibodies have been utilized in diverse studies:
Prostate Cancer: IRX4 knockdown suppresses PCa cell proliferation, migration, and invasion (LNCaP and C4-2B cells) .
Micropeptide Discovery: IRX4_PEP1, a micropeptide encoded by IRX4, promotes PCa stemness and docetaxel resistance via Wnt/β-catenin signaling .
Transcript Variants: Twelve IRX4 splice variants were identified in PCa cell lines, with differential expression linked to androgen regulation .
Cardiac Progenitors: Irx4-reporter mouse embryonic stem cells identified ventricular-specific progenitor cells, critical for heart development .
Western Blot: Antibodies like Invitrogen PA540481 detect IRX4 at ~54 kDa in human cell lysates .
Immunofluorescence: Cytoplasmic localization of IRX4_PEP1 was confirmed using anti-Flag antibodies in PCa cell lines .
Proteomic Analysis: IRX4_PEP1 overexpression in PC-3 cells altered Wnt pathway proteins (β-catenin, HNRNPK) .
Biomarker Potential: A short INDEL upstream of IRX4 disrupts androgen receptor (AR) binding and correlates with PCa risk, suggesting utility in prognosis .
Therapeutic Target: IRX4_PEP1’s role in chemoresistance highlights its potential as a therapeutic target .
The IRX4 antibody is designed to recognize the human IRX4 protein. This antibody was generated by immunizing a rabbit with recombinant human IRX4 protein (amino acids 287-363). The resulting immune response in the rabbit led to the production of a high concentration of antibodies against IRX4. The rabbit's blood was harvested and the anti-serum was isolated and purified using protein G, resulting in a polyclonal IRX4 antibody with a purity greater than 95%. This IRX4 antibody has been validated for its specificity in ELISA, Western blot, and immunofluorescence applications.
IRX4 plays a critical role in cardiac development and is essential for the proper functioning of the heart. Specifically, IRX4 regulates the differentiation of cardiac conduction cells, which are specialized cells that synchronize the rhythmic contraction of the heart muscle. Mutations in IRX4 have been associated with various heart conditions, including atrial fibrillation and ventricular tachycardia.
IRX4 (Iroquois Homeobox Protein 4) is a ventricle-specific transcription factor that plays a crucial role in cardiac chamber specification and development. It has gained significance in developmental biology research as it marks multipotent, ventricular-specific progenitor cells that contribute exclusively to ventricular myocardium. IRX4-positive cells exhibit cardiovascular potency, with the ability to generate endothelial cells, smooth muscle cells, and specifically ventricular myocytes. The ventricle-specific expression pattern makes IRX4 an invaluable marker for studying heart chamber differentiation and cardiac lineage specification during embryonic development . Recent studies have also implicated IRX4 in cancer biology, particularly in prostate cancer progression, suggesting its role extends beyond developmental contexts .
IRX4 antibodies are primarily utilized in three main experimental applications:
Western Blotting (WB): For detecting and quantifying IRX4 protein expression in tissue or cell lysates, with recommended dilutions typically ranging from 1:500-1:2000 .
Immunohistochemistry (IHC): For visualizing the spatial distribution of IRX4 in tissue sections, particularly useful for studying cardiac development and determining ventricle-specific expression patterns. Recommended dilutions generally range from 1:40-1:200 .
Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of IRX4 protein in solution, with higher dilutions typically used (1:5000-1:10000) .
Additionally, some specialized IRX4 antibodies have been validated for immunofluorescence (IF) applications, which are particularly valuable for co-localization studies with other cardiac markers .
When selecting an IRX4 antibody, researchers should consider their experimental model and the cross-reactivity profile of available antibodies. Based on current commercial offerings, IRX4 antibodies demonstrate varied species reactivity profiles:
For developmental studies spanning multiple model organisms, selecting antibodies with broader species reactivity can facilitate comparative analyses. For human-specific applications such as cancer research, antibodies with exclusive human reactivity might be preferred for greater specificity .
For optimal Western Blotting results with IRX4 antibodies, the following methodological considerations are recommended:
Sample Preparation:
For cardiac tissue/cells: Lyse samples in RIPA buffer containing protease inhibitors
For embryonic tissues: Consider using gentler lysis conditions to preserve protein integrity
Load 20-40μg of total protein per lane
Electrophoresis and Transfer:
Use 10-12% SDS-PAGE gels (IRX4 molecular weight: ~50-55 kDa)
Transfer to PVDF membrane (preferred over nitrocellulose for this application)
Transfer at 100V for 1 hour or 30V overnight at 4°C
Antibody Incubation:
Block with 5% non-fat milk in TBST for 1 hour at room temperature
Incubate overnight at 4°C with gentle rocking
Wash 3×10 minutes with TBST
Incubate with appropriate secondary antibody (typically anti-rabbit HRP at 1:5000-1:10000)
Wash 3×10 minutes with TBST
Detection:
Use enhanced chemiluminescence (ECL) detection system
Optimize exposure time based on signal intensity
For developmental studies tracking IRX4 expression during differentiation, consider running time-course experiments similar to those described in the literature where IRX4 expression was first detected at day 4.5 of embryoid body differentiation .
For effective immunohistochemical detection of IRX4 in cardiac tissues, the following protocol optimizations are recommended:
Tissue Preparation:
Fix tissues in 4% paraformaldehyde for 24 hours
For embryonic tissues, reduce fixation time to 6-12 hours to prevent over-fixation
Process and embed in paraffin, cutting sections at 5-7μm thickness
Antigen Retrieval (critical step):
Heat-mediated antigen retrieval in citrate buffer (pH 6.0)
Boil for 20 minutes, then cool slowly to room temperature
Antibody Application:
Block endogenous peroxidase with 3% H₂O₂ in methanol
Block non-specific binding with 5-10% normal serum matching the secondary antibody host
Incubate in a humidified chamber overnight at 4°C
For fluorescence detection, use appropriate fluorochrome-conjugated secondary antibodies
Counterstaining and Controls:
For brightfield IHC, counterstain with hematoxylin
For IF, counterstain nuclei with DAPI
Include both positive controls (ventricle tissue) and negative controls (antibody omission and non-cardiac tissue)
For dual staining to identify Irx4+ ventricular progenitor cells, co-stain with markers such as Tbx5 or Islet1 to identify first and second heart field populations, respectively, as demonstrated in published protocols .
Validating antibody specificity is critical for ensuring reliable research results. For IRX4 antibodies, employ these complementary validation approaches:
Western Blot Analysis:
Confirm single band at expected molecular weight (~50-55 kDa)
Compare expression in tissues known to express IRX4 (ventricle) versus non-expressing tissues
Include IRX4 knockout/knockdown samples as negative controls
Peptide Competition Assay:
Pre-incubate the antibody with excess immunizing peptide
Run parallel Western blots or IHC with blocked and unblocked antibody
Signal should be abolished or significantly reduced in the blocked sample
Orthogonal Validation:
Recombinant Expression System:
Overexpress tagged IRX4 in a cell line with low endogenous expression
Confirm antibody detection correlates with overexpression
Use tag-specific antibodies as controls
For reporter systems, validate co-expression of IRX4 and reporter genes by co-staining with antibodies to both IRX4 and the reporter, as demonstrated in the literature where researchers confirmed that reporter genes were restricted to Irx4+ cells .
IRX4 antibodies provide valuable tools for investigating ventricular specification during cardiomyocyte differentiation from stem cells:
Temporal Profiling of Ventricular Specification:
Conduct time-course experiments during cardiac differentiation
Use flow cytometry with IRX4 antibodies to quantify emergence of ventricular progenitors
Correlate IRX4 expression with other cardiac markers (MLC2v, cTnT) to track ventricular commitment
Purification of Ventricular Progenitors:
Employ IRX4 antibodies for fluorescence-activated cell sorting (FACS)
Combine with surface markers (Cxcr4, Pdgfr-alpha, Flk1, Flt1) for multi-parameter sorting
Culture sorted cells to assess proliferation capacity and differentiation potential
Co-expression Analysis:
Perform dual immunostaining of IRX4 with first heart field marker Tbx5 and second heart field marker Islet1
Quantify co-expression to identify distinctive ventricular subpopulations
Track maintenance or loss of IRX4 expression in differentiated progeny (ventricular cardiomyocytes maintain IRX4 expression, while endothelial and smooth muscle cells lose it)
Research has demonstrated that Irx4+ ventricular progenitor cells (VPCs) are particularly prevalent between days 4.5-6 of embryoid body differentiation, and these cells exhibit cardiovascular potency. When differentiated, approximately 64.4% become cTnT+/Mlc2v+ ventricular cardiomyocytes, 23% develop into smooth muscle cells, and 12.5% become endothelial cells . This quantitative distribution provides a benchmark for evaluating the efficiency of ventricular differentiation protocols.
Recent research has implicated IRX4 in cancer biology, particularly in relation to micropeptides and oncogenic signaling:
Detection of IRX4-Derived Micropeptides:
Use highly sensitive IRX4 antibodies capable of detecting small peptide fragments
Apply SWATH-MS/MS-based proteomic approaches to identify micropeptides (miPEPs) generated from IRX clusters
Investigate the 17 identified miPEPs from IRX clusters implicated in prostate, breast, endometrial, and ovarian cancers
Signaling Pathway Analysis:
Expression Analysis in Clinical Samples:
Use immunohistochemistry with IRX4 antibodies on tissue microarrays
Correlate expression levels with clinical outcomes and treatment response
Develop scoring systems based on staining intensity and distribution
The emerging role of IRX4 in cancer highlights its potential as a biomarker and therapeutic target. Genome-wide association studies have implicated Iroquois (IRX) gene clusters in cancer susceptibility, with IRX4-derived micropeptides potentially promoting prostate cancer progression through Wnt signaling dysregulation .
For single-cell analysis of IRX4 expression, researchers can employ several complementary approaches:
Single-Cell Flow Cytometry:
Optimize cell preparation protocols to maintain viability and antigen integrity
Perform intracellular staining for IRX4 with permeabilization steps
Combine with surface markers for multiparametric analysis
Consider using conjugated IRX4 antibodies (FITC, Alexa Fluor 647, Alexa Fluor 680) for multicolor experiments
Mass Cytometry (CyTOF):
Use metal-conjugated IRX4 antibodies
Combine with up to 40 additional markers for comprehensive phenotyping
Analyze data using dimensionality reduction techniques (t-SNE, UMAP)
Single-Cell RNA-seq Integration:
Correlate protein expression (from antibody-based methods) with transcriptomic data
Validate IRX4 transcript-to-protein relationships
Identify potential post-transcriptional regulation
Spatial Transcriptomics:
Combine IRX4 immunofluorescence with in situ hybridization techniques
Map spatial distribution of IRX4+ cells within tissue architecture
Correlate with neighboring cell populations and microenvironmental factors
Research has demonstrated that single Irx4+ ventricular progenitor cells exhibit cardiovascular potency, generating endothelial cells, smooth muscle cells, and ventricular myocytes in vitro . This finding, achieved through single-cell analysis techniques, established the multipotent nature of these progenitors and their specific contribution to ventricular myocardium development.
Researchers commonly encounter these challenges when working with IRX4 antibodies:
Low Signal Intensity:
Cause: Insufficient antigen, suboptimal antibody dilution, or ineffective antigen retrieval
Solution: Increase antibody concentration, extend incubation time, optimize antigen retrieval (try different buffers: citrate pH 6.0 vs. EDTA pH 9.0), or use signal amplification systems
High Background:
Cause: Excessive antibody concentration, insufficient blocking, or cross-reactivity
Solution: Increase blocking time/concentration, optimize antibody dilution, include additional washing steps, or use more specific detection systems
Inconsistent Results Between Experiments:
Cause: Variations in sample processing, antibody lots, or protocol execution
Solution: Standardize protocols, include positive controls in each experiment, and maintain consistent antibody lots for long-term studies
Non-specific Bands in Western Blot:
Cause: Cross-reactivity with related proteins, degradation products, or post-translational modifications
Solution: Increase stringency of washing conditions, use monoclonal antibodies if available, or validate with peptide competition assays
Epitope Masking in Fixed Tissues:
Cause: Over-fixation or protein-protein interactions blocking epitope access
Solution: Optimize fixation conditions, try alternative antigen retrieval methods, or consider using fresh-frozen sections
When working with IRX4 antibodies for ventricular progenitor cell identification, researchers have successfully addressed these challenges by implementing a reporter system that integrates luciferase, hygromycin resistance, and tdTomato reporter genes into the IRX4 locus, providing a complementary approach when antibody detection is challenging .
For detecting low abundance IRX4 or IRX4-derived peptides, consider these optimization strategies:
Signal Amplification Systems:
Sample Enrichment Techniques:
Perform subcellular fractionation to isolate nuclear fractions (where transcription factors typically localize)
Use immunoprecipitation to concentrate IRX4 protein before detection
Employ protein concentration methods for dilute samples
Alternative Detection Methods:
Technical Modifications:
For micropeptide research applications, where detection sensitivity is particularly critical, researchers have successfully identified 17 miPEPs generated from IRX clusters using specialized mass spectrometry techniques that can be complemented with targeted antibody-based validation .
Proper experimental controls are crucial for validating IRX4 antibody specificity and performance:
Positive Controls:
Tissue Controls: Ventricular myocardium (high endogenous expression)
Cell Line Controls: Cardiomyocyte cell lines with confirmed IRX4 expression
Overexpression Controls: Cells transfected with IRX4 expression constructs
Developmental Controls: Day 7-10 embryoid bodies from cardiac differentiation protocols
Negative Controls:
Tissue Controls: Non-cardiac tissues (e.g., liver, spleen)
Antibody Controls: Primary antibody omission, isotype controls
Genetic Controls: IRX4 knockout or knockdown samples
Peptide Competition: Antibody pre-incubated with immunizing peptide
Technical Controls:
Loading Controls: Housekeeping proteins (β-actin, GAPDH) for Western blots
Staining Controls: Nuclear counterstains (DAPI, Hoechst) for microscopy
Processing Controls: Samples processed identically except for the variable being tested
Differentiation Stage Controls:
Time course samples from different stages of cardiac differentiation
Multi-marker analysis to correlate IRX4 expression with known stage-specific markers
Research has established that Irx4+ cells should co-express specific markers (Cxcr4, Pdgfr-alpha, Flk1, Flt1) on the cell surface, and these can serve as validation markers when establishing new IRX4 antibody protocols .
IRX4 antibodies have significant potential in advancing regenerative medicine approaches for cardiac repair:
Ventricular-Specific Cell Therapy:
Use IRX4 antibodies to isolate and purify ventricular progenitors for transplantation
Develop sorting strategies based on IRX4 expression for clinical-grade cell preparation
Monitor engraftment and differentiation of transplanted cells in recipient tissue
Bioengineered Cardiac Tissues:
Employ IRX4 antibodies to characterize the ventricular specification of cells in engineered tissues
Validate chamber-specific organization in bioprinted cardiac constructs
Assess maturation state of ventricular cardiomyocytes in tissue engineered products
Disease Modeling:
Apply IRX4 antibodies to identify chamber-specific defects in patient-derived cardiac organoids
Track ventricular development in congenital heart disease models
Evaluate chamber-specific drug responses in personalized medicine applications
Research has demonstrated that Irx4+ ventricular progenitor cells injected into the cardiac crescent of developing mouse embryos specifically contribute to the nascent ventricle, highlighting their potential for targeted regenerative applications . This ventricular specificity could be leveraged to develop more precise approaches for treating ventricle-specific cardiac disorders.
Recent research has begun to uncover complex relationships between IRX4 expression and cancer biology:
IRX4-Derived Micropeptides in Oncogenesis:
Cancer Type Specificity:
Potential Therapeutic Implications:
Targeting IRX4 or its derived micropeptides might represent a novel therapeutic approach
Antibodies against IRX4 could help identify patients likely to benefit from Wnt pathway inhibitors
Monitoring IRX4 expression might predict chemotherapy resistance
This emerging area requires additional research to fully elucidate the mechanisms by which IRX4-derived micropeptides influence cancer progression and therapy resistance. Antibody-based detection of these micropeptides will be essential for advancing this field and developing potential therapeutic interventions .