The ESRRG (Estrogen-Related Receptor Gamma) antibody is a specialized tool designed to detect and study the ESRRG protein, a nuclear receptor encoded by the ESRRG gene. ESRRG belongs to the NR3B subgroup of nuclear hormone receptors and functions as a transcription factor regulating genes involved in metabolism, mitochondrial function, and cancer progression . ESRRG antibodies are critical for elucidating its roles in physiological and pathological processes, including tumor suppression, immune regulation, and metabolic reprogramming.
ESRRG antibodies are widely used in biomedical research for diverse experimental techniques:
Role in Wnt Signaling: ESRRG suppresses GC growth by inhibiting TCF4/LEF1 binding to the CCND1 promoter, reducing Wnt pathway activity. Overexpression of ESRRG decreased tumor volume by 60% in xenograft models .
Clinical Relevance: Low ESRRG expression correlates with poor prognosis in GC patients (hazard ratio: 2.1) .
Treg Modulation: ESRRG maintains mitochondrial fitness in regulatory T cells (Tregs). Esrrg deficiency impaired Treg differentiation, leading to autoimmunity in mice .
Human SLE: Reduced ESRRG levels in lupus patients’ CD4+ T cells suggest its role in disease pathology .
Glycolysis Inhibition: ESRRG downregulates PKM2, suppressing the Warburg effect. DY131 (ESRRG agonist) reduced ESCC cell proliferation by 45% .
Therapeutic Potential: Combining DY131 with anti-PD-1 therapy enhanced immune checkpoint inhibition efficacy .
Western Blot: Clear detection of ESRRG at 51 kDa in GC and ESCC cell lines .
IHC: Strong nuclear staining in normal gastric tissues vs. absent signal in tumors .
Storage: Lyophilized antibodies stable at -20°C for 1 year; reconstituted aliquots stable at 4°C for 1 month .
Dilution:
ESRRG antibodies will remain pivotal in exploring its dual role as a tumor suppressor and metabolic regulator. Ongoing studies focus on:
ESRRG (estrogen-related receptor gamma), also known as NR3B3 (nuclear receptor subfamily 3, group B, member 3), is a member of the nuclear hormone receptor family of steroid hormone receptors. As an orphan nuclear receptor, it binds specifically to estrogen response elements and activates reporter genes . ESRRG has significant research importance due to its roles in various biological processes, including renal development and metabolic regulation. Recent studies have demonstrated its involvement in suppressing esophageal squamous cell carcinoma (ESCC) through metabolic reprogramming . The protein's molecular weight is approximately 51 kDa, and it functions as a transcriptional regulator with implications in development, metabolism, and disease processes .
ESRRG antibodies have been validated for multiple research applications, with varying degrees of effectiveness depending on the specific antibody and target tissue. The primary validated applications include:
Western Blot (WB): Typically used at dilutions between 1:1000-1:4000, with positive detection in human spleen and kidney tissues
Immunohistochemistry (IHC): Effective at dilutions of 1:2000-1:8000, with positive detection in rat stomach tissue, mouse and rat brain tissue, and mouse and rat heart tissue
Immunofluorescence (IF): Validated for cellular localization studies
Chromatin Immunoprecipitation (ChIP): Some ESRRG antibodies are specifically validated for studying protein-DNA interactions
For optimal results, researchers should titrate these antibodies in their specific testing systems, as effectiveness can be sample-dependent .
For effective immunohistochemical detection of ESRRG, follow this methodological approach:
Sample preparation: Use 4-6 μm sections from paraformaldehyde-fixed, paraffin-embedded tissue samples.
Antigen retrieval:
Blocking: Apply 10% heat-inactivated serum (sheep or appropriate species) in PBS to reduce non-specific binding.
Primary antibody incubation: Dilute ESRRG antibody to 1:500-1:2000 (depending on specific antibody) and incubate at 4°C overnight in a humidified chamber .
Secondary antibody application: Apply biotinylated anti-rabbit IgG (1:1000) for 1 hour at room temperature.
Detection: Use either:
For more complex co-localization studies, consider dual immunofluorescence with appropriate tissue-specific markers to contextualize ESRRG expression patterns.
Non-specific binding is a common challenge when working with ESRRG antibodies. To address this issue, implement these methodological approaches:
Optimize blocking conditions: Increase blocking serum concentration to 10-15% and extend the blocking time to 1-2 hours at room temperature. Consider using the serum from the same species as your secondary antibody .
Antibody dilution optimization: Titrate your antibody across a broader range than recommended (e.g., 1:500 to 1:8000) to identify the optimal signal-to-noise ratio for your specific tissue or cell type .
Buffer modifications: Add 0.1-0.3% Triton X-100 to your antibody dilution buffer to reduce background in IHC/IF applications. For western blots, include 0.1% Tween-20 in washing buffers.
Include validated controls:
Cross-adsorption: If cross-reactivity is suspected, pre-adsorb the antibody with the immunizing peptide or irrelevant proteins to remove non-specific antibodies.
By systematically implementing these approaches, researchers can significantly improve the specificity of ESRRG antibody staining across different experimental platforms.
Failed detection of ESRRG in Western blot experiments can stem from multiple methodological issues. Consider these troubleshooting approaches:
Sample preparation concerns:
Ensure complete protein denaturation using appropriate SDS concentration and heating (95°C for 5 minutes)
Verify protein extraction efficiency from nuclear fractions, as ESRRG is a nuclear receptor
Add protease inhibitors freshly to prevent degradation
Transfer optimization:
For the 51 kDa ESRRG protein, use PVDF membranes with 0.45 μm pore size
Consider semi-dry transfer at 15V for 30-45 minutes or wet transfer at 30V overnight at 4°C
Verify transfer efficiency using reversible protein stains (Ponceau S)
Antibody considerations:
Detection system sensitivity:
If expression is low, switch to more sensitive chemiluminescent substrates
Consider amplification steps like biotin-streptavidin systems
Increase exposure time incrementally to detect weak signals
Control experiments:
Remember that the observed molecular weight of ESRRG is approximately 51 kDa , but post-translational modifications may alter the apparent molecular weight in different tissue contexts.
To investigate ESRRG's role in metabolic reprogramming, particularly in cancer contexts, design your experimental approach using these methodological strategies:
Genetic manipulation models:
Generate stable ESRRG-overexpressing cell lines using lentiviral vectors at a multiplicity of infection of 35 plaque-forming units per cell
Create ESRRG knockdown models using shRNA or siRNA approaches
Select stable transgenic cells with puromycin (1 μg/ml) and validate expression changes via Western blot and qRT-PCR
Metabolic phenotyping:
Molecular interaction studies:
Perform chromatin immunoprecipitation (ChIP) to identify ESRRG binding to promoters of metabolic genes
Use reporter assays to assess ESRRG's effect on promoter activity of target genes
Conduct co-immunoprecipitation experiments to identify protein-protein interactions in the metabolic regulatory network
Pharmacological approaches:
Transcriptomic profiling:
This comprehensive experimental approach will provide mechanistic insights into how ESRRG regulates metabolic reprogramming, particularly in disease contexts such as cancer.
To effectively study ESRRG's interaction with the PKM2 promoter, implement these specialized methodological approaches:
Chromatin Immunoprecipitation (ChIP):
Cross-link protein-DNA interactions with 1% formaldehyde for 10 minutes
Sonicate chromatin to 200-500 bp fragments
Immunoprecipitate using validated ESRRG antibodies
Design primers targeting the PKM2 promoter region for qPCR analysis
Include positive controls (known ESRRG targets) and negative controls (non-target regions)
Promoter reporter assays:
Clone the PKM2 promoter region into a luciferase reporter vector
Create deletion and point mutation constructs to identify specific ESRRG binding elements
Co-transfect with ESRRG expression vectors and measure luciferase activity
Include appropriate controls (empty vectors, unrelated transcription factors)
DNA-protein binding assays:
Perform Electrophoretic Mobility Shift Assays (EMSA) using nuclear extracts from ESRRG-expressing cells
Include competitive and non-competitive unlabeled probes to confirm binding specificity
Use supershift assays with ESRRG antibodies to verify complex formation
In vivo footprinting:
Apply in vivo DNase I footprinting to identify protected regions within the PKM2 promoter
Compare footprints between ESRRG-expressing and non-expressing cells
Integrative genomic approaches:
Combine ChIP-seq data for ESRRG with open chromatin assays (ATAC-seq)
Correlate with RNA-seq data to establish functional consequences of binding
Validate using targeted approaches like CRISPRi to disrupt specific binding sites
This multi-faceted approach will provide robust evidence of direct ESRRG regulation of the PKM2 promoter, as suggested by existing research showing that ESRRG directly interacts with the PKM2 promoter to inhibit its activity in esophageal squamous cell carcinoma .
For detecting ESRRG in renal development studies, optimize your experimental approach using these methodological considerations:
Developmental stage selection:
Tissue preparation optimizations:
Antigen retrieval specifics:
Antibody selection and dilution:
Co-localization studies:
Pair ESRRG staining with ureteric bud markers (e.g., Cdh1) and metanephric mesenchyme markers (e.g., Six2)
Use fluorescent secondary antibodies for multi-color analysis of expression domains
Consider confocal microscopy for detailed spatial resolution
Complementary techniques:
Validate IHC findings with in situ hybridization for ESRRG mRNA
Consider laser capture microdissection followed by qRT-PCR for quantitative analysis of expression in specific structures
These optimized conditions will facilitate accurate detection of ESRRG during renal development, enabling proper characterization of its role in ureteric bud branching and renal papilla formation .
For optimal detection of ESRRG in cardiac tissue samples, implement these methodological approaches tailored to the unique challenges of cardiac tissue:
Tissue preservation considerations:
Section preparation specifics:
For paraffin sections, cut at 4-6 μm thickness
For frozen sections, cut at 8-10 μm and fix briefly in acetone or 4% PFA
Mount on positively charged slides to prevent tissue loss during processing
Optimized antigen retrieval:
Blocking and permeabilization:
Block with 10% serum (matching secondary antibody species)
Add 0.1-0.3% Triton X-100 for improved antibody penetration
Consider adding 1% BSA to reduce background in cardiac tissue
Antibody application specifics:
Detection and counterstaining:
DAB development: Monitor closely as cardiac tissue can develop high background
Counterstain: Use hematoxylin briefly (30 seconds) to avoid masking specific signals
Mounting: Use permanent mounting media for long-term storage
Controls and validation:
These methodological refinements will enhance the specificity and sensitivity of ESRRG detection in cardiac tissues, facilitating accurate characterization of its expression patterns and functional significance in cardiac biology.
To investigate ESRRG's therapeutic potential through agonists like DY131, implement this comprehensive experimental design:
In vitro efficacy assessment:
Establish dose-response curves (typically 0.1-100 μM) in relevant cell lines
Measure effects on cell proliferation, metabolism, and specific pathway activation
Compare DY131 effects in ESRRG-overexpressing versus knockdown cells to confirm specificity
Assess combination effects with standard therapeutic agents
Mechanistic investigation:
Perform RNA-Seq on control versus DY131-treated cells to identify transcriptional changes
Conduct metabolomic profiling to characterize changes in glycolysis and other metabolic pathways
Use GSEA to identify enriched pathways after DY131 treatment
Validate key targets using qRT-PCR and Western blotting
Pharmacodynamic marker development:
Preclinical models:
Test DY131 in appropriate disease models (e.g., patient-derived xenografts for ESCC)
Determine optimal dosing regimens (dose, schedule, route of administration)
Measure tumor growth inhibition, metabolic changes, and immune microenvironment alterations
Assess toxicity and pharmacokinetic properties
Immune checkpoint interaction studies:
This comprehensive experimental approach will establish the therapeutic potential of DY131 and other ESRRG agonists, with particular emphasis on metabolic reprogramming and immune modulation in cancer contexts.
To investigate ESRRG's role in the cross-talk between metabolic regulation and immune response, implement these advanced methodological approaches:
Co-culture experimental systems:
Establish co-cultures of ESRRG-manipulated cancer cells with immune cells (T cells, macrophages)
Measure immune cell activation, proliferation, and effector functions
Analyze metabolite exchange using isotope tracing or metabolomic approaches
Test the impact of DY131 treatment on immune cell function in co-culture systems
Lactate pathway analysis:
Quantify lactate production in ESRRG-overexpressing versus control cells
Measure expression of lactate transporters (MCTs) and lactate receptor GPR81
Assess the impact of exogenous lactate on immune cell function
Test whether ESRRG-mediated suppression of PKM2 reduces lactate production and enhances immune cell activity
Immune checkpoint regulation:
Analyze the expression of immune checkpoint molecules (PD-L1, CTLA-4) in response to ESRRG modulation
Use flow cytometry to measure checkpoint molecule expression on tumor and immune cells
Test whether DY131 enhances the activity of immune checkpoint inhibitors
Perform RNA-Seq to identify transcriptional networks linking ESRRG to immune regulation
In vivo immune microenvironment studies:
Establish syngeneic mouse models with ESRRG-modulated tumors
Characterize tumor-infiltrating immune populations by flow cytometry and immunohistochemistry
Test combination therapies with DY131 and immune checkpoint inhibitors
Perform single-cell RNA-Seq to define cell-type-specific responses
Mechanistic dissection using CRISPR screens:
Conduct CRISPR screens targeting metabolic and immune pathway genes in ESRRG-modulated cells
Identify synthetic lethal interactions that could be therapeutically targeted
Validate key nodes using individual gene knockouts and rescue experiments
This multifaceted approach will elucidate how ESRRG-mediated metabolic reprogramming influences anti-tumor immunity, potentially revealing new therapeutic strategies that leverage this cross-talk for enhanced efficacy of immunotherapies .
Different ESRRG antibodies demonstrate variable performance characteristics based on their epitope recognition and production methods. This comparative analysis provides guidance for application-specific selection:
When selecting an ESRRG antibody:
Consider epitope accessibility: N-terminal antibodies often perform better in native protein detection, while middle or C-terminal antibodies may be superior for denatured applications.
Application-specific considerations:
Validation method alignment: Select antibodies validated using methods similar to your intended application, as performance can vary significantly across techniques.
This comparative analysis highlights the importance of selecting application-appropriate ESRRG antibodies based on experimental goals and target species.
When faced with conflicting ESRRG expression data across different experimental techniques, implement this systematic analytical approach:
Technical validation assessment:
Verify antibody specificity through knockout/knockdown controls for protein-based methods
For RNA-based methods, check primer specificity and efficiency
Consider whether each method measures different aspects (mRNA vs. protein, total vs. active protein)
Methodological sensitivity comparison:
Western blot: Typically detects ESRRG at expected 51 kDa molecular weight
IHC/IF: Provides spatial information but may have threshold detection limitations
qRT-PCR: Highly sensitive for transcript detection but doesn't reflect protein levels
RNA-Seq: Provides comprehensive transcriptomic context but requires proper normalization
Sample preparation differences:
Consider whether nuclear extraction was performed (critical for nuclear receptors like ESRRG)
Evaluate fixation effects on epitope availability in IHC/IF
Assess RNA quality metrics (RIN values) for transcript analyses
Biological context analysis:
Resolution framework:
Prioritize data from methods with appropriate controls
Consider orthogonal validation using alternative antibodies or detection methods
When possible, use genetic approaches (overexpression, knockdown) to validate functional relevance
Employ single-cell approaches to resolve potential cellular heterogeneity issues
By systematically analyzing these factors, researchers can resolve apparently conflicting data and develop a more nuanced understanding of ESRRG expression patterns across different experimental contexts.
Several cutting-edge methodologies are poised to revolutionize our understanding of ESRRG's tissue-specific functions:
Spatial transcriptomics and proteomics:
Technologies like Visium and GeoMx DSP can map ESRRG expression with spatial context
These approaches reveal microenvironmental influences on ESRRG function
Integration with single-cell data provides unprecedented resolution of heterogeneous responses
Particularly valuable for developmental contexts where ESRRG shows spatially restricted expression patterns
CRISPR-based functional genomics:
CRISPR activation/interference systems allow tissue-specific modulation of ESRRG
Base editing technologies enable precise modification of ESRRG binding sites
CRISPR screens can identify tissue-specific cofactors and downstream effectors
In vivo CRISPR delivery methods permit temporal control of ESRRG function
Organoid and tissue-on-chip models:
Organ-specific 3D cultures better recapitulate in vivo ESRRG function
Patient-derived organoids allow study of ESRRG in human disease contexts
Microfluidic organ-on-chip systems enable analysis of metabolic functions in controlled environments
Co-culture systems can investigate ESRRG's role in cellular cross-talk
Integrative multi-omics approaches:
Combined analysis of transcriptomics, proteomics, and metabolomics data
Chromatin conformation capture techniques (Hi-C, HiChIP) to map 3D genomic interactions
Computational modeling of ESRRG-regulated metabolic networks
Systems biology approaches to contextualize ESRRG within tissue-specific regulatory networks
Advanced imaging technologies:
CODEX multiplexed protein imaging for simultaneous detection of multiple markers
Intravital microscopy to observe ESRRG function in living tissues
Super-resolution techniques to visualize subcellular localization and dynamics
Label-free imaging of metabolites to directly visualize ESRRG's impact on metabolism
These emerging methodologies will provide unprecedented insights into ESRRG's tissue-specific functions, particularly in developmental contexts and metabolic regulation, advancing our understanding of this important nuclear receptor.
Several critical questions remain unanswered regarding ESRRG antibodies and their research applications, presenting opportunities for methodological advancement and biological discovery:
Isoform-specific detection challenges:
How can we develop and validate antibodies that specifically recognize distinct ESRRG isoforms?
What are the functional differences between ESRRG isoforms across tissues and developmental stages?
How do post-translational modifications affect epitope recognition and antibody performance?
Dynamic regulation visualization:
How can we develop tools to visualize ESRRG activity in live cells and tissues?
What approaches would enable temporal tracking of ESRRG nuclear translocation and DNA binding?
How might biosensor development advance our understanding of ESRRG activation dynamics?
Therapeutic targeting considerations:
Methodological standardization needs:
What reference standards should be adopted for ESRRG antibody validation?
How can we develop consensus protocols for tissue-specific applications?
What reporting standards would improve reproducibility in ESRRG antibody-based research?
Emerging application frontiers:
How might single-cell proteomics techniques be adapted for ESRRG detection?
What multiplex imaging approaches could contextualize ESRRG within regulatory networks?
How can proximity labeling approaches identify novel ESRRG interaction partners?
Cross-species considerations:
How conserved are ESRRG epitopes across species, and how does this affect antibody selection?
What reference tissues should be used for validation in different model organisms?
How can we better translate findings between model systems and human diseases?
Addressing these outstanding questions will advance not only our technical capabilities but also our fundamental understanding of ESRRG biology, potentially revealing new therapeutic opportunities in development, metabolism, and disease.