NR4A2 antibodies target the NR4A2 protein (also known as Nurr1 or NGFI-Bβ), a member of the nuclear hormone receptor family. This protein regulates gene expression by binding to specific DNA sequences, influencing processes such as dopamine neuron differentiation, inflammation modulation, and apoptosis .
NR4A2 antibodies are widely used in biomedical research, including:
Parkinson’s Disease: NR4A2 is essential for dopaminergic neuron survival. Mutations in NR4A2 correlate with late-onset Parkinson’s disease .
Alzheimer’s Disease: Elevated NR4A2 in hippocampal neurons may influence neuroinflammation .
Macrophage Polarization: NR4A2 promotes M2 macrophage polarization, reducing sepsis mortality .
Autoimmunity:
Myocardial Infarction (MI):
Transcriptional Regulation: NR4A2 binds to the arginase 1 promoter, inducing M2 macrophage genes .
Autophagy Modulation: Enhances autophagic flux in cardiomyocytes under ischemia, reducing apoptosis .
Immune Pathways: Represses NF-κB-mediated inflammation and modulates GATA3 in Th2 cells .
NR4A2 (also known as NURR1, NOT, IDLDP, RNR1, TINUR, and HZF-3) is a nuclear receptor transcription factor with a canonical protein length of 598 amino acid residues and a mass of 66.6 kDa in humans. It belongs to the Nuclear hormone receptor protein family and functions as a transcriptional regulator primarily localized in the nucleus and cytoplasm .
Specific antibodies are crucial for studying NR4A2 because:
They enable detection of different isoforms (up to 2 have been reported)
They allow differentiation between subcellular localizations
They can be used to identify specific cell types expressing NR4A2, including Medulla Oblongata Splatter Neurons, Ventral Excitatory Neurons, and T Follicular Helper Cells
They facilitate monitoring of expression changes during disease development or treatment
NR4A2 antibodies are widely used in neuroscience research due to the protein's critical role in neuronal development and function:
Dopaminergic neuron studies: NR4A2 regulates the differentiation and maintenance of meso-diencephalic dopaminergic (mdDA) neurons during development
Neurological disease investigations: NR4A2 mutations or dysregulation have been linked to Parkinson's disease, Alzheimer's disease, and Schizophrenia
Developmental neurobiology: NR4A2 is involved in axon outgrowth, neuronal patterning, and terminal differentiation
DNA repair mechanisms: NR4A2 is recruited to novel nuclear foci in response to UV irradiation and participates in nucleotide excision repair
For these applications, researchers typically employ Western blotting, immunofluorescence, and immunohistochemistry techniques with specific anti-NR4A2 antibodies.
Proper validation of NR4A2 antibodies is critical for experimental reliability. A comprehensive validation approach includes:
Specificity testing:
Application-specific validation:
For Western blotting: Optimize protein extraction (nuclear and cytoplasmic fractions)
For immunofluorescence: Test different fixation protocols (paraformaldehyde vs. methanol)
For immunohistochemistry: Compare paraffin-embedded vs. frozen sections
For ChIP assays: Verify with known NR4A2 binding sites (e.g., arginase 1 promoter)
Cross-reactivity assessment:
Based on published research protocols:
Sample preparation:
Include both nuclear and cytoplasmic fractions since NR4A2 localizes to both compartments
Use phosphatase inhibitors if studying phosphorylation-dependent regulation
For tissues with potential low expression, consider immunoprecipitation prior to Western blot
Running conditions:
Antibody conditions:
Primary antibody dilutions: typically 1:1000-1:2000 (optimize for each antibody)
Incubation: overnight at 4°C for best results
Secondary antibody selection: HRP-conjugated anti-host IgG at 1:5000-1:10000
Detection considerations:
Use enhanced chemiluminescence for standard detection
For quantitative analysis, consider fluorescent secondary antibodies and imaging
Multiple anti-NR4A2 antibodies have been successfully used in Western blot applications, including those targeting the N-terminal region (AA 13-42) , middle region, and C-terminal regions.
For optimal immunohistochemical detection of NR4A2:
Tissue preparation:
Both paraffin-embedded and frozen sections have been successfully used
For paraffin sections: use heat-induced epitope retrieval (citrate buffer pH 6.0)
For frozen sections: fix with 4% paraformaldehyde for 10-15 minutes
Blocking and antibody conditions:
Block with 5-10% normal serum (matching secondary antibody host) with 0.1-0.3% Triton X-100
Primary antibody incubation: 1:100-1:500 dilution overnight at 4°C
Secondary antibody: typically 1:200-1:500 for 1-2 hours at room temperature
Detection systems:
DAB (3,3'-diaminobenzidine) for brightfield microscopy
Fluorescent secondary antibodies for co-localization studies
Controls:
Positive tissue controls: brain sections (dopaminergic neurons), lymphoid tissues
Negative controls: primary antibody omission and isotype controls
Special considerations:
When studying neurological tissues, perfusion fixation gives superior results
For inflammatory conditions, consider antigen retrieval optimization as inflammation can alter epitope accessibility
NR4A2 has been identified as a potential pro-oncogenic factor in certain cancers, particularly glioblastoma (GBM). Researchers can employ NR4A2 antibodies in the following advanced applications:
Expression profiling in patient samples:
Immunohistochemistry and tissue microarray analysis to correlate NR4A2 expression with clinical outcomes
Western blot analysis of tumor vs. normal tissue samples
Functional studies in cancer cell lines:
ChIP assays to identify direct transcriptional targets in cancer cells
Co-immunoprecipitation to detect cancer-specific protein interactions
Immunofluorescence for subcellular localization changes during cancer progression
NR4A2 antagonist screening:
Western blot and immunofluorescence to monitor NR4A2 levels/localization after drug treatment
Combine with functional assays (proliferation, migration, invasion)
Research has shown that NR4A2 knockdown inhibits GBM cell and tumor growth, induces apoptosis, and inhibits migration and invasion of GBM cells. NR4A2 antagonists like DIM-C-pPhCl represent a potential new class of anti-cancer agents with applications for treating GBM .
To investigate NR4A2's function as a transcription factor:
Chromatin Immunoprecipitation (ChIP):
Electrophoretic Mobility Shift Assay (EMSA):
Reporter gene assays:
NR4A2 plays critical roles in immune cell function and inflammatory responses:
Macrophage polarization studies:
Immunofluorescence and flow cytometry to detect NR4A2 in different macrophage subsets
Western blotting to correlate NR4A2 expression with M1/M2 markers
ChIP analysis to identify NR4A2 binding to promoters of M2 characteristic genes
T-cell differentiation and function:
Monitor NR4A2 in regulatory T cells (Tregs) using flow cytometry
Co-staining with Foxp3 (NR4A2 can trans-activate Foxp3)
Immunoprecipitation to study protein interactions in T-cell subsets
Inflammatory disease models:
Tissue immunohistochemistry in arthritis, sepsis models
Correlation of NR4A2 with inflammatory markers
Adoptive transfer experiments with NR4A2-modified immune cells
Research has shown that NR4A2 functions as a transcription factor that induces expression of M2 characteristic genes, and adoptive transfer of macrophages overexpressing NR4A2 provides protection against septic mortality . Additionally, NR4A2 is expressed at elevated levels in inflamed joint tissues from patients with rheumatoid arthritis, osteoarthritis, and psoriatic arthritis .
Recent research has revealed NR4A2's protective role in cardiomyocytes during myocardial infarction. For investigating this function:
In vivo experimental design:
Animal models: Permanent coronary ligation models
NR4A2 manipulation approaches:
Lentiviral delivery of siRNA for knockdown (lv3-siNR4A2)
Overexpression vectors for rescue experiments
Outcome measures:
Heart size assessment
Apoptosis markers (PARP, caspase3)
Autophagy markers (LC3)
Functional measurements (echocardiography)
In vitro experimental design:
Cell models:
H9c2 cardiomyocytes with serum deprivation (mimics ischemia)
Neonatal rat cardiomyocytes (NRCMs) exposed to hypoxia
NR4A2 manipulation:
siRNA knockdown (verify with qPCR and western blot)
Overexpression plasmids for rescue
Assays:
Apoptosis detection (TUNEL staining, cleaved PARP, cleaved caspase3)
Autophagy assessment (LC3 conversion, using Bafilomycin A1, 3MA, rapamycin)
Protein interaction studies (Co-IP for NR4A2 and p53)
Data interpretation should consider that NR4A2 upregulation appears to be an adaptive response to ischemia that protects cardiomyocytes by enhancing autophagy and reducing p53/Bax-mediated apoptosis .
When investigating NR4A2's role in neurodevelopmental disorders:
Genotype controls:
Tissue/cellular controls:
Brain region specificity (dopaminergic regions vs. control regions)
Cell-type controls (neurons vs. glia)
Developmental stage controls (embryonic, postnatal, adult)
Technical controls for antibody validation:
Pre-absorption with immunizing peptide
Multiple antibodies targeting different epitopes
Knockdown validation in relevant cell types
Domain-specific considerations:
Mutations in DNA binding domain (DBD) vs. ligand binding domain (LBD)
When studying variants, consider antibodies that recognize regions outside the mutation
Research has shown that de novo variants of NR4A2 are associated with neurodevelopmental disorders and epilepsy, with patients presenting with developmental delay, hypotonia, and seizures .
NR4A2 exhibits context-dependent roles in inflammation, sometimes appearing pro-inflammatory and other times anti-inflammatory. To address these contradictions:
Systematic analysis approach:
Compare tissue/cell types across studies (microglia vs. T cells vs. synoviocytes)
Examine disease models (MS vs. arthritis vs. sepsis)
Assess NR4A2 modifications (phosphorylation status, binding partners)
Reconciliation strategies:
Perform time-course experiments (early vs. late inflammation responses)
Analyze all NR4A family members simultaneously (NR4A1, NR4A2, NR4A3)
Consider signaling context (NF-κB activity, cytokine milieu)
Mechanistic resolution experiments:
ChIP-seq under different inflammatory conditions
Protein interaction studies in various cell types
Domain mutation analysis to separate different functions
Research findings show that NR4A2 can play either a proinflammatory or anti-inflammatory role depending on the immune disorder. In multiple sclerosis models, NR4A2 is selectively upregulated in T cells and augments IL-17 and IFN-γ production. In contrast, NR4A2 exerts anti-inflammatory effects by docking to NF-κB/p65 on target inflammatory gene promoters in microglia and astrocytes .
NR4A2 has been shown to participate in nucleotide excision repair. To investigate this function:
Subcellular localization studies:
Use EYFP-tagged NR4A2 to track recruitment to nuclear foci after UV irradiation
Co-localization with known DNA repair proteins
Domain analysis to identify regions required for translocation (N-terminal domain is critical)
Signaling pathway investigation:
Inhibitor studies for p38 and PARP signaling (involved in NR4A2 translocation)
Assess impact of Ligand Binding Domain mutations
Test Histone Deacetylase Inhibitors to evaluate chromatin accessibility effects
Functional DNA repair assays:
Measure clearance of UV-induced cyclobutane pyrimidine dimers (CPDs)
Measure clearance of pyrimidine-(6-4)-pyrimidone photoproduct (6-4PP) lesions
Compare wild-type vs. NR4A2 overexpression or knockdown conditions
Research has shown that NR4A2 over-expression facilitates more efficient clearance of UVR-induced DNA lesions, revealing a novel role for NR4A nuclear receptors as direct facilitators of nucleotide excision repair .
NR4A2 antibodies can facilitate drug discovery for neurological disorders through several approaches:
Target validation strategies:
Immunohistochemistry to confirm NR4A2 expression in affected tissues
Western blot to quantify expression changes in disease states
ChIP to identify dysregulated transcriptional networks
High-throughput screening applications:
Develop cell-based assays using NR4A2 antibodies for imaging
Measure nuclear translocation in response to compounds
Quantify protein stability/degradation after drug treatment
Biomarker development:
Correlate NR4A2 levels with disease progression
Monitor treatment response using NR4A2 as a surrogate marker
Stratify patients based on NR4A2 expression patterns
Given NR4A2's essential role in dopaminergic neuron development and maintenance, compounds that modulate its activity may have therapeutic potential for Parkinson's disease, schizophrenia, and other neurological disorders .
For investigating NR4A2 as a cancer therapeutic target:
Target validation experiments:
Compare NR4A2 expression in patient-derived vs. established cancer cell lines
Utilize antisense oligonucleotides for selective knockdown
Measure effects on cell proliferation, invasion, and apoptosis markers
Antagonist development approaches:
Screen compounds like DIM-C-pPhCl that act as NR4A2 antagonists
Assess effects on NR4A2-regulated transactivation using reporter assays
Confirm specificity against other NR4A family members
In vivo efficacy evaluation:
Use xenograft models (e.g., U87-MG cells in athymic nude mice)
Measure tumor growth inhibition
Monitor apoptosis markers (Annexin V, caspase cleavage, PARP)
Research has demonstrated that NR4A2 is pro-oncogenic in glioblastoma, and bis-indole-derived NR4A2 antagonists represent a novel class of anti-cancer agents with potential future clinical applications .