IFRD1 antibodies are immunological reagents designed to bind specifically to the IFRD1 protein, enabling its detection and quantification in various experimental settings. These antibodies are critical for:
Western blot (WB): Identifying IFRD1 protein bands (~50–51 kDa) in cell lysates (e.g., HeLa, skeletal muscle) .
Immunohistochemistry (IHC): Localizing IFRD1 in tissue sections (e.g., intrahepatic cholangiocarcinoma, brain) .
Immunofluorescence (IF): Visualizing subcellular distribution in cultured cells .
IFRD1 antibodies have been pivotal in uncovering the protein’s roles in disease mechanisms:
IFRD1 polymorphisms modulate neutrophil effector function, impacting lung disease severity . Antibodies helped link IFRD1 expression to reactive oxygen species (ROS) production in CF neutrophils .
Prognostic Marker: High IFRD1 in colon cancer predicts poor survival .
Therapeutic Target: IFRD1 depletion synergizes with glutaminase inhibitors (e.g., CB-839) in hepatocellular carcinoma .
IFRD1 antibodies confirmed its role in enhancing muscle regeneration via MyoD/NF-κB pathways, suggesting therapeutic potential for muscular dystrophies .
Western Blot: Clear bands at ~51 kDa in HeLa cells and skeletal muscle lysates .
IHC: Nuclear/cytoplasmic staining in human brain and adrenal gland tissues .
Antigen Retrieval: Citrate buffer (pH 6.0) or TE buffer (pH 9.0) optimizes IHC staining .
Cross-Reactivity: Predicted in Pig, Bovine, and Sheep but requires empirical validation .
Biomarker Development: Validate IFRD1 as a prognostic marker in multicenter cancer cohorts.
Therapeutic Screening: Test IFRD1 inhibitors in preclinical models of CF and muscular dystrophy.
IFRD1 (interferon-related developmental regulator 1) is a transcriptional coactivator/repressor that controls gene expression by interacting with transcription factors or histone deacetylase (HDAC) complexes . It plays critical roles in multiple cellular processes including osteoclast differentiation, stress response regulation, and epithelial homeostasis . IFRD1 expression is upregulated in response to acute tissue injuries, various growth factors, and cytokines . Its importance in research stems from its implication in several pathological conditions, including cystic fibrosis through the regulation of neutrophil effector cells, and as a candidate gene for autosomal-dominant sensory/motor neuropathy with ataxia . IFRD1 is also involved in bone remodeling via an Ifrd1/NF-κB/NFATc1 axis, making it a potential therapeutic target for bone diseases .
IFRD1 antibodies are immunological reagents designed to specifically bind to IFRD1 protein epitopes. The commercially available antibody (12939-1-AP) is a rabbit polyclonal IgG that targets IFRD1 in Western blot (WB), immunohistochemistry (IHC), and ELISA applications . This antibody demonstrates reactivity with human samples and is generated using an IFRD1 fusion protein (Ag3988) as the immunogen . The antibody recognizes IFRD1 with a calculated molecular weight of 50 kDa (451 amino acids) and an observed molecular weight of 51 kDa in experimental settings . It is supplied in liquid form, purified through antigen affinity purification, and stored in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 .
For optimal preservation of antibody activity, IFRD1 antibodies should be stored at -20°C, where they remain stable for one year after shipment . Importantly, aliquoting is unnecessary for -20°C storage, which simplifies laboratory handling protocols . The 20μl size preparations contain 0.1% BSA as a stabilizer . When working with the antibody, avoid repeated freeze-thaw cycles as these can degrade antibody quality and compromise experimental results. During experiments, maintain the antibody on ice when in use and return it to -20°C promptly after completing procedures. When pipetting, use sterile technique to prevent contamination that could affect antibody performance and experimental outcomes.
The optimal working dilutions for IFRD1 antibody vary depending on the specific application:
| Application | Recommended Dilution Range |
|---|---|
| Western Blot (WB) | 1:500-1:2000 |
| Immunohistochemistry (IHC) | 1:50-1:500 |
It is essential to note that these dilutions are guidelines, and researchers should titrate the antibody in each testing system to obtain optimal results as the appropriate dilution may be sample-dependent . For Western blotting applications, researchers typically start with a 1:1000 dilution and adjust based on signal intensity. For IHC applications, antigen retrieval is suggested with TE buffer pH 9.0, though citrate buffer pH 6.0 may alternatively be used . Standardizing protocols within your laboratory for specific sample types will improve reproducibility across experiments.
Validating antibody specificity is critical for ensuring reliable experimental results. For IFRD1 antibody validation, employ multiple complementary approaches:
Positive controls: Use samples known to express IFRD1, such as HeLa cells or human skeletal muscle tissue, which have been confirmed to show positive Western blot results with this antibody .
Immunohistochemistry validation: Human intrahepatic cholangiocarcinoma tissue has been verified as a positive control for IHC applications .
Molecular weight verification: Confirm that the detected band appears at the expected molecular weight of approximately 51 kDa .
siRNA knockdown: Perform siRNA-mediated knockdown of IFRD1 in your experimental system and verify reduced antibody signal. This approach was successfully used in studies examining RANKL-induced IFRD1 expression .
Peptide competition assay: Pre-incubate the antibody with excess immunizing peptide to demonstrate signal reduction in Western blots.
Secondary antibody controls: Run controls omitting the primary antibody to identify potential non-specific binding of the secondary antibody.
Several cell lines and tissue samples have been validated for IFRD1 expression studies:
When selecting experimental systems, consider that IFRD1 expression can be significantly modulated by stress conditions or specific stimuli. For instance, tunicamycin treatment increases IFRD1 mRNA and protein levels in human kidney epithelial cells through stabilization of the mRNA . Similarly, RANKL treatment induces IFRD1 expression in preosteoclasts through the activator protein 1 (AP-1) pathway . These inducible expression systems can provide valuable experimental models for studying IFRD1 regulation and function.
Several challenges may arise when detecting IFRD1 via Western blot:
Weak or absent signal: This could be due to insufficient protein loading, antibody dilution that is too high, or degraded samples. Solutions include:
Multiple bands or non-specific binding: This may indicate cross-reactivity or protein degradation. Address by:
Including proper negative controls
Optimizing blocking conditions (5% non-fat milk or BSA)
Using freshly prepared samples with complete protease inhibitor cocktail
Increasing washing stringency between antibody incubations
High background: Can result from insufficient blocking or washing. Improve by:
Extending blocking time (1-2 hours at room temperature)
Using additional wash steps with increased TBST volume
Decreasing secondary antibody concentration
Inconsistent results: May stem from variable IFRD1 expression under different cellular conditions. Standardize by:
Following established protocols specific for IFRD1 antibody (12939-1-AP) and proper controls will significantly improve Western blot results .
Optimizing IHC detection of IFRD1 requires careful attention to several experimental parameters:
Antigen retrieval: The suggested protocol recommends TE buffer at pH 9.0, though citrate buffer at pH 6.0 may also be effective as an alternative . Comparing both methods for your specific tissue is advisable.
Antibody dilution: Start with a mid-range dilution (1:200) within the recommended range (1:50-1:500) , then adjust based on signal-to-noise ratio. Titration experiments across multiple dilutions are recommended for each new tissue type.
Incubation conditions:
Primary antibody: Incubate overnight at 4°C to maximize specific binding
Secondary antibody: 1-2 hours at room temperature
Allow sections to reach room temperature before starting the protocol
Controls:
Signal development:
Optimize DAB (3,3′-diaminobenzidine) exposure time
Consider signal amplification systems for low-abundance expression
Use hematoxylin counterstaining to provide cellular context
Troubleshooting specific issues:
High background: Increase blocking time or concentration
Weak signal: Extend antibody incubation time or increase concentration
Non-specific staining: Additional washing steps or more dilute antibody
Document all optimization steps methodically to establish a reproducible protocol for your specific tissue and research questions.
When designing co-immunoprecipitation (co-IP) experiments with IFRD1 antibody, several critical factors should be considered:
Antibody compatibility: The IFRD1 antibody (12939-1-AP) has been successfully used for co-IP studies in human bladder cancer cell line 5637 , demonstrating its utility for protein interaction studies.
Lysis buffer composition: Use a buffer that preserves protein-protein interactions while effectively solubilizing membrane-associated proteins. A typical buffer might include:
50 mM Tris-HCl (pH 7.5)
150 mM NaCl
1% NP-40 or 0.5% Triton X-100
1 mM EDTA
Protease inhibitor cocktail
Phosphatase inhibitors (if studying phosphorylation-dependent interactions)
Blocking non-specific interactions: Pre-clear lysates with Protein A/G beads and appropriate control IgG to reduce background.
Controls:
Detection methods:
Data interpretation: When analyzing MS/MS results, focus on proteins exclusively detected in IFRD1 precipitates but not in IgG controls. In one study, 943 proteins were found to exclusively interact with IFRD1 .
Validation of novel interactions: Confirm key interactions using complementary methods such as proximity ligation assay, FRET, or reciprocal co-IP.
The successful identification of IFRD1-interacting proteins provides valuable insights into its functional roles in various cellular processes.
IFRD1 expression exhibits dynamic regulation under various cellular stress conditions, serving as an important stress-response mediator:
ER stress response: Tunicamycin treatment, which induces endoplasmic reticulum stress through the unfolded protein response (UPR), significantly increases IFRD1 mRNA and protein levels in human kidney epithelial cells . This increase is detectable within 4 hours of treatment and persists through at least 8 hours .
Post-transcriptional regulation: The increase in IFRD1 mRNA during stress is not due to enhanced transcription but rather to mRNA stabilization. In unstressed cells, IFRD1 mRNA has a half-life of approximately 2 hours, which extends to almost 6 hours following tunicamycin treatment .
Translational control mechanism: IFRD1 mRNA stability is regulated through an upstream open reading frame (uORF) that represses translation of the major ORF in resting cells. During stress response, phosphorylation of eIF2α inhibits translational initiation, leading to mRNA stabilization .
Transcript-specific regulation: Two separate transcripts (TR1 and TR2) have been reported for the IFRD1 gene, differing in sequence within the 5'-leader region. Interestingly, only TR1 shows stress-dependent upregulation, while TR2 remains undetectable or unresponsive to stress stimuli .
Acute tissue injury response: Beyond chemical stressors, IFRD1 expression is upregulated in vivo by acute tissue injuries, including cerebral ischemia/reperfusion and muscle trauma .
These findings highlight IFRD1's role as a stress-sensitive regulator whose expression is controlled through sophisticated post-transcriptional mechanisms, providing a rapid response to cellular stress conditions.
IFRD1 plays a crucial role in osteoclast differentiation and bone remodeling through several molecular mechanisms:
Understanding IFRD1's role in osteoclast differentiation provides important insights into the molecular mechanisms controlling bone homeostasis and potential therapeutic approaches for bone disorders.
IFRD1 functions as a transcriptional modulator through extensive protein-protein interactions with various cellular components:
Interactome complexity: Proteomic analysis using co-immunoprecipitation followed by tandem mass spectrometry (MS/MS) in human bladder cancer cell line 5637 identified 943 proteins that exclusively interacted with IFRD1 . This extensive interaction network highlights IFRD1's involvement in multiple cellular processes.
HDAC interactions: IFRD1 interacts with histone deacetylase (HDAC) complexes to regulate gene expression . This interaction is functionally important, as IFRD1 deficiency inhibits HDAC-dependent deacetylation of the NF-κB subunit p65 at residues K122 and K123 .
Transcription factor interactions: IFRD1 can function as both a transcriptional coactivator and corepressor by interacting with various transcription factors . Its interaction with NF-κB is particularly important for regulating the expression of NFATc1, a critical regulator of osteoclastogenesis .
Cell type-specific interactions: The functional outcomes of IFRD1 interactions may vary depending on the cellular context:
In osteoclast precursors, IFRD1 promotes RANKL-induced differentiation through NF-κB/NFATc1 pathway modulation
In bladder epithelial cells, IFRD1 is required for maintaining epithelial homeostasis through interactions with components of the bladder interactome
In kidney epithelial cells, IFRD1 participates in stress response pathways
Regulatory mechanism: IFRD1 can modulate protein function through influencing post-translational modifications, as demonstrated by its effect on p65 acetylation status .
Temporal dynamics: The composition of the IFRD1 interactome may change in response to cellular conditions, such as stress or differentiation signals, allowing context-specific regulation of cellular processes.
Understanding this complex interactome provides insights into how a single protein can participate in diverse cellular processes and contribute to tissue-specific functions across different physiological contexts.
Investigating the post-transcriptional regulation of IFRD1 requires sophisticated experimental approaches targeting various aspects of RNA processing and translation:
mRNA half-life determination:
Actinomycin D chase experiments have revealed that IFRD1 mRNA half-life increases from approximately 2 hours in resting cells to almost 6 hours following tunicamycin treatment
Methodology: Treat cells with actinomycin D to block transcription, then measure residual IFRD1 mRNA levels at various time points using real-time RT-PCR
Transcript-specific analysis:
Primary transcript quantification:
Primers spanning intron-exon junctions can measure the abundance of primary transcripts as an indicator of transcription rate
This approach demonstrated that tunicamycin treatment did not increase IFRD1 primary transcript levels despite elevated mature mRNA, confirming post-transcriptional regulation
Transcription-independent expression systems:
Upstream ORF (uORF) analysis:
Translation mechanism studies:
These sophisticated approaches collectively provide a comprehensive understanding of the complex post-transcriptional mechanisms regulating IFRD1 expression during cellular stress responses.
IFRD1 antibodies are becoming increasingly valuable tools in investigating various disease contexts:
Bone disorders: Given IFRD1's critical role in osteoclast differentiation through the Ifrd1/NF-κB/NFATc1 axis, IFRD1 antibodies can help evaluate its expression and molecular interactions in pathological bone loss conditions, including osteoporosis and rheumatoid arthritis . The prevention of RANKL-induced bone loss in Ifrd1-deficient mice suggests potential therapeutic applications targeting this pathway .
Bladder epithelial dysfunction: Recent findings demonstrating IFRD1's requirement for bladder epithelial homeostasis highlight its potential involvement in bladder pathologies . IFRD1 antibodies could be instrumental in investigating changes in expression or localization in conditions affecting urothelial function.
Stress-related cellular pathologies: As a stress-sensitive regulator whose expression changes during the unfolded protein response, IFRD1 may participate in conditions involving cellular stress, such as neurodegenerative diseases, ischemia/reperfusion injury, and certain metabolic disorders . Antibody-based detection could track IFRD1 dynamics during disease progression.
Cystic fibrosis: IFRD1 has been implicated in the pathophysiology of cystic fibrosis through regulation of neutrophil effector cell function . Antibodies targeting IFRD1 could help elucidate its role in inflammatory processes associated with this condition.
Neurological disorders: As a candidate gene for autosomal-dominant sensory/motor neuropathy with ataxia, IFRD1 antibodies may contribute to understanding its involvement in neurological function and dysfunction .
Cancer research: The detection of IFRD1 in bladder cancer cell lines and its extensive protein interaction network suggest potential roles in cancer biology . Immunohistochemical analysis using IFRD1 antibodies could assess its expression patterns across tumor types and stages.
These emerging applications highlight the potential of IFRD1 antibodies to advance our understanding of disease mechanisms and identify novel therapeutic targets across multiple pathological conditions.
Several methodological advances could significantly enhance IFRD1 research:
Development of monoclonal antibodies: While polyclonal antibodies like 12939-1-AP provide valuable research tools, developing highly specific monoclonal antibodies against distinct epitopes of IFRD1 would enable more precise detection of different protein isoforms and post-translational modifications.
Phospho-specific antibodies: Given the importance of protein phosphorylation in cellular signaling, antibodies that specifically recognize phosphorylated forms of IFRD1 could provide insights into its activation status under different conditions.
CRISPR/Cas9-mediated genome editing:
Generation of IFRD1 knockout cell lines across different tissue types
Creation of cells expressing tagged endogenous IFRD1 for improved detection and purification
Introduction of specific mutations to evaluate the functional significance of particular domains or residues
Live-cell imaging techniques:
Development of fluorescently tagged IFRD1 constructs that maintain native function
Application of advanced microscopy techniques (FRAP, FRET, super-resolution) to study IFRD1 dynamics and interactions in real-time
Optogenetic approaches to precisely control IFRD1 activity in specific cellular compartments
Integrative multi-omics approaches:
Combining proteomics, transcriptomics, and ChIP-seq to comprehensively map IFRD1's regulatory networks
Analysis of IFRD1-associated ribonucleoprotein complexes to better understand its role in post-transcriptional regulation
Global mapping of histone modifications influenced by IFRD1-HDAC interactions
Structural biology approaches:
Determination of IFRD1 crystal structure alone and in complex with key interacting partners
Structure-guided design of small molecule modulators of IFRD1 function
Analysis of conformational changes associated with IFRD1 activation
High-throughput screening platforms:
Development of reporter assays to monitor IFRD1 activity
Screening for compounds that modulate IFRD1 expression or function
Identification of synthetic lethal interactions in IFRD1-deficient backgrounds
These methodological advances would significantly expand our understanding of IFRD1 biology and potentially lead to new therapeutic strategies targeting IFRD1-dependent pathways in various diseases.