ILDR1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to ILDR1 Antibody

ILDR1 (Immunoglobulin-like Domain-containing Receptor 1) antibodies are specialized tools used to detect and study the ILDR1 protein, a transmembrane receptor belonging to the immunoglobulin superfamily. ILDR1 is implicated in diverse biological processes, including tricellular tight junction (tTJ) formation, alternative splicing regulation, and viral pathogenesis . These antibodies enable researchers to investigate ILDR1's expression, localization, and functional roles in health and disease.

Table 1: Select ILDR1 Antibody Suppliers

SupplierCatalog NumberClonalityApplicationsCitations
Biossbs-11013RPolyclonalWB, ELISA, FCM
Thermo Fisher ScientificPA5-98555PolyclonalWB, IHC, ELISA
Antibodies-onlineABIN1386369PolyclonalWB, FACS, ELISA
Atlas AntibodiesHPA018207PolyclonalIHC, ICC-IF, WB

Viral Pathogenesis Studies

ILDR1 antibodies have been critical in uncovering ILDR1’s role in promoting influenza A virus (IAV) replication. Overexpression of ILDR1 in HEK293T cells increased viral RNA levels and reduced cell viability during H1N1 infection, suggesting ILDR1 competes with PLSCR1 (an antiviral protein) to facilitate viral NP protein nuclear import .

Tricellular Tight Junction Regulation

ILDR1 antibodies confirmed its localization at tTJs in epithelial cells. Studies using MDCK-II cells showed ILDR1 overexpression reduces transepithelial water permeability by 35% and solute permeability by 73%, highlighting its role as a paracellular barrier .

Alternative Splicing Modulation

ILDR1 antibodies helped identify its interaction with splicing factors (e.g., TRA2B, SRSF1). ILDR1 antagonizes TRA2B-mediated inclusion of exons in TUBD1 and IQCB1, linking it to post-transcriptional regulation .

Challenges and Future Directions

While ILDR1 antibodies are widely used, challenges include:

  • Specificity: Cross-reactivity with paralogs (e.g., ILDR2, LSR) requires careful validation .

  • Functional Complexity: ILDR1’s dual roles in tTJ integrity and viral replication necessitate context-dependent interpretation .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 working days of receipt. Delivery times may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery details.
Synonyms
ILDR1 antibody; Immunoglobulin-like domain-containing receptor 1 antibody
Target Names
ILDR1
Uniprot No.

Target Background

Function
ILDR1 is a putative membrane receptor.
Gene References Into Functions
  1. A novel p.G141R mutation in ILDR1 has been identified as the likely genetic cause of hearing impairment in two unrelated Chinese Han DFNB42 families. PMID: 29849566
  2. This study represents the first identification of ILDR1 and MYO6 mutations in southwest Iran. The findings expand the spectrum of mutations associated with these genes. PMID: 29224747
  3. This research reports the first ILDR1 gene mutation in a consanguineous family with hearing loss in the United Arab Emirates. The study confirms that whole-exome sequencing is a reliable tool for diagnosing monogenic diseases characterized by high levels of allelic and locus heterogeneity. PMID: 28945813
  4. Two genome-wide significant SNPs were discovered. The first was novel and located near ISG20. The second was in TRIOBP, a gene previously linked to prelingual nonsyndromic hearing loss. Based on the TRIOBP findings, the study examined exons in known hearing loss genes and identified two additional SNPs, rs2877561 in ILDR1 and rs9493672 in EYA4. PMID: 27764096
  5. Consanguineous deaf families with novel mutations in the ILDR1 gene were identified. PMID: 26440088
  6. Whole-exome sequencing of a Korean multiplex family with partial deafness identified a novel homozygous ILDR1 variant (p.P69H) within the Ig-like domain. PMID: 25668204
  7. Data suggests a mutation in immunoglobulin-like domain containing receptor 1 (ILDR1) as a causative gene for autosomal-recessive non-syndromic hearing loss (arNSHL) in a consanguineous Saudi family with three affected children. PMID: 24768815
  8. The findings highlight the heterogeneity of the molecular organization of tight junctions (TJs) in terms of the content of LSR, ILDR1 or ILDR2. The study suggests that ILDR1-mediated recruitment of tricellulin to TCs is crucial for hearing. PMID: 23239027
  9. Gene expression analysis extended to Refractory Anemia (RA) and Refractory Anemia with excess blasts (RAEB) cases revealed ILDR1 overexpression in 36% of the RAEB subgroup. PMID: 22365942
  10. Loss-of-function mutations of ILDR1 cause autosomal-recessive hearing impairment DFNB42. PMID: 21255762
  11. Clinical trial of gene-disease association and gene-environment interaction. (HuGE Navigator) PMID: 20379614

Show More

Hide All

Database Links

HGNC: 28741

OMIM: 609646

KEGG: hsa:286676

STRING: 9606.ENSP00000345667

UniGene: Hs.98484

Involvement In Disease
Deafness, autosomal recessive, 42 (DFNB42)
Protein Families
Immunoglobulin superfamily, LISCH7 family
Subcellular Location
Cell membrane; Single-pass type I membrane protein.; [Isoform 5]: Cytoplasm, cytosol.
Tissue Specificity
Mainly expressed in prostate and to a lower extent in testis, pancreas, kidney, heart and liver.

Q&A

What is ILDR1 and why is it an important research target?

ILDR1 (Immunoglobulin-like domain-containing receptor 1) is an evolutionarily conserved type I transmembrane protein containing an immunoglobulin (Ig)-like domain. It serves as a component of tricellular tight junctions (tTJs), which are specialized structures formed at the meeting points of three epithelial cells to establish a barrier within cellular sheets . ILDR1 has garnered significant research interest due to its diverse physiological roles across multiple systems. It is highly expressed in the lungs where it has been implicated in influenza virus infection responses . Additionally, ILDR1 has been identified as the causative gene for human deafness DFNB42, with knockout mice exhibiting profound hearing loss accompanied by tricellular tight junction destruction in the inner ear . The protein's involvement in both infectious disease pathways and structural cell junctions makes ILDR1 antibodies valuable tools for investigating multiple biological processes.

What types of ILDR1 antibodies are commonly used in research?

Researchers typically employ several types of ILDR1 antibodies depending on their experimental objectives. Polyclonal antibodies against ILDR1 are frequently used for general detection purposes in techniques like western blotting, immunohistochemistry, and immunoprecipitation, as demonstrated in studies examining ILDR1 expression across mouse tissues . Monoclonal antibodies provide greater specificity and are preferred for applications requiring precise epitope recognition. Custom antibodies targeting specific domains of ILDR1, particularly the immunoglobulin-like domain or the C-terminal region (ILDR1-Cter), have been utilized in interaction studies with proteins like PLSCR1 . For cellular localization studies, researchers have successfully employed anti-ILDR1 antibodies to examine its distribution in structures like the inner ear of mice . The selection of an appropriate antibody depends on the experimental context and the specific ILDR1 region of interest.

How is ILDR1 expression distributed across tissues?

ILDR1 exhibits a tissue-specific expression pattern that correlates with its diverse physiological functions. Western blot analyses performed on C57BL/6 mice have revealed that ILDR1 is present in multiple organs but shows particularly high expression in the lungs, which serves as a primary target organ for influenza virus infection . This pulmonary expression pattern is significant given ILDR1's identified role in influenza virus pathogenesis. Immunohistochemistry studies have confirmed the protein's presence in lung tissue, with expression levels dynamically changing during viral infection—increasing significantly (by more than 15-fold) one day after H1N1 swine influenza virus (SIV) infection . ILDR1 is also expressed in the inner ear, consistent with its critical function in hearing, as demonstrated by the profound deafness phenotype in ILDR1-deficient mice . Additional research has reported ILDR1 expression in kidneys, where it contributes to renal concentrating mechanisms, with knockout mice exhibiting polyuria due to renal concentrating defects . This diverse tissue distribution highlights ILDR1's multifunctional nature and the importance of tissue-specific antibody validation.

What are the optimal protocols for ILDR1 detection in western blotting?

For effective western blot detection of ILDR1, researchers should optimize several key parameters based on established protocols. Tissue or cell lysates should be prepared using RIPA buffer supplemented with protease inhibitors to prevent protein degradation. Approximately 20-40 μg of total protein per lane is typically sufficient for ILDR1 detection in most tissues, though higher amounts (50-60 μg) may be necessary for samples with lower expression levels. Proteins should be separated on 10-12% SDS-PAGE gels and transferred to PVDF membranes using standard wet transfer protocols (100V for 60-90 minutes) . For immunodetection, membranes should be blocked with 5% non-fat milk in TBST for 1 hour at room temperature, followed by overnight incubation at 4°C with primary anti-ILDR1 antibody (typically at 1:1000 dilution, though this should be optimized for each antibody) . After washing with TBST (3 × 10 minutes), HRP-conjugated secondary antibodies should be applied at 1:5000 dilution for 1 hour at room temperature. Signal development using enhanced chemiluminescence reagents generally produces clear ILDR1 bands at approximately 55-60 kDa, with GAPDH (36 kDa) serving as an effective loading control for cytoplasmic fractions and LaminB1 as a nuclear control in fractionation experiments .

How can researchers optimize immunohistochemistry protocols for ILDR1 detection in tissue sections?

Optimizing immunohistochemistry (IHC) for ILDR1 detection requires attention to fixation, antigen retrieval, and antibody incubation conditions. Tissues should be fixed in 4% paraformaldehyde for 24 hours, followed by paraffin embedding and sectioning at 4-5 μm thickness. Heat-induced epitope retrieval in citrate buffer (pH 6.0) for 15-20 minutes is generally effective for exposing ILDR1 epitopes. For primary antibody incubation, anti-ILDR1 antibodies should be applied at dilutions between 1:100 and 1:500 (optimized for specific antibody) and incubated overnight at 4°C in a humid chamber . Following thorough washing, HRP-conjugated secondary antibodies and DAB chromogen can be used for visualization. Counter-staining with hematoxylin provides cellular context. In murine lung tissues, ILDR1 shows weak basal expression before viral infection but exhibits substantially increased signal intensity after infection, with positive staining predominantly localized to bronchiolar epithelial cells . Quantification can be performed by calculating the percentage area stained using software like ImageJ, which has revealed 3-6 fold increases in staining area during peak viral infection periods in published studies .

What controls should be included when validating ILDR1 antibodies for research use?

Comprehensive validation of ILDR1 antibodies requires multiple controls to ensure specificity and reproducibility. Positive controls should include tissues with confirmed high ILDR1 expression, such as lung tissue from wildtype mice . Negative controls are crucial and should incorporate tissues from ILDR1 knockout mice (such as those generated using CRISPR/Cas9 technology) to confirm complete absence of signal with the antibody . Technical negative controls, omitting primary antibody while maintaining all other steps, help identify non-specific binding of secondary antibodies. For interaction studies using techniques like co-immunoprecipitation, researchers should include isotype control antibodies to distinguish specific from non-specific interactions. When examining viral infection studies, both mock-infected and virus-infected samples should be processed in parallel to document changes in ILDR1 expression . Finally, multiple detection methods (western blot, IHC, immunofluorescence) should be employed to cross-validate antibody performance across techniques. Published studies have successfully validated ILDR1 antibodies using these approaches, confirming antibody specificity through absence of signal in ILDR1 knockout mice by western blot, RT-PCR, and immunohistochemistry .

How does ILDR1 expression change during influenza virus infection?

ILDR1 expression undergoes significant dynamic regulation during influenza virus infection, exhibiting a temporal pattern that correlates with viral progression. In mouse models infected with H1N1 swine influenza virus (SIV), ILDR1 protein levels dramatically increase by more than 15-fold on the first day post-infection, as determined by western blot analysis . This rapid upregulation appears to be an early response to viral challenge. Immunohistochemical examination of infected lung tissues shows that ILDR1-positive cells increase substantially by day 3 post-infection, with quantitative analysis revealing a 3-6 fold increase in the percentage area stained in infected lungs compared to uninfected controls . This elevated expression is maintained during active infection but gradually returns to baseline levels as viral load decreases, with normal expression levels observed by day 14 post-infection . The expression pattern appears to be dose-dependent, as in vitro studies using HEK293T cells have demonstrated that ILDR1 mRNA expression increases in response to viral infection in both a dosage- and time-dependent manner . This temporal regulation suggests ILDR1 plays a specific role in the host response to influenza virus infection.

What is the functional relationship between ILDR1 and PLSCR1 in influenza virus replication?

ILDR1 and PLSCR1 (phospholipid scramblase 1) form a regulatory axis that modulates influenza A virus (IAV) replication through competitive interactions. PLSCR1 functions as a natural antiviral regulator that inhibits influenza virus replication by binding to the viral nucleoprotein (NP) . ILDR1 has been identified as a novel PLSCR1-binding partner through yeast two-hybrid screening, with their interaction confirmed by co-immunoprecipitation experiments . While ILDR1 cannot directly interact with viral NP protein, it competes with NP for binding to PLSCR1 . In experimental settings, increasing amounts of ILDR1 reduce the level of NP bound to PLSCR1, and vice versa, indicating a competitive mechanism . The functional consequence of this competition appears to be regulation of viral replication, as ILDR1 overexpression promotes viral replication by reducing the available PLSCR1 that would otherwise inhibit NP function . Cell fractionation experiments have demonstrated that PLSCR1 overexpression restricts NP nuclear localization, while ILDR1 overexpression promotes NP nuclear import . This regulatory pathway (PLSCR1-ILDR1-NP) represents a previously unidentified mechanism in IAV-host interactions that influences viral replication efficiency.

How can ILDR1 antibodies be used to study viral-host protein interactions?

ILDR1 antibodies serve as valuable tools for investigating viral-host protein interactions through multiple experimental approaches. Co-immunoprecipitation (co-IP) assays using anti-ILDR1 antibodies can effectively pull down protein complexes containing ILDR1 and its binding partners, such as PLSCR1 . In these experiments, cell lysates from virus-infected or transfected cells are incubated with anti-ILDR1 antibodies coupled to protein A/G beads, followed by western blot analysis to detect interacting proteins. This approach has successfully demonstrated the competitive binding between ILDR1 and viral NP protein for PLSCR1 . For localizing ILDR1 during viral infection, immunofluorescence microscopy using anti-ILDR1 antibodies can track changes in its cellular distribution. When combined with antibodies against viral proteins, this allows visualization of potential co-localization or spatial relationships. Cell fractionation experiments, where nuclear and cytoplasmic fractions are separated and probed with anti-ILDR1 antibodies, have provided insights into how ILDR1 affects the nuclear import of viral proteins like NP . For examining the temporal dynamics of ILDR1 expression during infection, western blot and immunohistochemistry with anti-ILDR1 antibodies can track protein levels at different time points post-infection, as demonstrated in studies showing ILDR1 upregulation following H1N1 SIV infection .

How are ILDR1 antibodies utilized in hearing loss research models?

ILDR1 antibodies have become essential tools in hearing loss research, particularly in studying the DFNB42 deafness model. Immunolocalization studies using anti-ILDR1 antibodies have been crucial for mapping the protein's distribution in the inner ear of both wildtype and ILDR1-deficient mice (Ildr1w−/−) . These antibodies have helped researchers identify ILDR1's precise localization at tricellular tight junctions (tTJs) within the cochlear structures, providing insight into its functional role in maintaining the epithelial barrier integrity essential for proper hearing . In investigative workflows, anti-ILDR1 antibodies are typically employed in immunofluorescence protocols on cochlear sections, where they reveal ILDR1's distribution pattern at cell junctions. When combined with antibodies against other tight junction proteins, these studies have elucidated the structural consequences of ILDR1 deficiency on junction formation and stability . Beyond localization studies, ILDR1 antibodies have been utilized in biochemical analyses of protein expression and interactions in the auditory system, helping to characterize the molecular mechanisms underlying ILDR1-associated hearing loss. The specificity of these antibodies has been validated using tissues from ILDR1 knockout mice as negative controls, ensuring reliable interpretation of experimental results .

What is the relationship between ILDR1 and cochlear tight junction integrity?

ILDR1 serves as a critical molecular component for maintaining cochlear tight junction integrity, particularly at tricellular tight junctions (tTJs) where three epithelial cells meet. Studies using immunolocalization with ILDR1 antibodies have demonstrated that ILDR1 is specifically expressed at these tricellular contact points in the inner ear . The functional significance of this localization is evident in ILDR1-deficient mouse models (Ildr1w−/−), which exhibit profound hearing loss accompanied by disruption of tTJ formation in the cochlea . The endocochlear potential (EP), which is maintained by the tight junction barrier of the stria vascularis, has been investigated in ILDR1 knockout models. While the positive EP was not significantly altered in Ildr1k mice at either P14 (102 ± 3 mV) or 10 weeks of age (114 ± 8 mV) compared to wildtype controls, changes in the negative EP recorded under anoxic conditions have been observed, suggesting subtle alterations in ion homeostasis . The relationship between ILDR1 and cochlear tight junctions highlights the protein's essential role in establishing the compartmentalization necessary for proper auditory function, with its absence leading to structural defects that compromise hearing capacity.

How can researchers quantify ILDR1 expression changes in inner ear tissues?

Quantifying ILDR1 expression changes in inner ear tissues requires specialized techniques adapted to the complex architecture of cochlear structures. For protein-level quantification, western blot analysis using validated anti-ILDR1 antibodies can be performed on cochlear tissue extracts, with careful normalization to housekeeping proteins like GAPDH or β-actin . Due to the limited amount of protein obtainable from inner ear samples, highly sensitive detection methods such as chemiluminescence with signal enhancement or fluorescent secondary antibodies are recommended. For spatial distribution analysis, immunofluorescence followed by confocal microscopy allows quantification of ILDR1 signal intensity at specific cochlear locations, with particular attention to tricellular tight junctions . Image analysis software can measure fluorescence intensity along cell boundaries, with statistical comparison between experimental groups. For transcript-level quantification, quantitative RT-PCR using ILDR1-specific primers provides sensitive detection of expression changes, with normalization to multiple reference genes improving reliability. Microdissection techniques can be employed to isolate specific cochlear regions for more localized expression analysis. When examining ILDR1 knockout models, researchers should utilize multiple detection methods to confirm complete absence of the protein, as demonstrated in studies that validated knockout models using RT-PCR, western blot, and immunohistochemistry approaches .

How can CRISPR/Cas9-generated ILDR1 knockout models be validated using antibodies?

Comprehensive validation of CRISPR/Cas9-generated ILDR1 knockout models requires a multi-faceted approach with anti-ILDR1 antibodies playing a central role. The validation process should begin with genomic verification through DNA sequencing to confirm the intended mutation, as demonstrated in studies where a 122 bp deletion was introduced into exon 4 of the mouse Ildr1 gene, resulting in a premature stop codon and a truncated protein of only 43 amino acids . Following genomic confirmation, protein-level validation using western blot analysis with anti-ILDR1 antibodies is essential to verify complete absence of the full-length protein. This western blot validation should include multiple tissue samples, particularly those with known high ILDR1 expression such as lung tissue . Immunohistochemical validation provides additional confirmation by demonstrating absence of ILDR1 staining in tissue sections from knockout animals compared to wildtype controls . RT-PCR analysis complements protein-level testing by confirming disruption of proper mRNA expression . To ensure antibody specificity, validation experiments should include appropriate positive controls (wildtype tissues) alongside the knockout samples. When properly implemented, this comprehensive validation approach has successfully confirmed complete ILDR1 disruption in CRISPR/Cas9-generated knockout mice, providing reliable models for studying ILDR1 function in various physiological contexts .

What are the best approaches for studying ILDR1 interactions with other tight junction proteins?

Investigating ILDR1 interactions with other tight junction proteins requires sophisticated methodological approaches that leverage antibody-based techniques. Co-immunoprecipitation (co-IP) experiments using anti-ILDR1 antibodies have proven effective for identifying and characterizing protein-protein interactions, as demonstrated in studies that discovered the interaction between ILDR1 and PLSCR1 . For these experiments, cell lysates should be prepared using gentle lysis buffers (containing 1% NP-40 or Triton X-100) that preserve protein-protein interactions, with pre-clearing steps to reduce non-specific binding. Proximity ligation assays (PLA) offer an alternative in situ approach where anti-ILDR1 antibodies are used in combination with antibodies against potential interaction partners to visualize protein complexes within intact cells, producing fluorescent signals only when proteins are in close proximity (< 40 nm). Advanced imaging techniques such as super-resolution microscopy (STORM or STED) using fluorescently labeled anti-ILDR1 antibodies can visualize the nanoscale organization of ILDR1 at tricellular tight junctions in relation to other junction components. For studying dynamic interactions, Förster resonance energy transfer (FRET) between fluorescently labeled antibodies or fusion proteins can detect molecular proximity in live cells. Biochemical approaches such as crosslinking followed by immunoprecipitation with anti-ILDR1 antibodies have also been successful in capturing transient or weak interactions that might be missed by conventional co-IP methods .

How can researchers investigate the role of ILDR1 in novel physiological systems beyond current applications?

Investigating ILDR1's role in novel physiological systems requires strategic application of antibody-based techniques combined with comprehensive experimental designs. Tissue expression profiling using anti-ILDR1 antibodies in immunohistochemistry or western blot analyses across diverse tissue panels can identify previously unrecognized sites of ILDR1 expression, guiding exploration of novel functions . For systems-level analysis, researchers can employ ILDR1 antibodies in proximity-based proteomics approaches such as BioID or APEX labeling, where proteins in close proximity to ILDR1 are biotinylated and subsequently identified by mass spectrometry, revealing potential interaction networks in specific physiological contexts. Conditional tissue-specific ILDR1 knockout models, validated using anti-ILDR1 antibodies, allow targeted investigation of ILDR1 function in specific organs or cell types while avoiding the confounding effects of global deletion. For temporal regulation studies, inducible knockout systems combined with antibody-based protein detection can elucidate ILDR1's role during specific developmental stages or disease processes. In studying regulatory mechanisms, chromatin immunoprecipitation using antibodies against transcription factors combined with ILDR1 promoter analysis can identify factors controlling ILDR1 expression in different physiological contexts. Single-cell approaches, including single-cell RNA sequencing followed by protein validation with anti-ILDR1 antibodies, can reveal cell-type-specific expression patterns and functions. These methodological approaches have supported discoveries such as ILDR1's role in regulating alternative splicing through interactions with splicing factors, and its involvement in renal concentrating mechanisms .

What are common issues in ILDR1 antibody-based experiments and how can they be resolved?

Researchers frequently encounter several technical challenges when working with ILDR1 antibodies that require systematic troubleshooting approaches. One common issue is weak or absent signal in western blot applications, which may be addressed by increasing protein loading (50-60 μg), optimizing antibody concentration (testing dilutions from 1:500 to 1:2000), extending primary antibody incubation time (overnight at 4°C), or employing signal enhancement systems such as biotin-streptavidin amplification . For immunoprecipitation experiments showing poor ILDR1 recovery, researchers should optimize lysis buffer conditions (testing different detergents like NP-40, Triton X-100, or CHAPS at varying concentrations), adjust antibody amounts, and extend incubation periods . High background in immunohistochemistry applications can be reduced by implementing additional blocking steps (using both serum and BSA), increasing washing frequency and duration, and titrating antibody concentrations more carefully. Cross-reactivity issues, which may appear as unexpected bands in western blots, can be addressed by utilizing ILDR1 knockout tissues as definitive negative controls and pre-absorbing antibodies with recombinant ILDR1 protein . For variable results across experiments, standardizing sample preparation protocols, employing quantitative internal controls, and maintaining consistent antibody lots can improve reproducibility. These troubleshooting approaches have been successfully implemented in published studies examining ILDR1 expression and interactions in various experimental contexts .

How should researchers interpret conflicting ILDR1 antibody results across different applications?

When researchers encounter conflicting results using ILDR1 antibodies across different experimental applications, systematic comparative analysis and validation approaches are essential for accurate interpretation. Epitope accessibility differences often explain discrepancies between applications, as some antibodies may recognize epitopes that are accessible in denatured proteins (western blot) but masked in native conformations (immunoprecipitation) or fixed tissues (immunohistochemistry) . Researchers should perform parallel validation using multiple antibodies targeting different ILDR1 epitopes to distinguish genuine signals from artifacts. Antibody specificity verification using ILDR1 knockout tissues as negative controls is critical for conclusively identifying true ILDR1 signals across applications, as demonstrated in studies that confirmed antibody specificity using tissues from CRISPR/Cas9-generated Ildr1−/− mice . Post-translational modifications may affect antibody recognition differently across applications, particularly if modifications alter epitope accessibility or antibody affinity. For applications yielding inconsistent results, researchers should systematically optimize protocol parameters specific to each method rather than applying identical conditions across different techniques. When studying ILDR1's dynamic expression during processes like viral infection, apparent conflicts may reflect genuine biological changes rather than technical artifacts, as demonstrated by the significant upregulation of ILDR1 following H1N1 SIV infection . Interpreting results in the context of existing literature on ILDR1 expression patterns and interacting partners can help distinguish genuine observations from technical anomalies.

What considerations are important when designing experiments to study the temporal dynamics of ILDR1 expression?

Designing experiments to accurately capture the temporal dynamics of ILDR1 expression requires careful consideration of multiple technical and biological factors. Based on published observations of ILDR1's rapid upregulation following viral infection, researchers should establish appropriate sampling timeframes, with early timepoints (e.g., 6, 12, 24 hours post-stimulus) to capture initial responses, followed by extended monitoring (days to weeks) to document resolution or persistence of expression changes . Multiple detection methods should be employed in parallel, with western blotting providing quantitative protein level changes, RT-qPCR measuring transcript dynamics, and immunohistochemistry revealing spatial distribution changes over time . Synchronized experimental systems are crucial for temporal studies, whether utilizing in vitro models (synchronized cell cultures) or in vivo systems (coordinated infection or treatment protocols) . Appropriate controls should include both treatment-matched controls at each timepoint and time-matched untreated samples to distinguish treatment effects from temporal variations in baseline expression. When studying dynamic processes like viral infection, researchers should correlate ILDR1 expression changes with pathogen burden or disease markers using techniques such as viral titer measurement, as demonstrated in studies correlating ILDR1 upregulation with H1N1 SIV infection progression . Statistical analysis should account for the temporal nature of the data, using repeated measures ANOVA or mixed models rather than simpler statistical approaches. These design considerations have successfully revealed ILDR1's dynamic expression pattern during influenza virus infection, demonstrating rapid upregulation within 24 hours post-infection and gradual return to baseline levels by 14 days post-infection .

What emerging techniques might enhance ILDR1 antibody-based research?

Several emerging technologies show promise for advancing ILDR1 antibody-based research beyond current capabilities. Multiplexed antibody imaging techniques, including Multiplexed Ion Beam Imaging (MIBI) and CO-Detection by indEXing (CODEX), could enable simultaneous visualization of ILDR1 alongside dozens of other proteins in the same tissue section, providing unprecedented insights into its spatial relationships within complex cellular networks. This approach would be particularly valuable for mapping ILDR1's associations within tricellular tight junctions and during responses to viral infection . Super-resolution microscopy methods like STORM, PALM, and STED, when combined with highly specific ILDR1 antibodies, offer nanoscale visualization of ILDR1 distribution and dynamics beyond the diffraction limit of conventional microscopy. For protein interaction studies, advanced proximity labeling techniques such as TurboID or splitBioID coupled with ILDR1 antibody validation could map the complete ILDR1 interactome with temporal and spatial resolution, potentially revealing additional interaction partners beyond the identified PLSCR1 association . Single-cell proteomics approaches using antibody-based detection methods would allow characterization of ILDR1 expression heterogeneity across individual cells within tissues, complementing transcriptomic data. For therapeutic applications, engineered antibody formats including bispecific antibodies or antibody-drug conjugates targeting ILDR1 could be explored for potential intervention in diseases where ILDR1 dysregulation contributes to pathology, such as influenza infection where ILDR1 promotes viral replication .

How might ILDR1 antibodies contribute to therapeutic developments for hearing loss or viral infections?

ILDR1 antibodies hold significant potential for advancing therapeutic approaches for both hearing loss and viral infections through multiple mechanisms. For DFNB42-associated hearing loss, antibody-based screening platforms could identify small molecules that modulate ILDR1 function or stability, potentially leading to therapeutic compounds for genetic hearing impairments linked to ILDR1 mutations . Therapeutic antibodies designed to recognize and stabilize mutant forms of ILDR1 could potentially rescue protein function in certain genetic variants. For viral infection applications, the discovered competitive interaction between ILDR1 and influenza NP protein for PLSCR1 binding suggests potential therapeutic strategies . Antibodies targeting specific ILDR1 domains involved in PLSCR1 binding could potentially disrupt this interaction, enhancing PLSCR1's antiviral activity. Additionally, ILDR1-specific antibodies could serve as research tools for high-throughput screening of compounds that modulate the ILDR1-PLSCR1-NP regulatory pathway, potentially identifying novel antiviral agents . In diagnostic applications, anti-ILDR1 antibodies could be developed into prognostic markers for influenza infection severity, given the significant upregulation of ILDR1 observed during infection . Beyond direct therapeutic applications, ILDR1 antibodies remain essential research tools for elucidating the fundamental biology of both hearing mechanisms and viral-host interactions, contributing to the knowledge foundation required for next-generation therapeutic development.

What are the most significant unresolved questions regarding ILDR1 that antibody-based research might address?

Several critical knowledge gaps regarding ILDR1 biology could be addressed through strategic application of antibody-based research approaches. The precise molecular mechanisms by which ILDR1 promotes influenza virus replication remain incompletely understood, despite evidence of its competition with viral NP for PLSCR1 binding . Antibody-based studies combining advanced imaging, proximity labeling, and interaction mapping could elucidate the complete regulatory pathway and identify additional components. The exact structural regions mediating the interaction between ILDR1 and PLSCR1 have not been fully mapped; epitope-specific antibodies could help determine which domains are critical for this interaction and how viral infection might modulate these binding interfaces . Beyond its established roles in viral infection and hearing, ILDR1's potential functions in other physiological contexts remain largely unexplored. Comprehensive tissue expression profiling using anti-ILDR1 antibodies could reveal previously unrecognized sites of expression and suggest novel functions . The regulatory mechanisms controlling ILDR1's rapid upregulation during viral infection are not well characterized; antibody-based chromatin immunoprecipitation studies could identify transcription factors driving this response . Additionally, ILDR1's evolutionary relationship with its paralogs ILDR2 and LSR suggests potential functional overlap or compensation; comparative antibody-based studies examining their expression and interactions could reveal integrated biological roles. Addressing these questions through antibody-based research approaches would significantly advance our understanding of ILDR1 biology and potentially reveal new therapeutic targets for both hearing disorders and viral infections .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.