HNRNPUL1 antibodies are immunoglobulin-based reagents designed to bind specifically to the HNRNPUL1 protein, which is encoded by the HNRNPUL1 gene. This protein belongs to the heterogeneous nuclear ribonucleoprotein (hnRNP) family and participates in RNA processing, DNA repair, and transcriptional regulation . Commercially available antibodies are typically produced in rabbits or other hosts using immunogens derived from HNRNPUL1 sequences.
HNRNPUL1 antibodies are pivotal in studying the protein's multifunctional roles:
HNRNPUL1 antibodies are rigorously validated for specificity and sensitivity:
HNRNPUL1 antibodies have facilitated critical discoveries:
HNRNPUL1 interacts with PARP1 and is recruited to DSB sites via poly(ADP-ribosyl)ation .
Depletion of HNRNPUL1 sensitizes cells to DNA damage and impairs homologous recombination repair .
Binds γH2A.X, RPA32, and Chk1 in nucleoli, suggesting roles in rDNA repair .
Represses replication-dependent histone genes during cell cycle arrest .
Overexpressed in uterine and gastric cancers, correlating with poor prognosis .
Fusions with MEF2D drive leukemogenesis by deregulating HDAC9 and RAG1 .
HNRNPUL1 antibodies can be utilized across multiple experimental techniques with varying dilution requirements. The polyclonal HNRNPUL1 rabbit antibody has been validated for Western blot (1:1000-1:10000), immunoprecipitation (1:1000-1:10000), immunohistochemistry (1:20-1:200), immunofluorescence (1:20-1:200), flow cytometry, and ELISA applications . For optimal results, researchers should conduct preliminary titration experiments to determine ideal concentrations for their specific experimental system.
For Western blot applications, HNRNPUL1 presents with an observed molecular weight of 90-115 kDa, which is consistent with its predicted mass and potential post-translational modifications . When conducting immunofluorescence, secondary antibodies such as Rhodamine-labeled goat anti-rabbit IgG have been successfully used for visualization of HNRNPUL1 localization in cellular compartments .
HNRNPUL1 antibodies have been validated across multiple human cell lines and mouse tissues, making them suitable for various experimental models. The following table summarizes validated systems:
| Sample Type | Validated Applications | Notes |
|---|---|---|
| A549 cells | Western blot | Human lung adenocarcinoma cells |
| HEK-293 cells | Western blot | Human embryonic kidney cells |
| HeLa cells | Western blot, Immunoprecipitation | Human cervical cancer cells |
| MCF7 cells | Western blot | Human breast cancer cells |
| HepG2 cells | Flow cytometry, Immunofluorescence | Human liver cancer cells |
| Mouse brain tissue | Western blot | Tissue homogenate |
| Human ovary tumor | Immunohistochemistry | Paraffin-embedded samples |
The antibody demonstrates cross-reactivity with human, mouse, and rat HNRNPUL1, although other species have not been thoroughly tested . This broad species reactivity makes the antibody suitable for comparative studies across mammalian models.
For maintaining antibody integrity and experimental reproducibility, HNRNPUL1 antibodies should be stored according to specific protocols. The polyclonal rabbit antibody is formulated in PBS with 0.1% sodium azide and 50% glycerol at pH 7.3 . This formulation ensures stability during storage and prevents microbial contamination.
Storage recommendations include:
Maintain at -20°C for long-term storage
Avoid repeated freeze-thaw cycles that can degrade antibody performance
Do not aliquot, as this can introduce contamination and stability issues
Allow the antibody to equilibrate to room temperature before opening the vial
When handling the antibody, appropriate safety precautions should be followed due to the presence of sodium azide, which is toxic and can form explosive compounds in metal plumbing.
While standard applications like Western blot and immunofluorescence are well-established for HNRNPUL1 antibodies, chromatin immunoprecipitation requires additional optimization due to the protein's dynamic association with both RNA and DNA. Research has identified HNRNPUL1 as highly enriched on small nuclear RNA (snRNA) genes, making it a valuable target for ChIP studies investigating transcriptional regulation .
For successful ChIP optimization:
Perform crosslinking optimization with different formaldehyde concentrations (0.5-1%) and incubation times (5-15 minutes)
Include RNase treatment controls to distinguish between direct DNA binding and indirect association through RNA
Use sonication parameters that generate 200-500bp fragments for optimal immunoprecipitation
Pre-clear chromatin with protein A/G beads to reduce non-specific binding
Include appropriate negative controls (IgG and non-target regions) and positive controls (known HNRNPUL1 binding sites like snRNA genes)
ChIP-seq analysis has revealed that HNRNPUL1 shows distinctive binding patterns at gene promoters and at sites downstream of transcription termination sites, particularly for histone genes and snRNA genes . This supports the protein's multifunctional role in RNA processing and transcription regulation.
HNRNPUL1 plays a critical role in DNA double-strand break (DSB) repair through its interaction with NBS1 as part of the MRE11-RAD50-NBS1 (MRN) complex . To effectively study this function, researchers can employ several complementary experimental approaches:
Laser microirradiation coupled with live-cell imaging:
Transfect cells with fluorescently-tagged HNRNPUL1 constructs
Apply laser microirradiation to induce localized DNA damage
Track recruitment kinetics of HNRNPUL1 to damage sites in real-time
Compare wild-type and mutant HNRNPUL1 recruitment dynamics
Proximity ligation assays (PLA):
Detect and quantify the interaction between HNRNPUL1 and DNA repair proteins (NBS1, MRE11)
Compare interaction frequencies before and after DNA damage induction
Assess the impact of HNRNPUL1 mutations on protein-protein interactions
CRISPR-Cas9 knockout models:
IP-MS following DNA damage induction:
Immunoprecipitate HNRNPUL1 from cells before and after DNA damage
Identify damage-specific interaction partners by mass spectrometry
Validate key interactions with co-immunoprecipitation and Western blotting
These approaches provide complementary data on HNRNPUL1's recruitment, interactions, and functional impact in the DNA damage response pathway.
HNRNPUL1 is a multifunctional RNA-binding protein with several domains involved in RNA interaction, including an RGG domain and a central domain comprising tightly juxtaposed SPRY and dead polynucleotide kinase (dPNK) folds . To comprehensively analyze its RNA-binding properties, researchers should consider these methodological approaches:
RNA Immunoprecipitation (RIP) followed by sequencing:
Cross-linking and Immunoprecipitation (CLIP) methods:
In vitro binding assays with recombinant protein domains:
Express and purify individual domains (RGG, SPRY, dPNK)
Perform electrophoretic mobility shift assays with candidate RNA targets
Determine binding affinities and specificities for different RNA structures
Mutational analysis of binding sites:
Create point mutations in key RNA binding domains
Assess the impact on binding affinity and specificity
Link binding properties to functional outcomes in cellular assays
Research has shown that HNRNPUL1 exhibits approximately 40-fold enrichment within the 3' end of U2 snRNA, specifically in a stem loop required for Integrator cleavage accuracy . This indicates a potential preference for specific RNA secondary structures rather than simple sequence motifs.
Recent research has identified heterozygous variants of HNRNPUL1 in amyotrophic lateral sclerosis (ALS) patients, including severe truncating mutations like R541X that suggest loss of HNRNPUL1 function may contribute to disease pathogenesis . When investigating these mutations, researchers should employ a comprehensive approach:
Patient-derived cell models:
Functional characterization of mutant proteins:
Clone wild-type and mutant HNRNPUL1 variants into expression vectors
Assess effects on known functions (RNA binding, splicing, DNA repair)
Investigate protein-protein interactions affected by mutations
iPSC-derived motor neuron models:
Generate induced pluripotent stem cells from patient samples
Differentiate into motor neurons for disease-relevant cellular context
Evaluate phenotypes such as cytoplasmic mislocalization, RNA processing defects, or stress granule dynamics
Animal models:
Create transgenic models expressing ALS-associated HNRNPUL1 mutations
Assess motor function, neurodegeneration, and molecular pathology
Test potential therapeutic approaches
HNRNPUL1 mutations in ALS patients should be studied in the context of other RNA-binding proteins implicated in ALS, as research suggests a common etiology with spinal muscular atrophy (SMA) through disruption of small nuclear ribonucleoprotein (snRNP) biogenesis .
HNRNPUL1 depletion studies require careful experimental design and controls to avoid misinterpretation of results. Several approaches have been used to deplete HNRNPUL1, including siRNA knockdown, CRISPR-Cas9 knockout, and auxin-inducible degron systems . Critical controls include:
Expression rescue controls:
Off-target effect controls:
Temporal controls:
Cell viability and proliferation monitoring:
When studying HNRNPUL1 depletion effects on splicing, researchers should combine RNA-seq approaches with mechanistic studies focusing on U4-U6 di-snRNP and tri-snRNP formation, as HNRNPUL1 has been shown to play a critical role in these processes .
HNRNPUL1 plays a critical role in ensuring efficient Integrator-mediated cleavage of nascent RNA downstream of snRNA genes . To investigate this function, researchers should implement these methodological approaches:
Comparative functional studies with Integrator components:
Compare the effects of HNRNPUL1 depletion with depletion of core Integrator subunits (e.g., INTS11)
Research has shown that both depletions result in comparable 2-5-fold increases in unprocessed U1, U2, and U4 snRNA transcripts
Use mNET-seq (mammalian native elongating transcript sequencing) to analyze nascent RNA processing
RNA-protein interaction mapping:
Biochemical reconstitution assays:
Purify recombinant HNRNPUL1 and Integrator components
Perform in vitro cleavage assays with model substrates
Test whether HNRNPUL1 enhances Integrator cleavage efficiency or specificity
Structural studies of HNRNPUL1-RNA complexes:
Use structural biology approaches (X-ray crystallography, cryo-EM) to determine how HNRNPUL1 binds terminal hairpins in snRNAs
Investigate how this binding might facilitate Integrator recruitment or activity
Examine the potential role of the RGG domain in destabilizing RNA secondary structures
Research has shown that HNRNPUL1 binding is centered approximately 25 bases upstream from the Integrator cleavage site in U2 snRNA, similar to the typical distance between a polyadenylation site and cleavage site in pre-mRNA . This suggests a potential mechanistic parallel worth investigating.
HNRNPUL1 contains multiple functional domains including an RGG RNA-binding domain, a central globular domain comprising tightly juxtaposed SPRY and dead polynucleotide kinase (dPNK) folds, and regions mediating protein-protein interactions . To investigate structure-function relationships, researchers should consider:
Domain-specific mutational analysis:
Structural biology approaches:
Biochemical characterization of dPNK activity:
Comparative analysis with related proteins:
Understanding these structure-function relationships is crucial for interpreting the impact of disease-associated mutations and for developing potential therapeutic approaches targeting specific HNRNPUL1 functions.
HNRNPUL1 plays a multifaceted role in snRNP biogenesis and recycling, particularly in the reformation of U4:U6 di-snRNPs for further rounds of pre-mRNA splicing . To effectively study these complex processes, researchers should implement these methodological approaches:
snRNP assembly assays:
Cajal body analysis:
Protein interaction network analysis:
Investigate HNRNPUL1's interactions with key snRNP assembly factors like SART3
Use proximity ligation assays or co-immunoprecipitation to confirm interactions
Determine whether these interactions are direct or RNA-mediated
Functional splicing assays:
These approaches should be integrated to develop a comprehensive understanding of how HNRNPUL1 contributes to the complex process of snRNP assembly, recycling, and function in the context of pre-mRNA splicing.
Non-specific binding is a common challenge when working with antibodies against RNA-binding proteins like HNRNPUL1. To minimize these issues and ensure experimental reproducibility, researchers should implement these methodological approaches:
Optimized blocking protocols:
Validation with genetic models:
Cross-reactivity assessment:
Test antibody specificity against closely related proteins (HNRNPUL2, HNRNPU)
Perform immunoprecipitation followed by mass spectrometry to identify all bound proteins
Evaluate species cross-reactivity if working with non-human models
Signal-to-noise optimization:
When troubleshooting HNRNPUL1 detection in Western blots, researchers should note that the protein typically presents with a molecular weight range of 90-115 kDa, which may vary depending on post-translational modifications and isoform expression .
Co-immunoprecipitation (co-IP) is essential for studying HNRNPUL1's interactions with proteins and RNA, but requires careful optimization. Based on validated protocols, researchers should consider these methodological details:
Lysis buffer optimization:
For protein-protein interactions, use buffers containing 150-300 mM NaCl, 0.5% NP-40 or Triton X-100
For RNA-dependent interactions, include RNase inhibitors
Compare results with and without RNase treatment to distinguish direct versus RNA-mediated interactions
Antibody orientation:
Controls for specificity:
Include isotype-matched IgG controls
Use HNRNPUL1 knockout cells as negative controls
Include input samples (typically 5-10% of IP material) for quantitative comparison
Detection strategies:
Successful co-IP of HNRNPUL1 has been demonstrated with HeLa cell lysates , making this cell line a good starting point for optimization. When investigating DNA damage-related interactions, researchers should compare results between untreated and DNA damage-induced conditions to identify damage-specific interactions.
RNA immunoprecipitation (RIP) is critical for understanding HNRNPUL1's RNA-binding properties in vivo. Based on successful studies of HNRNPUL1's interactions with specific RNAs like U4 snRNA , researchers should optimize their RIP protocols with these considerations:
Crosslinking strategies:
Compare UV crosslinking (254 nm for direct protein-RNA interactions) with formaldehyde crosslinking (captures indirect interactions)
Optimize crosslinking times to balance efficiency with potential damage to epitopes
Include non-crosslinked controls to assess background and specificity
Lysis and immunoprecipitation conditions:
Use lysis buffers with RNase inhibitors to prevent RNA degradation
Include appropriate detergent concentrations (0.1-0.5% NP-40) to maintain protein solubility while preserving interactions
Optimize wash stringency to balance between specificity and sensitivity
RNA recovery and analysis:
Implement careful RNA extraction procedures to maximize recovery
For known targets like U4 snRNA, use RT-qPCR for quantitative analysis
For discovery of novel targets, couple with RNA-seq
Include appropriate normalization controls (input RNA, non-target RNAs)
Validation strategies:
When investigating context-specific interactions, researchers should compare RIP results between different cellular conditions, such as before and after DNA damage induction or in different cell cycle phases, as HNRNPUL1 plays roles in multiple cellular processes including DNA repair and histone gene regulation .