HNRNPU Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Role of HNRNPU

HNRNPU is a ubiquitously expressed RNA-binding protein encoded by the HNRNPU gene. It regulates RNA splicing, stabilizes mRNA, and facilitates chromatin interactions . Aberrant HNRNPU expression is linked to triple-negative breast cancer (TNBC) progression, neural development defects, and immune-related DNA repair mechanisms .

Proteintech 14599-1-AP (Rabbit Polyclonal)

  • Applications: WB (1:2,000–1:10,000), IP (0.5–4.0 µg), IF/ICC (1:50–1:500) .

  • Reactivity: Validated in HEK-293, HeLa, and mouse lung tissues .

  • Citations: Used in 16+ WB and 3+ RIP studies .

Abcam ab10297 (Mouse Monoclonal, Clone 3G6)

  • Applications: WB, IHC-P, Flow Cytometry .

  • Reactivity: Human-specific; detects nuclear localization in cerebellar Purkinje cells .

  • Notable Use: Identified hnRNPU’s role in microRNA sorting into extracellular vesicles .

Proteintech 16365-1-AP (Rabbit Polyclonal)

  • Applications: WB (1:1,000–1:4,000), IHC (1:500–1:2,000), Flow Cytometry .

  • Reactivity: Confirmed in HepG2 cells and mouse kidney tissues .

Antibodies-Online ABIN108577 (Mouse Monoclonal, Clone 3G6)

  • Applications: WB, ELISA, IP, IF .

  • Cross-Reactivity: Human, Monkey; no reactivity with Chicken or Lizard .

Cancer Biology

  • TNBC Progression: HNRNPU promotes TNBC cell proliferation and metastasis by modulating Wnt/β-catenin and PI3K-Akt-mTOR pathways. Knockout reduces tumor growth in vivo .

  • Biomarker Potential: Overexpression correlates with poor prognosis in breast cancer (TCGA data) .

Neurological Disorders

  • Early-Onset Seizures: HNRNPU mutations disrupt neural progenitor survival and radial migration, linked to microcephaly and intellectual disability .

DNA Repair

  • Class-Switch Recombination (CSR): HNRNPU stabilizes R-loops and facilitates C-NHEJ-mediated DNA repair in B cells .

Protocols and Best Practices

  • Storage: -20°C in PBS with 50% glycerol; avoid freeze-thaw cycles .

  • Antigen Retrieval: For IHC, use TE buffer (pH 9.0) or citrate buffer (pH 6.0) .

  • Dilution Optimization: Titrate antibodies for specific cell lines (e.g., HeLa, HEK-293) .

Clinical and Experimental Relevance

HNRNPU antibodies are indispensable for:

  • Validating HNRNPU expression in cancer subtypes via IHC .

  • Studying RNA-protein interactions through RIP and IP .

  • Investigating chromatin dynamics in neurological development .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days after receiving them. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
Synonyms
Heterogeneous nuclear ribonucleoprotein U antibody; hnRNP U antibody; hnRNP U protein antibody; HNRNPU antibody; hnRNPU protein antibody; HNRPU antibody; HNRPU_HUMAN antibody; p120 antibody; p120 nuclear protein antibody; pp120 antibody; SAF A antibody; SAF-A antibody; SAFA antibody; Scaffold attachment factor A antibody; U21.1 antibody
Target Names
HNRNPU
Uniprot No.

Target Background

Function
HNRNPU is a DNA- and RNA-binding protein that plays a crucial role in a variety of cellular processes, including nuclear chromatin organization, telomere-length regulation, transcription, mRNA alternative splicing and stability, Xist-mediated transcriptional silencing, and mitotic cell progression. It contributes to the regulation of interphase large-scale gene-rich chromatin organization through chromatin-associated RNAs (caRNAs) in a transcription-dependent manner, thereby maintaining genomic stability. HNRNPU is essential for the localization of the long non-coding Xist RNA on the inactive chromosome X (Xi) and the subsequent initiation and maintenance of X-linked transcriptional gene silencing during X-inactivation. It acts as a RNA polymerase II (Pol II) holoenzyme transcription regulator. HNRNPU promotes transcription initiation by directly associating with the core-TFIIH basal transcription factor complex, facilitating the assembly of a functional pre-initiation complex with Pol II in an actin-dependent manner. However, it inhibits Pol II transcription elongation activity by preventing the C-terminal domain (CTD) phosphorylation of Pol II and dissociates from the Pol II pre-initiation complex prior to productive transcription elongation. HNRNPU positively regulates CBX5-induced transcriptional gene silencing and retention of CBX5 in the nucleus. It also negatively regulates glucocorticoid-mediated transcriptional activation. HNRNPU is a key regulator of transcription initiation and elongation in embryonic stem cells upon leukemia inhibitory factor (LIF) signaling. It is involved in the long non-coding RNA H19-mediated Pol II transcriptional repression. HNRNPU participates in the circadian regulation of the core clock component ARNTL/BMAL1 transcription. It plays a role in the regulation of telomere length. HNRNPU acts as a global pre-mRNA alternative splicing modulator by regulating U2 small nuclear ribonucleoprotein (snRNP) biogenesis. It also influences mRNA stability. HNRNPU is a component of the CRD-mediated complex that promotes MYC mRNA stabilization. It enhances the expression of specific genes, such as tumor necrosis factor TNFA, by regulating mRNA stability, potentially through binding to the 3'-untranslated region (UTR). HNRNPU plays a role in mitotic cell cycle regulation. It is involved in the formation of stable mitotic spindle microtubules (MTs) attachment to kinetochore, spindle organization, and chromosome congression. Phosphorylation at Ser-59 by PLK1 is required for chromosome alignment and segregation and progression through mitosis. HNRNPU also contributes to the targeting of AURKA to mitotic spindle MTs. It binds to double- and single-stranded DNA and RNA, poly(A), poly(C), and poly(G) oligoribonucleotides. HNRNPU binds to chromatin-associated RNAs (caRNAs) and associates with chromatin to scaffold/matrix attachment region (S/MAR) elements in a caRNAs-dependent manner. It binds to the Xist RNA and the long non-coding H19 RNA. HNRNPU also binds to SMN1/2 pre-mRNAs at G/U-rich regions, small nuclear RNAs (snRNAs), and the 3'-UTR of TNFA mRNA. Via its RNA-binding RGG-box region, HNRNPU binds directly to the long non-coding Xist RNA, bridging the Xist RNA and the inactive chromosome X (Xi). This binding negatively regulates embryonic stem cell differentiation upon LIF signaling. HNRNPU is essential for embryonic development. It binds to brown fat long non-coding RNA 1 (Blnc1), facilitating the recruitment of Blnc1 by ZBTB7B, which is necessary to drive brown and beige fat development and thermogenesis. In the context of microbial infection, HNRNPU negatively regulates immunodeficiency virus type 1 (HIV-1) replication by preventing the accumulation of viral mRNA transcripts in the cytoplasm.
Gene References Into Functions
  1. This study highlights common phenotypic features, including emerging dysmorphism, associated with heterozygous HNRNPU mutations. It defines a novel neurodevelopmental syndrome, likely resulting from haploinsufficiency. PMID: 28944577
  2. SAF-A, in conjunction with Ku, temporally regulates base damage repair in the irradiated cell genome. PMID: 27303920
  3. HNRPU deletion is linked to neurodevelopmental disorders. PMID: 28815871
  4. This research expands the clinical and mutational spectrum associated with HNRNPU, demonstrating that heterozygous HNRNPU variants cause epilepsy, severe intellectual disability with pronounced speech impairment, and variable central nervous system, cardiac, and renal abnormalities. PMID: 28393272
  5. Findings indicate that SAF-A and caRNAs form a dynamic, transcriptionally responsive chromatin mesh that organizes large-scale chromosome structures and protects the genome from instability. PMID: 28622508
  6. These results confirm and refine the complex genotype-phenotype correlations present in the 1qter microdeletion syndrome and define more precisely the neurodevelopmental phenotypes associated with genetic alterations of AKT3, ZBTB18, and HNRNPU in humans. PMID: 28283832
  7. Mutual regulatory mechanisms exist between PP4 and SAF-A. Interactions between PP4 and SAF-A play a role in prometaphase/metaphase transition. PMID: 27041735
  8. CENP-W interacts with hnRNPU and may contribute to kinetochore-microtubule attachment in mitotic cells. PMID: 26881882
  9. Nuclear TDP-43 becomes neurotoxic by escaping the inhibitory regulation by hnRNP-U or hnRNP-A2. hnRNP-U inhibits TDP-43-mediated alterations in splicing of POLDIP3 mRNA. PMID: 25378556
  10. The results suggest that NCRNA00201 is not a major gene for microcephaly and corpus callosum abnormalities but is a good candidate for intellectual disability and seizures. PMID: 22678713
  11. Both phosphorylation and dephosphorylation of SAF-A serine 59 by PLK1 and PP2A, respectively, are required for accurate and timely exit from mitosis. PMID: 25986610
  12. This research demonstrates that H19 inhibits RNA Pol II-mediated transcription by disrupting the hnRNP U-actin complex. PMID: 23811339
  13. The hnRNP-U protection of cells after oxidative stress is largely due to the enhancement of NEIL1-mediated repair. PMID: 22902625
  14. This study experimentally verified the targets heterogeneous nuclear ribonucleoprotein U, phosphatidylinositol-3-OH kinase, the WNK (with-no-lysine) kinase family, and USP19 (ubiquitin-specific peptidase 19) as vulnerable nodes in the host cellular defense system against viruses. PMID: 22810585
  15. These findings suggest that HNRNPU, FAM36A, and NCRNA00201 are not major genes for microcephaly and corpus callosum abnormalities but are good candidates for intellectual disability (ID) and seizures. PMID: 22678713
  16. SAF-A interacts with BRG1, and both components are required for RNA Polymerase II-mediated transcription. PMID: 22162999
  17. hnRNP U is a regulator of SMN2 splicing. PMID: 22325991
  18. The nuclear scaffold protein (SAF-A) is a novel spindle regulator that plays an essential role in kinetochore-microtubules (MT) attachment and mitotic spindle organization. PMID: 21242313
  19. Single nucleotide polymorphism in the HNRPU gene is associated with speech delay, seizures, and variable corpus callosum thickness. PMID: 20382278
  20. hnRNP U/SAF-A/SP120 regulates the enzyme DNA Topoisomerase IIbeta in dual ways. PMID: 20554522
  21. Ash2l and Saf-A are recruited to the inactive X chromosome at the onset of stable X inactivation. PMID: 20150277
  22. hnRNP-U engages a highly neddlylated active SCF beta-TRCP which dissociates in the presence of a high-affinity substrate, resulting in the ubiquitination of the latter. PMID: 11850407
  23. Scaffold/matrix attachment region elements interact with a p300-scaffold attachment factor A complex and are bound by acetylated nucleosomes. PMID: 11909954
  24. Heterogeneous nuclear ribonuclear protein U associates with YAP and regulates its co-activation of Bax transcription. PMID: 15096513
  25. PRMT1 plays a role in arginine methylation of SAF-A. PMID: 15364944
  26. The results suggest that HIV-1 requires machinery for the nuclear export of viral mRNAs that can be specifically blocked by an interfering gene. PMID: 16916646
  27. These findings enhance our understanding of how WT1 exerts its transcriptional regulatory role and suggest that hnRNP-U may be a candidate Wilms' tumor gene at 1q44. PMID: 16924231
  28. This study demonstrates the spatial proximities among a constellation of functionally related sites found within euchromatic regions of the cell nucleus, including: HP1gamma, RNA polymerase II, matrin 3, and SAF-A sites. PMID: 18618731
  29. The histone acetyltransferase (HAT) PCAF associates with actin and hnRNP U. PMID: 18710935
  30. hnRNP-U is phosphorylated at Ser59 by DNA-PK in vitro and in cells in response to DNA double-strand breaks. PMID: 19351595
  31. Data demonstrate that hnRNP U is involved in HP1alpha function, shedding new light on the mode of action of HP1alpha and the function of hnRNP U. PMID: 19617346

Show More

Hide All

Database Links

HGNC: 5048

OMIM: 602869

KEGG: hsa:3192

STRING: 9606.ENSP00000283179

UniGene: Hs.106212

Involvement In Disease
Epileptic encephalopathy, early infantile, 54 (EIEE54)
Subcellular Location
Nucleus. Nucleus matrix. Chromosome. Nucleus speckle. Cytoplasm, cytoskeleton, microtubule organizing center, centrosome. Chromosome, centromere, kinetochore. Cytoplasm, cytoskeleton, spindle. Cytoplasm, cytoskeleton, spindle pole. Midbody. Cytoplasm. Cell surface. Cytoplasmic granule.
Tissue Specificity
Widely expressed.

Q&A

What is HNRNPU and what cellular functions does it perform?

HNRNPU, also known as HNRPU, SAFA, and U21.1, is an 825 amino acid protein that is extensively phosphorylated. It functions as a component of ribonucleosomes and is localized in cytoplasmic mRNP granules containing untranslated mRNAs . HNRNPU was originally identified as a component of heterogeneous ribonucleoprotein (hnRNP) complexes and is also known as nuclear scaffold attachment factor A (SAF-A) .

HNRNPU possesses dual binding capabilities - it has a DNA binding domain at the N-terminus and an RNA-binding domain (RGG domain) at the C-terminus. This dual binding ability enables HNRNPU to perform multiple cellular functions including:

  • Transcriptional regulation

  • Nuclear matrix/scaffold attachment

  • Alternative splicing

  • mRNA stability control (particularly for inflammatory cytokines like IL-6 and IL-1β)

  • Post-transcriptional regulation

Though primarily nuclear, HNRNPU is a nucleocytoplasmic shuttling protein that can be detected in cytoplasmic fractions, suggesting distinct roles in different cellular compartments .

Why do HNRNPU antibodies show different applications across research platforms?

HNRNPU antibodies demonstrate versatility across multiple experimental applications, with each application providing unique insights into protein function, localization, or interactions. Based on validated research data, HNRNPU antibodies can be reliably used in the following applications:

ApplicationDetection CapabilitiesKey Considerations
Western Blot (WB)Detect HNRNPU in HEK-293, HeLa, Jurkat, K-562 cells, and mouse lung tissueShows a band at approximately 120 kDa
Immunoprecipitation (IP)Successfully precipitates HNRNPU from HeLa cellsUseful for protein-protein interaction studies
Immunofluorescence (IF)/ICCDetects HNRNPU in HeLa and HepG2 cellsPrimarily shows nuclear localization pattern
Immunohistochemistry (IHC)Detects HNRNPU in mouse kidney tissueRequires optimization of antigen retrieval methods
Flow Cytometry (FC)Detects HNRNPU in HepG2 cellsUseful for quantitative analysis at single-cell level

Each application requires specific antibody dilutions and sample preparation techniques to achieve optimal results .

What are the recommended dilution ratios for different experimental applications?

Proper antibody dilution is critical for experimental success, as it affects signal-to-noise ratio, specificity, and reproducibility. The recommended dilutions for HNRNPU antibodies vary by application:

ApplicationAntibody 14599-1-AP DilutionAntibody 16365-1-AP Dilution
Western Blot (WB)1:2000-1:100001:1000-1:4000
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg of total protein lysate0.5-4.0 μg for 1.0-3.0 mg of total protein lysate
Immunofluorescence (IF)/ICC1:50-1:5001:500-1:2000
Immunohistochemistry (IHC)Not specified1:500-1:2000
Flow Cytometry (FC)Not specified0.20 μg per 10^6 cells in 100 μl suspension

It is important to note that these dilutions serve as starting points, and researchers should optimize the dilution for their specific experimental system to obtain optimal results . Sample-dependent variations may require further titration.

How should HNRNPU antibodies be stored and handled for optimal performance?

Proper storage and handling of HNRNPU antibodies are essential for maintaining their reactivity and specificity. Based on manufacturer recommendations:

HNRNPU antibodies are typically supplied in liquid form containing PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 . For optimal preservation:

  • Store antibodies at -20°C where they remain stable for one year after shipment

  • Aliquoting is typically unnecessary for -20°C storage

  • Some antibody preparations (e.g., 20μl sizes) may contain 0.1% BSA as a stabilizer

  • Avoid repeated freeze-thaw cycles as they can denature antibodies and reduce activity

  • Allow antibodies to equilibrate to room temperature before opening the vial

  • For long-term storage beyond one year, consider making small aliquots to minimize freeze-thaw cycles

Proper handling during experiments is equally important:

  • Keep antibodies on ice when in use

  • Return to -20°C storage promptly after use

  • Avoid contamination by using clean pipette tips when handling antibody stocks

How can I optimize CLIP experiments to study HNRNPU RNA binding properties?

An optimized approach based on recent research includes:

  • Improved BrdU-CLIP method: This modified protocol produces approximately 10-fold greater yield of pre-amplified CLIP library, resulting in lower duplicate rate of CLIP-tag reads and reduced PCR cycle requirements .

  • Subcellular fractionation: Since HNRNPU shuttles between nucleus and cytoplasm, separate CLIP experiments on nuclear and cytoplasmic fractions can reveal compartment-specific binding patterns. Research has shown that cytoplasmic CLIP (cyto-CLIP) identifies more differentially expressed genes than whole-cell CLIP for HNRNPU .

  • Controls and validation:

    • Include universal control RNAs

    • Use a non-shuttling RBP (like HNRNPC) as a control

    • Validate binding sites with RNA immunoprecipitation (RIP) assays

    • Confirm subcellular localization by immunofluorescence and biochemical fractionation

  • Technical considerations:

    • While radioisotope labeling has been traditional in CLIP, newer methods like eCLIP and irCLIP may be performed without it

    • When first performing CLIP with HNRNPU, it's advisable to check RNA-RBP complex positions using radioisotope labeling before transitioning to non-radioisotope methods

    • Optimize RNase concentration, lysate volume, and antibody selection

This optimized approach has successfully identified HNRNPU binding to the 3'-UTR of IL-6 mRNA, providing new insights into its role in cytokine regulation .

What are the most effective strategies to study cytoplasmic versus nuclear functions of HNRNPU?

HNRNPU predominantly localizes to the nucleus but also has important cytoplasmic functions. Distinguishing between these compartment-specific roles requires specialized experimental approaches:

Recent research has demonstrated that cytoplasmic HNRNPU interacts with specific mRNAs like IL-6, suggesting an important role in controlling mRNA stability in the cytoplasm despite its predominant nuclear localization by immunofluorescence .

How can I validate HNRNPU's direct binding to specific target mRNAs?

Validating direct binding of HNRNPU to specific target mRNAs requires a multi-faceted approach that combines various RNA-protein interaction assays:

  • RNA Immunoprecipitation (RIP):

    • RIP assays can confirm that specific mRNAs (e.g., IL-6) are co-immunoprecipitated with HNRNPU but not with control IgG

    • Include housekeeping genes (e.g., RPLP0) as specificity controls

    • Quantitative PCR of immunoprecipitated RNA can demonstrate enrichment of target mRNAs

  • Optimized CLIP approaches:

    • Cytoplasmic CLIP (cyto-CLIP) has been shown to identify more HNRNPU direct targets than whole-cell CLIP

    • For example, analysis combining RNA sequencing data from HNRNPU-knockdown cells with cyto-CLIP identified 214 differentially expressed genes as direct targets, compared to only 44 with whole-cell CLIP

  • Functional validation:

    • siRNA-mediated knockdown of HNRNPU followed by measurement of target mRNA and protein levels

    • Experimental example: HNRNPU knockdown in HeLa cells significantly decreased IL-6 mRNA levels after PMA/ionophore stimulation, and reduced IL-6 protein secretion

  • Subcellular localization analysis:

    • For cytoplasmic functions like mRNA stability control, confirm binding occurs in the cytoplasmic fraction

    • Example: HNRNPU interaction with IL-6 mRNA was specifically observed in the cytoplasmic fraction, not in the nuclear fraction

  • Binding site identification:

    • Map the precise binding regions (e.g., 3'-UTR) through deletion constructs and reporter assays

    • Identify specific binding motifs through mutational analysis

This comprehensive approach provides strong evidence for direct, functionally relevant interactions between HNRNPU and target mRNAs in specific cellular compartments.

What controls should be included when studying HNRNPU's role in inflammatory responses?

  • Knockdown/expression controls:

    • Include non-targeting siRNA controls alongside HNRNPU siRNA

    • Validate knockdown efficiency by Western blot and RT-qPCR

    • In overexpression studies, use empty vector controls

  • Stimulation controls:

    • Include time-course analysis of stimulation (e.g., 2, 4, and 24 hours post-stimulation)

    • Appropriate unstimulated controls under identical conditions

    • Consider multiple stimulation methods (e.g., PMA/ionophore versus LPS) as HNRNPU's effects may be stimulus-specific

  • Target specificity controls:

    • Measure multiple cytokines beyond your target of interest

    • Include housekeeping genes (e.g., RPLP0) as negative controls

    • For RIP experiments, include immunoprecipitation with normal IgG as a specificity control

  • Subcellular localization controls:

    • Validate subcellular fractionation using established markers:

      • Cytoplasmic marker: GAPDH

      • Nuclear marker: Lamin B1

    • Perform immunocytochemistry to confirm HNRNPU localization before and after stimulation

  • Experimental validation controls:

    • Measure both mRNA levels (by RT-qPCR) and protein levels (by ELISA or Western blot)

    • Include positive controls known to be affected by HNRNPU (e.g., IL-6)

    • For cytoplasmic roles, confirm minimal nuclear contamination in cytoplasmic fractions

Research has demonstrated that HNRNPU regulates IL-6 expression in HeLa cells stimulated with PMA and calcium ionophore, with knockdown significantly decreasing both mRNA and secreted protein levels, highlighting its importance in inflammatory responses .

What are the advantages and limitations of different antibody types for HNRNPU detection?

Different types of antibodies have distinct characteristics that affect their performance in various applications. For HNRNPU detection, researchers should consider:

Antibody TypeAdvantagesLimitationsApplication Suitability
Polyclonal (e.g., 14599-1-AP, 16365-1-AP)- Recognize multiple epitopes
- Higher sensitivity
- Better for detecting denatured proteins
- More tolerant to minor antigen changes
- Batch-to-batch variation
- Higher background potential
- Less specificity than monoclonals
- Excellent for WB
- Good for IP and IF
- Useful for detecting low-abundance proteins
Monoclonal (e.g., sc-32315)- Consistent reproducibility
- High specificity for single epitope
- Lower background
- Batch consistency
- May miss proteins with modified epitopes
- Sometimes less sensitive
- May perform poorly if epitope is masked
- Superior for IP and IF
- Good for applications requiring high specificity
- Preferred for CLIP experiments

Experimental considerations when choosing antibodies:

  • Application-specific selection:

    • For immunoprecipitation in CLIP or RIP: Monoclonal antibodies like sc-32315 have been successfully used

    • For Western blot: Both types work well, with polyclonals offering potentially higher sensitivity

  • Verification approaches:

    • Knockdown/knockout validation to confirm specificity

    • Multiple antibodies targeting different epitopes for confirmation

    • Cross-validation with tagged proteins

  • Target-specific considerations:

    • HNRNPU's observed molecular weight (120 kDa) differs from calculated weight (91 kDa), requiring antibodies validated to detect the correct band

    • Consideration of potential post-translational modifications

    • Ability to detect both nuclear and cytoplasmic forms

The choice between polyclonal and monoclonal antibodies should be guided by the specific experimental requirements, with consideration of sensitivity, specificity, and application needs.

How should I interpret discrepancies between immunofluorescence and fractionation results for HNRNPU localization?

A common challenge in HNRNPU research is reconciling the predominantly nuclear localization observed by immunofluorescence with the detection of HNRNPU in cytoplasmic fractions. This apparent discrepancy requires careful interpretation:

This discrepancy highlights the importance of using complementary techniques when studying nucleocytoplasmic shuttling proteins like HNRNPU, as different methods may reveal distinct aspects of their biology.

What strategies can optimize HNRNPU antibody performance in challenging experimental contexts?

Optimizing HNRNPU antibody performance in challenging experimental settings requires careful consideration of multiple factors:

  • Western Blot optimization:

    • Sample preparation: Complete protein denaturation is crucial as HNRNPU is a large protein (120 kDa observed)

    • Transfer conditions: Extend transfer time or use specialized buffers for high molecular weight proteins

    • Blocking optimization: Test different blocking agents (BSA vs. milk) to reduce background

    • Primary antibody concentration: Titrate within recommended ranges (1:1000-1:10000) for optimal signal-to-noise ratio

    • Secondary antibody selection: Choose high-sensitivity detection systems for challenging samples

  • Immunoprecipitation enhancements:

    • Pre-clear lysates to reduce non-specific binding

    • Optimize antibody amount (0.5-4.0 μg per 1.0-3.0 mg protein lysate)

    • Consider protein A vs. protein G beads based on antibody isotype

    • Adjust wash stringency to balance between specificity and yield

    • For RNA-protein interactions, optimize crosslinking conditions

  • Immunofluorescence/Immunohistochemistry refinements:

    • Fixation method: Compare paraformaldehyde vs. methanol fixation

    • Antigen retrieval: For IHC, test TE buffer pH 9.0 vs. citrate buffer pH 6.0

    • Permeabilization optimization: Adjust detergent type and concentration

    • Signal amplification: Consider tyramide signal amplification for low-abundance detection

    • Mounting media selection: Use anti-fade reagents to preserve signal during imaging

  • Flow cytometry considerations:

    • Cell fixation and permeabilization protocol optimization

    • Titrate antibody concentration (starting with 0.20 μg per 10^6 cells)

    • Include appropriate isotype controls

    • Optimize compensation when using multiple fluorophores

    • Consider using cell cycle phase markers when analyzing nuclear proteins

These optimization strategies should be systematically tested and documented to establish reliable protocols for specific experimental contexts, ensuring reproducible results across different applications of HNRNPU antibodies.

How can HNRNPU antibodies be used to investigate its role in disease mechanisms?

HNRNPU has been implicated in various disease processes, and antibodies against this protein serve as valuable tools for mechanistic investigations:

  • Inflammatory disorders:

    • HNRNPU regulates expression of inflammatory cytokines including IL-6 and IL-1β

    • Antibody-based techniques can track HNRNPU's interactions with cytokine mRNAs during inflammation

    • CLIP and RIP assays using HNRNPU antibodies can identify direct binding to inflammatory mRNA targets

    • Changes in HNRNPU expression or localization can be monitored during inflammatory responses

  • Cancer research:

    • Recent research indicates that proteins encoded by circHNRNPU promote multiple myeloma progression

    • HNRNPU antibodies can be used to:

      • Assess expression levels across cancer types

      • Investigate alterations in subcellular localization

      • Study interaction with cancer-associated RNAs

      • Examine alternative splicing regulation in tumors

  • Neurological disorders:

    • RNA-binding proteins like HNRNPU are often associated with neurological diseases

    • Antibodies enable the study of:

      • Protein aggregation patterns

      • Altered nucleocytoplasmic shuttling

      • Dysregulated RNA processing

      • Post-translational modifications in disease states

  • Methodological approaches:

    • Immunohistochemistry of patient samples to assess expression patterns

    • Co-immunoprecipitation to identify altered protein-protein interactions

    • ChIP assays to study altered chromatin interactions

    • CLIP-seq to map disease-specific RNA binding profiles

    • Western blot analysis of patient-derived samples to detect altered expression or post-translational modifications

HNRNPU antibodies are thus essential tools for uncovering the diverse roles of this multifunctional protein in pathological processes, potentially leading to new therapeutic targets and biomarkers.

What emerging techniques might enhance our understanding of HNRNPU biology?

Recent technological advances offer new opportunities to study HNRNPU's complex biology:

  • Advanced RNA-protein interaction methodologies:

    • Optimized CLIP methods with greater sensitivity for nucleocytoplasmic shuttling proteins

    • Proximity-dependent RNA labeling techniques for spatial transcriptomics

    • Single-molecule imaging to visualize dynamic RNA-protein interactions

    • CRISPR-based RNA targeting to manipulate specific HNRNPU-RNA interactions

  • High-resolution localization techniques:

    • Super-resolution microscopy to better visualize nuclear vs. cytoplasmic distribution

    • Live-cell imaging with tagged HNRNPU to track shuttling dynamics

    • Correlative light and electron microscopy for ultrastructural context

    • Lattice light-sheet microscopy for 4D tracking of HNRNPU movement

  • Functional genomics approaches:

    • CRISPR screening to systematically identify HNRNPU functional partners

    • RNA-seq combined with HNRNPU perturbation to map global regulatory networks

    • Ribosome profiling to assess translational impacts of HNRNPU regulation

    • Single-cell multiomics to understand cell-type specific functions

  • Structural biology integration:

    • Cryo-EM structures of HNRNPU-RNA complexes

    • Hydrogen-deuterium exchange mass spectrometry for conformational dynamics

    • Integrative structural modeling combining multiple data types

    • AlphaFold-based prediction of protein-RNA interaction interfaces

These emerging approaches, when combined with traditional antibody-based methods, promise to provide unprecedented insights into HNRNPU's diverse cellular functions and disease associations. The integration of multiple techniques will be particularly powerful for understanding the compartment-specific roles of this nucleocytoplasmic shuttling protein.

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.