HNRNPD Antibody

Shipped with Ice Packs
In Stock

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 order within 1-3 business days of receiving it. Delivery times may vary depending on your location and preferred shipping method. Please consult your local distributor for specific delivery timelines.
Synonyms
ARE binding protein AUFI type A antibody; AU-rich element RNA-binding protein 1 antibody; AUF antibody; AUF1 antibody; AUF1A antibody; Heterogeneous nuclear ribonucleoprotein D0 antibody; hnRNP D antibody; hnRNP D0 antibody; Hnrnpd antibody; hnRNPD0 antibody; HNRPD antibody; HNRPD_HUMAN antibody; P37 antibody
Target Names
HNRNPD
Uniprot No.

Target Background

Function
HNRNPD (Heterogeneous nuclear ribonucleoprotein D) is a protein that binds with high affinity to RNA molecules containing AU-rich elements (AREs), commonly found within the 3'-UTR of many proto-oncogenes and cytokine mRNAs. It also exhibits specific binding to double- and single-stranded DNA sequences, functioning as a transcription factor. Its RNA-binding domains exhibit a preference for single-stranded non-monotonous 5'-UUAG-3' sequences, also showing weaker binding to the single-stranded 5'-TTAGGG-3' telomeric DNA repeat. HNRNPD binds RNA oligonucleotides with 5'-UUAGGG-3' repeats more tightly than the telomeric single-stranded DNA 5'-TTAGGG-3' repeats. Binding of HNRNPD to DNA inhibits the formation of DNA quadruplex structure, potentially playing a role in telomere elongation. It may be involved in translationally coupled mRNA turnover. Additionally, HNRNPD collaborates with other RNA-binding proteins in the cytoplasmic deadenylation/translational and decay interplay of the FOS mRNA mediated by the major coding-region determinant of instability (mCRD) domain. This protein may play a role in the regulation of the rhythmic expression of circadian clock core genes. It directly binds to the 3'UTR of CRY1 mRNA, inducing CRY1 rhythmic translation. HNRNPD might also be involved in the regulation of PER2 translation.
Gene References Into Functions
  1. Our research offers fresh insights into the regulation of APP pre-mRNA processing. It supports the role of nELAVLs as neuron-specific splicing regulators and unveils a novel function for HNRNPD in alternative splicing. PMID: 28291226
  2. Both hnRNP D and DL possess the capability to control their own expression through alternative splicing of cassette exons within their 3'UTRs. Exon inclusion leads to the production of mRNAs that are degraded via nonsense-mediated decay. Moreover, hnRNP D and DL exert control over each other's expression through the same mechanism. PMID: 29263134
  3. Lnc_ASNR interacts with the protein ARE/poly(U)-binding/degradation factor 1 (AUF1), known to promote rapid degradation of Bcl-2 mRNA, an apoptosis inhibitor. Lnc_ASNR binds to AUFI in the nucleus, reducing the cytoplasmic proportion of AUF1, which targets B-cell lymphoma-2 (Bcl-2) mRNA. PMID: 27578251
  4. In the present case, the identified mutations in HNRNPD and risk polymorphisms are plausible molecular players in the manifestation of CD. PMID: 28300425
  5. AUF1 p45 triggers the RNA switch in the flaviviral genome, which is crucial for viral replication. These findings provide a significant example of how cellular (host) factors facilitate the propagation of RNA viruses. PMID: 29263261
  6. HNRNPD might be a potential player in renal tubulointerstitial fibrosis through modulation of TGF-beta signal transduction via posttranscriptional regulation of Nedd4L. PMID: 28986222
  7. Results indicate that the IL-6/STAT3/NF-kappaB positive feedback loop includes AUF1 and is responsible for the sustained active status of cancer-associated fibroblasts. PMID: 27248826
  8. Depletion of HNRNPD eliminates the global interaction of miRNAs and AGO2. HNRNPD functions in promoting miRNA-mediated mRNA decay globally. PMID: 28334781
  9. High HNRNPD expression is associated with esophageal squamous cell carcinoma. PMID: 27826622
  10. Arginine methylation enhances the viral RNA chaperone activity of AUF1 p45. PMID: 27520967
  11. These results suggest that the post-transcriptional regulation of ATX expression by HuR and AUF1 modulates cancer cell migration. PMID: 27784781
  12. Analysis of the effect of the N-terminal RNA recognition motif on AUF1. PMID: 27437398
  13. We identified a C-rich element (CRE) in mu-opioid receptor (MOR) 3'-untranslated region (UTR) to which poly(rC) binding protein 1 (PCBP1) binds, resulting in MOR mRNA stabilization. AUF1 phosphorylation also led to an increased interaction with PCBP1. PMID: 27836661
  14. Findings indicate that hnRNP D and arginine methylation play important roles in the regulation of Flt-1 mRNA alternative polyadenylation. PMID: 26728997
  15. Down-regulation of hnRNPD inhibits the proliferation of esophageal squamous cell carcinoma cells by promoting cell apoptosis. PMID: 26648300
  16. In human ovarian, esophageal, and pancreatic cancer tissues, the expression of SOD1 was significantly correlated with that of AUF-1, further supporting the importance of AUF-1 in regulating SOD1 gene expression. PMID: 25908445
  17. hnRNPD plays roles in cellular proliferation and survival, in addition to RNA splicing and stability in oral cancer. PMID: 26318153
  18. These data specify a post-transcriptional mechanism through which AUF1 and YB1 contribute to the normal development of erythropoietic cells, and to non-hematopoietic tissues in which AUF1- and YB1-based regulatory mRNPs assemble on heterologous mRNAs. PMID: 25720531
  19. HNRNPD preserves genomic integrity through its actions on target RNAs. PMID: 25366541
  20. HNRNPD may play a role in the elimination of oxidized RNA, which is required for the maintenance of proper gene expression under conditions of oxidative stress. PMID: 25486179
  21. Taken together, these findings demonstrated the inhibitory effect of TP-1 on the growth and metastasis of SMMC-7721 cells, suggesting TP-1 as a potential chemopreventive agent against hepatocellular carcinoma (HCC) metastasis. PMID: 25787750
  22. HNRNPD interacts with the EV71 IRES to negatively regulate viral translation and replication. PMID: 25077793
  23. A novel mechanism by which HNRNPD binding and transfer of microRNA let-7 to AGO2 facilitates let-7-elicited gene silencing. PMID: 26253535
  24. Functional analysis of selected regulated proteins revealed that knockdown of HNRNPD, PHB2, and UB2V2 can increase HCMV replication, while knockdown of A4 and KSRP resulted in decreased HCMV replication. PMID: 25910425
  25. HNRNPD has been implicated in controlling a variety of physiological functions through its ability to regulate the expression of numerous mRNAs containing 3'-UTR AREs, thereby coordinating functionally related pathways. PMID: 24687816
  26. MicroRNA-141 and microRNA-146b-5p play a role in inhibiting the prometastatic mesenchymal characteristics through the RNA-binding protein HNRNPD targeting the transcription factor ZEB1 and the protein kinase AKT. PMID: 25261470
  27. IL-6 plays a major role in activating breast stromal fibroblasts through STAT3-dependent HNRNPD induction. PMID: 25231991
  28. AUF1 p45 promotes West Nile virus replication by an RNA chaperone activity that supports cyclization of the viral genome. PMID: 25078689
  29. The prolyl isomerase pin1 regulates mRNA levels of genes with short half-lives by targeting specific RNA binding proteins, such as HuR and AUF1. PMID: 24416409
  30. Our findings suggest that the HNRNPD gene may play an important role in hepatocellular carcinoma progression. PMID: 24213928
  31. hnRNP D is critically involved in LDLR mRNA degradation in liver tissue in vivo. PMID: 24158514
  32. Analysis of how HNRNPD targets mRNAs and how HNRNPD binding possibly regulates protein and/or microRNA binding events at adjacent sites. PMID: 23940053
  33. Results suggest that cleavage of HNRNPD may be a strategy employed by coxsackievirus B3 (CVB3) to enhance the stability of its viral genome. PMID: 23572232
  34. Hsp27 and F-box protein beta-TrCP promote degradation of mRNA decay factor HNRNPD. PMID: 23530064
  35. ING4 may regulate c-MYC translation by its association with HNRNPD. PMID: 23603392
  36. Here, authors describe experiments suggesting that HNRNPD, a host RNA binding protein involved in mRNA decay, plays a role in the infectious cycle of picornaviruses such as poliovirus and human rhinovirus. PMID: 23131833
  37. EBER1 may disturb the normal homeostasis between HNRNPD and ARE-containing mRNAs or compete with other HNRNPD-interacting targets in cells latently infected by Epstein-Barr virus. PMID: 23012480
  38. Findings point to a contribution of HNRNPD to the deleterious effects of cytokines on beta cell functions and suggest a role for this RNA-binding protein in the early phases of type 1 diabetes. PMID: 22159912
  39. The binding sites for HuR and HNRNPD present in androgen receptor mRNA, defined by their exact target sequences, are the same sequence for both proteins. PMID: 22368252
  40. HNRNPD and HuR bind to VEGFA ARE RNA under both normoxic and hypoxic conditions, and a pVHL-RNP complex determines VEGFA mRNA decay. PMID: 22086907
  41. The ability of HNRNPD isoforms to regulate the mRNA binding and decay-promoting activities of TTP. PMID: 22203679
  42. Data suggest that HNRNPD and YB-1 are required for normal accumulation of beta-globin mRNA in erythroid cells; YB-1 and HNRNPD exhibit sequence-specific binding to 3-prime-untranslated region of beta-globin mRNA. PMID: 22134169
  43. This review briefly describes the roles of mRNA decay in gene expression in general and ARE-mediated decay (AMD) in particular, with a focus on HNRNPD and the different modes of regulation that govern HNRNPD involvement in AMD. PMID: 21956942
  44. Knockdown of HNRNPD mRNA expression by HNRNPD siRNA in MKP-1 WT bone marrow macrophages significantly delayed degradation of IL-6, IL-10, and TNF-alpha mRNAs compared with controls. PMID: 21733716
  45. p16(INK4a) is also a modulator of transcription and apoptosis through controlling the expression of two major transcription regulators, HNRNPD and E2F1. PMID: 21799732
  46. These results suggest that the p38 MAP kinase (MAPK)-MK2-Hsp27 signaling axis may target HNRNPD destruction by proteasomes, thereby promoting AU-rich element mRNA stabilization. PMID: 21245386
  47. p40(AUF1) regulates a critical node within the NF-kappaB signaling pathway to permit IL10 induction for the anti-inflammatory arm of an innate immune response. PMID: 21135123
  48. Alternatively expressed domains of AU-rich element RNA-binding protein 1 (AUF1) regulate RNA-binding affinity, RNA-induced protein oligomerization, and the local conformation of bound RNA ligands. PMID: 20926381
  49. Leukotriene B(4) BLT receptor signaling regulates the level and stability of cyclooxygenase-2 (COX-2) mRNA through restricted activation of the Ras/Raf/ERK/p42 AUF1 pathway. PMID: 20489206
  50. The degradation of bcl-2 mRNA induced by AS1411 results from both interference with nucleolin protection of bcl-2 mRNA and recruitment of the exosome by HNRNPD. PMID: 20571027

Show More

Hide All

Database Links

HGNC: 5036

OMIM: 601324

KEGG: hsa:3184

STRING: 9606.ENSP00000313199

UniGene: Hs.480073

Subcellular Location
Nucleus. Cytoplasm. Note=Localized in cytoplasmic mRNP granules containing untranslated mRNAs. Component of ribonucleosomes. Cytoplasmic localization oscillates diurnally.

Q&A

What are the key functions of HNRNPD/AUF1 in cellular processes?

HNRNPD/AUF1 is a multifunctional RNA binding protein that plays critical roles in several cellular processes. It primarily binds to AU-rich elements (AREs) found within the 3'-UTR of many proto-oncogenes and cytokine mRNAs . HNRNPD functions include mRNA decay, telomere maintenance, translation initiation, and mCRD-mediated mRNA turnover . Recent research has expanded our understanding of HNRNPD's role beyond traditional RNA processing to include DNA damage repair pathways. Specifically, HNRNPD has been identified as a novel player in DNA double-strand break (DSB) repair through homologous recombination (HR) . Though HNRNPD binds chromatin independently of DNA damage, upon damage it relocates to γH2Ax foci and participates in the DNA end resection process that is crucial for HR .

To study these functions, researchers should consider experimental approaches that combine both RNA-protein and DNA-protein interaction analyses, including:

  • RNA immunoprecipitation to study native RNA targets

  • Chromatin immunoprecipitation to assess DNA binding

  • Immunofluorescence to track subcellular localization before and after DNA damage

  • Functional assays measuring mRNA stability and translation efficiency

What are the different isoforms of HNRNPD and how should antibody selection account for isoform specificity?

HNRNPD exists in four isoforms of different molecular weights (p37, p40, p42, and p45), all produced by alternate splicing of a single transcript . When selecting antibodies for HNRNPD research, researchers must consider which isoform(s) they wish to target, as the functions of these isoforms may differ in specific cellular contexts.

HNRNPD IsoformMolecular WeightExon CompositionReported Functions
p3737 kDaLacks exon 2, 7mRNA decay, base level function
p4040 kDaLacks exon 7mRNA turnover, enhanced binding to ARE
p4242 kDaLacks exon 2Translation enhancement
p4545 kDaContains all exonsStrongest ITAF activity for HCV IRES

Methodological approach for isoform-specific studies:

  • Examine the antibody epitope information to determine which isoforms will be recognized

  • For isoform-specific detection, choose antibodies raised against unique exon junctions

  • Validate antibody specificity using cell lines with known isoform expression patterns

  • Consider using knockout/knockdown systems followed by isoform-specific reconstitution, as demonstrated in research where HNRNPD knockout cells were generated via CRISPR-Cas9 targeting exon 2

What experimental techniques are most effective for studying HNRNPD's interactions with nucleic acids?

HNRNPD interacts with both RNA and DNA, requiring diverse experimental approaches to fully characterize these interactions. Based on the research literature, these techniques have proven effective:

For RNA interactions:

  • UV cross-linking and immunoprecipitation - This technique has successfully demonstrated direct HNRNPD binding to HCV IRES RNA

  • RNA pull-down assays using biotinylated RNA and purified HNRNPD protein - Effective for mapping binding sites, as shown in studies identifying stem-loop II of HCV 5' NTR as an interaction region

  • CLIP-seq (Cross-linking immunoprecipitation followed by sequencing) - For genome-wide identification of RNA binding sites

For DNA/chromatin interactions:

  • Chromatin immunoprecipitation - To study HNRNPD association with chromatin

  • DNA damage-induced foci formation assays - Using immunofluorescence to monitor HNRNPD relocalization to γH2Ax foci upon DNA damage

  • Synthetic DNA structure approaches - Using DNA structures mimicking resection intermediates as baits to identify protein interactions

How does HNRNPD contribute to DNA double-strand break repair, and what methodologies best measure this function?

HNRNPD has been identified as an important factor in DNA double-strand break (DSB) repair through homologous recombination. Research indicates that while HNRNPD binds to chromatin independently of DNA damage, it relocates to γH2Ax foci upon damage . Its silencing impairs critical processes in the DNA damage response cascade, including:

  • CHK1 S345 phosphorylation

  • The DNA end resection process

  • Single-strand DNA formation upon camptothecin treatment

  • AsiSI-induced DSB resection

  • RPA32 S4/8 phosphorylation

Methodological approaches to measure HNRNPD's role in DSB repair:

TechniqueApplicationReadoutReference
ImmunofluorescenceTrack HNRNPD relocalizationCo-localization with γH2Ax
Western blottingMonitor damage response markersPhosphorylation of CHK1 S345, RPA32 S4/8
CRISPR/Cas9 knockoutCreate HNRNPD-deficient cellsGenerated using guide RNAs targeting exon 2
In vitro DNA resection assayMeasure resection capacityQuantification of ssDNA generation
Drug sensitivity assaysAssess functional relevanceSensitivity to camptothecin and olaparib
DNA:RNA hybrid detectionAnalyze R-loop formationIncreased R-loops in HNRNPD-depleted cells

For researchers investigating this function, it's crucial to combine multiple methodologies to build a comprehensive understanding. When using HNRNPD antibodies for these studies, ensure they are validated for immunofluorescence applications with minimal background staining to accurately detect relocalization events.

What controls and validation steps are essential when using HNRNPD antibodies in RNA:DNA hybrid resolution studies?

HNRNPD has been implicated in RNA:DNA hybrid (R-loop) resolution during DNA damage repair. When using antibodies to study this process, proper controls and validation are essential:

Critical controls:

  • Positive control: Include known R-loop forming regions (e.g., highly transcribed genes)

  • Negative control: Use RNase H-treated samples to eliminate RNA:DNA hybrids

  • HNRNPD knockout/knockdown verification: Confirm antibody specificity by showing loss of signal

  • Reconstitution experiments: Test function restoration using HNRNPD-expressing constructs with PAM-resistant mutations for CRISPR knockout models

Validation methodology:

  • DNA:RNA hybrid immunoprecipitation (DRIP) using antibodies specific to RNA:DNA hybrids (like S9.6)

  • Complementary technique verification: Combine immunofluorescence with in situ proximity ligation assays

  • Functional validation: RNase H1 expression and RNA polymerase II inhibition should rescue the ability to phosphorylate RPA32 S4/8 in HNRNPD knockout cells upon DNA damage

  • Co-immunoprecipitation controls: Verify interaction with known partners like heterogeneous nuclear ribonucleoprotein SAF-A

When publishing results, researchers should clearly document these validation steps to ensure reproducibility and establish confidence in the antibody's performance for this specific application.

How can researchers effectively use HNRNPD antibodies to study its role in translation regulation of viral and cellular mRNAs?

HNRNPD has been identified as an important internal ribosome entry site (IRES) trans-acting factor (ITAF) that modulates translation of viral RNAs like HCV and potentially cellular mRNAs. When using HNRNPD antibodies to study these functions, researchers should consider:

Experimental design approaches:

  • Reporter systems: Use dicistronic or monocistronic reporter constructs containing the IRES of interest (e.g., HCV IRES)

  • HNRNPD manipulation: Apply overexpression, knockdown (siRNA/shRNA), or knockout (CRISPR-Cas9) strategies

  • Ribosome profiling: Monitor shifts in HCV mRNA distribution across polysome fractions upon HNRNPD depletion

  • Direct interaction studies: Perform UV cross-linking and immunoprecipitation to confirm HNRNPD binding to the RNA of interest

Translational effect quantification:

  • In reporter systems, normalize luciferase activities to RNA levels to accurately measure translation efficiency

  • Use replicon systems containing viral proteins to study effects in more physiological contexts

  • For infection studies, employ infectious clones containing reporter genes fused to viral proteins

For accurate results when studying HNRNPD's effect on translation, researchers must:

  • Include controls for RNA levels, as HNRNPD can affect both translation and RNA stability

  • Account for potential isoform-specific effects (the p45 isoform shows strongest ITAF activity for HCV IRES)

  • Verify antibody specificity for detecting the relevant HNRNPD isoforms

  • Consider cell-type specific effects, as HNRNPD's function may vary between cell lines

What are the optimal conditions for using HNRNPD antibodies in different experimental applications?

Based on the available research data, HNRNPD antibodies have been successfully employed in multiple experimental techniques. Here are the optimal conditions for various applications:

ApplicationRecommended DilutionIncubation ConditionsSpecial Considerations
Western Blotting1:1000-1:5000Overnight at 4°CMultiple antibodies used in research: 07-260 (Millipore), D6O4F (Cell Signaling)
Immunofluorescence1:200-1:6001-2 hours at room temperatureCo-staining with γH2Ax (1:600, 05-636, Millipore) to detect damage foci
ImmunoprecipitationPer manufacturer protocolOvernight at 4°C with rotationFlag-tagged constructs and anti-Flag antibodies have been used successfully
ChIP (Chromatin IP)1:200Overnight at 4°CPre-clearing with protein A/G beads recommended
RNA-IP1:2004 hours at 4°CRNase inhibitors must be included

Methodological notes:

  • For chromatin-associated studies, optimize fixation conditions (typically 1% formaldehyde for 10 minutes)

  • When detecting multiple isoforms, use gradient gels (10-15%) for optimal separation

  • For co-immunoprecipitation of interacting partners, gentler lysis conditions are preferable

  • Blocking with 5% BSA instead of milk may improve results for phospho-specific antibody detection following HNRNPD studies

  • For immunofluorescence, permeabilization conditions affect nuclear signal intensity

What troubleshooting approaches are recommended when working with HNRNPD antibodies in chromatin studies?

When studying HNRNPD's association with chromatin and its role in DNA damage response, researchers may encounter several technical challenges. Here are recommended troubleshooting approaches:

Challenge: Poor signal in chromatin fractions
Solutions:

  • Optimize extraction buffers - Use specialized chromatin extraction protocols with increasing salt concentrations

  • Adjust fixation conditions - Over-fixation can mask epitopes; try reducing formaldehyde concentration or fixation time

  • Include phosphatase inhibitors - Critical for preserving damage-induced phosphorylation events

  • Test multiple HNRNPD antibodies - Different epitopes may be differentially accessible in chromatin contexts

Challenge: Non-specific bands in Western blots
Solutions:

  • Increase blocking stringency - Use 5% BSA with 0.1% Tween-20

  • Optimize antibody concentration - Titrate to find minimal concentration giving specific signal

  • Include knockout/knockdown controls - CRISPR-Cas9 knockout cells as negative controls

  • Use isoform-specific detection strategies - Target unique exon junctions

Challenge: Inconsistent co-localization with γH2Ax foci
Solutions:

  • Standardize damage induction - Control dosage and timing of DNA damaging agents

  • Sequential immunostaining - Apply primary antibodies sequentially rather than simultaneously

  • Optimize image acquisition settings - Use identical exposure settings for comparative analyses

  • Quantify co-localization using software - Apply algorithms to eliminate confirmation bias

How should researchers adapt protocols when using HNRNPD antibodies to study different isoforms in various experimental systems?

HNRNPD exists as four isoforms (p37, p40, p42, and p45), requiring careful experimental design when studying isoform-specific functions. Researchers should modify their protocols as follows:

For Western blot analysis:

  • Use longer SDS-PAGE gels (preferably gradient gels) to achieve better separation of closely migrating isoforms

  • Employ longer run times at lower voltage to enhance resolution

  • Consider using isoform-specific antibodies when available, or antibodies targeting common regions to detect all isoforms

  • Include positive controls with known isoform expression patterns

For knockdown/knockout studies:

  • Design siRNAs targeting specific exons to selectively deplete certain isoforms:

    • Target exon 7 to deplete p42/p45 but retain p37/p40

    • Use siRNAs targeting conserved regions to deplete all isoforms simultaneously

  • For CRISPR-Cas9 knockout, target early exons (e.g., exon 2) for complete knockout

  • Design reconstitution experiments with PAM-resistant mutants to verify specificity

For functional studies:

  • Express individual isoforms in knockout backgrounds to assess isoform-specific functions

  • Use tagged constructs (e.g., Flag-tagged p45) for clean immunoprecipitation

  • Consider tissue-specific expression patterns when selecting appropriate experimental systems

  • For viral translation studies, note that p45 shows the strongest ITAF activity for HCV IRES

How can HNRNPD antibodies be utilized to study the protein's role in R-loop resolution during DNA repair?

HNRNPD has been implicated in resolving RNA:DNA hybrids (R-loops) during DNA repair, a critical process for maintaining genomic stability. Researchers can utilize HNRNPD antibodies to investigate this role through the following approaches:

Experimental strategy:

  • DNA damage induction: Treat cells with agents like camptothecin that are known to generate R-loops

  • HNRNPD visualization: Use validated antibodies for immunofluorescence to track HNRNPD localization to damage sites

  • R-loop detection: Apply S9.6 antibody (specific for RNA:DNA hybrids) in parallel studies

  • Co-localization analysis: Assess overlap between HNRNPD, R-loops, and γH2Ax foci

Functional validation experiments:

  • HNRNPD depletion: Generate knockout cells using CRISPR-Cas9 or knockdown using siRNA

  • R-loop quantification: Measure R-loop accumulation via DRIP-qPCR or immunofluorescence

  • Resolution pathway manipulation: Express RNase H1 or inhibit RNA polymerase II to rescue defects

  • Interaction studies: Assess HNRNPD association with known R-loop processing factors like heterogeneous nuclear ribonucleoprotein SAF-A

Research has demonstrated that HNRNPD depletion results in increased RNA:DNA hybrids upon DNA damage, and that expression of RNase H1 or RNA polymerase II inhibition rescues the ability to phosphorylate RPA32 S4/8 in HNRNPD knockout cells . These findings suggest that R-loop resolution is one mechanism by which HNRNPD facilitates the DNA damage response.

What are the critical considerations when designing experiments to investigate HNRNPD's role in viral translation and replication?

HNRNPD has been identified as a modulator of viral translation, particularly for Hepatitis C virus (HCV), making it an important research target for understanding viral pathogenesis. When designing experiments to investigate this role, researchers should consider:

Experimental system selection:

  • Reporter constructs: Use dicistronic reporters containing viral IRES elements driving reporter genes

  • Replicon systems: Employ subgenomic or full-length viral replicons for more physiological context

  • Infectious virus systems: When available, use reporter-tagged infectious clones

HNRNPD manipulation strategies:

  • Overexpression: Express individual isoforms (p37, p40, p42, p45) to identify those with strongest effect

  • Knockdown: Use siRNA targeting all isoforms or isoform-specific regions

  • Knockout: Generate stable CRISPR-Cas9 knockout cell lines for complete depletion

Critical controls and measurements:

  • RNA level normalization: Measure viral RNA levels alongside protein expression, as HNRNPD affects both translation and RNA stability

  • Polysome profiling: Analyze redistribution of viral RNA across polysome fractions to confirm translation effects

  • Direct binding verification: Use UV cross-linking and immunoprecipitation to confirm protein-RNA interactions

  • Viral protein quantification: Western blotting for viral proteins and reporter assays should be performed in parallel

Key insights from published research:

  • HNRNPD (particularly p45) enhances HCV IRES-dependent translation

  • The protein interacts directly with stem-loop II of HCV 5' NTR

  • Knockdown of HNRNPD results in increased HCV RNA replication but decreased translation

  • In infection models, HNRNPD depletion significantly hampers HCV infection

These findings suggest HNRNPD may function as a switch between viral translation and replication phases, similar to mechanisms described for other viruses.

How should researchers integrate multi-omics approaches with HNRNPD antibody-based studies to comprehensively understand its cellular functions?

As research on HNRNPD expands beyond individual pathways to systems-level understanding, integration of antibody-based studies with multi-omics approaches becomes increasingly valuable. Here's a methodological framework for such integration:

Transcriptomics integration:

  • RNA-seq after HNRNPD manipulation: Identify differentially expressed genes following knockout/knockdown

  • RIP-seq (RNA immunoprecipitation sequencing): Use validated HNRNPD antibodies to identify direct RNA targets

  • CLIP-seq: Map precise HNRNPD binding sites on RNAs with single-nucleotide resolution

  • Correlation analysis: Compare HNRNPD binding patterns with changes in mRNA stability and translation efficiency

Proteomics integration:

  • IP-MS: Use HNRNPD antibodies for immunoprecipitation followed by mass spectrometry to identify protein interactors

  • BioID or proximity labeling: Identify proteins in close proximity to HNRNPD in different cellular compartments

  • Comparative proteomics: Analyze protein expression changes in HNRNPD-depleted vs. control cells

  • PTM analysis: Examine how DNA damage affects HNRNPD post-translational modifications

Genomics integration:

  • ChIP-seq: Map HNRNPD chromatin binding sites genome-wide

  • DRIP-seq: Correlate R-loop formation with HNRNPD binding

  • DNA damage mapping: Integrate γH2AX ChIP-seq with HNRNPD binding data

  • Chromatin accessibility: Compare ATAC-seq profiles before and after HNRNPD depletion

Data integration strategies:

  • Multi-factor analysis: Correlate HNRNPD binding with RNA stability, translation efficiency, and R-loop formation

  • Network modeling: Build protein-protein and protein-RNA interaction networks centered on HNRNPD

  • Pathway enrichment: Identify biological processes overrepresented in HNRNPD-regulated genes

  • Temporal analyses: Examine how HNRNPD functions change during cellular stress response timecourse

This integrated approach can help resolve seemingly contradictory functions of HNRNPD, such as its roles in both promoting HCV translation while inhibiting viral replication , or its context-dependent effects on mRNA stability versus translation.

What quality control measures should be implemented when validating new lots of HNRNPD antibodies?

Antibody lot-to-lot variation can significantly impact experimental reproducibility. When validating new lots of HNRNPD antibodies, researchers should implement these quality control measures:

Basic validation tests:

  • Western blot comparison: Run side-by-side blots with old and new antibody lots on:

    • Positive control lysates (cells known to express HNRNPD)

    • Negative control lysates (HNRNPD knockout cells generated via CRISPR-Cas9)

    • Multiple cell lines to assess cross-reactivity patterns

  • Dilution series: Test multiple antibody concentrations to determine optimal working dilution

  • Blocking peptide competition: Confirm specificity by pre-incubating antibody with immunizing peptide

Advanced validation for specific applications:

  • Immunoprecipitation efficiency: Compare protein recovery between antibody lots

  • Immunofluorescence pattern consistency: Assess nuclear localization and damage-induced foci formation

  • ChIP-qPCR reproducibility: Test enrichment at known binding sites

  • Cross-reactivity assessment: Test antibody against recombinant HNRNPD isoforms and related hnRNP family members

Documentation requirements:

  • Generate validation reports including images of control experiments

  • Record lot number, dilution, incubation conditions, and buffer compositions

  • Document detection of all four HNRNPD isoforms (p37, p40, p42, p45)

  • Maintain positive control lysates as reference standards

For researchers studying HNRNPD's role in DNA repair, additional validation steps should include testing the antibody's ability to detect HNRNPD relocalization to γH2Ax foci after DNA damage , as this is a critical functional readout.

How can researchers distinguish between direct and indirect effects when studying HNRNPD in complex cellular processes?

HNRNPD participates in multiple cellular processes, making it challenging to distinguish direct from indirect effects when using antibody-based detection methods. Researchers should apply these methodological approaches:

Causality establishment strategies:

  • Temporal resolution: Perform time-course experiments to establish order of events

    • Example: Track HNRNPD relocalization to damage sites relative to RPA32 phosphorylation

  • Rescue experiments: Reconstitute HNRNPD-depleted cells with:

    • Wild-type HNRNPD

    • RNA-binding deficient mutants

    • DNA-binding deficient mutants

    • Isoform-specific constructs

  • Domain mapping: Express individual HNRNPD domains to identify minimal functional units

  • Direct interaction verification: Use purified components in vitro to confirm direct biochemical activities

Controls for off-target effects:

  • Use multiple knockdown/knockout strategies targeting different regions of HNRNPD

  • Include non-targeting controls in all experiments

  • Validate phenotypes with multiple HNRNPD antibodies targeting different epitopes

  • Consider compensatory effects from other hnRNP family members

For HCV translation studies, researchers demonstrated direct effects by:

  • Showing direct binding of purified HNRNPD to HCV IRES RNA

  • Demonstrating that ribosomal profiles shift when HNRNPD is depleted

  • Confirming that effects on viral translation occur independently of effects on viral replication

For DNA repair studies, direct effects were established by:

  • Demonstrating HNRNPD relocalization to damage foci

  • Showing that HNRNPD knockout directly impairs in vitro DNA resection

  • Establishing that RNA:DNA hybrid accumulation in HNRNPD-depleted cells can be rescued by RNase H1 expression

What are the most effective experimental designs for resolving contradictory findings about HNRNPD functions in different cellular contexts?

Research on HNRNPD has revealed seemingly contradictory functions, such as promoting mRNA decay in some contexts while enhancing translation in others. To resolve such contradictions, consider these experimental design strategies:

Contextual variation analysis:

  • Cell type comparative studies: Test HNRNPD function across multiple cell types using identical assays

  • Stress-dependent analysis: Examine HNRNPD activity under normal conditions versus various stress stimuli (DNA damage, viral infection, etc.)

  • Isoform-specific investigation: Systematically test each HNRNPD isoform (p37, p40, p42, p45) in identical experimental settings

  • Substrate-specific effects: Compare HNRNPD's activity on different target RNAs or DNA structures

Mechanistic resolution approaches:

  • Competitive binding studies: Assess whether DNA damage affects HNRNPD's RNA binding capacity

  • Interactome analysis under different conditions: Compare HNRNPD binding partners before and after stress

  • Post-translational modification mapping: Identify condition-specific modifications that might alter HNRNPD function

  • Subcellular fractionation: Track HNRNPD distribution between nucleus, cytoplasm, and chromatin across conditions

Example resolution framework for HCV translation versus replication contradiction:
The research indicates HNRNPD enhances HCV translation but suppresses viral RNA replication . This apparent contradiction was addressed by:

  • Separating translation from replication measurements

  • Using both dicistronic and monocistronic reporter systems

  • Normalizing protein expression to RNA levels

  • Analyzing polysome profiles to directly assess translation efficiency

For DNA repair function studies, researchers might resolve contradictions by:

  • Distinguishing between HNRNPD's roles in different repair pathways (HR vs. NHEJ)

  • Examining temporal dynamics of HNRNPD recruitment to damage sites

  • Investigating the interplay between HNRNPD's RNA binding and DNA repair functions

  • Testing whether R-loop resolution is a primary or secondary function in the repair process

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.