RPRD1A Human

Regulation Of Nuclear Pre-MRNA Domain Containing 1A Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of RPRD1A Human

RPRD1A (Regulation of Nuclear Pre-mRNA Domain-Containing Protein 1A) is a 312-amino acid nuclear protein critical for transcriptional regulation and cell cycle control. It interacts with the phosphorylated C-terminal domain (CTD) of RNA polymerase II (RNAPII) subunit POLR2A, serving as a scaffold for phosphatase recruitment and CTD dephosphorylation . RPRD1A also regulates cyclin-dependent kinase inhibitors and antioxidant pathways, with implications in cancer progression .

Protein Interactions

RPRD1A forms homodimers and heterodimers with RPRD1B via coiled-coil domains and binds phosphorylated CTD repeats (e.g., phospho-S2 and phospho-S7) through its CTD-interaction domain (CID) . This interaction facilitates recruitment of RPAP2 phosphatase, enabling dephosphorylation of CTD phospho-S5 residues .

Table 1: Functional Partners of RPRD1A

PartnerRole in RPRD1A FunctionalityInteraction Score
RPRD1BForms heterodimers; enhances CTD binding0.982
RPAP2Phosphatase for CTD dephosphorylation0.980
POLR2ARNAPII subunit; CTD phosphorylation site0.975
POLR2JRNAPII subunit; core transcription machinery0.796

Transcriptional Regulation

RPRD1A modulates transcription elongation and termination by:

  • Scaffolding RPAP2 to dephosphorylate CTD phospho-S5, critical for snRNA gene transcription .

  • Regulating cyclin-D1 (CCND1) and cyclin-E (CCNE1), acting as a potential negative regulator of G1/S phase progression .

Clinical Correlations

RPRD1A overexpression is linked to aggressive HCC features:

Mechanisms in HCC

  • NRF2 Stabilization: RPRD1A inhibits Keap1-mediated ubiquitination of NRF2, enhancing antioxidant gene expression (e.g., NQO1, HO-1) and reducing reactive oxygen species (ROS) .

  • Oxidative Stress Resistance: RPRD1A knockdown increases ROS accumulation and sensitizes HCC cells to platinum-based therapies .

Table 2: Experimental Models and Outcomes

ModelInterventionOutcomeSource
MHCCLM3/Huh7 (in vitro)RPRD1A knockdown↓ Proliferation; ↓ Colony formation
Nude mice (in vivo)RPRD1A depletion↓ Tumor growth; ↓ Pulmonary metastases
Transwell assaysRPRD1A overexpression↑ Cell invasion

Transcriptional Modulation in Human Cells

RPRD1A inversely correlates with nascent RNA synthesis, acting as a transcriptional repressor:

  • RPRD1A Overexpression: ↓ RNA levels of snRNA genes (e.g., U1, U2) and stress-response genes .

  • RPRD1A Knockdown: ↑ Transcription of inflammation-related genes (e.g., IL6, CXCL8) .

Table 3: Transcriptional Effects of RPRD1A Modulation

ConditionAffected GenesBiological ImpactSource
RPRD1A overexpression↓ snRNA genesImpaired RNA processing
RPRD1A knockdown↑ Inflammatory genesAltered stress response

Product Specs

Introduction
RPRD1A (Regulation of Nuclear Pre-MRNA Domain Containing 1A) shows increased expression in cells with elevated levels of cyclin-dependent kinase inhibitor p15(INK4b), suggesting a potential role in cell cycle control. This protein interacts with the phosphorylated C-terminal heptapeptide repeat domain (CTD) of RNA polymerase II subunit POLR2A, the largest subunit, and participates in the dephosphorylation of the CTD. RPRD1A may act as a negative regulator of cyclin-D1 (CCND1) and cyclin-E (CCNE1) within the cell cycle.
Description
Recombinant human RPRD1A, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 337 amino acids (residues 1-312) with a molecular weight of 38.4 kDa. The protein includes a 25 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The RPRD1A solution is provided at a concentration of 0.5 mg/ml in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, 1 mM DTT, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 95.0% by SDS-PAGE analysis.
Synonyms
Regulation of Nuclear Pre-MRNA Domain Containing 1A, RPRD1A, P15RS, Cyclin-Dependent Kinase 2B-Inhibitor-Related Protein, Cyclin-Dependent Kinase Inhibitor 2B-Related Protein (P15INK4B-Related Protein), P15INK4B-Related Protein, HsT3101, Regulation of Nuclear Pre-MRNA Domain-Containing Protein 1A, Cyclin-Dependent Kinase Inhibitor 2B-Related Protein, FLJ10656.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSEFMSAFS EAALEKKLSE LSNSQQSVQT LSLWLIHHRK HSRPIVTVWE RELRKAKPNR KLTFLYLAND VIQNSKRKGP EFTKDFAPVI VEAFKHVSSE TDESCKKHLG RVLSIWEERS VYENDVLEQL KQALYGDKKP RKRTYEQIKV DENENCSSLG SPSEPPQTLD LVRALQDLEN AASGDAAVHQ RIASLPVEVQ EVSLLDKITD KESGERLSKM VEDACMLLAD YNGRLAAEID DRKQLTRMLA DFLRCQKEAL AEKEHKLEEY KRKLARVSLV RKELRSRIQS LPDLSRLPNV TGSHMHLPFA GDIYSED.

Q&A

What is RPRD1A and what domains characterize its structure?

RPRD1A is a CTD-interacting protein that binds to the C-terminal domain (CTD) of RNA Polymerase II's largest subunit, RPB1. Structurally, RPRD1A contains a CTD-interacting domain (CID) that mediates its binding to the phosphorylated serine-5 residue of RNA Pol II CTD. Additionally, RPRD1A features coiled-coil (CC) domains that facilitate its interactions with other proteins, including its paralog RPRD1B, with which it shares approximately 80% similarity . When designing experiments to study RPRD1A, researchers should consider these structural features for protein-protein interaction studies, as the CID domain is critical for its functional interactions with transcriptional machinery.

How does RPRD1A participate in transcriptional regulation?

RPRD1A exhibits complex effects on transcriptional regulation with potentially context-dependent functions. Research indicates that RPRD1A can form a heterodimer with RPRD1B to recruit RPAP2 (CTD S5 phosphatase) and HDAC1 (CTD deacetylase) during early transcription stages . While some studies suggest this heterodimer promotes transcription, other findings indicate RPRD1A can act as a negative regulator of transcription. When investigating RPRD1A's transcriptional effects, researchers should employ RNA metabolic labeling techniques such as 4sU-seq (thiouridine labeling followed by sequencing) to measure nascent RNA production, as demonstrated in studies showing that RPRD proteins' depletion increased RNA synthesis while their overexpression downregulated transcription . Additionally, researchers should note that RPRD1A appears to have less clear effects on transcription compared to its family members RPRD1B and RPRD2.

What experimental approaches are recommended for modulating RPRD1A expression in cellular models?

To effectively study RPRD1A function, researchers can employ both knockdown and overexpression approaches. For knockdown experiments, siRNA treatment has proven effective in HEK293T cells . For stable knockdown, shRNA expressing lentiviral vectors have been successfully used in HCC cell lines like MHCCLM3 and Huh7 . For overexpression studies, the Flp-In T-REx 293 system provides an inducible expression platform that has been successfully employed . When assessing knockdown or overexpression efficiency, both RT-qPCR and western blotting should be performed to confirm altered expression at both RNA and protein levels. Researchers should be aware that RPRD1A knockdown may initially cause increased cell death during the first week post-transduction, potentially due to disrupted redox homeostasis .

How can researchers effectively measure RPRD1A's impact on transcription?

To comprehensively assess RPRD1A's effects on transcription, researchers should employ nascent RNA labeling techniques like 4sU-seq, which captures newly synthesized RNA. This approach allows for the detection of immediate transcriptional changes rather than steady-state RNA levels that may be affected by processing and degradation. Previous studies have shown that metabolic labeling followed by sequencing can effectively reveal that RPRD1A depletion results in altered accumulation of nascent RNAs across different gene classes . When analyzing results, researchers should segregate findings by RNA class (mRNAs, lncRNAs, snRNAs) as RPRD1A may differentially affect various RNA species. Additionally, visualization tools like Integrated Genome Viewer (IGV) are useful for examining specific gene examples to confirm global trends observed in statistical analyses.

What protein complexes does RPRD1A form, and how do they influence its function?

RPRD1A forms mutually exclusive complexes with RNA Polymerase II to coordinate transcriptional control. Most notably, RPRD1A forms heterodimers with its paralog RPRD1B through their coiled-coil domains . This heterodimer recruits RPAP2 and HDAC1, which function as CTD S5 phosphatase and CTD deacetylase, respectively. To investigate these complexes, researchers should employ co-immunoprecipitation followed by mass spectrometry or western blotting to identify interacting partners. Additionally, proximity ligation assays can visualize these interactions in situ. Researchers should note that despite biochemical evidence suggesting the RPRD1A-RPRD1B heterodimer promotes transcription, functional studies show discrepancies in phenotypes associated with altering levels of either protein , indicating complex regulatory mechanisms that warrant careful experimental design when studying these interactions.

How does RPRD1A's role in transcription differ from other RPRD family proteins?

Among the RPRD protein family (RPRD1A, RPRD1B, and RPRD2), RPRD1A exhibits distinct effects on transcriptional regulation. While all three proteins can negatively regulate transcription, research indicates that RPRD2 exerts the most substantial impact, followed by RPRD1B, while RPRD1A shows more variable effects . To properly differentiate the functions of these family members, researchers should conduct comparative studies using selective knockdown or overexpression of each protein individually. When analyzing RPRD1A's specific functions, calculated ratios of upregulated to downregulated transcriptional units provide valuable metrics - previous research demonstrated that RPRD1A knockdown resulted in a ratio of 1.7 (compared to 5.8 for RPRD2 and 4.1 for RPRD1B) . These differences highlight the importance of studying each family member separately while also considering their potential compensatory mechanisms.

What methodologies should be used to investigate RPRD1A's role in tumor progression?

To comprehensively examine RPRD1A's role in tumor progression, researchers should employ both in vitro and in vivo approaches. In vitro, cell proliferation assays (CCK-8 or MTT), colony formation assays, and transwell invasion assays have effectively demonstrated that RPRD1A knockdown inhibits HCC cell proliferation, colony formation, and invasion, while its overexpression promotes these processes . For in vivo studies, subcutaneous xenograft models using stable RPRD1A-depleted cells have shown significant inhibition of tumor growth . To assess metastatic potential, tail vein injection models followed by quantification of pulmonary metastatic lesions can demonstrate RPRD1A's influence on metastasis, with H&E and IHC staining confirming HCC cell characteristics in metastatic sites . Researchers should also consider orthotopic liver implantation models for more physiologically relevant assessments of HCC progression and metastasis in the context of RPRD1A modulation.

How does RPRD1A affect cellular redox homeostasis?

RPRD1A plays a crucial role in maintaining cellular redox homeostasis, with its depletion significantly increasing reactive oxygen species (ROS) production . Research has demonstrated that RPRD1A knockdown decreases the GSH/GSSG ratio (an indicator of cellular antioxidant capacity), while its overexpression increases this ratio . To investigate RPRD1A's effects on redox balance, researchers should employ fluorescent ROS indicators (e.g., DCFDA) to measure cellular ROS levels under basal conditions and after oxidative stress induction (e.g., H₂O₂ treatment). Glutathione assays measuring GSH/GSSG ratios provide quantitative assessment of antioxidant capacity. Cell viability assays following oxidative stress challenge can further demonstrate RPRD1A's protective role, as RPRD1A knockdown causes increased cell death upon H₂O₂ treatment that can be rescued by antioxidants like N-acetyl-L-cysteine (NAC) . These methodological approaches provide complementary evidence of RPRD1A's function in oxidative stress response.

What is the relationship between RPRD1A and NRF2 signaling?

RPRD1A enhances the nuclear translocation of NRF2 (Nuclear Factor Erythroid 2-Related Factor 2), a master regulator of antioxidative responses . This interaction appears critical for RPRD1A's role in oxidative stress defense. To investigate this relationship, researchers should examine NRF2 protein accumulation and subcellular localization following RPRD1A modulation using western blotting and immunofluorescence microscopy. Functional analysis through ARE (Antioxidant Response Element)-luciferase reporter assays can assess NRF2 transcriptional activity, which is enhanced by RPRD1A . Researchers should also examine phosphorylation of p38 MAPK (a biosensor of oxidative stress) that increases with RPRD1A knockdown under H₂O₂ treatment . For mechanistic studies, co-immunoprecipitation experiments can determine whether RPRD1A directly interacts with NRF2 or affects its stability through other mechanisms, providing insight into how RPRD1A regulates this critical antioxidant pathway.

How does RPRD1A regulate antioxidant enzyme expression?

RPRD1A positively regulates the expression of multiple antioxidant enzymes, including SOD1, SOD2, GCLC, GCLM, ANT, NQO1, HO1, PRDX4, and PRDX6 . This regulation appears to be mediated through RPRD1A's effect on NRF2 activity. To investigate this regulatory network, researchers should employ RT-qPCR to measure mRNA levels of these antioxidant genes following RPRD1A knockdown or overexpression. Western blotting should confirm changes at the protein level for key enzymes like GCLC, GCLM, and NQO1 . ChIP-seq analysis of NRF2 binding sites can determine whether RPRD1A affects NRF2 occupancy at the promoters of these antioxidant genes. For a comprehensive assessment, researchers should measure enzymatic activities of these antioxidant enzymes in addition to their expression levels, as post-translational modifications might affect their function independently of expression changes. This multi-level analysis would provide a complete picture of how RPRD1A influences the antioxidant defense system.

What cutting-edge techniques can be applied to study RPRD1A-mediated transcriptional regulation?

For advanced investigation of RPRD1A's role in transcriptional regulation, researchers should consider employing PRO-seq (Precision Nuclear Run-On sequencing) or NET-seq (Native Elongating Transcript sequencing) to capture RNA polymerase II positions genome-wide with single-nucleotide resolution. These techniques provide more precise information about transcriptional dynamics than conventional RNA-seq. Additionally, ChIP-seq for RNA Polymerase II and its phosphorylated forms (Ser5P, Ser2P) can reveal how RPRD1A affects polymerase recruitment and processivity across the genome. CUT&RUN or CUT&Tag methods offer higher signal-to-noise ratios than traditional ChIP-seq for studying RPRD1A chromatin associations. For protein-protein interactions, BioID or APEX2 proximity labeling can identify the RPRD1A interactome in living cells, while cryo-EM studies might elucidate the structural basis of RPRD1A-RNA Pol II interactions. These advanced approaches would provide mechanistic insights beyond what has been revealed by conventional transcriptomic analyses .

How can researchers investigate the potential therapeutic targeting of RPRD1A in cancer?

To explore RPRD1A as a therapeutic target in cancer, researchers should employ multiple complementary approaches. First, CRISPR-Cas9 knockout of RPRD1A in patient-derived xenograft (PDX) models would provide physiologically relevant insights into its requirement for tumor maintenance. Second, researchers should investigate synergistic interactions between RPRD1A depletion and conventional chemotherapeutics, as studies have shown that RPRD1A knockdown sensitizes cancer cells to platinum-induced cell death by disrupting redox homeostasis . Third, high-throughput screening for small molecule inhibitors of RPRD1A or its interaction with NRF2 could identify lead compounds for drug development. Fourth, combination therapies targeting both RPRD1A and oxidative stress pathways might prove especially effective. Finally, analysis of patient samples before and after treatment with conventional therapies could reveal whether RPRD1A expression correlates with treatment resistance, providing clinical rationale for targeting this protein in refractory disease.

What approaches can resolve the discrepancy between RPRD1A's reported transcriptional effects?

The literature presents conflicting reports regarding RPRD1A's role in transcription, with some studies suggesting it promotes transcription while others indicate negative regulatory effects . To resolve this discrepancy, researchers should employ stage-specific analyses throughout the transcription cycle using techniques like ChIP-seq with antibodies against initiation, elongation, and termination factors to determine if RPRD1A affects specific transcriptional phases differently. Gene-specific effects should be classified through genome-wide analyses to identify whether RPRD1A's impact varies across gene categories, potentially explaining the variable effects observed. Context-dependent functions should be examined by studying RPRD1A under various cellular conditions (stress, cell cycle phases, differentiation states) to determine if its role changes with cellular context. Additionally, single-cell approaches could reveal cell-to-cell variability in RPRD1A function that might be masked in bulk analyses. Finally, biochemical characterization of different RPRD1A-containing complexes may identify distinct complexes with opposing transcriptional effects.

How do the functions of RPRD proteins differ between normal and cancer cells?

While RPRD proteins act as negative regulators of transcription in normal human cells , in cancer contexts like HCC, RPRD1A appears to promote tumor progression through multiple mechanisms . To investigate this apparent dichotomy, researchers should conduct parallel studies in matched normal and cancer cell lines, examining RPRD1A's impact on transcription, cell proliferation, and oxidative stress response. RNA-seq and 4sU-seq comparisons between normal hepatocytes and HCC cells following RPRD1A modulation would reveal differentially affected pathways. Researchers should examine whether RPRD1A forms different protein complexes in normal versus cancer cells using immunoprecipitation followed by mass spectrometry. Post-translational modifications of RPRD1A should be profiled, as these might differ between normal and cancer contexts, potentially explaining functional differences. This comparative approach would help determine whether RPRD1A's seemingly contradictory roles reflect tissue-specific functions, cancer-specific adaptations, or context-dependent regulation that could be therapeutically exploited.

What is the relationship between RPRD1A expression and therapy resistance in cancer?

Given RPRD1A's role in oxidative stress defense and NRF2 pathway activation , it may contribute to therapy resistance in cancer. Many cancer therapies induce oxidative stress, and enhanced antioxidant capacity is associated with treatment resistance. To investigate this relationship, researchers should analyze RPRD1A expression in paired pre- and post-treatment tumor samples from patients receiving standard-of-care therapies. Cell line models with modulated RPRD1A expression should be challenged with various therapeutic agents to determine if RPRD1A alters sensitivity. Combination approaches targeting RPRD1A (via genetic knockdown) alongside conventional therapies should be tested in preclinical models. The table below summarizes potential experimental approaches:

Experimental ApproachPurposeExpected Outcome if RPRD1A Mediates Resistance
Patient sample analysisCorrelate RPRD1A with treatment responseHigher RPRD1A in non-responders
Cell viability assaysTest therapy sensitivity with/without RPRD1ARPRD1A knockdown increases sensitivity
Oxidative stress assaysMeasure ROS after treatmentRPRD1A reduces therapy-induced ROS
In vivo combination therapyTest synergy with RPRD1A targetingEnhanced efficacy with combination

This systematic approach would determine RPRD1A's potential as a therapeutic target to overcome resistance.

How does the interplay between RPRD1A and other transcriptional regulators affect gene expression programs?

RPRD1A interacts with multiple regulatory factors including RPAP2, HDAC1, DSIF, and PAF1C , suggesting it functions within larger regulatory networks. To map this interplay, researchers should perform integrative analyses combining RNA-seq, ChIP-seq for RPRD1A and associated factors, and protein interaction studies. Sequential ChIP (Re-ChIP) could identify genomic regions co-occupied by RPRD1A and other factors. CRISPR-based epigenome editing approaches could test the functional consequences of disrupting specific interactions. Global Run-On sequencing (GRO-seq) following perturbation of individual components would reveal their contributions to transcriptional regulation. Network analysis algorithms applied to these datasets could identify regulatory hubs and feedback mechanisms. For mechanistic studies, in vitro transcription systems reconstituted with purified components would allow systematic testing of how different factor combinations affect transcription initiation, pausing, and elongation. This integrative approach would place RPRD1A within its broader regulatory context and potentially identify critical nodes for therapeutic intervention.

Product Science Overview

Gene and Protein Structure

The RPRD1A gene is located on chromosome 18q12.2 and spans approximately 78 kb, containing 7 exons . The protein encoded by this gene consists of 312 amino acids with a predicted molecular mass of 35.7 kDa . It contains several important domains and motifs, including:

  • RAR domain
  • Leucine zipper motif
  • Multiple phosphorylation sites (4 cAMP/cGMP-dependent protein kinase sites, 3 PKC sites, and 8 casein kinase II sites) .
Function and Mechanism

RPRD1A is involved in the regulation of the cell cycle and transcription. It interacts with the cell cycle inhibitor cyclin-dependent kinase 4 inhibitor B (CDKN2B) and may function as a negative regulator of the G1/S phase progression . The protein forms homo- and heterodimers with Regulation Of Nuclear Pre-MRNA Domain Containing 1B (RPRD1B) to create a scaffold that interacts with the C-terminal domain of RNA polymerase II subunit B1 (POLR2A) . This interaction regulates several aspects of transcription, including the dephosphorylation of the C-terminal heptapeptide repeat domain (CTD) of RNA polymerase II by RPAP2 .

Expression and Regulation

RPRD1A is upregulated in cells overexpressing cyclin-dependent kinase inhibitor p15 (INK4b) and may have a role in cell cycle regulation . It is highly expressed in the prefrontal cortex, with much lower expression in other adult and fetal tissues . The protein enhances the expression of cyclin D1 (CCND1) and promotes the binding of RNA polymerase II to the CCND1 promoter .

Clinical Significance

RPRD1A has been implicated in various cellular processes and diseases. For instance, it is highly expressed in hepatocellular carcinoma (HCC) tumors and is correlated with aggressive clinicopathological features . Additionally, antisense expression of RPRD1A upregulates cyclin D1 and cyclin E in melanoma cells overexpressing transfected CDKN2B, suggesting its role as a negative regulator of these cyclins in the cell cycle .

Research and Applications

The study of RPRD1A is ongoing, with researchers exploring its potential roles in various biological processes and diseases. Understanding the function and regulation of this protein could provide insights into cell cycle control and transcription regulation, potentially leading to new therapeutic targets for cancer and other diseases.

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 2024 Thebiotek. All Rights Reserved.