RRM2 Human

Ribonucleotide Reductase M2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to RRM2 Human

Ribonucleotide reductase regulatory subunit M2 (RRM2) is a 389-amino acid protein encoded by the RRM2 gene located on chromosome 2p25-p24 . It is a critical component of ribonucleotide reductase (RR), an enzyme responsible for converting ribonucleotides to deoxynucleotides, which are essential for DNA synthesis, repair, and replication . RRM2’s expression is tightly regulated during the cell cycle, peaking in the S phase and degrading post-mitosis .

Protein Structure

FeatureDescriptionSource
Amino Acid Length389 residues
Molecular Weight44,883 Da
Key Domains- Dinuclear iron center
- 13 helices and 2 β-sheets
- Conserved C-terminal (aa 69–389)
Iron BindingNon-heme iron center stabilized by Tyr162 and hydrophilic residues

The N-terminal region (aa 1–68) exhibits low homology across species, while the C-terminal (aa 69–389) is conserved, indicating its critical role in enzymatic activity .

Enzymatic and Non-Enzymatic Functions

  1. Deoxyribonucleotide Synthesis

    • Converts ribonucleotides (e.g., GDP, CDP, ADP) to deoxynucleotides via RR activity .

    • Rate-limiting enzyme for DNA replication and repair .

  2. Non-Enzymatic Roles

    • Regulates Wnt signaling via Tyr phosphorylation at Ser-20 .

    • Modulates cell cycle progression and apoptosis .

Transcriptional Control

Regulatory MechanismDescriptionSource
Alternative PromotersTwo transcripts (same ORF) with distinct promoter regions
S-Phase SpecificityDriven by CCAAT sequences and TATA-box-like elements in the promoter
Cell Cycle DependencyAccumulates during G1/S transition, degraded in G2/M via cyclin F

Post-Translational Modifications

Modification TypeEnzyme InvolvedEffect on RR ActivitySource
AcetylationKAT7 (HBO1)Inhibits RR activity
DeacetylationSIRT2Activates RR activity
PhosphorylationATR (Thr-33)Stabilizes RRM2 100

Prognostic Biomarker

Cancer TypeRRM2 ExpressionPrognostic CorrelationSource
Hepatocellular Carcinoma (HCC)↑ Tumor vs. normalWorse OS, PFS, DSS; Linked to T stage, histologic grade, and AFP levels
Glioblastoma (GBM)↑ High-grade tumorsPoorer survival; Correlates with Ki-67 (proliferation marker)
HER-2+ Breast Cancer↑ Tumor vs. normalAssociated with lymph node metastasis and reduced survival

Mechanisms in Tumorigenesis

  • DNA Synthesis and Proliferation: Sustains dNTP pools for rapid cell division .

  • Drug Resistance: Overexpression linked to chemoresistance in lung and breast cancers .

  • Immune Modulation: Inversely correlates with CD8+ T cells and neutrophils in HCC .

Inhibitors and Strategies

InhibitorMechanism of ActionClinical ImpactSource
GTI2040Antisense oligonucleotide targeting RRM2 mRNAEnhances gemcitabine efficacy in solid tumors
GW8510Small-molecule inhibitor of RR activityReverses gemcitabine resistance in NSCLC
SIRT2 InhibitorsBlocks deacetylation-dependent RR activationSynergizes with DNA-damaging agents

Preclinical Evidence

  • Glioblastoma Models: RRM2 knockdown reduces tumor size and proliferation via downregulation of cyclin B1/D1 and N-cadherin .

  • Hepatoblastoma: Combining RRM2 inhibitors with chemotherapy delays tumor relapse .

Product Specs

Introduction
RRM2 is an enzyme that catalyzes the conversion of ribonucleotides to deoxyribonucleotides, a crucial step in DNA synthesis. The enzyme's activity is tightly regulated throughout the cell cycle, ensuring the availability of DNA building blocks when needed. RRM2's role in DNA synthesis makes it essential for cell division and proliferation. Additionally, RRM2 has been found to inhibit Wnt signaling, a pathway involved in cell fate determination and development.
Description
Recombinant Human RRM2 is a purified protein produced in E. coli. This 47 kDa protein consists of a single, non-glycosylated polypeptide chain of 409 amino acids (amino acids 1-389) and includes a 20 amino acid His-Tag at the N-terminus. The protein is purified using proprietary chromatographic techniques to ensure high purity.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The RRM2 protein is supplied as a 1 mg/ml solution in a buffer containing 20mM Tris-HCl (pH 8), 0.1M NaCl, and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the protein can be stored at 4°C. For long-term storage, it is recommended to store the protein frozen at -20°C. To further enhance stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is advisable. Repeated freezing and thawing of the protein should be avoided.
Purity
The purity of the RRM2 protein is greater than 85%, as determined by SDS-PAGE analysis.
Synonyms
EC 1.17.4.1, RR2M, RR2, Ribonucleotide Reductase M2, R2, RRM2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MLSLRVPLAP ITDPQQLQLS PLKGLSLVDK ENTPPALSGT RVLASKTARR IFQEPTEPKT KAAAPGVEDE PLLRENPRRF VIFPIEYHDI WQMYKKAEAS FWTAEEVDLS KDIQHWESLK PEERYFISHV LAFFAASDGI VNENLVERFS QEVQITEARC FYGFQIAMEN IHSEMYSLLI DTYIKDPKER EFLFNAIETM PCVKKKADWA LRWIGDKEAT YGERVVAFAA VEGIFFSGSF ASIFWLKKRG LMPGLTFSNE LISRDEGLHC DFACLMFKHL VHKPSEERVR EIIINAVRIE QEFLTEALPV KLIGMNCTLM KQYIEFVADR LMLELGFSKV FRVENPFDFM ENISLEGKTN FFEKRVGEYQ RMGVMSSPTE NSFTLDADF.

Q&A

What is RRM2 and what is its primary function in human cells?

RRM2 is the regulatory subunit of ribonucleotide reductase (RNR), a heterotetrameric holoenzyme that catalyzes the rate-limiting conversion of ribonucleoside diphosphates to deoxyribonucleoside diphosphates. This enzyme plays a crucial role in DNA synthesis and repair, particularly during the S-phase of the cell cycle. The RNR holoenzyme is composed of two distinct subunits: the catalytic subunit (RRM1) and either the regulatory subunit RRM2 or its paralog RRM2B. The RRM1:RRM2 complex primarily contributes to DNA synthesis and repair during S-phase, making it essential for cellular proliferation and division .

How does RRM2 differ from RRM2B in cellular function?

While both RRM2 and RRM2B can form functional complexes with RRM1, they serve distinct biological roles. RRM1:RRM2 predominantly supports DNA synthesis and repair during the active S-phase of proliferating cells. In contrast, RRM1:RRM2B is primarily responsible for DNA repair in quiescent cells and for mitochondrial DNA synthesis or repair . This functional separation becomes particularly relevant in cancer contexts, where computational analyses have revealed that RRM2 supports cell proliferation pathways while RRM2B participates heavily in stress response mechanisms . Understanding this distinction is crucial when interpreting research data and designing targeted interventions.

What techniques are commonly used to measure RRM2 expression in research settings?

Researchers employ multiple complementary techniques to assess RRM2 expression:

  • For mRNA quantification:

    • Reverse transcription quantitative PCR (RT-qPCR) is frequently used to measure RRM2 transcript levels in paired tumor and normal tissues

    • RNA-seq and microarray analysis of datasets from repositories like GEO (Gene Expression Omnibus) provide broader expression patterns

  • For protein detection:

    • Enzyme-linked immunosorbent assay (ELISA) for measuring serum RRM2 levels

    • Immunohistochemistry (IHC) on tissue microarrays (TMAs) for spatial expression analysis

    • Western blotting for semi-quantitative protein assessment

When designing experiments, researchers should consider using multiple approaches to validate findings across methodological platforms.

How does RRM2 overexpression contribute to cancer progression mechanisms?

RRM2 overexpression promotes cancer progression through multiple interrelated mechanisms:

  • Cell cycle promotion: Gene ontology enrichment analysis of RRM2 co-expressed genes in cancer tissues reveals that high RRM2 expression correlates strongly with cell cycle and cell division gene signatures . This suggests RRM2 supports the accelerated proliferation characteristic of malignant cells.

  • Metastatic potential: High RRM2 expression significantly associates with metastatic disease at diagnosis (p=0.0004) and occurrence of metastatic and/or local relapse (p=0.0095) in Ewing sarcoma patients . This indicates RRM2 may enhance cancer cell motility and invasion capabilities.

  • Therapy resistance: In hepatoblastoma models, when RRM2 is suppressed by standard chemotherapies, compensatory upregulation of RRM2B occurs, promoting cell survival and subsequent relapse . This dynamic switching between RRM2 and RRM2B represents a sophisticated adaptation mechanism that allows cancer cells to evade therapeutic intervention.

  • Immune microenvironment modulation: RRM2 expression correlates significantly with immune cell infiltration, including CD8 T cells, cytotoxic cells, dendritic cells, neutrophils, NK cells, and T helper cells in hepatocellular carcinoma . This suggests RRM2 may influence the tumor immune microenvironment, potentially contributing to immune evasion.

These diverse mechanisms highlight why RRM2 overexpression consistently correlates with poor clinical outcomes across multiple cancer types.

What signaling pathways does RRM2 interact with in cancer contexts?

RRM2 interacts with several critical signaling pathways in cancer:

  • p53 signaling pathway: Gene Ontology/KEGG analysis demonstrates that RRM2 expression influences p53 signaling, which is central to cellular responses to stress and DNA damage . This interaction may help explain how elevated RRM2 allows cancer cells to evade apoptotic mechanisms.

  • Cell cycle regulation: Gene Set Enrichment Analysis (GSEA) shows that high RRM2 expression correlates with cell cycle pathway activation and P27 pathway modulation . These connections provide mechanistic insight into how RRM2 promotes uncontrolled proliferation.

  • Immune signaling: RRM2 expression correlates with PD-1 signaling and T cell receptor signaling pathways , suggesting RRM2 may influence how cancer cells interact with the immune system.

  • Chromosome condensation: GO analysis indicates RRM2 influences chromosome condensation processes , potentially affecting genomic stability in cancer cells.

Understanding these pathway interactions provides potential avenues for combination therapy approaches targeting RRM2 alongside these interacting pathways.

How can researchers effectively target RRM2 in experimental models?

Effective RRM2 targeting in experimental models requires consideration of several methodological approaches:

  • Genetic inhibition strategies:

    • RNA interference (RNAi) using siRNA or shRNA constructs targeting RRM2

    • CRISPR-Cas9 mediated knockout or knockdown

    • Antisense oligonucleotides targeting RRM2 mRNA

  • Pharmacological approaches:

    • Specific RRM2 inhibitors (several are in development but require validation for specificity)

    • Consideration of combination approaches, particularly with cell cycle checkpoint inhibitors targeting CHEK1 or WEE1

  • Experimental design considerations:

    • Monitor both RRM2 and RRM2B expression simultaneously, as suppression of RRM2 often leads to compensatory RRM2B upregulation

    • Include longitudinal sampling to capture the dynamic switching between RRM2 and RRM2B during treatment and relapse phases

    • Validate findings across multiple cancer cell lines to ensure generalizability of results

When designing RRM2 targeting experiments, researchers should anticipate potential compensatory mechanisms and include appropriate controls to detect these adaptations.

What is the evidence for RRM2 as a diagnostic biomarker across cancer types?

Evidence supporting RRM2 as a diagnostic biomarker varies across cancer types but shows promising consistency:

  • Non-small cell lung cancer (NSCLC):

    • RRM2 mRNA expression is significantly increased in NSCLC lesions compared to para-adjacent tissues

    • Serum RRM2 levels are significantly elevated in NSCLC patients compared to healthy controls

    • Receiver operating characteristic (ROC) curve analysis demonstrates a higher diagnostic ratio for NSCLC using RRM2 alone compared to traditional tumor markers like CYFRA21-1, ProGRP, CEA, and NSE

  • Hepatocellular carcinoma (HCC):

    • RRM2 expression is significantly higher in tumor tissues than in normal tissues (p<0.001)

    • Elevated RRM2 expression correlates with clinical parameters including T stage, pathologic stage, tumor status, histologic grade, and AFP levels

  • Ewing sarcoma (EwS):

    • RRM2 is specifically overexpressed in EwS tissues compared to normal tissues

    • High RRM2 expression is associated with aggressive disease features

Cancer TypeSample TypeDetection MethodDiagnostic Performance
NSCLCTissueRT-qPCRSignificantly elevated in tumor vs. normal
NSCLCSerumELISASuperior to traditional markers
HCCTissueMultiple platformsCorrelates with multiple clinical parameters
EwSTissueIHCAssociates with aggressive features

How reliable is RRM2 as a prognostic biomarker?

RRM2 has demonstrated consistent prognostic value across multiple cancer types:

These findings are particularly notable because they have been validated at both mRNA and protein levels and across independent patient cohorts, suggesting robust reliability as a prognostic indicator. Researchers should consider incorporating RRM2 assessment into clinical studies evaluating novel therapeutic approaches, particularly for stratifying patients by risk category.

What methodological considerations apply when studying RRM2 in patient samples?

When investigating RRM2 in clinical samples, researchers should consider:

  • Sample collection and processing:

    • For tissue analysis: Paired tumor and adjacent normal tissue sampling is optimal for establishing baseline expression patterns

    • For liquid biopsies: Serum is preferable to plasma for RRM2 protein detection using ELISA

  • Analytical approaches:

    • Multiple detection methods should be employed (RT-qPCR for mRNA, ELISA for protein, IHC for spatial distribution)

    • Compare RRM2 with established biomarkers (e.g., CEA, CYFRA21-1 for NSCLC; AFP for HCC) to establish relative diagnostic value

  • Clinical correlation:

    • Comprehensive clinical data collection including stage, grade, treatment history, and outcome measures is essential for meaningful biomarker validation

    • Statistical approaches should include both univariate and multivariate analyses to establish independent prognostic value

  • RRM2B consideration:

    • Parallel assessment of RRM2B is advisable, particularly in treatment response studies, due to the documented subunit switching phenomenon

What evidence supports targeting RRM2 as a therapeutic strategy?

Several lines of evidence support RRM2 as a promising therapeutic target:

  • Preclinical mechanistic evidence:

    • RRM2 inhibition reduces cancer cell proliferation and growth in multiple tumor models

    • RRM2 targeting can overcome acquired chemoresistance in some models

  • Combination therapy potential:

    • RRM2 inhibitors show synergistic effects when combined with cell cycle checkpoint inhibitors targeting CHEK1 or WEE1

    • Combining RRM2 inhibitors with conventional chemotherapy effectively delays tumor relapse in hepatoblastoma models

  • Specificity to cancer cells:

    • The substantial overexpression of RRM2 in multiple cancer types compared to normal tissues suggests a potential therapeutic window

    • RRM2's role in rapidly dividing cells makes it particularly relevant in cancer contexts

Future research should focus on developing highly specific RRM2 inhibitors and identifying the optimal combination strategies and cancer subtypes most likely to benefit from RRM2-targeted therapeutic approaches.

How might RRM2 and RRM2B switching affect treatment response?

The dynamic switching between RRM2 and RRM2B represents a sophisticated adaptation mechanism with important therapeutic implications:

  • Treatment response patterns:

    • Standard chemotherapies can effectively suppress RRM2 in cancer cells but simultaneously induce significant upregulation of RRM2B

    • RRM2B upregulation promotes cancer cell survival during treatment stress and facilitates subsequent relapse

    • During relapse phases, RRM2B is gradually replaced back by RRM2, completing an adaptive cycle

  • Strategic therapeutic considerations:

    • Sequential or simultaneous targeting of both RRM2 and RRM2B may be necessary to prevent this compensatory mechanism

    • Monitoring the relative expression of both subunits during treatment could provide early indications of developing resistance

    • Timing of RRM2 inhibitor administration may be critical, potentially being most effective when administered concurrently with initial chemotherapy

This subunit switching phenomenon highlights the importance of understanding dynamic molecular adaptations in cancer cells and designing treatment strategies that anticipate these changes.

What are the most promising areas for future RRM2 research?

Several research directions show particular promise:

  • Comprehensive mechanism studies:

    • Further elucidation of how RRM2 interacts with immune signaling pathways to potentially influence immunotherapy responses

    • Deeper investigation into the molecular triggers and consequences of RRM2/RRM2B switching

  • Biomarker refinement:

    • Development of standardized protocols for RRM2 detection in liquid biopsies to facilitate clinical implementation

    • Integration of RRM2 with other biomarkers to create more powerful prognostic panels

  • Therapeutic development:

    • Design of highly specific RRM2 inhibitors with favorable pharmacokinetic profiles

    • Clinical trials investigating RRM2 inhibition in combination with standard treatments or novel targeted agents

    • Exploration of RRM2 as a potential target in cancers beyond those already studied

  • RRM2 in special populations:

    • Investigation of RRM2's role in pediatric cancers, particularly given its emerging importance in hepatoblastoma and Ewing sarcoma

    • Assessment of RRM2 expression patterns and significance in therapy-resistant and metastatic disease settings

The consistent importance of RRM2 across diverse cancer types suggests its targeting could have broad applicability in oncology research and practice.

Product Science Overview

Introduction

Ribonucleotide Reductase M2 (RRM2) is a critical enzyme involved in the synthesis of deoxyribonucleotides from ribonucleotides, which are essential precursors for DNA synthesis and repair. This enzyme plays a pivotal role in maintaining the balance of the deoxyribonucleotide pool within cells, ensuring proper DNA replication and cell division.

Structure and Function

RRM2 is one of the two non-identical subunits of ribonucleotide reductase, the other being RRM1. The RRM2 subunit is regulated in a cell-cycle-dependent manner, with its expression peaking during the S phase of the cell cycle. This regulation ensures that deoxyribonucleotide production is synchronized with DNA replication demands .

The enzyme catalyzes the reduction of ribonucleotides to deoxyribonucleotides through a complex mechanism involving radical generation and transfer. The active site of RRM2 contains a diiron-oxygen cluster, which is essential for the generation of a tyrosyl radical necessary for the reduction process .

Biological Significance

RRM2 is crucial for DNA synthesis and repair, making it indispensable for cell proliferation and survival. Its activity is tightly regulated to prevent imbalances in the deoxyribonucleotide pool, which can lead to genomic instability and contribute to carcinogenesis .

Expression Patterns and Tissue Distribution

RRM2 is ubiquitously expressed in proliferating cells, with higher expression levels observed in tissues with high proliferative rates, such as the bone marrow, thymus, and gastrointestinal tract . Its expression is also upregulated in various cancers, where it is often associated with poor prognosis .

Regulatory Mechanisms

The expression of RRM2 is regulated at multiple levels, including transcriptional, post-transcriptional, and post-translational modifications. Transcriptional regulation is mediated by cell cycle-dependent transcription factors, while post-transcriptional regulation involves microRNAs that target RRM2 mRNA for degradation . Post-translational modifications, such as phosphorylation, can also modulate the activity and stability of the RRM2 protein .

Clinical Implications

Given its essential role in DNA synthesis and repair, RRM2 is a potential target for cancer therapy. Inhibitors of ribonucleotide reductase, such as hydroxyurea, have been used to treat certain cancers by disrupting DNA synthesis in rapidly proliferating tumor cells . Additionally, the overexpression of RRM2 in cancers makes it a valuable biomarker for prognosis and a potential target for therapeutic intervention .

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.