RNF7 Human

Ring Finger Protein 7 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to RNF7 Human

RNF7 Human (also known as ROC2, SAG, or RBX2) is a highly conserved ring finger protein critical for E3 ubiquitin ligase activity. It serves as an essential subunit of SCF (SKP1-CUL1-F-box protein) complexes, which regulate protein degradation, cell cycle progression, and signal transduction . RNF7 is phosphorylated by casein kinase II (CSNK2A1), promoting the degradation of proteins like IkappaBalpha and p27Kip1 . Recombinant RNF7 Human (PRO-1671) is produced in E. coli as a 15.1 kDa polypeptide (136 amino acids) with a His-tag for purification .

Molecular Properties

PropertyDescription
Amino Acid SequenceMGSSHHHHHHSSGLVPRGSHMGSMADVEDG EETCALASHSGSSGSKSGGD KMFSLKKWNA VAMWSWDVEC DTCAICRVQV MDACLRCQAE NKQEDCVVVW GECNHSFHNC CMSLWVKQNN RCPLCQQDWV VQRIGK
Molecular Weight15.1 kDa
PurificationProprietary chromatography; fused with 23-aa His-tag at N-terminus
Storage4°C (short-term) or -20°C (long-term) with 0.1% HSA/BSA carrier protein

Functional Partners

RNF7 interacts with core components of E3 ligase complexes and regulatory proteins:

ProteinRole in Complex/PathwayInteraction Score (STRING-DB)
CUL1Scaffold for SCF complexes0.999
SKP1Links F-box proteins to CUL10.992
UBE2FNEDD8-conjugating enzyme0.998
NEDD8Activates cullin ligases0.996
CSNK2A1Phosphorylates RNF7

Prostate Cancer and Chemoresistance

RNF7 overexpression in castration-resistant prostate cancer (CRPC) cells (DU145, PC3) enhances proliferation and cisplatin resistance. Knockdown of RNF7:

  • Reduces proliferation via accumulation of tumor suppressors (p21, p27, NOXA) .

  • Inactivates ERK1/2 signaling, impairing tumor invasion .

ParameterRNF7 Knockdown (vs. Control)Source
Proliferation↓ (DU145: 26.2%, PC3: 24.9%)
Cisplatin Sensitivity↑ (Apoptosis ↑)

Sepsis-Induced Skeletal Muscle Injury

In CLP rat models, RNF7:

  • Promotes apoptosis via THBS1 upregulation and PI3K/Akt inhibition .

  • Exacerbates oxidative stress (↑ NO, MDA; ↓ GSH-Px, SOD) .

BiomarkerRNF7 OverexpressionRNF7 Silencing
THBS1
Bax
LC3-II/LC3-I

Renal Cell Carcinoma (RCC)

RNF7 overexpression in RCC cells:

  • Inhibits apoptosis and enhances glycolysis via JAK/STAT3 activation .

  • Degrades SOCS1, sustaining STAT3 signaling and sunitinib resistance .

Applications and Therapeutic Implications

  • Research Tool: Recombinant RNF7 (PRO-1671) is used to study ubiquitination mechanisms .

  • Therapeutic Target: Silencing RNF7 shows promise in CRPC and RCC by restoring tumor suppressor activity and chemosensitivity .

References

  1. ProspecBio: RNF7 Human Recombinant Protein (PRO-1671) .

  2. STRING-DB: RNF7 Interaction Network .

  3. Infection and Immunity: RNF7 in sepsis-induced muscle injury .

  4. Scientific Reports: RNF7 knockdown in prostate cancer .

  5. NCBI Gene: RNF7 Gene Entry .

  6. Wikipedia: RNF7 Overview .

  7. Oncotarget: RNF7 in renal cell carcinoma .

Product Specs

Introduction
Ring Finger Protein 7, also known as RNF7, is a highly conserved protein characterized by the presence of a ring finger domain. This protein plays a crucial role as a component of SKP1-cullin/CDC53-F box protein ubiquitin ligases, which are integral to the protein degradation machinery. These ligases are essential for regulating cell cycle progression and signal transduction pathways. RNF7 is known to be a substrate of casein kinase II (CSNK2A1/CKII) and also interacts with it. Notably, the phosphorylation of RNF7 by CSNK2A1 facilitates the degradation of IkappaBalpha (CHUK/IKK-alpha/IKBKA) and p27Kip1 (CDKN1B).
Description
This product consists of the recombinant human RNF7 protein, produced in E. coli. It is a single, non-glycosylated polypeptide chain comprising 136 amino acids (specifically, amino acids 1 through 113). The protein has a molecular weight of 15.1 kDa. For purification and detection purposes, the RNF7 protein is fused to a 23 amino acid His-tag at its N-terminus. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
The product is a clear, colorless solution that has been sterilized by filtration.
Formulation
The RNF7 protein is supplied in a solution at a concentration of 1 mg/ml. The solution buffer consists of 20mM Tris-HCl at pH 8.0, 10% glycerol, and 0.4M Urea.
Stability
For short-term storage (up to 2-4 weeks), the product should be kept at 4°C. For longer storage periods, it is recommended to store the product in a frozen state at -20°C. To further enhance stability during long-term storage, the addition of a carrier protein such as HSA or BSA to a final concentration of 0.1% is advised. It's important to avoid repeated cycles of freezing and thawing the product.
Purity
The purity of the RNF7 protein in this product is greater than 90.0%, as determined by SDS-PAGE analysis.
Synonyms
RING-box protein 2 isoform 1, Ring finger protein 7, CKBBP1, ROC2, SAG, RING-box protein 2, Rbx2, CKII beta-binding protein 1, Regulator of cullins 2, Sensitive to apoptosis gene protein, RBX2, RNF7.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMADVEDG EETCALASHS GSSGSKSGGD KMFSLKKWNA VAMWSWDVEC DTCAICRVQV MDACLRCQAE NKQEDCVVVW GECNHSFHNC CMSLWVKQNN RCPLCQQDWV VQRIGK.

Q&A

What is RNF7 and what are its primary molecular characteristics?

RNF7, also known as SAG (sensitive to apoptosis gene), ROC2 (regulator of cullins 2), or Rbx2 (RING-box 2), is a highly conserved ring finger protein consisting of 113 amino acids with a molecular weight of 12.6 kDa. It was originally identified as a redox-inducible anti-oxidant protein . RNF7 functions as an essential subunit of SKP1-cullin/CDC53-F box protein ubiquitin ligases, which are integral parts of the protein degradation machinery important for cell cycle progression and signal transduction . The protein contains zinc ion binding domains and demonstrates interaction with several substrate proteins. RNF7 plays crucial roles in the ubiquitin-proteasome pathway by facilitating the degradation of various regulatory proteins involved in cell proliferation and apoptosis .

What protein interactions does RNF7 participate in to execute its cellular functions?

RNF7 engages in several critical protein-protein interactions that facilitate its role in cellular homeostasis. One of the most well-documented interactions is with casein kinase II (CSNK2B), which phosphorylates RNF7 . This phosphorylation promotes the degradation of important regulatory proteins including IkappaBalpha (CHUK/IKK-alpha/IKBKA) and p27Kip1 (CDKN1B) . In cancer contexts, RNF7 interacts with components of the PI3K/AKT signaling pathway to promote cell proliferation and survival . Furthermore, RNF7 interacts with cullin proteins as part of E3 ubiquitin ligase complexes that target various tumor suppressive proteins including p21, p27, and NOXA for proteasomal degradation . These interactions collectively contribute to RNF7's role in cell cycle regulation, apoptosis resistance, and cancer progression.

How does RNF7 contribute to prostate cancer progression and chemoresistance?

RNF7 plays a significant role in prostate cancer progression, particularly in castration-resistant prostate cancer (CRPC). Mechanistically, RNF7 functions by preventing the accumulation of tumor suppressive proteins such as p21, p27, and NOXA in prostate cancer cells . When RNF7 is silenced through shRNA interference in CRPC cell lines DU145 and PC3, there is a marked attenuation of proliferation and enhanced sensitivity to cisplatin treatment . The underlying mechanism involves two major pathways: (1) accumulation of tumor suppressive proteins p21, p27, and NOXA following RNF7 knockdown, and (2) inactivation of the ERK1/2 pathway, which normally promotes cell survival and proliferation . Additionally, RNF7 silencing attenuates the invasive properties of DU145 and PC3 cells, suggesting its role in promoting metastatic potential in prostate cancer. These findings collectively indicate that RNF7 contributes to prostate cancer progression by inhibiting apoptosis, promoting proliferation, enhancing invasion, and conferring chemoresistance .

What is the relationship between RNF7 expression and glioma malignancy?

RNF7 expression demonstrates a strong positive correlation with glioma malignancy and patient outcomes. High-throughput sequencing has identified RNF7 among the top 15 upregulated genes in high-grade glioma (HGG) compared to low-grade glioma (LGG), with expression altered more than five-fold . This upregulation was confirmed at both mRNA and protein levels using qRT-PCR, Western blotting, and immunohistochemistry . Clinical data analysis reveals that high RNF7 expression is significantly associated with several adverse clinicopathological features:

FeaturesRNF7 Expressionp-Value
LowHigh
Tumor size, cm
<222130.003
>=2517
Karnofsky Performance Scale
<9010200.026
>=901710
WHO grade
Low-grade (I + II)1580.027
High-grade (III + IV)1222

Multivariate Cox regression analysis confirms that high RNF7 expression is an independent prognostic factor for poor survival (HR=1.738, 95% CI: 1.243-4.214, p<0.001) . The predictive value of RNF7 is further enhanced when combined with WHO staging (AUC=0.732) compared to WHO staging alone (AUC=0.604) . These findings establish RNF7 as both a marker of glioma malignancy and an independent predictor of poor prognosis.

Through which signaling pathways does RNF7 promote cancer cell proliferation?

RNF7 promotes cancer cell proliferation through multiple signaling pathways, with the PI3K/AKT pathway being particularly significant. In glioma cells, Gene Set Enrichment Analysis (GSEA) demonstrated that RNF7 expression is significantly associated with PI3K/AKT signaling . This was further confirmed using Cignal Finder Cancer 10-Pathway Reporter Kits, which showed that PI3K/AKT signaling was specifically inhibited by RNF7 knockdown in T98G and U-87MG glioma cells, while other signaling pathways remained largely unaffected .

In prostate cancer, RNF7 knockdown led to inactivation of the ERK1/2 pathway, suggesting this as another mechanism through which RNF7 promotes cancer progression . RNF7 also influences cell cycle progression, with overexpression promoting G1 to S-phase transition and accelerating cell proliferation in glioma cells . Conversely, RNF7 knockdown induces cell cycle arrest at the G1 phase .

These pathways converge on critical cellular processes including apoptosis inhibition, cell cycle progression, and enhanced invasion capability, all contributing to the pro-tumorigenic role of RNF7 .

What methods are most effective for modulating RNF7 expression in cancer cell lines?

Several effective methodologies have been established for modulating RNF7 expression in cancer research. For knockdown experiments, short hairpin RNA (shRNA) interference has proven particularly effective. In prostate cancer studies, researchers successfully silenced RNF7 using shRNA in DU145 and PC3 cell lines, achieving significant reduction in RNF7 protein levels confirmed by Western blotting . Similarly, in glioma research, RNF7 knockdown in T98G and U-87MG cells was achieved using targeted approaches with knockdown efficiency verified through Western blotting .

For overexpression experiments, researchers have successfully established stable RNF7-overexpressing cell lines in T98G and U-87MG glioma cells, with verification of increased expression by Western blotting . When designing these experiments, it is crucial to include appropriate controls and to verify the modulation efficiency through robust protein quantification methods. Additionally, researchers should consider the potential compensatory mechanisms that might be activated upon RNF7 modulation, as well as cell line-specific variations in transfection or transduction efficiency .

What in vivo models are appropriate for studying RNF7 function in cancer progression?

Xenograft models have proven valuable for studying RNF7 function in cancer progression in vivo. In glioma research, subcutaneous tumor xenograft models using immunodeficient mice have successfully demonstrated that RNF7 overexpression significantly enhances tumor growth, while RNF7 knockdown inhibits tumor growth . These models allow for direct measurement of tumor volume and weight, as well as histological analysis of proliferation markers such as Ki-67 .

When establishing these models, researchers should consider several methodological aspects: (1) cell number optimization for injection, (2) appropriate immunodeficient mouse strain selection, (3) tumor growth monitoring frequency, (4) endpoint selection criteria, and (5) post-sacrifice analysis methodologies. For more advanced studies, orthotropic models (where tumors are implanted in their organ of origin) may provide more physiologically relevant insights into RNF7's role in tumor-microenvironment interactions .

Additionally, in the context of sepsis research, cecal ligation and puncture (CLP) rat models have been utilized to study RNF7's role in sepsis-induced skeletal muscle injury . These diverse animal models allow researchers to investigate RNF7 function across different pathological contexts.

What techniques are most reliable for quantifying RNF7 expression in clinical samples?

Multiple complementary techniques are recommended for reliable RNF7 quantification in clinical samples. Quantitative real-time PCR (qRT-PCR) provides sensitive measurement of RNF7 mRNA levels and has been successfully employed to demonstrate RNF7 upregulation in glioma tissues compared to normal brain tissues . Western blotting offers protein-level confirmation and has been used in both prostate cancer and glioma studies to verify differential RNF7 expression .

Immunohistochemistry (IHC) is particularly valuable for clinical samples as it preserves tissue architecture and allows assessment of RNF7 expression patterns within the tumor microenvironment . This technique has successfully demonstrated higher RNF7 protein levels in high-grade gliomas compared to low-grade gliomas and normal brain tissues .

For localization studies, fluorescence in situ hybridization (FISH) can be employed, as demonstrated in skeletal muscle research . When analyzing clinical samples, researchers should implement standardized protocols with appropriate positive and negative controls, and consider using tissue microarrays for high-throughput analysis when available. Quantification should employ digital image analysis software to minimize subjective interpretation .

What is the evidence supporting RNF7 as a therapeutic target in cancer treatment?

Multiple lines of evidence support RNF7 as a promising therapeutic target. First, RNF7 is consistently overexpressed across multiple cancer types including prostate cancer, glioma, and carcinomas of lung, colon, stomach, and liver . This broad overexpression pattern suggests RNF7 may be a common driver of malignant progression rather than a passenger alteration.

Second, functional studies demonstrate that RNF7 knockdown produces significant anti-cancer effects across multiple experimental systems. In prostate cancer, RNF7 silencing attenuates proliferation, enhances chemosensitivity to cisplatin, and reduces invasive properties of castration-resistant prostate cancer cells . In glioma, RNF7 knockdown suppresses cell proliferation, promotes apoptosis, inhibits invasion, and induces cell cycle arrest at the G1 phase both in vitro and in vivo .

Third, mechanistic insights reveal that RNF7 inhibition simultaneously affects multiple cancer-promoting pathways, including PI3K/AKT and ERK1/2 signaling . This multi-pathway impact suggests RNF7 inhibition might be less susceptible to resistance development compared to single-pathway targeted therapies. Finally, multivariate Cox regression analysis identifies high RNF7 expression as an independent prognostic factor for poor survival in glioma patients (HR=1.738, p<0.001), highlighting its clinical relevance .

What molecular strategies might be effective for pharmacologically targeting RNF7?

Several molecular strategies show potential for pharmacologically targeting RNF7. As RNF7 functions as part of E3 ubiquitin ligase complexes, designing small molecule inhibitors that disrupt protein-protein interactions between RNF7 and its binding partners represents one promising approach. Specifically, compounds that interfere with RNF7's interaction with cullin proteins or with substrate recognition components could impair its ubiquitin ligase activity .

Leveraging RNF7's dependence on phosphorylation by casein kinase II (CSNK2B) offers another potential strategy . Compounds that prevent this phosphorylation event could potentially reduce RNF7's ability to promote degradation of tumor suppressive proteins. Additionally, as RNF7 contains zinc-binding domains, zinc chelators specifically designed to target these domains might disrupt RNF7's structural integrity and function .

For RNA-based approaches, the successful application of shRNA in experimental settings suggests that siRNA-based therapeutics or antisense oligonucleotides targeting RNF7 mRNA could be developed for clinical applications . These approaches would require appropriate delivery systems to ensure adequate tissue distribution and cellular uptake. When developing these strategies, researchers should consider potential off-target effects and compensatory mechanisms that might limit therapeutic efficacy.

How might RNF7 inhibition synergize with existing cancer therapies?

RNF7 inhibition shows potential for synergistic interactions with several existing cancer therapies. Most notably, RNF7 silencing enhances sensitivity of prostate cancer cells to cisplatin treatment, suggesting combination with platinum-based chemotherapies as a promising approach . This chemosensitization effect likely results from RNF7 inhibition allowing accumulation of pro-apoptotic proteins that would otherwise be degraded through RNF7-mediated ubiquitination .

For glioma, RNF7 knockdown leads to G1 phase cell cycle arrest . This effect could potentially synergize with radiation therapy, which is often more effective against cells in specific cell cycle phases. Additionally, as RNF7 promotes PI3K/AKT signaling in glioma, combining RNF7 inhibition with existing PI3K/AKT/mTOR pathway inhibitors might produce enhanced anti-tumor effects through vertical pathway inhibition .

Since RNF7 knockdown leads to accumulation of tumor suppressive proteins including p21 and p27 , combining RNF7 inhibition with CDK inhibitors might produce synergistic effects on cell cycle arrest. When designing combination strategies, researchers should carefully evaluate potential overlapping toxicities and conduct thorough pre-clinical assessment of therapeutic indices for each combination approach.

How does RNF7 expression correlate with other molecular markers in cancer?

Understanding RNF7's correlation with other molecular markers remains an important area for further investigation. In glioma, preliminary evidence suggests that RNF7 expression correlates with clinical outcomes when combined with WHO grading . The combination model of RNF7 expression and WHO stage (AUC=0.732) outperformed the WHO-based model alone (AUC=0.604) in predicting clinical outcomes . This suggests potential interactions between RNF7 and other molecular features that define WHO glioma classifications.

For comprehensive correlation analysis, researchers should employ multi-omic approaches including transcriptomics, proteomics, and phosphoproteomics to identify molecular patterns associated with RNF7 expression across different cancer types. Particular attention should be given to correlations with established biomarkers such as p53 status, PTEN expression, IDH mutation status in gliomas, and androgen receptor status in prostate cancers .

Methodologically, researchers should utilize publicly available databases such as The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC) to conduct large-scale correlation analyses. These analyses should be complemented with experimental validation in cell line panels and patient-derived samples to confirm biologically significant associations.

What are the tissue-specific functions of RNF7 in non-cancerous pathological conditions?

While RNF7's role in cancer has been extensively studied, its tissue-specific functions in non-cancerous pathological conditions require further investigation. Emerging evidence suggests RNF7 plays a role in sepsis-induced skeletal muscle injury, where it appears to induce skeletal muscle cell apoptosis and arrest cell cycle progression . This finding indicates that RNF7 may have context-dependent functions across different tissues and disease states.

As RNF7 was originally identified as a redox-inducible anti-oxidant protein , investigating its role in oxidative stress-related pathologies such as ischemia-reperfusion injury, neurodegenerative disorders, and inflammatory conditions presents valuable research opportunities. Researchers should employ tissue-specific conditional knockout models to elucidate RNF7's function in different organ systems under both physiological and pathological conditions.

Methodologically, single-cell RNA sequencing could reveal cell type-specific expression patterns and functions of RNF7 within complex tissues. Additionally, phosphoproteomic analysis following tissue-specific RNF7 modulation could identify downstream effectors that may vary between tissues, providing insights into tissue-specific regulatory networks involving RNF7.

What epigenetic mechanisms regulate RNF7 expression in different cellular contexts?

The epigenetic regulation of RNF7 remains largely unexplored and represents an important frontier for future research. Given RNF7's differential expression across normal and malignant tissues, epigenetic mechanisms likely play a crucial role in controlling its expression . Researchers should investigate several potential regulatory mechanisms including DNA methylation at the RNF7 promoter, histone modifications affecting chromatin accessibility, and non-coding RNAs that might post-transcriptionally regulate RNF7 expression.

Methodologically, researchers can employ bisulfite sequencing to characterize the methylation status of the RNF7 promoter across different tissues and disease states. ChIP-seq for various histone modifications (H3K4me3, H3K27ac, H3K27me3) could reveal the chromatin state at the RNF7 locus. RNA-seq coupled with bioinformatic prediction tools can identify potential microRNAs targeting RNF7 mRNA, which can then be validated through luciferase reporter assays.

For functional validation, researchers should utilize epigenetic modifying drugs such as DNA methyltransferase inhibitors and histone deacetylase inhibitors to assess their impact on RNF7 expression. Understanding these epigenetic regulatory mechanisms could potentially identify new approaches to therapeutically modulate RNF7 expression in various pathological contexts.

Product Science Overview

Introduction

Ring Finger Protein 7 (RNF7), also known as Sensitive to Apoptosis Gene (SAG), is a highly conserved protein that functions as an E3 ubiquitin ligase. This protein plays a crucial role in the ubiquitination process, which is a post-translational modification that regulates the degradation of proteins within the cell. RNF7 is involved in various cellular processes, including cell cycle regulation, apoptosis, and signal transduction.

Structure and Function

RNF7 contains a RING (Really Interesting New Gene) finger domain, which is essential for its function as an E3 ubiquitin ligase. The RING domain facilitates the transfer of ubiquitin from an E2 ubiquitin-conjugating enzyme to the target substrate, marking it for degradation by the proteasome. This process is vital for maintaining cellular homeostasis and regulating protein levels within the cell .

Role in Cancer

Overexpression of RNF7 has been observed in multiple human cancers, including renal cell carcinoma (RCC). In RCC, RNF7 inhibits apoptosis and promotes glycolysis, contributing to tumor growth and progression. The elevated expression of RNF7 in tumor tissues is correlated with poor survival outcomes in patients. RNF7 achieves these effects by activating the JAK/STAT3 signaling pathway through the ubiquitination of SOCS1 (Suppressor of Cytokine Signaling 1) .

Mechanisms of Action

RNF7’s role in cancer is primarily mediated through its function as an E3 ubiquitin ligase. By ubiquitinating specific target proteins, RNF7 can regulate various signaling pathways that control cell proliferation, apoptosis, and metabolism. For example, in RCC, RNF7 promotes tumor growth by inhibiting apoptosis and enhancing glycolysis. This is achieved through the activation of the JAK/STAT3 signaling pathway, which is a critical regulator of cell survival and proliferation .

Therapeutic Potential

Given its role in cancer progression, RNF7 represents a potential therapeutic target for cancer treatment. Inhibiting RNF7’s activity could restore normal cellular processes and reduce tumor growth. Research is ongoing to develop specific inhibitors that can target RNF7 and disrupt its function in cancer cells .

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.