RNASE2 Human

Ribonuclease 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to RNASE2 Human

RNASE2 Human, encoded by the RNASE2 gene, is a non-secretory ribonuclease belonging to the pancreatic ribonuclease A superfamily. Synonyms include eosinophil-derived neurotoxin (EDN), RNS2, and RAF3 . This enzyme is primarily stored in eosinophil secondary granules but is also expressed in liver tissue and induced in macrophages under inflammatory conditions . RNASE2 exhibits broad-spectrum antiviral and antimicrobial activities, making it critical in innate immune responses .

Immune and Antiviral Functions

RNASE2 plays multifaceted roles in immunity:

Antiviral Mechanisms

  • Broad-spectrum activity: Inhibits replication of RSV, HIV-1, and hepatitis B virus by degrading viral RNA .

  • TLR7 activation: Mediates immune responses via TLR7 signaling in antiviral immunity .

  • Eosinophil-mediated defense: Released during eosinophil degranulation to target pathogens .

Antimicrobial and Immunomodulatory Roles

  • Bacterial agglutination: Aggregates pathogens via cationic charge (shared with RNASE3) .

  • Tissue repair: Participates in RNA scavenging and extracellular matrix remodeling .

  • Chemotaxis: Attracts dendritic cells and macrophages to inflammatory sites .

Expression Patterns and Disease Associations

Tissue/Cell TypeExpression LevelInducing StimuliDisease Links
EosinophilsHigh (secondary granules)Viral infections, cytokines Asthma, parasitic infections
MacrophagesInducedProinflammatory signals SLE, autoimmune disorders
LiverConstitutiveBasal expression Liver pathologies (unexplored)
Cancer TissuesVariableTumor microenvironment Prognostic marker (unclear)

Key Findings

  • SLE correlation: Elevated RNASE2 mRNA in lupus patients correlates with disease activity, autoantibody levels, and age-associated B cell (ABC) expansion .

  • IL-10 dependency: RNASE2 silencing reduces IL-10 secretion in monocytes, impairing ABC differentiation .

  • Cancer expression: Detected in breast, lung, and colorectal cancers, though prognostic significance remains unclear .

Systemic Lupus Erythematosus (SLE)

  • Mechanism: Monocyte-derived RNASE2 promotes IL-10 secretion, driving ABC proliferation .

  • Biomarker potential: High RNASE2 correlates with SLEDAI scores, proteinuria, and anti-dsDNA antibodies .

Antiviral Therapy

  • HIV-1 treatment: Recombinant RNASE2 administration is proposed to inhibit viral replication .

  • Immunotoxins: Engineered RNase constructs evade ribonuclease inhibitor (RI) to target cancer cells .

Research and Therapeutic Applications

ApplicationApproachOutcome
Antiviral DrugsRNASE2 protein therapyInhibition of RSV, HIV-1 replication
Cancer ImmunotherapyRNase-based immunotoxinsSelective tumor cell RNA degradation
Autoimmune DiseaseRNASE2 silencing in monocytesReduced ABCs and IL-10 levels

Future Directions

  • SLE biomarkers: Investigating RNASE2 as a predictive marker for disease flare-ups .

  • Therapeutic delivery: Optimizing recombinant RNASE2 formulations for clinical use .

Product Specs

Introduction
RNASE2, also known as EDN, is one of the four major proteins found in the large specific granules of human eosinophilic leukocytes. It is also expressed in human liver tissue and can be induced in macrophages by proinflammatory stimuli. RNASE2 is a non-secretory ribonuclease belonging to the pancreatic ribonuclease family, a subset of the ribonuclease A superfamily. Diseases associated with EDN include Atopic Dermatitis and Pulmonary Eosinophilia.
Description
RNASE2 Human produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 143 amino acids (28-161 a.a.) and having a molecular mass of 16.5 kDa. It is fused to a 6 amino acid His tag at the C-Terminus and purified by proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered colorless solution.
Formulation
RNASE2 Human protein (1 mg/ml) is formulated in 20% glycerol and Phosphate-Buffered Saline (pH 7.4).
Stability
Store at 4°C if the entire vial will be used within 2-4 weeks. Store, frozen at -20°C for longer periods. For long-term storage, it is recommended to add a carrier protein (0.1% HSA or BSA). Avoid multiple freeze-thaw cycles.
Purity
Greater than 90.0% as determined by SDS-PAGE.
Synonyms

EDN, RNS2, ribonuclease A family member 2, RAF3, non-secretory ribonuclease, Eosinophil-derived neurotoxin, RNase UpI-2, Ribonuclease 2, RNase 2, Ribonuclease US.

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

ADPKPPQFTW AQWFETQHIN MTSQQCTNAM QVINNYQRRC KNQNTFLLTT FANVVNVCGN PNMTCPSNKT RKNCHHSGSQ VPLIHCNLTT PSPQNISNCR YAQTPANMFY IVACDNRDQR RDPPQYPVVP VHLDRIIHHH HHH

Q&A

What is human RNASE2 and what are its primary functions?

RNASE2, also known as Eosinophil Derived Neurotoxin (EDN), belongs to the ribonuclease A superfamily. It is a secretory protein primarily expressed in leukocytes that participates in host defense responses. RNASE2 combines direct action against pathogens (particularly single-stranded RNA viruses) with diverse immunomodulatory properties . Its high catalytic activity against single-stranded RNA makes it effective against several viral types, including rhinoviruses, adenoviruses, and retroviruses including HIV . Recent research has also correlated the presence of eosinophils and their associated RNases with the prognosis of COVID-19 patients .

In which cell types and tissues is RNASE2 predominantly expressed?

RNASE2 is one of the main components of the eosinophil secondary granule matrix. Beyond eosinophils, it is also expressed in other leukocyte cell types such as neutrophils and monocytes, as well as epithelial cells, liver, and spleen . Human monocyte-derived macrophages can produce RNASE2 after stimulation . Additionally, the THP1 monocytic cell line has been shown to abundantly express RNASE2, making it a useful model for studying this protein's functions .

How does RNASE2 expression change during viral infection?

During viral infection, RNASE2 expression is significantly upregulated in responsive cells. For example, in THP1-derived macrophages infected with Respiratory Syncytial Virus (RSV), RNASE2 gene expression is upregulated in a time-dependent manner, with significant increases detectable as early as 4 hours post-infection and reaching a 7-fold increase at 72 hours . The secreted protein levels also increase, reaching maximum concentration at 48 hours post-infection . This temporal expression pattern correlates with viral population dynamics, suggesting a direct response to viral presence.

What techniques are recommended for measuring RNASE2 expression levels?

For comprehensive assessment of RNASE2 expression, multiple complementary techniques should be employed:

  • Transcriptional analysis: Real-time PCR using GAPDH as a housekeeping gene control is effective for detecting mRNA levels .

  • Protein detection:

    • ELISA for quantifying secreted RNASE2 in culture medium or serum

    • Western blotting for detecting intracellular RNASE2 protein

    • Flow cytometry using intracellular staining protocols (requires FcR blocking, surface staining with markers like CD14, fixation/permeabilization, and staining with RNASE2-specific antibodies)

These methods should be used in combination to distinguish between changes in transcription, translation, and secretion of RNASE2.

How can researchers effectively manipulate RNASE2 expression for functional studies?

Several approaches have proven effective for modulating RNASE2 expression:

  • Gene knockout: CRISPR/Cas9 gene editing has been successfully used to create RNASE2-knockout cell lines, such as the RNase2-KO THP1 monocyte model .

  • RNA interference:

    • siRNA-mediated silencing using targeted sequences at concentrations of approximately 1μM for 3 days has shown efficacy in reducing RNASE2 expression .

  • Lentiviral-based modification:

    • For overexpression: Cloning the RNASE2 coding sequence into vectors like pLVX-mCMV-ZsGreen-puro using appropriate restriction enzymes (EcoRI and NotI) .

    • For silencing: Cloning specific shRNA sequences targeting RNASE2 into vectors like pLent-U6-GFP-Puro .

    • Puromycin selection is recommended to establish stable cell lines .

Each approach has specific applications depending on research goals, with CRISPR providing complete knockout, siRNA offering transient reduction, and lentiviral systems enabling stable manipulation of expression levels.

What functional assays are appropriate for studying RNASE2's biological activities?

To comprehensively assess RNASE2 functions, researchers should consider multiple complementary assays:

  • Antiviral activity assessment:

    • Viral infection models (e.g., RSV infection of macrophages at MOI 1:1)

    • Comparison of viral titers between wild-type and RNASE2-modified cells

    • Time-course measurements of viral replication and cell survival

  • RNA cleavage analysis:

    • Total non-coding RNA population analysis using cp-RNAseq methodology

    • Screening for tRNA-derived fragments using library arrays

    • Mapping of selective RNA cleavage patterns

  • Cell proliferation and survival:

    • CCK-8 assay measuring optical density at 450nm over multiple days

    • Annexin V-APC/7-AAD apoptosis detection after 48 hours of culture

  • Migration and invasion:

    • Transwell and Transwell-Matrigel assays for assessing cellular movement

  • In vivo models:

    • Subcutaneous tumor growth measurement for cancer-related studies

    • Animal infection models for antiviral research

How is RNASE2 implicated in autoimmune disorders?

RNASE2 shows significant dysregulation in autoimmune conditions, particularly Systemic Lupus Erythematosus (SLE):

  • Expression patterns: RNASE2 mRNA is highly expressed in peripheral blood mononuclear cells (PBMCs) from SLE patients compared to healthy controls and patients with other autoimmune conditions like rheumatoid arthritis (RA) and primary Sjögren's syndrome .

  • Clinical correlations: RNASE2 expression positively correlates with:

    • SLEDAI score (disease activity)

    • 24-hour proteinuria levels

    • Serum creatinine levels

  • Autoantibody associations: Higher RNASE2 levels are found in seropositive SLE patients with anti-Sm, anti-dsDNA, and anti-SSB antibodies (but not anti-SSA), suggesting a link to autoantibody production mechanisms .

  • Cellular mechanisms: RNASE2 may mediate age-associated B cell expansion and affect immune sensing through Toll-like receptor 8 (TLR8) , serving as a bridge between innate and adaptive immunity in autoimmune contexts.

What role does RNASE2 play in cancer progression?

Research has identified RNASE2 as a significant factor in cancer, particularly glioma:

These findings position RNASE2 as both a potential biomarker and therapeutic target in glioma treatment strategies.

How does RNASE2 contribute to antiviral defense mechanisms?

RNASE2 plays multiple roles in antiviral defense:

  • Direct viral RNA degradation: RNASE2 directly targets viral RNA genomes, as shown in studies with RSV where its ribonucleolytic activity is required to remove the viral genome .

  • Structural specificity: Despite other RNaseA family members having higher catalytic activity, they lack RNASE2's antiviral properties, indicating that structural specificity beyond mere catalytic ability is crucial .

  • Response to infection: RSV infection induces both RNASE2 protein expression and secretion in human THP1-derived macrophages, with intracellular viral replication correlating with RNASE2 upregulation .

  • Impact on viral replication: Knockout of RNASE2 in THP1-derived macrophages results in higher RSV titers and reduced cell survival, confirming its protective role .

How does RNASE2 achieve selectivity for viral versus cellular RNA?

This represents one of the most intriguing aspects of RNASE2 biology:

  • Substrate preference: RNASE2 exhibits selective patterns of non-coding RNA cleavage, suggesting recognition of specific RNA structures or sequences .

  • Comparative enzymology: Despite other RNaseA family members having higher general catalytic activity, they lack RNASE2's specific antiviral properties, indicating unique target recognition mechanisms .

  • Mechanistic considerations: For methodological investigation, researchers should:

    • Compare cleavage sites on viral versus cellular RNAs using techniques like cp-RNAseq

    • Analyze the structural features of preferred substrates

    • Perform mutagenesis studies to identify residues involved in substrate discrimination

    • Compare the activity of RNASE2 against RNAs with different secondary structures

Understanding this selectivity mechanism could enable the development of more targeted antiviral therapeutics.

What is the relationship between RNASE2's enzymatic and immunomodulatory functions?

The dual functionality of RNASE2 raises important mechanistic questions:

  • Catalytic dependence: Some functions, like RSV genome degradation, require RNASE2's ribonucleolytic activity, while others may depend on protein-protein interactions .

  • Immune signaling: RNASE2 treatment induces dendritic cell maturation and stimulates production of pro-inflammatory cytokines and chemokines .

  • Pattern recognition: RNASE2 participates in immune sensing of pathogens through Toll-like receptor 8 (TLR8), potentially linking nucleic acid recognition to immune activation .

  • Experimental approaches:

    • Generate catalytically inactive RNASE2 mutants to dissect enzymatic versus structural roles

    • Map interaction partners in different immune cell types

    • Compare transcriptional responses to native versus enzymatically inactive RNASE2

    • Analyze downstream signaling pathway activation in various immunological contexts

How do post-translational modifications affect RNASE2 function?

While the search results don't directly address post-translational modifications, this represents an important advanced research question:

  • Potential modifications: As a secreted protein, RNASE2 likely undergoes various modifications including glycosylation, phosphorylation, or proteolytic processing.

  • Functional impact: These modifications could affect:

    • Protein stability and half-life

    • Substrate recognition and catalytic efficiency

    • Cellular localization and trafficking

    • Interactions with immune receptors or viral components

  • Methodological approaches:

    • Mass spectrometry to identify and map modifications

    • Site-directed mutagenesis of modification sites

    • Comparison of recombinant versus naturally produced RNASE2

    • Analysis of modification patterns in different disease states

What are the primary challenges in developing RNASE2-based therapeutics?

The therapeutic potential of RNASE2 faces several research challenges:

  • Context-dependent effects: RNASE2 shows beneficial effects in viral infections but potentially detrimental roles in autoimmunity and cancer, requiring careful targeting approaches.

  • Delivery considerations:

    • Tissue-specific delivery systems

    • Maintaining stability of protein-based therapeutics

    • Achieving appropriate biodistribution (particularly for CNS applications in glioma)

  • Mode of intervention:

    • For antiviral applications: Methods to enhance endogenous RNASE2 activity

    • For autoimmunity/cancer: Specific inhibitors targeting RNASE2 without affecting related RNases

    • Potential for catalytically modified RNASE2 variants with altered substrate specificity

  • Patient stratification: Identifying biomarkers to determine which patients might benefit from RNASE2-targeting approaches based on expression levels or disease subtypes.

How can researchers effectively study RNASE2 interactions with the microbiome?

The potential interaction between RNASE2 and the microbiome represents an unexplored frontier:

  • Research questions:

    • Does RNASE2 affect bacterial RNA or bacteriophage populations?

    • Do microbial products modulate RNASE2 expression?

    • Is there cross-talk between RNASE2, the microbiome, and host immunity?

  • Methodological approaches:

    • Gnotobiotic models with defined microbial communities

    • Metagenomic and metatranscriptomic analysis in the presence/absence of RNASE2

    • Ex vivo co-culture systems with primary human cells and microbial communities

    • Comparative analysis of microbiome composition in disease states with altered RNASE2 expression

  • Technical challenges:

    • Distinguishing direct versus indirect effects on microbial communities

    • Accounting for variable RNASE2 expression across tissue microenvironments

    • Controlling for effects of other antimicrobial proteins co-expressed with RNASE2

What are the emerging technologies for studying RNASE2 at single-cell resolution?

Advanced technologies offer new opportunities for RNASE2 research:

  • Single-cell approaches:

    • scRNA-seq to map cell-specific expression patterns

    • Single-cell proteomics to detect RNASE2 protein levels

    • Spatial transcriptomics to analyze tissue distribution

    • CRISPR screens at single-cell resolution to identify regulators

  • Live-cell imaging:

    • Fluorescently tagged RNASE2 to track cellular localization

    • FRET-based sensors for monitoring enzymatic activity in real-time

    • Correlative light and electron microscopy to detect subcellular compartmentalization

  • Computational methods:

    • Machine learning approaches to predict RNASE2 targets

    • Network analysis to identify pathway interactions

    • Multi-omics integration to correlate RNASE2 expression with cellular phenotypes

These emerging approaches will provide unprecedented resolution for understanding RNASE2 biology in health and disease.

Product Science Overview

Introduction

Ribonuclease 2 (RNase 2), also known as eosinophil-derived neurotoxin (EDN), is a member of the RNase A superfamily. This enzyme is primarily produced by eosinophils, a type of white blood cell involved in the body’s immune response. RNase 2 has garnered significant interest due to its diverse biological functions, including its role in immune response and its potential therapeutic applications.

Structure and Function

RNase 2 is a small, cationic protein with a molecular weight of approximately 18 kDa. It shares structural similarities with other members of the RNase A superfamily, characterized by a conserved catalytic triad essential for its ribonucleolytic activity. The enzyme cleaves RNA molecules, contributing to the degradation of RNA in various biological processes.

One of the unique features of RNase 2 is its ability to bind to and degrade single-stranded RNA, which is crucial for its antiviral and antimicrobial activities. This enzyme has been shown to possess potent antiviral properties, particularly against respiratory syncytial virus (RSV) and other RNA viruses .

Biological Roles

RNase 2 plays a multifaceted role in the immune system. It is stored in the granules of eosinophils and released upon activation. Once released, RNase 2 can exert several effects:

  1. Antiviral Activity: RNase 2 has been demonstrated to inhibit the replication of various RNA viruses, including RSV. This antiviral activity is attributed to its ability to degrade viral RNA, thereby preventing the virus from replicating and spreading .

  2. Immune Modulation: RNase 2 can activate dendritic cells (DCs), leading to the production of inflammatory cytokines, chemokines, and growth factors. This activation enhances the immune response by promoting the maturation and activation of DCs, which are essential for initiating adaptive immune responses .

  3. Alarmin Function: RNase 2 is classified as an alarmin, a type of endogenous molecule released by damaged or stressed cells to alert the immune system. As an alarmin, RNase 2 helps to amplify immune responses and maintain tissue homeostasis .

Recombinant Production

The recombinant production of RNase 2 involves the expression of the human RNase 2 gene in a suitable host system, such as Escherichia coli or yeast. This process allows for the large-scale production of RNase 2 for research and therapeutic purposes. Recombinant RNase 2 retains the enzymatic and biological properties of the native protein, making it a valuable tool for studying its functions and potential applications.

Therapeutic Potential

Given its antiviral and immune-modulating properties, RNase 2 holds promise as a therapeutic agent. Its ability to degrade viral RNA and activate immune cells makes it a potential candidate for the treatment of viral infections and immune-related disorders. Further research is needed to explore its therapeutic potential and develop effective RNase 2-based therapies.

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.