RNASE1 is expressed in multiple tissues, with primary roles in RNA degradation and immune defense:
In endothelial cells, RNASE1 is stored in Weibel-Palade bodies (WPBs) and released during inflammation to degrade extracellular RNA (eRNA), mitigating vascular damage .
RNASE1 acts as a "RNA scavenger" in the extracellular space, preventing eRNA-induced inflammation . During acute inflammation, its release from WPBs neutralizes pro-inflammatory eRNA, while chronic inflammation suppresses RNASE1 expression via MAPK/CK2/HDAC2 signaling, exacerbating vascular dysfunction .
Recent studies reveal contradictory roles in malignancy:
In HCC, RNASE1 upregulation in non-responders to immunotherapy suggests its utility as a prognostic biomarker . Conversely, in breast cancer, RNASE1 enhances Th1/Th17 responses and NK cell activity, promoting tumor clearance .
Human RNASE1 exhibits distinct biochemical traits compared to other vertebrate homologs:
Property | Human RNASE1 | Bovine RNase A | Mouse RNASE1 | Source |
---|---|---|---|---|
pH Optimum | ~8.0 | ~7.5 | ~7.0 | |
dsRNA Degradation | High | Moderate | Low | |
Cell-Surface Glycan Binding | High | Low | Moderate |
These differences highlight evolutionary divergence in RNA degradation efficiency and substrate specificity .
HCC: High RNASE1 expression in tumors and plasma predicts poor survival and resistance to anti-PD-1 therapy .
Breast Cancer: RNASE1 overexpression correlates with enhanced T-cell infiltration and improved therapeutic response .
Ribonuclease 1R (NASE1), a member of the pancreatic ribonuclease enzyme family, is a small protein. It possesses four disulfide bonds in its natural state and exhibits specific cleavage activity after pyrimidine nucleotides. The cleavage process unfolds in two steps: initially, the 3',5'-phosphodiester bond undergoes cleavage, yielding a 2',3'-cyclic phosphodiester intermediate; subsequently, this cyclic phosphodiester is hydrolyzed to generate a 3'-monophosphate. NASE1 exhibits heightened activation with single-stranded RNA. Its activity is inhibited by alkylation of His12 and His119 residues, while potassium and sodium salts enhance its activity. NASE1 is utilized to hydrolyze RNA present in protein samples.
Recombinant human RNASE1, produced in HEK cells, is a single, glycosylated polypeptide chain encompassing amino acids 29-156. With a total of 134 amino acids, it has a molecular weight of 15.3 kDa. A 6-amino acid His-tag is fused to the C-terminus of RNASE1, and it undergoes purification using proprietary chromatographic methods.
The RNASE1 solution is provided at a concentration of 1 mg/ml. It is formulated in a buffer containing 10% glycerol and Phosphate-Buffered Saline (pH 7.4).
For short-term storage (2-4 weeks), the RNASE1 vial can be stored at 4°C. For extended storage, it is recommended to store the protein in frozen aliquots at -20°C. To further enhance long-term stability during freezing, the addition of a carrier protein (0.1% HSA or BSA) is advised. Repeated freeze-thaw cycles should be minimized.
The purity of RNASE1 is determined to be greater than 90.0% using SDS-PAGE analysis.
The specific activity of RNASE1 is determined to be greater than 3 x 10^6 units/mg. This value represents the quantity of enzyme required to hydrolyze 1 nanomole of RNA per minute at a temperature of 25°C.
RNASE1, ribonuclease A family member 1, pancreatic, RAC1, RIB1, RNS1, ribonuclease pancreatic, HP-RNase, RIB-1, RNase UpI-1, RNase 1, Ribonuclease A, RNase A, Ribonuclease 1.
HEK293 Cells.
KESRAKKFQR QHMDSDSSPS SSSTYCNQMM RRRNMTQGRC KPVNTFVHEP LVDVQNVCFQ EKVTCKNGQG NCYKSNSSMH ITDCRLTNGS RYPNCAYRTS PKERHIIVAC EGSPYVPVHF DASVEDSTHH HHHH
Human RNASE1 is ubiquitously expressed throughout the body, similar to its murine counterpart which has been detected in at least 18 different tissue types via qPCR analysis . The widespread expression pattern suggests broad physiological functions beyond digestive processes. Although traditionally considered a pancreatic enzyme, significant expression occurs in non-pancreatic tissues, indicating diverse biological roles across multiple organ systems . When studying RNASE1 expression patterns, researchers should employ multiple detection methods including qPCR for mRNA analysis and immunohistochemical techniques with polyclonal antibodies against RNASE1 to detect protein-level expression in tissue samples .
Several complementary techniques can be employed to detect and quantify RNASE1:
Immunohistochemical analysis: Using polyclonal antibodies against RNASE1 to detect expression at the cellular level in tissue samples. This technique is particularly valuable for examining expression patterns in pathological specimens .
In situ hybridization (ISH): Utilizing digoxigenin-labeled RNASE1 probes to detect RNASE1 mRNA in tissues. This method was successfully employed to detect RNASE1, establishing that neoplastic cells of ductal type pancreatic adenocarcinomas express this enzyme .
Zymogram assays: These detect RNASE1 by its ribonucleolytic activity and electrophoretic mobility. In murine models, this technique revealed significant reduction of ribonucleolytic activity in RNASE1-deficient samples (approximately 69% loss compared to wild-type) .
Solution-phase assays: Used to measure ribonucleolytic activity in plasma samples, these assays demonstrated an estimated 47-52% reduction in total ribonucleolytic activity in RNASE1-knockout mice compared to wild-type controls .
Site-specific fluorophore-labeling: For advanced research applications, cysteine residues can be introduced via site-directed mutagenesis into loop regions distal to the enzymatic active site, allowing for fluorophore conjugation and tracking of RNASE1 in cellular contexts .
Despite historical assumptions of functional equivalence, human RNASE1 exhibits distinct structural and functional properties compared to bovine RNASE A:
Substrate specificity: Human RNASE1 demonstrates high catalytic activity against double-stranded RNA substrates, a rare quality among ribonucleases and a feature not shared with bovine RNASE A .
Cellular interactions: Human RNASE1 is readily endocytosed by mammalian cells, facilitated by tight interactions with cell surface glycans. This property suggests important intracellular functions beyond extracellular RNA degradation .
Evolutionary homology: Bovine brain ribonuclease, not RNASE A, represents the true functional homolog of human RNASE1. Both enzymes share remarkable similarities in catalytic properties against double-stranded RNA and cellular uptake mechanisms .
Physiological roles: While bovine RNASE A appears to function primarily in the ruminant digestive system, human RNASE1 likely serves broader physiological functions including possible roles in blood clotting regulation, inflammation modulation, and tumor biology .
RNASE1 plays a critical role in regulating extracellular RNA (eRNA) levels in circulation, with significant implications for blood coagulation and inflammatory processes:
Plasma RNA regulation: RNASE1-knockout mice exhibit significantly elevated levels of RNA in plasma compared to wild-type mice, demonstrating RNASE1's physiological function in degrading circulating RNA .
Coagulation effects: Plasma from RNASE1-deficient mice clots more rapidly than wild-type plasma, likely due to increased levels of active factor XII (FXIIa). This phenotype is consistent with known effects of eRNA on factor XII activation .
Factor XI activation: The apparent activity of factor XI in RNASE1-knockout mice plasma is 1000-fold higher when measured in assays triggered by low tissue factor concentration, suggesting eRNA enhances factor XI activation by thrombin .
Inflammatory modulation: By controlling eRNA levels, RNASE1 may help regulate inflammatory responses, as eRNA can function as a danger-associated molecular pattern (DAMP) that triggers immune activation .
These findings indicate that one of RNASE1's primary physiological functions is to degrade eRNA in blood plasma, potentially preventing inappropriate activation of coagulation and inflammatory pathways. Researchers investigating RNASE1 should consider these functions when designing experiments and interpreting results, particularly in disease models with altered RNA homeostasis.
Comparative analysis of RNASE1 homologs from diverse mammalian species reveals important functional differences and evolutionary adaptations:
Experimental approach: Effective comparative studies have employed recombinant expression systems with site-directed mutagenesis to introduce cysteine residues (P19C human RNASE1, S19C mouse RNASE1, A19C cow RNASE1, etc.) for site-specific fluorophore labeling .
Cellular uptake differences: Flow cytometry analysis of fluorescently labeled RNASE1 homologs reveals species-specific variations in cellular internalization efficiency, potentially reflecting adaptations to different physiological requirements .
Substrate specificity: Comparative enzymatic assays demonstrate variable activity against different RNA substrates, including single-stranded and double-stranded RNAs, across species .
Experimental methodology for uptake studies:
These comparative approaches provide fundamental insight into the ancestral roles of RNASE1 and its evolutionary adaptations across different mammalian lineages. Researchers should consider these methodological approaches when designing cross-species studies of ribonuclease function.
The generation of RNASE1-knockout mice has provided valuable insights into the physiological functions of this enzyme:
Generation methodology: RNASE1-deficient mice were created using Cre-LoxP recombination, with the entire RNASE1 protein-coding exon flanked by loxP sites, allowing for targeted excision .
Viability and general health: RNASE1-knockout mice are viable, healthy, and fertile, though notably larger than wild-type counterparts, suggesting a role in growth regulation .
Plasma RNA levels: Deficient mice exhibit significantly elevated plasma RNA compared to wild-type mice, demonstrating RNASE1's role in degrading extracellular RNA .
Coagulation phenotype: RNASE1-knockout plasma clots more rapidly than wild-type plasma, with increased levels of activated factor XII (FXIIa) and enhanced factor XI activity (1000-fold higher when measured with low tissue factor-triggered assays) .
Ribonucleolytic activity: Analysis of plasma samples shows a 47-69% reduction in total ribonucleolytic activity in knockout mice, indicating that RNASE1 contributes approximately half of the plasma ribonucleolytic activity in mice .
These findings suggest that while RNASE1 is not essential for survival, it plays important roles in RNA homeostasis and blood coagulation regulation. RNASE1-knockout mice represent a valuable tool for evaluating various hypotheses about RNASE1 and extracellular RNA functions in vivo.
RNASE1 exhibits distinctive expression patterns in pancreatic adenocarcinoma that may have diagnostic and pathophysiological significance:
Elevated serum levels: RNASE1 is present at high levels in the serum of most patients with pancreatic adenocarcinoma, suggesting potential utility as a biomarker .
Cellular expression: Immunohistochemical analysis and in situ hybridization revealed that 15 of 18 pancreatic adenocarcinoma samples were positive for RNASE1, confirming expression by neoplastic cells themselves rather than contaminating acinar cells .
Expression in precursor lesions: RNASE1 was detected in metaplastic ducts and atrophic islets in 4 of 6 chronic pancreatitis samples, as well as in hyperplastic ducts adjacent to well-differentiated adenocarcinomas, suggesting abnormal expression may be an early event in pancreatic tumorigenesis .
Altered gene expression patterns: RNASE1, traditionally considered an acinar protein, is abnormally expressed in pancreatic ductal adenocarcinoma cells, demonstrating that these cancer cells exhibit gene expression patterns distinct from normal pancreatic duct cells .
These findings indicate that RNASE1 may play roles in pancreatic cancer pathogenesis and progression, potentially contributing to the characteristic inflammatory and hypercoagulable states associated with this malignancy. Researchers investigating pancreatic cancer should consider RNASE1 expression analysis in their experimental designs.
Recent research has uncovered RNASE1's role in shaping the tumor immune microenvironment, with significant implications for immunotherapy response:
Association with immunotherapy resistance: Non-responders to Nivolumab (anti-PD-1 antibody) in hepatocellular carcinoma (HCC) exhibit high abundance of secreted RNASE1, correlating with poor prognosis across various cancer types .
Macrophage polarization: RNASE1 induces polarization of macrophages toward a tumor growth-promoting phenotype through activation of the anaplastic lymphoma kinase (ALK) signaling pathway .
Experimental evidence: Mice implanted with HCC cells overexpressing RNASE1 develop immunosuppressive tumor microenvironments and demonstrate diminished response to anti-PD-1 therapy .
Therapeutic targeting: Inhibition of the RNASE1/ALK axis can reprogram macrophage polarization, leading to increased recruitment of CD8+ T-cells and Th1 cells to the tumor microenvironment .
Combination therapy potential: Combined treatment with an ALK inhibitor and anti-PD-1 antibody exhibits enhanced tumor regression and facilitates development of long-term anti-tumor immunity .
These findings reveal RNASE1 as a potential therapeutic target in cancer immunotherapy, particularly in combination with immune checkpoint inhibitors. Researchers developing cancer therapeutics should consider targeting the RNASE1/ALK axis as a strategy to overcome immunotherapy resistance.
To effectively investigate RNASE1's contributions to tumor biology, researchers should consider these methodological approaches:
Comparative secretome analysis: Retrospective comparison of proteins secreted by cancer cells from responders versus non-responders to immunotherapy can identify RNASE1 as a differentially expressed factor .
Genetic manipulation models: Establishing cancer cell lines that overexpress RNASE1 through stable transfection allows evaluation of its effects on tumor growth, immune infiltration, and therapy response in vivo .
Immune cell phenotyping: Flow cytometry and immunohistochemistry to characterize changes in tumor-infiltrating immune populations, particularly focusing on macrophage polarization states (M1 vs. M2) and T-cell subsets .
Signaling pathway analysis: Western blotting and phospho-specific antibodies to assess activation of the ALK pathway and downstream signaling mediators in response to RNASE1 exposure .
Combination therapy testing: In vivo models comparing single-agent versus combination approaches (e.g., ALK inhibitors plus anti-PD-1) to evaluate synergistic effects on tumor regression and long-term immunity .
These methodological approaches provide a comprehensive framework for investigating RNASE1's roles in cancer biology and developing targeted therapeutic strategies to overcome RNASE1-mediated immunosuppression.
Successful production of functional RNASE1 requires specific experimental considerations:
Expression system selection: Bacterial expression systems can yield inclusion bodies that contain recombinant RNASE1, requiring subsequent refolding steps to obtain active enzyme .
Site-directed mutagenesis: Introduction of cysteine residues (e.g., P19C human RNASE1) in loop regions distal to the active site enables site-specific labeling with fluorophores or other chemical probes without disrupting catalytic activity .
Purification methodology: Successful purification protocols typically involve:
Activity verification: Zymogram assays and solution-phase ribonucleolytic activity measurements should be employed to confirm proper folding and catalytic function of purified RNASE1 .
Labeling strategies: For cellular uptake and localization studies, conjugation with appropriate fluorophores (e.g., BODIPY) followed by spectroscopic characterization ensures proper labeling without compromising enzymatic activity .
These technical considerations are essential for researchers aiming to produce high-quality recombinant RNASE1 for structural, functional, and cellular studies.
Investigating RNASE1's effects on extracellular RNA requires multiple complementary approaches:
These methodological approaches provide a comprehensive toolkit for researchers investigating RNASE1's role in RNA homeostasis and its impact on physiological processes like coagulation.
The discovery of RNASE1's role in immunosuppression opens several promising therapeutic avenues:
Combination immunotherapy approaches: Co-targeting ALK (to block RNASE1 signaling) and immune checkpoints (anti-PD-1) has demonstrated enhanced tumor regression and development of long-term immunity in preclinical models .
Biomarker development: RNASE1 levels could potentially serve as a predictive biomarker for immunotherapy response, helping identify patients who might benefit from combination approaches targeting the RNASE1/ALK axis .
Macrophage reprogramming strategies: Since RNASE1 promotes tumor-supporting macrophage polarization, inhibiting this pathway represents a promising approach to convert tumor-associated macrophages toward an anti-tumor phenotype .
Novel ALK inhibitors: Development of ALK inhibitors specifically optimized to block RNASE1-induced signaling in immune cells rather than targeting ALK fusion proteins in tumor cells represents an opportunity for selective immunomodulation .
RNASE1 neutralization approaches: Direct targeting of RNASE1 through neutralizing antibodies or aptamers could provide an alternative strategy to prevent its immunosuppressive effects while potentially preserving its physiological functions in RNA homeostasis .
These emerging therapeutic strategies highlight the potential clinical significance of understanding RNASE1 biology in cancer and other diseases.
Despite significant progress, several aspects of RNASE1 biology remain poorly understood:
Regulation of expression: The molecular mechanisms controlling RNASE1 expression in normal and pathological conditions remain largely unexplored, particularly the factors driving its aberrant expression in cancer cells .
Extracellular RNA identity: The specific RNA species degraded by RNASE1 in circulation and their potential functional roles before degradation represent an important knowledge gap .
Non-catalytic functions: Potential protein-protein interactions and signaling activities of RNASE1 that may be independent of its ribonucleolytic activity warrant investigation .
Tissue-specific roles: Despite ubiquitous expression, RNASE1 may serve different functions in specific tissues that have yet to be characterized .
Evolutionary significance: Further comparative studies across species could provide insights into why RNASE1 has been evolutionary conserved and how its functions have adapted to different physiological requirements .
Interplay with other ribonucleases: The functional relationship and potential redundancy between RNASE1 and other secreted ribonucleases in mammals represents an important area for future research .
These unexplored aspects highlight the continuing need for fundamental research on RNASE1 biology alongside translational applications.
Ribonuclease 1 (RNase 1), also known as human pancreatic ribonuclease, is an extracellular enzyme that belongs to the ribonuclease A (RNase A) superfamily. This superfamily consists of eight members in humans, all of which are encoded on chromosome 14 . RNase 1 is a basic protein composed of 128 amino acid residues and is characterized by its high catalytic activity towards both single-stranded and double-stranded RNA .
RNase 1 is found in various organs, including the exocrine pancreas, blood, milk, saliva, urine, and seminal plasma . It plays a crucial role in the digestion of dietary RNA and has been implicated in antiviral host defense . The enzyme’s relevance to host defense is supported by reports of its antiviral activity against human immunodeficiency virus (HIV)-1 .
RNase 1 shares 70% sequence identity with the well-known bovine pancreatic RNase A . Despite this high degree of sequence identity, RNase 1 differentiates from the bovine enzyme by exhibiting a very high activity towards double-stranded RNA (dsRNA) . The enzyme cleaves RNA specifically on the 3′ side of pyrimidine bases . Under physiological-like conditions, RNase 1’s hybridase activity is maximal at neutral pH, increases with lowering ionic strength, and is fully inhibited by the cytosolic RNase inhibitor .