Recombinant Macaca nemestrina Ribonuclease-like protein 9 (RNASE9) is a protein belonging to the ribonuclease A (RNase A) family. RNases are a superfamily of enzymes that catalyze the degradation of RNA and participate in various biological processes, including RNA turnover, defense against pathogens, and sperm maturation . RNASE9 is notable for its restricted expression pattern and potential roles in male reproductive biology .
The rat Rnase9 gene was cloned using in silico methods and genome walking, based on its homology to the Macaca mulatta (rhesus monkey) epididymal Rnase9 . The gene is located on chromosome 15p14, spanning two exons . The rat Rnase9 gene contains 1279 bp and encodes 182 amino acids, including a 24-amino acid signal peptide .
The Macaca nemestrina genome annotation reveals several key features of RNASE9:
Gene Location: The RNASE9 gene is located within the Macaca nemestrina genome .
Protein-Coding Potential: It is annotated as a protein-coding gene, indicating its role in producing a functional protein .
mRNA Transcripts: The gene is transcribed into mRNA, which serves as a template for protein synthesis .
Non-secretory ribonuclease: RNASE9 is a non-secretory ribonuclease. It is a pyrimidine-specific nuclease with a slight preference for U. It is also a cytotoxin and helminthotoxin .
RNASE9 exhibits a specific expression pattern, particularly in the male reproductive system :
Tissue Specificity: In rats, Rnase9 mRNA is specifically expressed in the epididymis, especially in the caput and corpus .
Androgen Dependence: The expression of Rnase9 is androgen-dependent, suggesting hormonal regulation .
Cellular Localization: RNASE9 is expressed in a principal cell-specific pattern within the epididymis .
Sperm Association: RNASE9 has been observed to bind to the acrosomal domain of sperm, hinting at a role in sperm maturation .
Expression in Mice: Studies in mice confirm that RNASE9 expression is restricted to the epididymis and is first detected in the midcaput, persisting through the distal caput and corpus, and waning in the cauda .
Studies using Rnase9 knockout mice have provided insights into the protein's function:
Normal Development and Fertility: Rnase9−/− mice are born at the expected Mendelian ratio, have normal postnatal growth and development, and have no outwardly apparent phenotype .
Spermatogenesis: Spermatogenesis is normal, and Rnase9-null sperm are morphologically normal .
Fertility: Rnase9 −/− males have normal fertility in unrestricted mating trials, and fertilization rates in in vitro fertilization assays are indistinguishable from wild-type mice .
Sperm Motility: Motility of Rnase9-null sperm is significantly impaired .
The precise function of RNASE9 is still under investigation, but evidence suggests roles related to sperm maturation :
Sperm Maturation: Given its expression in the epididymis and association with sperm, RNASE9 may be involved in sperm maturation processes .
RNA Processing: As a member of the RNase A family, RNASE9 could be involved in RNA degradation or processing within the epididymis, potentially affecting sperm function .
Non-enzymatic Roles: RNASE9 may have non-enzymatic functions, such as binding to other proteins or molecules, to influence sperm function .
| Feature | Description |
|---|---|
| Assembly Name | Mnem_1.0 |
| Assembly Accession | GCF_000956065.1 |
| Gene and Pseudogene Count | 41,011 |
| Protein-Coding Genes | 21,017 |
| Non-Coding Genes | 13,163 |
| Transcribed Pseudogenes | 1,403 |
| Non-Transcribed Pseudogenes | 5,190 |
| Genes with Variants | 14,741 |
| mRNAs | 66,484 |
| Non-Coding RNAs | 23,429 |
| Pseudo Transcripts | 1,438 |
| CDSs | 66,722 |
| Percentage Masked Sequence | RepeatMasker: 48.48% |
RNASE9 belongs to the ribonuclease A (RNase A) superfamily, a group of secreted proteins with diverse biological functions. Despite classification in this family, RNASE9 lacks the conventional ribonucleolytic activity that characterizes many RNase A members. Studies on human RNASE9 demonstrate no detectable ribonucleolytic activity against yeast tRNA .
The RNase A superfamily traditionally functions in RNA degradation, but several members, including RNASE9, have evolved distinct functions beyond RNA catalysis. RNASE9 belongs to a subset that has lost catalytic activity but gained new functional roles in reproductive biology and host defense .
Based on human RNASE9 studies, this protein exhibits a highly specific expression pattern primarily in the male reproductive tract. In humans, RNASE9 expression is predominantly localized to the epididymis, with immunofluorescence analyses revealing its presence throughout this tissue .
By extension, it is reasonable to hypothesize that Macaca nemestrina RNASE9 may exhibit a similar tissue-specific expression pattern, primarily in the epididymis and potentially other regions of the male reproductive tract. In the RNase A family, tissue-specific expression patterns are often conserved across related species.
To characterize the expression pattern of RNASE9 in Macaca nemestrina, researchers should perform:
RT-PCR analysis of RNA extracted from various tissues
Immunohistochemistry using specific antibodies against Macaca nemestrina RNASE9
Western blot analysis of protein extracts from different tissues
Human RNASE9 is present on the head and neck regions of ejaculated spermatozoa and in vitro capacitated spermatozoa, suggesting similar localization might be expected in Macaca nemestrina .
Based on available information, particularly from human RNASE9 studies, several physiological roles have been proposed:
Sperm Maturation: RNASE9 may contribute to biochemical modifications that spermatozoa undergo during epididymal transit. Its presence on sperm heads suggests possible involvement in membrane remodeling during maturation .
Host Defense: Human RNASE9 exhibits antibacterial activity against E. coli, suggesting a protective role in the male reproductive tract against bacterial infections . This antimicrobial property may help maintain a sterile environment for sperm maturation and storage.
Male Fertility: Multiple RNase family members, including RNases 9 and 10, have been implicated in sperm maturation and male fertility . RNASE9 may similarly contribute to fertility through direct interactions with spermatozoa.
Cell Surface Interactions: The localization on sperm surfaces suggests possible roles in sperm-oocyte recognition and binding, sperm migration through the female reproductive tract, or protection from immune surveillance.
The retention of RNASE9 across primate species despite the loss of ribonucleolytic activity suggests important non-canonical functions. The evolutionary pressure to maintain this protein implies its critical role in reproductive biology.
Based on successful approaches used for human RNASE9 and standard recombinant protein production practices, the following comprehensive protocol is recommended:
Cloning Strategy:
RNA Extraction and cDNA Synthesis:
PCR Amplification:
Vector Construction:
Expression Systems:
| System | Cell Line | Vector | Induction | Culture Conditions | Expected Yield |
|---|---|---|---|---|---|
| Mammalian | HEK293T | pcDNA-RNASE9-His | N/A | DMEM, 10% FBS, 48-72h | 0.5-2 mg/L |
| Bacterial | E. coli BL21(DE3) | pET25b(+)-RNASE9 | 0.5-1 mM IPTG | LB, 16-30°C, 4-16h | 5-20 mg/L |
Purification Methods:
For His-tagged Proteins:
Use Ni-NTA affinity chromatography
Apply imidazole gradient elution (20-250 mM)
For Non-tagged Proteins:
Additional Purification Steps:
Size exclusion chromatography to remove aggregates
Endotoxin removal for immunological studies
Quality Control:
Western blot for identity confirmation
Mass spectrometry for molecular weight verification
N-terminal sequencing to confirm correct processing
Functional assays (e.g., antimicrobial activity testing)
This approach, adapted from methods used for human RNASE9 , provides a reliable framework for producing recombinant Macaca nemestrina RNASE9 for research purposes.
Given that human RNASE9 demonstrates antibacterial activity against E. coli , a comprehensive assessment of Macaca nemestrina RNASE9's antimicrobial properties should include:
1. Radial Diffusion Assay:
Prepare agarose plates containing target microorganisms
Create wells in the agarose and add different concentrations of purified RNASE9
Incubate for 18-24 hours and measure zones of inhibition
Include appropriate positive controls (known antimicrobial peptides) and negative controls
2. Broth Microdilution Assay:
Prepare serial dilutions of RNASE9 in microplate wells
Add standardized bacterial suspensions (e.g., E. coli, Staphylococcus aureus)
Incubate for 16-24 hours and measure optical density
Calculate minimum inhibitory concentration (MIC) values
3. Time-Kill Kinetics:
Incubate bacteria with RNASE9 at different concentrations
Remove aliquots at various time points (0, 30, 60, 120, 180 minutes)
Plate dilutions on appropriate agar media and count colonies
This approach reveals whether RNASE9 exhibits concentration/time-dependent antibacterial activity as observed with human RNASE9
4. Membrane Permeabilization Assays:
Use fluorescent dyes like propidium iodide or SYTOX Green
Treat bacteria with RNASE9 and monitor dye uptake
Measure fluorescence to quantify membrane disruption
5. Comparative Analysis:
Test activity against a panel of microorganisms (Gram-positive bacteria, Gram-negative bacteria, fungi)
Compare activity spectrum with human RNASE9
Analyze the effect of ionic strength, pH, and temperature on antimicrobial activity
These methodologies provide comprehensive insights into Macaca nemestrina RNASE9's antimicrobial potential and its possible role in host defense within the male reproductive tract.
To elucidate the three-dimensional structure of Macaca nemestrina RNASE9, researchers should employ complementary structural biology techniques:
1. X-ray Crystallography:
Express and purify large quantities (>10 mg) of homogeneous RNASE9
Screen various conditions to obtain protein crystals
Collect diffraction data at synchrotron radiation facilities
Process data and solve the structure by molecular replacement using human RNASE structures as templates
Refine the structure to generate a high-resolution model
2. Nuclear Magnetic Resonance (NMR) Spectroscopy:
Prepare isotopically labeled RNASE9 (15N, 13C)
Collect multi-dimensional NMR spectra
Assign backbone and side-chain resonances
Generate distance restraints and calculate the solution structure
This approach is particularly useful for analyzing flexible regions
3. Computational Approaches:
Homology Modeling:
Use human RNASE9 or other RNase A family members as templates
Generate multiple models and evaluate using energy minimization
Validate models using molecular dynamics simulations
Ab initio Protein Structure Prediction:
Use methods like AlphaFold2 or RoseTTAFold
Generate predictions based solely on the amino acid sequence
4. Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS):
Expose RNASE9 to deuterium-containing buffers
Analyze the rate of hydrogen-deuterium exchange by mass spectrometry
Map regions of structural flexibility and solvent accessibility
5. Circular Dichroism (CD) Spectroscopy:
Analyze secondary structure content (α-helices, β-sheets)
Study thermal stability and folding properties
Compare with human RNASE9 to identify structural differences
The combination of these methodologies would provide comprehensive insights into Macaca nemestrina RNASE9's three-dimensional structure, revealing important features that may correlate with its functional properties and evolutionary adaptations.
Post-translational modifications (PTMs) can significantly influence protein structure, localization, stability, and function. For RNASE9, potential PTMs and their functional implications include:
1. N-Glycosylation:
Many secreted proteins, including RNases, undergo N-glycosylation
Potential N-glycosylation sites can be predicted from the amino acid sequence (Asn-X-Ser/Thr motifs)
Glycosylation affects:
Protein folding and stability
Protection from proteolytic degradation
Recognition by cell surface receptors
2. Disulfide Bond Formation:
RNase A family members typically contain multiple disulfide bonds
These are critical for maintaining tertiary structure
Correct disulfide bond formation is essential for proper folding and function
3. Proteolytic Processing:
Signal peptide cleavage is essential for secretion
Additional proteolytic processing may occur in the epididymal environment
The N-terminal amino acid sequences of mature RNASE9 provide important information about processing events
Methodological Approaches to Study PTMs in RNASE9:
Mass Spectrometry-Based Approaches:
Liquid chromatography-tandem mass spectrometry (LC-MS/MS)
Glycoproteomics for detailed glycan structure analysis
Phosphoproteomics for identification of phosphorylation sites
Site-Directed Mutagenesis:
Mutate potential modification sites
Compare functional properties of wild-type and mutant proteins
Assess the impact on antimicrobial activity and sperm binding
Enzymatic Deglycosylation:
Treat native RNASE9 with enzymes like PNGase F or Endo H
Compare properties of glycosylated and deglycosylated forms
Understanding PTMs of RNASE9 in different primate species would provide valuable insights into the evolution of this protein and how structural modifications contribute to species-specific functions in reproductive biology and host defense.
Based on successful approaches used for human RNASE9 , the following comprehensive protocol is recommended for generating antibodies against Macaca nemestrina RNASE9:
1. Antigen Preparation:
A. Recombinant Full-Length Protein:
Express recombinant Macaca nemestrina RNASE9 in E. coli or mammalian cells
Purify to >95% homogeneity using appropriate chromatography techniques
Verify protein identity by mass spectrometry or N-terminal sequencing
B. Synthetic Peptides:
Design 15-20 amino acid peptides from predicted antigenic regions
Consider using multiple peptides targeting different regions
Conjugate to carrier proteins (KLH or BSA) to enhance immunogenicity
2. Immunization Protocol:
| Step | Timepoint | Procedure | Adjuvant |
|---|---|---|---|
| Primary immunization | Day 0 | 25μg antigen, subcutaneous | Freund's complete adjuvant |
| First boost | Day 14 | 25μg antigen, subcutaneous | Freund's incomplete adjuvant |
| Second boost | Day 28 | 25μg antigen, subcutaneous | Freund's incomplete adjuvant |
| Final boost | Day 42 | 25μg antigen, intraperitoneal | None |
| Serum collection | Day 56 | Cardiac puncture | N/A |
This immunization schedule follows the protocol successfully used for human RNASE9 .
3. Antibody Characterization:
A. Specificity Testing:
Western blot analysis against recombinant RNASE9
Cross-reactivity assessment with other RNase family members
Pre-absorption controls with immunizing antigen
B. Sensitivity Determination:
Limit of detection in ELISA and Western blot
Immunoprecipitation efficiency
C. Functional Validation:
Immunofluorescence on known RNASE9-expressing tissues
Blocking experiments to confirm specificity
4. Antibody Purification:
A. Polyclonal Antibodies:
Protein A/G affinity purification
Antigen-specific affinity purification for highest specificity
B. Monoclonal Antibodies (if developed):
Protein A/G chromatography
Ion exchange chromatography for additional purification
This approach should yield high-quality antibodies suitable for immunolocalization studies of Macaca nemestrina RNASE9, allowing researchers to map its distribution in reproductive tissues and on spermatozoa.
Investigating the evolutionary conservation of RNASE9 requires a multifaceted approach combining comparative genomics, biochemistry, structural biology, and functional assays:
1. Comparative Sequence Analysis:
A. Sequence Collection and Alignment:
Retrieve RNASE9 sequences from various primate species
Perform multiple sequence alignment using tools like MUSCLE or CLUSTAL
Calculate sequence identity and similarity percentages
B. Evolutionary Rate Analysis:
Calculate dN/dS ratios to identify sites under positive or purifying selection
Identify lineage-specific accelerated evolution
Map evolutionary changes onto the protein structure
2. Functional Comparison:
A. Recombinant Protein Production:
Express and purify RNASE9 from multiple primate species
Ensure comparable purity and structural integrity
B. Comparative Antimicrobial Assays:
Test antimicrobial activity against the same panel of microorganisms
Compare minimum inhibitory concentrations and killing kinetics
Evaluate the spectrum of antimicrobial activity
C. Sperm Binding Assays:
Develop cross-species sperm binding assays
Assess whether RNASE9 from one species can bind to sperm from other species
Quantify binding affinities and specificities
3. Expression Pattern Comparison:
A. Tissue Distribution Analysis:
Compare RNASE9 expression patterns across primate species using:
RT-PCR or RNA-Seq for transcript detection
Immunohistochemistry for protein localization
Focus on reproductive tract tissues and potential extragonadal sites
4. Interspecies Complementation Experiments:
Express RNASE9 from different species in the same cellular background
Compare subcellular localization and secretion efficiency
Assess functional readouts (e.g., antimicrobial protection)
This comprehensive approach would provide significant insights into the evolutionary conservation and divergence of RNASE9 function across primate species, highlighting both conserved ancestral functions and species-specific adaptations.
The RNase A superfamily consists of diverse members with varying functions. Comparing Macaca nemestrina RNASE9 to other family members reveals important structural and functional relationships:
Structural Comparisons:
Catalytic Residues:
Traditional RNases (RNase 1-5) contain conserved catalytic residues (His12, Lys41, His119 in RNase A numbering)
RNASE9 lacks these conserved catalytic residues, explaining its absence of ribonucleolytic activity
This loss of catalytic function is similar to RNases 9-13, which have evolved alternative functions
Disulfide Bonding Pattern:
RNase A family members typically contain conserved disulfide bonds
Analysis of disulfide bonding patterns can reveal structural constraints maintained during evolution
Functional Comparisons:
This comparative analysis highlights how RNASE9 represents an example of functional diversification within the RNase A superfamily, evolving specialized roles in reproduction and host defense despite losing ancestral catalytic function.
To investigate the interaction between Macaca nemestrina RNASE9 and spermatozoa, researchers should consider the following experimental approaches:
1. Binding Studies:
A. Direct Binding Assays:
Label recombinant RNASE9 with fluorescent tags or biotin
Incubate with spermatozoa under various conditions
Analyze binding using flow cytometry or fluorescence microscopy
Quantify binding parameters (affinity, saturation)
B. Competition Assays:
Use unlabeled RNASE9 to compete with labeled RNASE9
Identify specific binding sites through selective inhibition
Test fragments or mutants of RNASE9 to map binding domains
2. Immunolocalization:
A. Confocal Microscopy:
Incubate fixed sperm with anti-RNASE9 antibodies
Use fluorescently-labeled secondary antibodies for detection
Perform z-stack imaging to precisely locate RNASE9 on sperm surfaces
Co-stain with markers for different sperm compartments (acrosome, mitochondria)
B. Immunoelectron Microscopy:
Localize RNASE9 at ultrastructural level using gold-labeled antibodies
Identify potential membrane microdomains associated with RNASE9
3. Functional Studies:
A. Sperm Function Assays:
Assess the effect of RNASE9 on:
Sperm motility parameters using computer-assisted sperm analysis
Capacitation using chlortetracycline staining or tyrosine phosphorylation
Acrosome reaction using fluorescent lectins
B. Antibody Blocking Studies:
Use anti-RNASE9 antibodies to block endogenous RNASE9
Assess the impact on sperm function parameters
4. Receptor Identification:
A. Cross-linking Studies:
Use chemical cross-linkers to capture RNASE9-receptor complexes
Identify binding partners by mass spectrometry
B. Yeast Two-Hybrid or Mammalian Two-Hybrid Screens:
Identify proteins that interact with RNASE9
Confirm interactions using co-immunoprecipitation
5. Real-time Interaction Analysis:
A. Surface Plasmon Resonance (SPR):
Immobilize RNASE9 or potential binding partners
Measure binding kinetics and affinity constants
Determine the effect of pH, ionic strength, and temperature on binding
These methodological approaches would provide comprehensive insights into the interaction between Macaca nemestrina RNASE9 and spermatozoa, elucidating both the binding mechanisms and functional consequences of this interaction.
To investigate the role of Macaca nemestrina RNASE9 in host defense against reproductive tract infections, researchers should consider the following comprehensive approaches:
1. In Vitro Antimicrobial Assays:
A. Activity Against Reproductive Tract Pathogens:
Test antimicrobial activity against common reproductive tract pathogens:
Bacteria (Escherichia coli, Staphylococcus aureus, Neisseria gonorrhoeae)
Fungi (Candida albicans)
Viruses (Herpes simplex virus)
Determine minimum inhibitory concentrations (MICs) and minimum bactericidal concentrations (MBCs)
B. Mechanism of Action Studies:
Membrane permeabilization assays using fluorescent dyes
Electron microscopy to visualize microbial cell damage
Transcriptomics to identify affected pathways in microorganisms
2. Ex Vivo Studies:
A. Reproductive Tract Explant Models:
Culture epididymal tissue explants from Macaca nemestrina
Challenge with pathogens in the presence or absence of RNASE9
Assess microbial growth and tissue inflammation markers
B. Sperm Protection Assays:
Incubate spermatozoa with pathogens with or without RNASE9
Evaluate sperm viability, motility, and functional parameters
Determine if RNASE9 protects sperm from pathogen-induced damage
3. Expression Studies During Infection:
A. Infection Models:
Analyze RNASE9 expression in response to:
Bacterial lipopolysaccharide (LPS) stimulation
Pro-inflammatory cytokines (TNF-α, IL-1β, IL-6)
Viral mimetics (poly I:C)
Use both cell culture and tissue explant models
B. Clinical Samples (if available):
Compare RNASE9 levels in healthy vs. infected reproductive tract samples
Correlate RNASE9 expression with pathogen load or inflammatory markers
4. Cell Culture Models:
A. Epithelial Cell Defense:
Transfect reproductive tract epithelial cells to express RNASE9
Challenge with pathogens and assess survival/infection rates
Measure inflammatory cytokine production
B. Immune Cell Interaction:
Study the effect of RNASE9 on neutrophil and macrophage function
Assess chemotactic properties and enhancement of phagocytosis
Evaluate modulation of immune cell cytokine production
5. In Vivo Models (where ethically appropriate):
A. Expression Analysis:
Induce experimental infections in the reproductive tract
Monitor RNASE9 expression using qPCR and immunohistochemistry
Correlate with immune cell infiltration and pathogen clearance
These methodological approaches would provide comprehensive insights into the potential role of Macaca nemestrina RNASE9 in host defense against reproductive tract infections, particularly its direct antimicrobial activities and potential immunomodulatory functions.