PRSS41, also known as Testis Serine Protease 1 (TESSP1), is a member of the serine protease family. It is a GPI-anchored protein primarily expressed in the testis with significant roles in reproductive biology. Research has also detected expression in the minor salivary gland and lungs . PRSS41 is encoded by a gene located on chromosome 16p13.3, specifically at coordinates 2848485-2855133 in the GRCh37 reference genome . Functionally, PRSS41 is required for the progression of meiosis during spermatogenesis, making it crucial for male reproductive research .
Gene-specific Copy Number Variation (CNV) probes are available for PRSS41 detection using Fluorescence In Situ Hybridization (FISH) techniques. These probes typically target the flanking regions of the PRSS41 gene and come with hybridization reagents . The methodology involves:
Using a labeled probe with selected dye color (available options include OR, RE, GO, GR, and AQ with different absorbance and emission spectra)
Hybridizing the probe to chromosome preparations
Visualizing signals to detect amplifications or deletions
These probes can detect both amplification and deletion events affecting the PRSS41 gene, providing valuable data on genetic alterations in research samples .
While the search results don't specifically address PRSS41 expression systems, we can draw parallels from methodologies used for other serine proteases. For bacterial expression systems, E. coli BL-21(DE3)pLysE has been successfully used for high-level expression of serine proteases in the same family . The recommended procedure includes:
Transformation of the expression vector containing the PRSS41 coding sequence into the host strain
Culture growth at 37°C with appropriate antibiotics (typically ampicillin at 50 μg/ml and chloramphenicol at 34 μg/ml)
Induction with IPTG (2 mM) at OD560 of approximately 0.3
Additional growth for 3 hours post-induction
Purification using affinity chromatography, such as Ni-NTA agarose for His-tagged recombinant proteins
This approach typically yields sufficient quantities of recombinant protein for research applications, though optimization may be necessary for specific experimental needs.
Investigating PRSS41's role in spermatogenesis requires a multi-faceted approach combining molecular, cellular, and physiological techniques:
Gene Expression Analysis: Implement RNA sequencing to identify genes with increased transcript levels and altered DNA methylation patterns that may interact with PRSS41
Protein-Protein Interaction Studies: Identify binding partners of PRSS41 using techniques such as:
Yeast two-hybrid screening
Co-immunoprecipitation followed by mass spectrometry
Proximity labeling approaches
Functional Assays for Meiotic Progression:
Immunohistochemistry of testicular sections to localize PRSS41 during different stages of spermatogenesis
Assessment of meiotic markers in PRSS41-deficient models
Protease activity assays using fluorogenic substrates to characterize enzymatic function
Genetic Modification Approaches:
CRISPR/Cas9-mediated knockout or knockin models
Conditional gene targeting to study stage-specific effects
Rescue experiments with wild-type and mutant forms of PRSS41
Since PRSS41 is specifically required for meiotic progression during spermatogenesis , these approaches would help elucidate its precise molecular mechanisms and potential therapeutic applications.
Characterizing the enzymatic activity of recombinant PRSS41 requires specialized assays that account for its serine protease nature:
Substrate Specificity Profiling:
Utilize peptide libraries with diverse sequences
Employ fluorogenic or chromogenic substrates with different cleavage sites
Analyze cleavage products by mass spectrometry to determine preferred recognition motifs
Kinetic Parameters Determination:
Measure initial velocities at varying substrate concentrations
Calculate Km, kcat, and catalytic efficiency (kcat/Km)
Assess the effects of pH, temperature, and ionic strength on activity
Inhibitor Screening:
Test broad-spectrum serine protease inhibitors (e.g., PMSF, aprotinin)
Evaluate substrate-based inhibitors
Screen for specific small-molecule inhibitors
Post-translational Modification Analysis:
Investigate the role of the GPI anchor in enzymatic activity
Assess potential regulatory phosphorylation or glycosylation sites
Compare native versus recombinant protein activity profiles
These methodological approaches would provide comprehensive characterization of PRSS41's enzymatic properties, enabling further functional studies in reproductive biology.
Correlating PRSS41 copy number variations (CNVs) with male fertility phenotypes requires a systematic approach integrating genomic, clinical, and functional data:
CNV Detection Using FISH Probes:
Apply gene-specific PRSS41 probes available in multiple dye colors (as shown in the table below)
Perform quantitative analysis of signal numbers and intensities
| Number | Dye Color | Order Name | Absorbance Maximum | Emission Maximum |
|---|---|---|---|---|
| 1 | OR | CNVFP-PRSS41-16476-OR | 573nm | 548nm |
| 2 | RE | CNVFP-PRSS41-16476-RE | 599nm | 580nm |
| 3 | GO | CNVFP-PRSS41-16476-GO | 551nm | 525nm |
| 4 | GR | CNVFP-PRSS41-16476-GR | 515nm | 491nm |
| 5 | AQ | CNVFP-PRSS41-16476-AQ | 467nm | 418nm |
Clinical Correlation Studies:
Collect comprehensive semen analysis data from subjects
Evaluate testicular biopsies for histological abnormalities
Track fertility outcomes in patients with identified CNVs
Functional Validation in Model Systems:
Generate models with varying PRSS41 copy numbers
Assess spermatogenesis progression at cellular and molecular levels
Evaluate fertility outcomes in animal models
Multi-omics Integration:
This integrative approach would establish meaningful connections between PRSS41 genetic variations and male fertility, potentially revealing diagnostic markers or therapeutic targets.
Purifying recombinant PRSS41 to maintain its native conformation and enzymatic activity requires careful consideration of its biochemical properties:
Affinity Chromatography Approaches:
Optimization Protocol:
Assess solubility in bacterial expression systems (soluble supernatant vs. inclusion bodies)
For inclusion bodies: Implement proper refolding protocols using step-wise dialysis
Test different buffer compositions to maintain stability and activity
Quality Control Assays:
SDS-PAGE and Western blotting with specific antibodies to confirm purity and identity
Size exclusion chromatography to ensure monomeric state
Circular dichroism to verify proper protein folding
Activity assays with model substrates to confirm functional integrity
Storage Considerations:
Determine optimal pH, buffer composition, and additives for long-term stability
Evaluate freeze-thaw stability and appropriate storage temperature
Consider lyophilization options if applicable
These methodological considerations are essential for obtaining functional recombinant PRSS41 suitable for downstream applications in reproductive biology research.
Developing specific antibodies against human PRSS41 requires strategic planning to ensure specificity, especially considering the homology with other serine proteases:
Antigen Design Strategies:
Select unique epitopes with low homology to other serine proteases
Consider both full-length recombinant protein and synthetic peptides
Use bioinformatics tools to identify surface-exposed regions
Antibody Production Protocol:
Immunize rabbits with purified recombinant PRSS41 (50-100 μg per immunization)
Follow a standard immunization schedule with appropriate adjuvants
Collect sera and evaluate antibody titers by ELISA
Specificity Testing:
Application-Specific Validation:
Verify antibody performance in immunohistochemistry, immunofluorescence, and flow cytometry
Test functionality in immunoprecipitation assays
Validate in knockout models or with siRNA knockdown controls
Despite potential sequence homology with other serine proteases, proper antigen design can yield specific antibodies, as demonstrated in related studies where antisera against homologous proteins (66% sequence identity) showed specificity for their respective immunizing antigens .
Designing effective CRISPR/Cas9 knockout strategies for PRSS41 requires careful consideration of several factors:
Target Site Selection:
Analyze the PRSS41 gene structure (exon-intron boundaries)
Select early exons encoding critical domains of the protein
Use in silico tools to identify guide RNAs with high on-target and low off-target scores
Consider targeting conserved catalytic residues of the serine protease domain
Delivery Methods for Reproductive Research:
For cell lines: Standard transfection or viral delivery methods
For animal models: Zygote microinjection or embryonic stem cell modification
For primary testicular cells: Optimize nucleofection or viral transduction protocols
Validation Strategy:
Design PCR-based genotyping assays to detect intended modifications
Sequence the target region to confirm exact genetic alterations
Verify protein knockout by Western blotting using validated antibodies
Assess functional consequences through spermatogenesis analysis
Phenotypic Analysis Plan:
Comprehensive evaluation of male reproductive parameters
Histological assessment of testicular architecture
Stage-specific analysis of meiotic progression
Fertility testing in animal models
Since PRSS41 is required for meiotic progression during spermatogenesis , knockout models would be particularly valuable for understanding its precise role in reproductive biology and potentially informing male fertility treatments.
PRSS41 research has significant potential to advance our understanding of male infertility, particularly in cases with unexplained meiotic arrest:
Diagnostic Applications:
Development of genetic screening panels including PRSS41 CNVs
Identification of PRSS41 mutations or variants in infertile men
Correlation of PRSS41 expression levels with specific infertility phenotypes
Mechanistic Insights:
Elucidation of molecular pathways regulated by PRSS41 during spermatogenesis
Identification of PRSS41 substrates essential for meiotic progression
Understanding the role of proteolytic processing in sperm development
Translational Potential:
Design of targeted therapies for specific forms of male infertility
Development of non-hormonal male contraceptives targeting PRSS41
Creation of diagnostic biomarkers based on PRSS41 activity or levels
Comparative Biology Approaches:
Exploration of PRSS41 function across species with different reproductive strategies
Investigation of evolutionary conservation of PRSS41-dependent pathways
Identification of compensatory mechanisms in species with PRSS41 variants
Given that PRSS41 is required for the progression of meiosis during spermatogenesis , these research directions could significantly impact our approach to diagnosing and treating certain forms of male infertility.
Researchers face several technical challenges when studying PRSS41, each requiring specific methodological solutions:
Protein Expression and Purification Challenges:
Challenge: Maintaining the native conformation of GPI-anchored PRSS41
Solution: Develop mammalian expression systems with proper post-translational modification machinery or use truncated constructs focusing on the catalytic domain
Specificity in Functional Assays:
Challenge: Distinguishing PRSS41 activity from other serine proteases
Solution: Design highly specific substrates based on unique cleavage preferences and use PRSS41 knockout controls
Tissue-Specific Expression Limitations:
Challenge: Restricted expression pattern primarily in testis
Solution: Develop organoid models of testicular tissue or stage-specific isolation of spermatogenic cells
In Vivo Functional Analysis:
Challenge: Complexity of studying meiosis in living systems
Solution: Combine advanced imaging techniques with cell-specific reporters to track PRSS41 activity during spermatogenesis
Structural Biology Hurdles:
Challenge: Obtaining crystal structures of membrane-associated proteases
Solution: Use cryo-electron microscopy or computational modeling approaches validated with experimental data
Addressing these technical challenges will accelerate progress in understanding PRSS41 biology and its potential applications in reproductive medicine.