Recombinant Human Serine protease 42 (PRSS42)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates. Note: Our proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing. The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
PRSS42P; PRSS42; TESSP2; Putative serine protease 42; EC 3.4.21.-; Serine protease 42; pseudogene; Testis serine protease 2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
27-293
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Homo sapiens (Human)
Target Names
PRSS42
Target Protein Sequence
GMAA PRLPSPLLSE EGGENPEASP APGPEAGPPL NLFTSFPGDS LLCGRTPLRI VGGVDAEEGR WPWQVSVRTK GRHICGGTLV TATWVLTAGH CISSRFHYSV KMGDRSVYNE NTSVVVSVQR AFVHPKFSTV TTIRNDLALL QLQHPVNFTS NIQPICIPQE NFQVEGRTRC WVTGWGKTPE REKLASEILQ DVDQYIMCYE ECNKIIQKAL SSTKDVIIKG MVCGYKEQGK DSCQGDSGGR LACEYNDTWV QVGIVSWGIG CGR
Uniprot No.

Target Background

Function
Plays a crucial role in spermatogenesis and germ cell survival during meiosis.
Database Links

HGNC: 30716

KEGG: hsa:339906

UniGene: Hs.585127

Protein Families
Peptidase S1 family
Subcellular Location
Cytoplasm. Cell membrane; Lipid-anchor, GPI-anchor.

Q&A

What is the genomic location and structure of the PRSS42 gene?

The human PRSS42 gene (also known as TESSP2) is located on chromosome 3 and belongs to the serine protease family. Based on structural analysis, PRSS42 contains characteristic domains common to serine proteases, including a catalytic triad essential for its enzymatic activity. The gene shows significant homology to the mouse Prss42/Tessp-2 gene, which has been more extensively studied . Mouse Prss42/Tessp-2 is part of a cluster of genes known as the Prss/Tessp locus, which includes other serine proteases expressed predominantly in testicular tissue. The promoter region of Prss42/Tessp-2 contains important regulatory elements that interact with enhancers and non-coding RNAs for transcriptional activation during spermatogenesis .

What is the tissue-specific expression pattern of PRSS42?

PRSS42 exhibits a highly tissue-specific expression pattern, predominantly expressed in testicular tissue. Studies in mice have demonstrated that Prss42/Tessp-2 expression is tightly regulated during spermatogenesis, with expression primarily detected in pachytene spermatocytes and secondary spermatocytes . This tissue-specific expression pattern suggests specialized roles in reproduction and fertility. The expression is regulated by both enhancer elements and a testis-specific long non-coding RNA called Tesra, which binds to the Prss42/Tessp-2 promoter region to increase transcriptional activity . Limited expression in other tissues indicates that PRSS42 likely serves highly specialized functions rather than housekeeping roles.

What are the known substrates and enzymatic properties of PRSS42?

As a member of the serine protease family, PRSS42 is expected to function as a proteolytic enzyme with a catalytic triad consisting of histidine, aspartic acid, and serine residues. While specific substrates for human PRSS42 have not been definitively characterized in the provided literature, its structural similarity to other serine proteases suggests it may cleave peptide bonds at specific recognition sites. By comparison, other serine proteases like matriptase have been shown to cleave substrates such as the amyloid-beta peptide at specific arginine and lysine residues (Arg-5, Lys-16, and Lys-28) . Determining the specific substrates and cleavage sites for PRSS42 would require targeted proteomic approaches, including substrate phage display libraries or mass spectrometry-based identification of cleavage products.

What methods are commonly used for detecting PRSS42 expression?

Several methods can be employed to detect PRSS42 expression at both mRNA and protein levels:

  • For mRNA detection:

    • Quantitative real-time PCR (qRT-PCR) using specific primers targeting PRSS42 transcript

    • RNA-seq analysis for genome-wide expression profiling

    • Northern blotting for detecting transcript size and abundance

    • In situ hybridization for spatial expression in tissue sections

  • For protein detection:

    • Western blotting using specific antibodies against PRSS42

    • Immunohistochemistry for localization in tissue sections

    • Mass spectrometry for proteomic identification and quantification

In mouse models, researchers have successfully used qRT-PCR to quantify Prss42/Tessp-2 expression levels in various cell types and experimental conditions, demonstrating significant changes in expression during different stages of spermatogenesis and in response to various treatments .

How is PRSS42 transcriptionally regulated, and what factors influence its expression?

PRSS42 transcriptional regulation involves complex mechanisms including enhancer elements and non-coding RNAs. Based on studies of the mouse ortholog Prss42/Tessp-2, we can identify several key regulatory factors:

  • Long non-coding RNA regulation: A testis-specific long non-coding RNA called Tesra (4,435 nucleotides) binds to the Prss42/Tessp-2 promoter region and significantly increases its transcriptional activity . Overexpression of Tesra in hepatic tumor cells (Hepa1-6) resulted in a significant increase in endogenous Prss42/Tessp-2 expression .

  • Enhancer activity: Both upstream and downstream sequences of the Prss42/Tessp-2 locus possess enhancer activity. Specifically:

    • The downstream enhancer showed approximately 1.7-fold increase in promoter activity

    • The upstream enhancer showed approximately 1.3-fold increase in promoter activity

  • Cooperative activation: Tesra and the downstream enhancer can cooperatively but independently increase Prss42/Tessp-2 promoter activity. When Tesra expression was induced in cells containing the Prss42/Tessp-2 promoter with the downstream enhancer, promoter activity increased significantly by 2.2-fold .

  • Chromatin interactions: During spermatogenesis, the chromatin at the Prss42/Tessp-2 promoter interacts with the 3' end of another non-coding RNA region (lncRNA-HSVIII), facilitating transcriptional activation .

The table below summarizes the effects of different regulatory elements on Prss42/Tessp-2 promoter activity:

Regulatory ElementEffect on Promoter ActivityFold Increase
Upstream EnhancerIncreases activity~1.3-fold
Downstream EnhancerIncreases activity~1.7-fold
Tesra lncRNAIncreases activitySignificant increase
Tesra + Downstream EnhancerCooperative increase~2.2-fold
Tesra + Upstream EnhancerNo significant cooperative effectNo significant change

This complex regulatory mechanism ensures the precise temporal and spatial expression of Prss42/Tessp-2 during spermatogenesis .

What experimental approaches can be used to identify PRSS42 substrates and cleavage sites?

Identifying PRSS42 substrates and cleavage sites requires specialized proteomic approaches:

  • Recombinant protein production:

    • Express and purify recombinant PRSS42 protease domain using bacterial, insect, or mammalian expression systems

    • Validate protease activity using generic substrates for serine proteases

  • Substrate identification strategies:

    • Proteomic approach: Incubate recombinant PRSS42 with cellular lysates or purified candidate substrates, followed by SDS-PAGE and mass spectrometry analysis to identify cleaved proteins

    • Peptide library screening: Use synthetic peptide libraries to identify preferred cleavage motifs

    • Substrate phage display: Screen phage-displayed peptide libraries to identify preferred substrate sequences

  • Cleavage site determination:

    • MALDI-TOF mass spectrometry: Similar to the approach used for matriptase cleavage site identification in Aβ1-42 peptide , MALDI-TOF MS can precisely identify the masses of peptide fragments generated after PRSS42 digestion

    • N-terminal sequencing: Edman degradation to identify the new N-termini generated after cleavage

    • Mutagenesis studies: Mutate candidate cleavage sites in putative substrates to confirm specificity

  • Validation experiments:

    • Co-expression studies: Express PRSS42 with candidate substrates in cell culture systems

    • Enzyme kinetics: Determine catalytic efficiency (kcat/KM) for confirmed substrates

    • Domain mapping: Identify which domains of PRSS42 are required for substrate recognition and cleavage

Drawing from similar studies with matriptase, researchers identified specific cleavage sites in the amyloid-beta peptide at Arg-5, Lys-16, and Lys-28 using recombinant protease domain and MALDI-TOF mass spectrometry . A similar experimental pipeline could be applied to characterize PRSS42 substrates and cleavage sites.

What are the molecular mechanisms by which the Tesra lncRNA regulates PRSS42 expression?

The Tesra long non-coding RNA employs several molecular mechanisms to regulate Prss42/Tessp-2 expression:

  • Direct chromatin binding: Chromatin immunoprecipitation (ChIP) experiments have demonstrated that Tesra directly binds to the Prss42/Tessp-2 promoter region . This binding is specific, as control experiments with RNase treatment eliminated the binding signal, confirming the RNA-dependent nature of this interaction.

  • Promoter activation: Functional studies using luciferase reporter assays confirmed that Tesra significantly increases Prss42/Tessp-2 promoter activity . When Tesra was overexpressed in Hepa1-6 cells transfected with a Prss42/Tessp-2 promoter-driven luciferase construct, a significant increase in luciferase activity was observed compared to control cells.

  • Independence from enhancer activity: While both Tesra and downstream enhancer elements can increase Prss42/Tessp-2 promoter activity, they appear to function independently through distinct mechanisms. When combined, they show cooperative effects, suggesting complementary modes of action .

  • Temporal regulation during spermatogenesis: The expression pattern of Tesra coincides with the timing of Prss42/Tessp-2 activation during spermatocyte development, suggesting a coordinated regulatory mechanism specific to meiotic processes .

To visualize the proposed model for Prss42/Tessp-2 activation through Tesra:

Stage of SpermatogenesisMolecular Events
Spermatogonia to Primary SpermatocytesInitial chromatin interaction between Prss42/Tessp-2 promoter and lncRNA-HSVIII region; beginning of Prss42/Tessp-2 transcription
Pachytene SpermatocytesAccumulation of Tesra transcripts in nuclei; binding of Tesra to Prss42/Tessp-2 promoter; strengthening of chromatin interactions
Late Pachytene/Secondary SpermatocytesDownstream enhancer physically contacts Prss42/Tessp-2 promoter; cooperative action of Tesra and enhancer elements; high-level Prss42/Tessp-2 transcription

This coordinated regulatory mechanism ensures the precise temporal expression of Prss42/Tessp-2 during specific stages of spermatogenesis .

What methods can be used to study PRSS42 protease activity in vitro and in vivo?

Comprehensive investigation of PRSS42 protease activity requires both in vitro and in vivo approaches:

In Vitro Methods:

  • Enzyme activity assays:

    • Fluorogenic peptide substrates: Design peptides containing fluorophore/quencher pairs that emit fluorescence upon cleavage

    • Colorimetric assays: Use substrates that change color when cleaved (e.g., p-nitroaniline-conjugated peptides)

    • Gel-based assays: Incubate recombinant PRSS42 with candidate protein substrates and analyze by SDS-PAGE to detect cleavage products

  • Kinetic parameters determination:

    • Measure initial reaction velocities at varying substrate concentrations

    • Calculate Michaelis-Menten parameters (KM, Vmax, kcat)

    • Determine specificity constants (kcat/KM) for different substrates

  • Inhibitor screening:

    • Test natural and synthetic serine protease inhibitors

    • Perform structure-activity relationship studies

    • Use protease-dead mutants (e.g., mutation in the catalytic serine) as negative controls

In Vivo Methods:

  • Cell-based assays:

    • Overexpression studies: Express wild-type PRSS42 or catalytically inactive mutants in cell lines

    • CRISPR/Cas9 knockout: Generate PRSS42-deficient cells to study loss-of-function effects

    • Substrate processing: Monitor processing of co-expressed substrates in cell culture

  • Animal models:

    • Knockout mice: Generate PRSS42-deficient mice to study physiological roles

    • Tissue-specific expression: Use testis-specific promoters to drive expression of PRSS42 variants

    • Phenotypic analysis: Evaluate fertility, sperm development, and testicular function

  • Quantification techniques:

    • Western blotting: Monitor substrate cleavage using antibodies specific to intact proteins or cleavage products

    • Immunohistochemistry: Visualize PRSS42 and substrate localization in tissue sections

    • Mass spectrometry: Identify and quantify protease-generated peptides in complex biological samples

Similar to studies with matriptase, where researchers used a recombinant protease domain to cleave synthetic peptides and analyzed the products by MALDI-TOF MS , these methods would provide comprehensive characterization of PRSS42 activity and substrate specificity.

What are the potential roles of PRSS42 in reproductive biology and fertility?

Given its predominant expression in testicular tissue and specific regulation during spermatogenesis, PRSS42 likely plays critical roles in reproductive biology:

  • Spermatogenesis regulation:

    • The tightly controlled expression pattern during specific stages of spermatocyte development suggests roles in meiotic progression

    • Potential involvement in chromatin remodeling, nuclear protein processing, or intercellular signaling during sperm development

  • Sperm maturation:

    • As a serine protease, PRSS42 may process proteins essential for sperm maturation

    • Potential roles in remodeling sperm cell surface proteins or extracellular matrix components in the seminiferous tubules

  • Fertilization:

    • Possible involvement in sperm-egg interaction processes

    • May participate in proteolytic cascades similar to the acrosome reaction

  • Pathophysiological implications:

    • Mutations or expression alterations could contribute to male infertility

    • Potential biomarker for specific types of male reproductive disorders

To fully elucidate PRSS42's role in reproduction, comprehensive studies would be needed, including:

  • Detailed phenotypic analysis of PRSS42-deficient animal models

  • Identification of physiological substrates in reproductive tissues

  • Correlation studies between PRSS42 variants/expression levels and fertility parameters

A proposed research pipeline for investigating PRSS42's reproductive functions would include:

Research PhaseApproachesExpected Outcomes
Expression AnalysisSingle-cell RNA-seq of testicular cells; ImmunohistochemistryPrecise cellular and subcellular localization
Functional GenomicsCRISPR/Cas9 knockout in mice; Tissue-specific conditional knockoutsReproductive phenotypes; Fertility assessment
Substrate IdentificationProteomic analysis of testicular tissues from wild-type vs. knockout; Proximity labelingPhysiological substrates in reproductive context
Clinical CorrelationAnalysis of PRSS42 variants in infertile men; Expression studies in testicular biopsiesAssociation with specific fertility disorders

What are the optimal conditions for expressing recombinant PRSS42 protein?

Producing functional recombinant PRSS42 requires careful consideration of expression systems and conditions:

  • Expression systems selection:

    • Bacterial expression (E. coli):

      • Advantages: High yield, cost-effective, rapid production

      • Limitations: Lack of post-translational modifications, potential inclusion body formation

      • Recommended for: Protease domain expression for initial substrate screening

    • Insect cell expression (Baculovirus):

      • Advantages: Proper folding, some post-translational modifications

      • Limitations: More complex than bacterial systems, moderate yield

      • Recommended for: Full-length PRSS42 for structural and functional studies

    • Mammalian cell expression (HEK293, CHO):

      • Advantages: Proper folding and authentic post-translational modifications

      • Limitations: Lower yield, higher cost, longer production time

      • Recommended for: Studies requiring native-like protein activity

  • Construct design considerations:

    • Include purification tags (His, GST, MBP) for efficient purification

    • For active protease production, consider expressing as a zymogen (inactive precursor) to prevent self-digestion

    • Engineer constructs with removable tags using specific protease sites (TEV, thrombin)

    • For structural studies, optimize construct boundaries based on domain predictions

  • Purification strategy:

    • Two-step purification recommended (affinity chromatography followed by size exclusion)

    • Consider ion exchange chromatography for higher purity

    • Optimize buffer conditions (pH, salt concentration) to maintain stability

    • Include protease inhibitors during purification if expressing active protease

  • Activity verification:

    • Develop a simple activity assay using generic serine protease substrates

    • Verify proper folding using circular dichroism or thermal shift assays

    • Test stability under different storage conditions (temperature, buffer composition)

Based on approaches used for other serine proteases like matriptase , a recombinant PRSS42 protease domain could be produced and used for initial activity and substrate specificity studies, while full-length protein may be required for understanding regulatory mechanisms.

How can RNA-protein interactions between Tesra and PRSS42 promoter be experimentally validated?

Validating RNA-protein interactions between Tesra lncRNA and the PRSS42 promoter requires multiple complementary approaches:

  • Chromatin Isolation by RNA Purification (ChIRP):

    • Design biotinylated antisense oligos tiling the Tesra sequence

    • Crosslink RNA-chromatin complexes in cells expressing Tesra

    • Pull down Tesra and associated chromatin using streptavidin beads

    • Analyze associated DNA by qPCR or sequencing to confirm PRSS42 promoter enrichment

  • RNA Immunoprecipitation (RIP):

    • Identify potential RNA-binding proteins that interact with both Tesra and the PRSS42 promoter

    • Immunoprecipitate candidate proteins and analyze associated RNA by qRT-PCR

    • Confirm presence of Tesra in the immunoprecipitated material

  • RNA tiling experiments:

    • Generate a series of truncated Tesra constructs

    • Test each construct for ability to activate PRSS42 promoter using reporter assays

    • Identify minimal RNA regions required for PRSS42 activation

  • In vitro binding assays:

    • Perform electrophoretic mobility shift assays (EMSA) with labeled Tesra RNA and PRSS42 promoter DNA

    • Use competition assays with unlabeled RNA to confirm specificity

    • Test mutant versions of Tesra to identify critical binding motifs

  • Functional validation:

    • Perform luciferase reporter assays with wild-type and mutated PRSS42 promoter constructs

    • Test the effect of Tesra overexpression or knockdown on reporter activity

    • Quantify changes in endogenous PRSS42 expression upon Tesra manipulation

Similar to the approaches used in studying Tesra's interaction with the mouse Prss42/Tessp-2 promoter , these methods would provide comprehensive validation of the RNA-promoter interaction and identify the molecular mechanisms involved.

What are the challenges in developing specific antibodies against PRSS42 and how can they be addressed?

Developing specific antibodies against PRSS42 presents several challenges due to its membership in the serine protease family, which shows high sequence conservation:

  • Challenges in antibody development:

    • Sequence similarity with other serine proteases causing cross-reactivity

    • Potential post-translational modifications affecting epitope recognition

    • Conformational epitopes that may be lost in denatured protein detection methods

    • Limited availability of purified native protein for immunization

  • Strategic approach to antibody development:

    • Epitope selection:

      • Identify unique peptide sequences specific to PRSS42 using bioinformatics analysis

      • Target regions with low homology to other serine proteases

      • Consider both N-terminal and C-terminal regions outside the conserved catalytic domain

      • Design multiple candidate epitopes (8-20 amino acids each)

    • Immunization strategies:

      • Use multiple host species (rabbit, mouse, goat) for broader epitope recognition

      • Consider both peptide immunization and recombinant protein approaches

      • Implement prime-boost protocols for enhanced immune response

      • Use adjuvants appropriate for inducing high-affinity antibodies

  • Screening and validation methods:

    • Primary screening:

      • ELISA against immunizing peptides/proteins

      • Western blot against recombinant PRSS42

      • Competitive ELISA to confirm specificity

    • Secondary validation:

      • Western blot against tissue lysates from tissues known to express PRSS42

      • Immunoprecipitation followed by mass spectrometry

      • Immunohistochemistry on tissues with known expression patterns

      • Testing on samples from PRSS42 knockout models as negative controls

  • Antibody formats and applications:

    • Develop both polyclonal and monoclonal antibodies for different applications

    • Consider recombinant antibody technologies for enhanced reproducibility

    • Generate application-specific antibodies (Western blot, IHC, IP, FACS)

By implementing this comprehensive strategy, researchers can develop and validate specific antibodies against PRSS42 that avoid cross-reactivity with other serine proteases while maintaining high sensitivity for the target protein.

What approaches can be used to identify physiological regulators of PRSS42 expression beyond Tesra lncRNA?

Identifying additional physiological regulators of PRSS42 expression requires a multi-faceted approach:

  • Transcription factor binding site analysis:

    • Perform in silico analysis of the PRSS42 promoter region to identify putative transcription factor binding sites

    • Conduct ChIP-seq experiments to identify transcription factors that bind to the PRSS42 promoter in relevant tissues

    • Validate predicted binding using reporter assays with wild-type and mutated promoter constructs

  • Epigenetic regulation assessment:

    • Analyze DNA methylation patterns at the PRSS42 promoter using bisulfite sequencing

    • Perform ChIP-seq for histone modifications (H3K4me3, H3K27ac, H3K27me3) to assess chromatin state

    • Investigate the effects of HDAC inhibitors and DNA methyltransferase inhibitors on PRSS42 expression

  • Other non-coding RNA interactions:

    • Perform RNA-seq of nuclear and cytoplasmic fractions to identify additional lncRNAs expressed in PRSS42-expressing tissues

    • Conduct RNA pulldown assays followed by mass spectrometry to identify proteins that interact with candidate regulatory RNAs

    • Investigate miRNA binding sites in PRSS42 mRNA using prediction algorithms and functional validation

  • Signaling pathway analysis:

    • Treat relevant cell lines with pathway activators/inhibitors and monitor effects on PRSS42 expression

    • Perform phosphoproteomics to identify signaling pathways active in PRSS42-expressing cells

    • Use CRISPR/Cas9 screening to identify genes that regulate PRSS42 expression

  • Hormonal regulation:

    • Given the testis-specific expression, investigate effects of reproductive hormones (testosterone, FSH, LH) on PRSS42 expression

    • Perform hormone treatment experiments in appropriate cell models or ex vivo tissue cultures

    • Analyze PRSS42 expression in animal models with altered hormonal states

A comparison table of expected regulatory mechanisms based on studies of the mouse ortholog:

Regulatory MechanismEvidence from Mouse StudiesPotential Methods for Human PRSS42
lncRNA (Tesra)Direct binding to promoter; increased expression RNA-seq, ChIRP, reporter assays
Enhancer ElementsUpstream and downstream enhancers identified ATAC-seq, 4C-seq, reporter assays
Chromatin LoopingInteraction between promoter and enhancer regions Hi-C, 3C, 4C-seq
Developmental TimingStage-specific expression during spermatogenesis Single-cell RNA-seq, in situ hybridization

This comprehensive approach would provide a detailed understanding of the complex regulatory network controlling PRSS42 expression beyond the already identified Tesra lncRNA mechanism.

How do I interpret conflicting data on PRSS42 expression in different tissues or experimental conditions?

Interpreting conflicting data on PRSS42 expression requires systematic analysis of multiple factors:

  • Technical considerations:

    • Detection method specificity:

      • Antibody cross-reactivity with other serine proteases

      • PCR primer specificity and potential amplification of homologs

      • RNA-seq depth and mapping quality to distinguish closely related genes

    • Sample preparation differences:

      • Cell isolation methods affecting cell type purity

      • RNA/protein extraction protocols influencing yield from different tissues

      • Fixation methods for histological samples affecting epitope accessibility

  • Biological variability factors:

    • Developmental stages: Expression may vary significantly during development

    • Disease states: Pathological conditions might alter normal expression patterns

    • Individual variation: Genetic or environmental factors affecting expression levels

    • Splice variants: Different isoforms may be expressed in different tissues

  • Analytical approach to resolve conflicts:

    • Methodological triangulation:

      • Combine multiple detection methods (qRT-PCR, Western blot, immunohistochemistry)

      • Use orthogonal approaches (e.g., reporter assays, in situ hybridization)

      • Employ negative controls (knockout tissues/cells) to confirm specificity

    • Quantitative assessment:

      • Establish detection thresholds relevant to biological function

      • Use absolute quantification methods where possible

      • Consider relative expression levels compared to housekeeping genes

  • Integration of contextual information:

    • Consider species differences when comparing human and mouse data

    • Evaluate effects of experimental manipulations on expression regulation

    • Assess correlation with expression of known interacting partners or regulators

  • Resolution strategies for specific conflict types:

Conflict TypePossible ExplanationsResolution Strategy
Different tissues showing expressionCell type heterogeneity; ContaminationSingle-cell RNA-seq; Laser capture microdissection
Different expression levels in same tissueTechnical variability; Biological conditionsStandardized protocols; Increased biological replicates
Detection in unexpected tissuesLow-level expression; Cross-reactivityValidation with multiple methods; Functional testing
Inconsistent effects of regulatorsContext-dependent regulation; Indirect effectsMechanistic studies; Time-course experiments

By systematically addressing these factors, researchers can resolve apparent conflicts in PRSS42 expression data and develop a more accurate understanding of its true expression pattern and regulation.

What are the implications of PRSS42 substrate specificity for therapeutic development?

Understanding PRSS42 substrate specificity has important implications for therapeutic development:

  • Target validation considerations:

    • Specificity assessment:

      • Determine whether PRSS42 has unique substrates or shares targets with other proteases

      • Evaluate redundancy with other serine proteases in key pathways

      • Assess phenotypic consequences of PRSS42 inhibition in model systems

    • Therapeutic relevance:

      • Evaluate whether PRSS42 substrates are involved in disease processes

      • Determine if substrate processing is rate-limiting in pathological conditions

      • Assess potential on-target side effects based on physiological substrates

  • Inhibitor design strategies:

    • Structure-based approaches:

      • Design competitive inhibitors that mimic substrate binding sites

      • Target unique features of PRSS42 active site to achieve selectivity

      • Consider allosteric inhibitors targeting non-conserved regulatory domains

    • Substrate-guided approaches:

      • Develop transition-state analogs based on preferred cleavage sequences

      • Create substrate-derived inhibitory peptides with enhanced stability

      • Design prodrugs activated by PRSS42 for targeted drug delivery

  • Screening methodologies:

    • Biochemical assays:

      • Develop high-throughput fluorogenic substrate assays

      • Implement counter-screens against related proteases to ensure selectivity

      • Include physiologically relevant substrate cleavage assays

    • Cell-based assays:

      • Monitor cellular substrate processing in PRSS42-expressing cells

      • Develop reporter systems for PRSS42 activity in intact cells

      • Assess effects on downstream signaling pathways

  • Therapeutic modality considerations:

    • Small molecule inhibitors:

      • Advantages: Oral bioavailability, potential tissue penetration

      • Challenges: Achieving selectivity among serine proteases

    • Biologics (antibodies, peptides):

      • Advantages: Higher specificity, longer half-life

      • Challenges: Delivery to relevant tissues, manufacturing complexity

    • RNA therapeutics:

      • Advantages: High specificity for PRSS42 gene, adaptable chemistry

      • Challenges: Delivery, stability, potential off-target effects

  • Development pathway considerations:

    • Establish correlation between substrate processing and disease biomarkers

    • Develop pharmacodynamic assays based on substrate processing

    • Consider reproductive implications given PRSS42's role in testicular function

By carefully considering these aspects of PRSS42 substrate specificity, researchers can develop more effective and selective therapeutic strategies while anticipating potential challenges in the development process.

What emerging technologies could advance our understanding of PRSS42 function and regulation?

Several cutting-edge technologies hold promise for elucidating PRSS42 function and regulation:

  • Single-cell multi-omics approaches:

    • Single-cell RNA-seq: Map PRSS42 expression at unprecedented cellular resolution

    • Single-cell ATAC-seq: Identify cell-specific chromatin accessibility at the PRSS42 locus

    • Single-cell proteomics: Detect low-abundance PRSS42 protein in specific cell populations

    • Multi-omics integration: Correlate PRSS42 expression with global cellular state

  • Advanced genome editing technologies:

    • Base editing: Introduce precise point mutations in PRSS42 or its regulatory elements

    • Prime editing: Engineer specific mutations without double-strand breaks

    • CRISPR activation/inhibition (CRISPRa/CRISPRi): Modulate PRSS42 expression without altering DNA sequence

    • CRISPR screening: Identify genes affecting PRSS42 expression or function

  • Spatial biology technologies:

    • Spatial transcriptomics: Map PRSS42 mRNA expression within tissue architecture

    • Multiplexed immunofluorescence: Visualize PRSS42 protein alongside multiple markers

    • Mass cytometry imaging: Quantify PRSS42 and interacting proteins with subcellular resolution

    • Expansion microscopy: Achieve super-resolution imaging of PRSS42 localization

  • Protein structure and interaction technologies:

    • Cryo-electron microscopy: Determine PRSS42 structure at atomic resolution

    • AlphaFold2/RoseTTAFold: Predict PRSS42 structure and substrate interactions computationally

    • Hydrogen-deuterium exchange mass spectrometry: Map dynamic protein-protein interactions

    • Proximity labeling (BioID, APEX): Identify proteins in close proximity to PRSS42 in living cells

  • Organoid and advanced cell culture systems:

    • Testicular organoids: Recapitulate spermatogenesis in vitro for functional studies

    • Microfluidic systems: Model dynamic cellular environments and cell-cell interactions

    • Organs-on-chips: Study PRSS42 function in physiologically relevant conditions

    • Patient-derived organoids: Investigate PRSS42 variants in personalized disease models

The application of these technologies could transform our understanding of PRSS42 biology, as summarized in this potential research roadmap:

TechnologyApplication to PRSS42 ResearchExpected Impact
Single-cell RNA-seqDefine exact cell populations expressing PRSS42Precise cellular context for functional studies
CRISPR base editingCreate catalytic mutants or regulatory element variantsDissection of structure-function relationships
Spatial transcriptomicsMap PRSS42 expression in relation to tissue architectureUnderstanding of spatial regulation and function
AlphaFold2 structural predictionModel PRSS42-substrate interactionsRational design of selective inhibitors
Testicular organoidsRecapitulate PRSS42 expression during in vitro spermatogenesisControlled system for functional studies

These emerging technologies, often used in combination, will enable unprecedented insights into PRSS42 biology that were previously inaccessible with conventional approaches.

How can multi-omics data integration improve our understanding of PRSS42 function in reproductive biology?

Multi-omics data integration offers a comprehensive approach to understanding PRSS42's role in reproductive biology:

  • Integration of diverse data types:

    • Genomics: Identify genetic variants affecting PRSS42 function or expression

    • Transcriptomics: Map expression patterns across cell types, developmental stages, and conditions

    • Proteomics: Identify post-translational modifications and protein interactions

    • Metabolomics: Assess downstream effects of PRSS42 activity on cellular metabolism

    • Epigenomics: Characterize chromatin state and regulatory mechanisms

  • Computational integration strategies:

    • Network analysis: Construct protein-protein interaction networks centered on PRSS42

    • Pathway enrichment: Identify biological processes affected by PRSS42 activity

    • Machine learning approaches: Predict PRSS42 function from integrated datasets

    • Bayesian integration: Combine evidence from multiple sources with uncertainty quantification

  • Reproductive biology-specific applications:

    • Developmental trajectory analysis: Map PRSS42 function across spermatogenesis stages

    • Cell-cell communication inference: Identify potential paracrine signaling involving PRSS42

    • Fertility phenotype correlation: Link PRSS42 variants/expression to reproductive outcomes

  • Case study example: Multi-omics approach to PRSS42 in male fertility

Omics LayerMethodological ApproachExpected Insights
GenomicsExome/genome sequencing of fertile vs. infertile menPRSS42 variants associated with fertility phenotypes
TranscriptomicsSingle-cell RNA-seq of testicular biopsiesCell type-specific expression and co-expression networks
ProteomicsTesticular tissue proteomics; PRSS42 interactome analysisProtein interactions and activation state
PhosphoproteomicsPhosphorylation profiling of PRSS42-expressing cellsSignaling pathways affected by PRSS42 activity
EpigenomicsATAC-seq and ChIP-seq of testicular cellsRegulatory landscape controlling PRSS42 expression
MetabolomicsMetabolite profiling of PRSS42-deficient vs. normal tissueDownstream metabolic effects
  • Data integration challenges and solutions:

    • Challenge: Different data types have varying scales, noise levels, and biological meanings

    • Solution: Apply normalization techniques and weighted integration methods

    • Challenge: Sample availability from human reproductive tissues

    • Solution: Leverage model organisms and in vitro systems with careful cross-species mapping

    • Challenge: Temporal dynamics of spermatogenesis

    • Solution: Design studies with appropriate time-series sampling and pseudo-time analysis

This multi-omics approach would provide unprecedented insights into PRSS42's role in reproductive biology, potentially identifying novel biomarkers for fertility assessment and targets for therapeutic intervention in reproductive disorders.

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.