PRSS35 is a trypsin-like serine protease activated by proprotein convertases (e.g., FURIN) through cleavage of its N-terminal domain . Key characteristics include:
Function: Degrades chemokines like CXCL2 via KK motif cleavage, suppressing neutrophil recruitment and extracellular trap formation in hepatocellular carcinoma (HCC) .
Regulation: Transcriptionally controlled by HNF4A and post-translationally modulated by hyperosmotic stress via NFAT5/ATF2 pathways .
Clinical Relevance: Downregulated in HCC lesions and patient serum, correlating with poor prognosis .
Commercial PRSS35 antibodies are predominantly rabbit polyclonal IgG formulations validated for specificity and reproducibility.
PRSS35 suppresses HCC progression by degrading CXCL2, reducing neutrophil infiltration, and inhibiting tumor growth in murine models .
Serum PRSS35 levels are significantly lower in HCC patients vs. healthy controls (ELISA; n = 222) .
PRSS35 modulates collagen deposition in fibroblasts under hyperosmotic stress, influencing wound healing and tumor microenvironments .
NFAT5 knockdown reduces PRSS35 expression by 70–90% in osmostressed cells (WB, luciferase assays) .
IHC using C-terminal PRSS35 antibodies reveals progressive loss of PRSS35 in advanced HCC stages .
Custom ELISA kits detect PRSS35 at sensitivities sufficient for clinical biomarker applications .
Western Blot: Detects FL-PRSS35 (~47 kDa) and cleaved forms (e.g., D1 domain) .
IHC: Validated in HCC patient tissues, showing reduced staining in tumors vs. adjacent normal liver .
Cross-Reactivity: Mouse and rat homologs share 85% sequence identity with human PRSS35, enabling translational studies .
PRSS35, or serine protease 35, functions as a key player in proteolytic pathways that regulate cellular function and disease progression. It has emerged as a promising research target for studies involving cancer, inflammation, and neurodegenerative disorders. Recent evidence specifically identifies PRSS35 as a tumor suppressor in hepatocellular carcinoma (HCC), where it suppresses CXCL2-mediated neutrophil extracellular traps . As a secreted protease, PRSS35 undergoes processing via cleavage by proprotein convertases to become active, after which it targets proteins containing tandem lysine (KK) recognition motifs . This activity positions PRSS35 as an important regulatory element in the extracellular microenvironment.
PRSS35 antibodies are primarily employed in Western blotting, ELISA, and immunohistochemistry applications . Western blotting represents the most validated application, with recommended dilutions typically ranging from 1:1000 to 1:5000 . These antibodies enable accurate detection and quantification of PRSS35 in various cellular contexts, making them valuable for investigating molecular biology, cell signaling pathways, and cancer research . For immunohistochemistry, dilutions between 1:20 and 1:200 are generally recommended to achieve optimal staining with minimal background .
PRSS35 antibodies have demonstrated successful detection in multiple sample types. Mouse and rat testis tissues serve as positive control samples for validating antibody performance . In human samples, PRSS35 has been successfully detected in liver tissues and serum samples . When working with clinical specimens, researchers should note that PRSS35 shows differential expression between healthy and diseased states - particularly in HCC, where both tissue and serum levels are significantly reduced compared to normal controls . This differential expression pattern makes PRSS35 a potential biomarker candidate for certain pathological conditions.
For optimal Western blot detection of PRSS35, consider the following methodological approach:
Sample preparation: Given PRSS35's presence in both cellular and secreted forms, analyze both cell lysates and conditioned media
Protein separation: Use gradient gels (4-15%) to effectively resolve the full-length PRSS35 (~45 kDa) and its shorter variants
Transfer conditions: Employ wet transfer methods with methanol-containing buffer for efficient transfer of all PRSS35 forms
Antibody selection: Choose antibodies based on your target region of interest, as different antibodies detect distinct forms:
N-terminal antibodies detect full-length PRSS35
C-terminal antibodies capture processed forms
Blocking and incubation: Use 5% non-fat milk in TBST for blocking, with primary antibody dilutions at 1:1000-1:5000
Detection: For enhanced sensitivity, particularly with secreted forms, consider using chemiluminescent substrates with extended exposure times
When interpreting results, note that PRSS35 appears in multiple molecular weight forms due to post-translational processing, with the secreted form (SF-PRSS35) having significantly lower molecular weight than full-length (FL-PRSS35) .
When performing immunohistochemistry for PRSS35, researchers should implement the following methodological approach:
Fixation: Use 10% neutral buffered formalin for tissue fixation to preserve antigen integrity
Antigen retrieval: Employ heat-induced epitope retrieval in citrate buffer (pH 6.0) to expose PRSS35 epitopes
Antibody selection: Choose antibodies targeting specific regions based on research objectives:
Antibody dilution: Start with 1:20-1:200 dilutions and optimize based on signal-to-noise ratio
Detection system: Utilize polymer-based detection systems for enhanced sensitivity
Controls: Include both positive (testis tissue) and negative controls in each experiment
Counterstaining: Use light hematoxylin counterstaining to maintain visibility of PRSS35 signal
For quantitative analysis, consider using digital pathology tools to measure staining intensity across tissue sections, particularly when comparing expression levels between normal and pathological samples, as PRSS35 shows gradual reduction with increasing stages of HCC development .
Distinguishing between full-length and processed PRSS35 forms requires a strategic approach:
Antibody panel selection: Utilize multiple antibodies targeting different regions:
Molecular weight analysis: Compare observed bands with expected molecular weights:
PRSS35 Form | Approximate MW | Detected By |
---|---|---|
Full-length (FL-PRSS35) | ~45 kDa | N, M, C antibodies |
Secreted form (SF-PRSS35) | Significantly lower | Primarily C antibodies |
Processed variants | Multiple bands | Varies by fragment |
Subcellular fractionation: Separate cellular compartments to determine localization:
Mass spectrometry validation: Confirm band identity through peptide sequencing, which has verified that shorter bands detected by SDS-PAGE contain PRSS35 peptides
Western blot interpretation: When analyzing samples, note that FL-PRSS35 is predominantly found in cell lysates, while SF-PRSS35 accumulates in culture medium of PRSS35-expressing cells .
To investigate PRSS35's tumor suppressor functions in cancer models, implement these methodological strategies:
Expression analysis in clinical samples:
In vitro functional studies:
In vivo tumor models:
Mechanism investigation:
Therapeutic potential assessment:
Test recombinant PRSS35 administration effects on established tumors
Develop strategies to enhance endogenous PRSS35 expression
This comprehensive approach has revealed that PRSS35 overexpression significantly suppresses HCC growth in multiple mouse models, while PRSS35 knockout accelerates liver cancer development .
Developing a robust ELISA for PRSS35 quantification in clinical samples involves these methodological steps:
Antibody pair selection:
ELISA protocol optimization:
Validation parameters:
Analytical sensitivity: Determine limit of detection (LOD) and quantification (LOQ)
Specificity: Test for cross-reactivity with related proteases
Precision: Assess intra- and inter-assay coefficient of variation (CV)
Recovery: Spike known amounts of PRSS35 into samples
Parallelism: Verify similar detection across dilutions
Clinical sample analysis:
Establish reference ranges in healthy controls
Compare levels in disease states (e.g., HCC patients)
Correlate with clinical parameters and outcomes
This approach has been successfully implemented to demonstrate significantly lower serum PRSS35 levels in HCC patients compared to normal subjects, suggesting its potential as a prognostic biomarker .
The observation of multiple bands during PRSS35 Western blotting is an expected phenomenon with specific biological significance. Multiple bands result from:
Post-translational processing: PRSS35 undergoes cleavage by proprotein convertases, generating various shorter forms
Antibody specificity: Different antibodies (N-terminal, middle-region, or C-terminal) detect distinct fragments
Sample preparation: Different extraction methods may preserve or disrupt various processed forms
To properly interpret these patterns:
Full-length PRSS35 appears at approximately 45 kDa in cell lysates
Secreted PRSS35 forms have significantly lower molecular weights
Multiple short forms enrich in culture medium of PRSS35-expressing cells
This is not a technical artifact but reflects the biological reality of PRSS35 processing. Mass spectrometry analysis has confirmed that these shorter bands indeed contain PRSS35 peptides . When troubleshooting, compare your observed pattern with expected processing forms and consider using antibodies targeting different regions to gain a complete picture of PRSS35 expression and processing.
When studying PRSS35 across experimental models, implement a comprehensive control strategy:
Positive tissue controls:
Cell line controls:
Expression controls:
PRSS35 overexpression system (positive control)
PRSS35 knockout or knockdown system (negative control)
Empty vector transfection (baseline control)
Antibody controls:
Processing controls:
Recombinant PRSS35 protein standards
Protease inhibitor treatment to block processing
Cross-species consideration:
This comprehensive control strategy ensures reliable data interpretation across different experimental systems and helps distinguish biological variations from technical artifacts.
Integrating PRSS35 antibody techniques with proteomics to identify novel substrates involves this methodological workflow:
Substrate prediction:
Differential secretome analysis:
Immunoprecipitation-mass spectrometry:
Use PRSS35 antibodies to pull down PRSS35 and interacting proteins
Perform mass spectrometry to identify co-precipitated proteins
Validate interactions through reverse immunoprecipitation
In vitro cleavage assays:
Express and purify recombinant candidate substrates
Incubate with active PRSS35
Analyze cleavage products by SDS-PAGE and mass spectrometry
Functional validation:
Mutate potential cleavage sites in candidate substrates
Assess resistance to PRSS35-mediated degradation
Evaluate functional consequences of blocked processing
This integrated approach has successfully identified CXCL2 as a PRSS35 substrate, demonstrating how PRSS35 cleaves CXCL2 at its tandem lysine recognition motif, ultimately suppressing neutrophil recruitment and tumor progression .
To comprehensively investigate PRSS35 regulation and expression across tissues, implement this multi-modal approach:
Transcriptional regulation analysis:
Tissue expression profiling:
Single-cell analysis:
Implement single-cell RNA sequencing to identify cell types expressing PRSS35
Validate with immunofluorescence co-staining using cell-type markers
Map expression changes during disease progression
Epigenetic regulation:
Analyze DNA methylation patterns at PRSS35 promoter
Assess histone modifications using ChIP-seq
Evaluate effects of epigenetic modifiers on PRSS35 expression
Post-transcriptional regulation:
Identify miRNAs targeting PRSS35 mRNA
Validate through luciferase reporter assays with miRNA binding site mutations
Assess mRNA stability and translation efficiency
The regulation of PRSS35 by HNF4A represents an example of this approach, where HNF4A response elements in the PRSS35 promoter were predicted and functionally validated through luciferase assays, connecting low HNF4A expression in HCC with reduced PRSS35 levels .
PRSS35's potential as a disease biomarker can be explored through these methodological approaches:
Clinical sample analysis strategy:
Develop standardized ELISA protocols for serum PRSS35 quantification
Establish reference ranges in healthy populations
Compare levels across disease states and stages
Performance metrics assessment:
Multi-marker panel integration:
Combine PRSS35 with established biomarkers
Develop algorithms to enhance diagnostic accuracy
Validate in independent patient cohorts
Form-specific biomarker analysis:
Therapeutic monitoring applications:
Track PRSS35 levels during treatment
Correlate changes with treatment response
Evaluate potential as a predictive biomarker
Research has demonstrated that PRSS35 levels are markedly decreased in HCC lesions compared to adjacent non-cancerous tissues, with patients expressing high PRSS35 exhibiting longer survival times. Additionally, truncated PRSS35 forms show marked reduction in HCC patient serum compared to normal subjects .
Developing therapeutic strategies targeting the PRSS35 pathway requires careful consideration of these methodological aspects:
Mechanism-based therapeutic approaches:
Model system selection for preclinical evaluation:
Cell culture systems: Limited value due to PRSS35's microenvironment-dependent effects
Xenograft models: Useful for human PRSS35 studies in immunocompromised hosts
Immunocompetent models: Essential for studying immune microenvironment interactions
PRSS35-knockout mice: Valuable for pathway validation studies
Delivery system optimization:
Targeted delivery to specific tissues (e.g., liver for HCC applications)
Protection of recombinant PRSS35 from degradation
Controlled release systems for sustained activity
Combination therapy strategies:
Integration with conventional treatments (chemotherapy, immunotherapy)
Sequential treatment approaches
Biomarker-guided patient selection
Safety and efficacy monitoring:
Development of antibodies for detecting therapeutic PRSS35
Distinguishing endogenous from exogenous PRSS35
Monitoring immune responses to PRSS35-based therapeutics
The tumor suppressive effects observed in multiple mouse models (xenograft HCC, spontaneous YAP-5SA-induced HCC, and Hepa1-6 murine HCC) suggest therapeutic potential, with the interesting observation that PRSS35 exhibits tumor suppressive effects in vivo but not in cultured cells, highlighting the importance of the tumor microenvironment in PRSS35 function .